relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in...

269
Relationships between nuclear lamins and telomere biology in progeroid laminopathies and cancer Thesis submitted for the degree of Doctor of Philosophy Rachel Turner (BSc) Department of Genetics & Department of Biochemistry University of Leicester December 2014

Upload: duongkhanh

Post on 20-May-2018

217 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

Relationships between nuclear lamins and telomere

biology in progeroid laminopathies and cancer

Thesis submitted for the degree of

Doctor of Philosophy

Rachel Turner (BSc)

Department of Genetics & Department of Biochemistry

University of Leicester

December 2014

Page 2: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …
Page 3: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

i

Abstract

Telomeres are essential for maintaining the integrity of the genome. Shortening

and dysfunction of telomeres initiates cellular senescence, halting further cell

division, and instigating alterations in biological processes which contribute to

ageing. Progeroid syndromes are disorders of ageing. Patients exhibit not only

an early external appearance of old age, but several age-related diseases

including osteoporosis, muscle wasting and cancer.

Laminopathies are diseases caused by mutations in LMNA, the gene encoding

the key nuclear matrix component lamin A. Some mutations result in progeroid

phenotypes that are associated with elevated levels of senescent cells. The rate

of telomere shortening in progeria cells was found to be accelerated relative to

healthy controls, confirming a role for lamin A function in telomere biology.

In order for a cell to bypass the protective mechanism of senescence, it must

initiate a telomere maintenance mechanism (TMM), resulting in cancer. Several

biological changes accompanying the alternative lengthening of telomeres

(ALT) TMM overlap with mechanisms affected by changes in lamin biology. As

such, lamins and related nuclear proteins were investigated in ALT cells and

found to differ from tumour-derived cells which activate the telomerase-based

TMM. It was found that although nuclear levels of lamins differed between cells

of the two TMMs, the interaction of lamin A with chromatin was not altered.

Localisation studies of lamin A revealed different distribution of lamins in the

nucleus. An excess of intranuclear lamin observed in ALT cells is proposed to be

permissive for recombination-based processes required for telomere elongation

through ALT, whereas laminopathy lines exhibit a reduced intranuclear lamin,

and an accompanying DNA damage repair deficiency. These findings reinforce

the importance of lamin organisation and intranuclear network stability in

chromatin maintenance, and highlight a new role for lamins in ALT cancers.

Page 4: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

ii

Acknowledgements

Firstly I would like to thank my supervisors, Dr Nicola Royle and Dr Sue

Shackleton for their invaluable expertise and guidance throughout this project.

Thanks are also due to Dr Maria Viskaduraki for her input into my statistical

analyses and Dr Kees Straatman for his guidance and assistance with cell

imaging. I would also like to thank members of the G18/19 laboratory past and

present for their support, in particular Maria Pakendorf and Frances Tippins, for

providing much-needed good company during my last few months in the lab.

Thanks go to my family for their understanding and support, and for still being

there at the other end of the phone regardless of how long it’s been. A special

thanks to my mum and my gran for their long-standing belief in me, and for

getting some of that to rub off.

Finally, to Jake, who has provided unwavering encouragement. Thank you for

your love and company in these last four years. Never surrender.

Page 5: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

iii

Table of Contents

Abstract ............................................................................................................................................. i

Acknowledgements ...................................................................................................................... ii

Table of Contents ........................................................................................................................ iii

Abbreviations ................................................................................................................................. x

Chapter 1 Introduction ........................................................................................................... 1

1.1 The nuclear lamina ...................................................................................................... 1

1.1.1 Overview ................................................................................................................. 1

1.1.2 Properties of lamins ............................................................................................ 2

1.1.2.1 Lamin processing and localisation ......................................................................... 6

1.1.3 Additional lamina components and lamin-interacting partners ........ 9

1.1.3.1 The LINC complex ........................................................................................................ 9

1.1.3.2 LEM domain-containing proteins and BAF ...................................................... 11

1.1.3.3 Other binding partners ............................................................................................ 12

1.1.4 Lamin A dysfunction and laminopathies ................................................... 14

1.1.4.1 Non-progeroid laminopathies .............................................................................. 14

1.1.5 Hutchinson-Gilford progeria syndrome .................................................... 17

1.1.5.1 Lamin A processing and progerin expression in classical HGPS ............. 18

1.1.5.2 Impact of progerin on nuclear lamin organisation ....................................... 19

1.1.5.3 Impact of progerin on chromatin ........................................................................ 21

1.1.5.4 Franesyl transferase treatment ............................................................................. 22

1.1.6 Atypical HGPS and other progeroid laminopathies .............................. 23

1.2 Telomeres ...................................................................................................................... 28

1.2.1 Overview ............................................................................................................... 28

1.2.2 Telomere structure ............................................................................................ 29

1.2.2.1 Shelterin components .............................................................................................. 31

1.2.2.2 The 3’ overhang .......................................................................................................... 32

1.2.3 Telomeres, DNA damage and ageing ........................................................ 33

Page 6: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

iv

1.2.3.1 The mitotic clock and telomere-associated senescence ............................ 34

1.2.3.2 DNA damage response ........................................................................................... 35

1.2.3.3 Repair via homologous recombination ............................................................. 36

1.2.3.4 Repair via non-homologous end-joining ......................................................... 37

1.2.3.5 Telomere dysfunction induces senescence through a DNA damage

response pathway ........................................................................................................................ 39

1.2.3.6 Werner syndrome ...................................................................................................... 40

1.2.4 Telomere maintenance mechanisms and cancer ................................... 41

1.2.4.1 Telomerase ................................................................................................................... 42

1.2.4.2 The ALT mechanism .................................................................................................. 44

1.3 Links between telomere and lamin biology ..................................................... 46

1.3.1 DNA damage and lamin deficiencies ......................................................... 46

1.3.2 Telomeres, heterochromatin, lamins and nuclear localisation .......... 48

1.3.3 Telomere length is reduced by lamin A deficiencies ............................ 49

1.4 Objectives ..................................................................................................................... 51

Chapter 2 Materials and Methods .................................................................................... 53

2.1 Cell culture .................................................................................................................... 53

2.1.1 Subculturing cell lines ...................................................................................... 53

2.1.2 Population growth measurement ................................................................ 53

2.1.3 Trypan Blue staining ......................................................................................... 54

2.1.4 -galactosidase Senescence assay .............................................................. 54

2.1.5 Cell lines ................................................................................................................ 54

2.1.6 DNA/protein pellet preparation ................................................................... 56

2.2 Genomic DNA extractions ....................................................................................... 56

2.2.1 DNA extractions ................................................................................................. 56

2.2.2 Hirt DNA extractions ......................................................................................... 57

2.3 Single Telomere Length Analysis (STELA) ......................................................... 57

2.3.1 PCR cycling conditions ..................................................................................... 58

Page 7: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

v

2.4 MS32 analysis .............................................................................................................. 59

2.4.1 PCR cycling conditions ..................................................................................... 59

2.5 DSN overhang assay ................................................................................................. 59

2.5.1 Digests ................................................................................................................... 59

2.5.2 Gel Electrophoresis ............................................................................................ 60

2.5.3 Gel transfer ........................................................................................................... 61

2.6 Chromatin Immunoprecipitation (ChIP) ............................................................. 62

2.6.1 Cross-link and cell harvest .............................................................................. 62

2.6.2 Bead preparation ............................................................................................... 62

2.6.3 Cell lysis and chromatin preparation .......................................................... 63

2.6.3.1 Gel confirmation of chromatin shearing ........................................................... 64

2.6.4 Washes ................................................................................................................... 64

2.6.5 Sample purification ........................................................................................... 65

2.6.6 Semi-quantitative PCR ..................................................................................... 65

2.6.7 Antibody verification ........................................................................................ 65

2.7 Southern Blotting ....................................................................................................... 66

2.8 DNA Dot blot preparation ...................................................................................... 67

2.8.1 Denaturing dot blots for ChIP analysis ...................................................... 67

2.8.2 Native dot blots for overhang analysis ...................................................... 67

2.9 DNA radiolabelling .................................................................................................... 67

2.9.1 Random-primed labelling of DNA double stranded probes ............. 67

2.9.1.1 Telomere probe .......................................................................................................... 68

2.9.2 Oligonucleotide probe end-labelling ......................................................... 68

2.9.2.1 Oligonucleotides ........................................................................................................ 69

2.10 Hybridisation conditions ..................................................................................... 69

Page 8: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

vi

2.10.1 STELA and MS32 ............................................................................................ 69

2.10.2 Native and denaturing dot blots.............................................................. 69

2.10.3 DSN overhang assay ..................................................................................... 70

2.11 Analysis and quantification of hybridisation assays .................................. 70

2.11.1 STELA .................................................................................................................. 70

2.11.2 Dot Blots ........................................................................................................... 70

2.11.3 DSN overhang assay ..................................................................................... 71

2.12 Subtelomere genotyping .................................................................................... 73

2.12.1 12q ...................................................................................................................... 73

2.12.2 Xp/Yp ................................................................................................................. 74

2.13 Western Blotting .................................................................................................... 75

2.13.1 Gel preparation............................................................................................... 75

2.13.2 Electrophoresis conditions and transfer ................................................ 75

2.13.3 Immunoblotting ............................................................................................. 76

2.13.4 Quantification ................................................................................................. 77

2.14 Immunofluorescence ............................................................................................ 77

2.15 Primers ....................................................................................................................... 78

2.16 Antibodies ................................................................................................................. 79

PART A • Telomere shortening in progeria cell lines ..................................................... 80

Chapter 3 Telomere shortening rate in progeroid laminopathy cell lines

established by STELA ................................................................................................................. 80

3.1 Background .................................................................................................................. 80

3.1.1 Measuring telomere length: STELA ............................................................. 81

3.1.2 Telomere length in progeria cell lines ........................................................ 84

Page 9: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

vii

3.1.3 Cell lines ................................................................................................................ 85

3.2 Results ............................................................................................................................ 88

3.2.1 Growth rates and senescence levels ........................................................... 88

3.2.2 Telomere shortening in laminopathy and control fibroblasts ........... 93

3.2.2.1 NHDF .............................................................................................................................. 93

3.2.2.2 155-BR ........................................................................................................................... 98

3.2.2.3 HGPS ............................................................................................................................. 101

3.2.2.4 RD .................................................................................................................................. 103

3.2.2.5 T623S ............................................................................................................................ 105

3.2.2.6 E578V ........................................................................................................................... 107

3.3 Discussion .................................................................................................................. 108

3.3.1 Exploring the effects of population dynamics and long molecule

amplification efficiency on shortening rate .......................................................... 109

3.3.2 Telomere shortening rate over lifespan.................................................. 117

3.3.3 Cell line differences in telomere shortening rate ................................ 123

3.3.4 Potential sources for increased telomere shortening in laminopathy

patient cells ....................................................................................................................... 128

3.3.5 Summary ............................................................................................................ 132

Chapter 4 Exploring mechanisms behind accelerated telomeres shortening

in laminopathy patient cells ................................................................................................. 134

4.1 Background ............................................................................................................... 134

4.2 Results ......................................................................................................................... 137

4.2.1 Initial observations of DSN method ......................................................... 137

4.2.2 Troubleshooting .............................................................................................. 138

4.2.3 Native dot blots ............................................................................................... 141

4.3 Discussion .................................................................................................................. 144

4.3.1 Alternative methods for overhang analysis ........................................... 144

Page 10: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

viii

PART B • The nuclear lamina and telomere biology in ALT and Telomerase

positive cancer cells ................................................................................................................ 146

Chapter 5 Differential expression of lamins and related proteins in ALT and

telomerase positive cell lines ............................................................................................... 146

5.1 Background ............................................................................................................... 146

5.2 Results ......................................................................................................................... 148

5.2.1 Protein quantification by Western blot................................................... 148

5.2.2 Protein quantification and identification of lamin localisation by

immunofluorescence microscopy ............................................................................. 153

5.3 Discussion .................................................................................................................. 160

Chapter 6 Interactions between chromatin and lamins in ALT and telomerase

positive cells............................................................................................................................... 166

6.1 Background ............................................................................................................... 166

6.2 Results ......................................................................................................................... 167

6.2.1 Optimisation and verification of ChIP ..................................................... 167

6.2.2 A-type lamins do not associate differently with telomeric chromatin

in ALT versus telomerase positive cells................................................................... 170

6.2.3 Discussion .......................................................................................................... 175

Chapter 7 Final Discussion ............................................................................................... 180

7.1 Accelerated telomere shortening in patient cells expressing farnesylated

lamin A forms ....................................................................................................................... 180

7.1.1 Future work ....................................................................................................... 182

7.1.1.1 The role of 53BP1 .................................................................................................... 182

7.1.1.2 Cohesion ..................................................................................................................... 185

7.2 Altered lamin state in ALT cells may enable the ALT phenotype .......... 187

7.2.1 Future work ....................................................................................................... 187

Page 11: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

ix

7.3 Concluding remarks ............................................................................................... 189

Appendices................................................................................................................................. 192

Appendix A Materials and Methods supplemental ............................................ 192

Appendix B STELA .......................................................................................................... 194

B.1 STELA Southern blots ........................................................................................ 194

B.2 HT1080 control data .......................................................................................... 197

Appendix C Confirmation of cell line sub-telomere genotypes .................... 199

C.1 12q genotyping ................................................................................................... 199

C.2 XpYp genotyping ................................................................................................ 201

Appendix D Confirmation of cell line purity with MS32 analysis ................... 202

Appendix E NT2D1 exclusion ..................................................................................... 203

Appendix F ChIP ............................................................................................................. 204

References .................................................................................................................................. 206

Page 12: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

x

Abbreviations

ALT Alternative lengthening of telomeres

APS Ammonium persulphate

BSA Bovine serum albumin

ChIP Chromatin immunoprecipitaion

CSR Class switch recombination

DNA Deoxyribonucleic acid

DSB Double-strand break

dsDNA Double-stranded DNA

EDTA Ethylenediaminetetraacetic acid

FC Final concentration

FISH Fluorescent in situ hybridisation

FTI Farnesyl transferase inhibitor

g Gram

HGPS Hutchinson-Gilford progeria syndrome

HMW High molecular weight

hTERT Human telomerase reverse transcriptase

IF Immunofluorescence

IF Intermediate filament

IMS Industrial methylated spirit

INM Inner nuclear membrane

IP Immunoprecipitation

IQR Inter-quartile range

kb Kilobase

kDa Kilodalton

LA Lamin A

Lamin A/C Lamins A and C

LMW Low molecular weight

Page 13: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

xi

M Molar

MBq Mega Becquerel

mg Milligram

ml Millilitre

mM Millimolar

NPC Nuclear pore complex

nt Nucleotides

ONM Outer nuclear membrane

PAGE Polyacrylamide gel electrophoresis

PBS Phosphate buffered saline

PCR Polymerase chain reaction

PD Population doubling

pg Picogram

PI Propidium iodide

PNA Peptide nucleic acid

PNS Perinuclear space

Q-FISH Quantitative-fluorescent in situ hybridisation

Q-PCR Quantitative-polymerase chain reaction

rcf Relative centrifugal force

RD Restrictive dermopathy

RNA Ribonucleic acid

rpm Revolutions per minute

SDS Sodium dodecyl sulphate

SSC Saline-sodium citrate

ssDNA Single-stranded DNA

STELA Single telomere length analysis

TBE Tris-borate-EDTA

Tel+ Telomerase positive

TEMED Tetramethylethylenediamine

Page 14: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

xii

TERC Telomerase RNA component

TRF Terminal restriction fragment

WS Werner syndrome

WT Wild type

μg Microgram

μl Microlitre

μM Micromolar

Page 15: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

1

Chapter 1 Introduction

1.1 The nuclear lamina

1.1.1 Overview

In eukaryotic organisms, the nucleus serves to compartmentalise essential

genetic processes away from the other activities of the cell. This segregation is

made possible by the nuclear envelope, a structure comprising of a double

membrane, nuclear pore complexes (NPCs) and the lamina. The outer nuclear

membrane (ONM) is contiguous with the endoplasmic reticulum and is resident

to ribosomes. NPCs span the nuclear membranes, providing a channel which

allows the passage of RNA and proteins in and out of the nucleus. The inner

nuclear membrane (INM) is home to a large variety of integrated proteins, the

composition of which varies between tissue- and cell-type (Schirmer and

Gerace, 2005; Schirmer et al., 2003). The nuclear lamina is a fibrous meshwork

composed of several proteins, primarily the lamins from which it derives its

name, as well as numerous INM proteins which interact with lamins and anchor

the lamina to the INM and perform several other nuclear functions. Two types

of lamins, A- and B-types, form the core of the lamina, which provides the

nucleus with its structural stability and shape, while interactions with binding-

partners allow the anchorage of chromatin to the lamina, and of the nucleus

within the cell. At least one B-type lamin is present constitutively in metazoan

cells, while A-type lamins are regulated developmentally, with particular

importance in skeletal and cardiac development (Ho et al., 2013; Solovei et al.,

2013; Swift et al., 2013).

Mutations in the lamin A gene may have dire consequences for the cell and the

individual, as is observed in the numerous and diverse disorders that arise from

Page 16: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

2

lamin A dysfunction, termed laminopathies. The structure of the lamina serves

to protect the integrity of the nucleus and its contents, and contributes to the

co-ordination of nuclear membrane break-down and re-assembly during cell

division (Patel et al., 2014). The lamina is essential for the organisational

distribution of chromatin throughout the nucleus, and chromatin attachment to

the INM (Dechat et al., 2008). Lamins are also thought to form an intranuclear

network, providing a scaffolding for the anchorage of replication, transcription

and splicing factors (Broers and Ramaekers, 2004).

The discovery of mutations in LMNA leading to progeroid disorders has led to

considerable research efforts into characterising the molecular basis of rapid

ageing in these individuals. As patients are not exposed to a life-time’s worth of

environmental factors, progeria is considered as an ideal model for the

molecular processes behind the normal ageing process (Olive et al., 2010).

1.1.2 Properties of lamins

Nuclear lamins comprise the family of type V intermediate filament (IF) proteins

present in all metazoans. Vertebrates have three lamin genes encoding seven

distinct isoforms (Cohen et al., 2001), which may be classified into two major

types of lamin, A- and B-type, according to sequence and biochemistry (Dechat

et al., 2008). In humans, the B-type lamins are encoded at two separate loci,

LMNB1 on chromosome 5q which encodes lamin B1, and LMNB2 located on

chromosome 19p which encodes lamins B2 and B3. Lamins B1 and B2 are

expressed throughout the cell lifespan (Stewart and Burke, 1987). A-type lamins

are encoded by the LMNA gene located at chromosome 1q and are

differentially expressed through alternative splicing (Figure 1.1:1a). Lamin A and

lamin C are the most abundant and are expressed in nearly all differentiated

cells (Dechat et al., 2008). Lamin C2, an additional splice variant, is germ-cell

specific (Furukawa et al., 1994; Nakajima and Abe, 1995), as is lamin B3

(Furukawa and Hotta, 1993), and it is likely that both contribute to

Page 17: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

3

spermatogenesis. A minor isoform, lamin AΔ10, is also produced by LMNA in

small amounts in several somatic cell types (Machiels et al., 1996). Evolutionarily,

it would seem that lamins hold the ancestral origin of IF proteins (Weber et al.,

1989), and that lamin B originated first, as invertebrates, which express only a

single lamin, express only B-type lamins (Erber et al., 1999).

Caenorhabditis elegans have one lamin gene, lmn-1, that resembles a B-type

lamin (Riemer et al., 1993). Reduction of lmn-1 results in chromosomal

abnormalities, nuclear deformities, and embryonic lethality (Liu et al., 2000).

Figure 1.1:1 Schematic representations of A-type lamins a LMNA gene transcript

including exon numbers and alternative splice sites used for the three major A-type

lamin isoforms. Lamin AΔ10 is initiated within intron 1 b Lamin A translated protein

indicating the relative positions of the head, rod and tail domains, the nuclear

localisation signal (NLS), immunoglobulin-like fold (Ig-LF), and known regions of

interaction with the proteins indicated. Adapted from Scharner et al. (2010).

Page 18: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

4

While several invertebrates express only one lamin, Drosophila melanogaster

express two forms of lamin: lamin Dm0, a B-type lamin that is processed at the

nuclear periphery into two forms, Dm1 and Dm2 (Gruenbaum et al., 1988), and

the developmentally regulated A-type DmC (Bossie and Sanders, 1993; Riemer

et al., 1995). Fungi and plants do not express lamins.

Despite significant sequence homology and functional overlap, A- and B-type

lamins are distinguishable by several differing biological factors. Lamin A and C

function is associated with developed cells (Stewart and Burke, 1987), while

lamin B is required for development. Targetted mutations in functional regions

of Lmnb1 results in neo-natal death and severe cellular abnormalities including

dysmorphic nuclei, differentiation problems and accelerated senescence in mice

(Vergnes et al., 2004). Unlike lamin A/C, B-type lamins are essential for viability

in mammalian cells (Harborth et al., 2001). Lamin A/C expression correlates with

the later stages of development (Constantinescu et al., 2006; Riemer et al., 1995;

Rober et al., 1989), although this may not be the case in all organisms (Foster et

al., 2007; Hall et al., 2005). Lamin B expression decreases during the lifespan of

cultured cells (Shimi et al., 2011) and, upon the induction of lamin A/C

expression, lamin B1 expression is reduced in chicken embryo development

(Lehner et al., 1987). In mitosis, B-type lamins remain associated with the

nuclear membrane while A-type lamins solubilise and disperse during mitosis

prior to re-assembly (Dechat et al., 2007; Gerace and Blobel, 1980; Lee et al.,

2000). Post-translational processing also differs between different lamins (see

section 1.1.2.1).

All lamins consist of a small N-terminal globular head excluding lamin C2

(Furukawa et al., 1994), a large C-terminal tail, and a central rod domain (Figure

1.1:1), through which lamins may homodimerise to form the basic lamin unit

(Taniura et al., 1995). The rod domain consists of four α-helical coils (1A, 1B, 2A

and 2B), joined by three linkers (L1, L12 and L2). Sequences at the rod ends

Page 19: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

5

mediate head-to tail polymer formation, which may then associate to form the

characteristic 10nm filaments of IFs (Stuurman et al., 1998, 1996), although the

globular head domain may also have a regulatory role in A-type lamin polymer

formation (Izumi et al., 2000). IF proteins form of a wide array of insoluble and

complex higher-order structures which have been observed in vitro for several

IF proteins, including keratin and vimentin filaments and aggregates (Coulombe

et al., 1990; Hatzfeld and Weber, 1991). This organisation creates high tensile

strength, allowing IF proteins to absorb mechanical stress, thus protecting cells

from damage. IF proteins may also transduce mechanical pressure to signalling

and gene regulation (Herrmann et al., 2009).

IF structures, including the lamina, are highly insoluble (Goldman et al., 1986),

and investigation of higher-order lamin organisation has been made difficult by

the propensity of lamins to form large paracrystal structures in vitro, which do

not occur naturally in vivo. Although the meshwork of A- and B-type lamins

form the core framework of the lamina, the inability in vitro to form higher-

order structures suggest a dependence on the association of lamina-associated

proteins for organisation (Gant et al., 1999). Although visualisation of higher-

order lamin structures has not been possible in mammals, head-to-tail dimers

have been observed in vitro assembling into long protofilaments in a staggered,

anti-parallel manner, which may then associate laterally and form 10nm

filaments in Caenorhabditis elegans (Ben-Harush et al., 2009) and an extremely

organised lattice structure has been observed in Xenopus laevis oocytes

(Stuurman et al., 1998).

The structure and stability of the lamina network conserves the integrity of the

nucleus. The lamina is also crucial for sensing and responding to mechanical

stress. The ability of the cell to sense mechanical strain is crucial to many

biological processes. Mechanical stress initiates a response cascade, whereby

physical forces induce conformation change in mechano-sensitive proteins of

Page 20: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

6

the cytoskeleton, exposing or altering molecular recognition sites, thereby

affecting biochemical signal transduction (Vogel and Sheetz, 2006). Mouse

embryonic fibroblasts (MEFs) lacking lamin A (Lmna-/-) exhibit altered

mechanotransduction, a loss of nuclear integrity and stiffness, and lowered

viability in response to stress (Broers et al., 2004; Lammerding et al., 2004).

Recent evidence suggests that the natural variation in amounts of lamin A in the

nuclear volume could be attributable to developmental regulation in relation to

mechanical forces. The expression level of lamin A responds to mechanical

stress during differentiation of tissue-types, and is proportionally increased in

cells that are exposed to high levels of mechanical strain, for example bone and

muscle tissue (Swift et al., 2013). High levels of lamin A/C result in a stiff nuclear

envelope that is less responsive to conformational change, while lamin B does

not contribute (Lammerding et al., 2006; Swift et al., 2013).

1.1.2.1 Lamin processing and localisation

Lamins are extensively decorated with post-translational modifications,

including SUMOylation, acetylation and phosphorylation (Simon and Wilson,

2013), however, most research interest is focused at the C-terminus, where a

series of processing events must be carried out in order to produce mature

lamin A, as illustrated in Figure 1.1:2a. Translocation to the nucleus after

translation is mediated via a C-terminal nuclear localisation signal (NLS)

consensus sequence resembling that of the SV40 large T-antigen (Loewinger

and Mckeon, 1988; Wu et al., 2014). However, localisation is also highly

dependent on a C-terminal CaaX box and the processing stages that modify the

C-terminus. With the exception of lamin C, all lamins are translated as precursor

molecules, terminating with CaaX (one cysteine, followed by two aliphatic

residues and any amino acid; CSIM in lamin A and CAIM in B-type lamins (Holtz

et al., 1989)). This sequence serves to mark several proteins for farnesylation via

farnesyltransferase (FTase) activity (Beck et al., 1990; Casey and Seabra, 1996;

Page 21: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

7

Farnsworth et al., 1989). After farnesylation, proteolytic cleavage between the

cysteine and first aliphatic residue is carried out by RCE or ZMPSTE24 (Maske et

al., 2003; Young et al., 2005) and the exposed cysteine residue is

carboxymethylated by isoprenylcysteine carboxyl methyltransferase (ICM)

(Clarke et al., 1988; Dai et al., 1998).

This form of lamin A, termed prelamin A, is then processed further, in an

apparently unique additional cleavage at the C-terminus (Figure 1.1:2a). The

second cleavage step, carried out by ZMPSTE24, releases the terminal fifteen

amino acids along with the farnesyl and methyl modifications (Corrigan et al.,

2005). As lamin C does not contain the CaaX motif, and is therefore never

farnesylated, this final maturation step of lamin A represents the major

difference between A- and B-type lamins, as B-type lamins remain farnesylated.

Lamin A processing is also unique among farnesylated proteins in that

processing steps may occur in the nucleus, rather than at the ER (Clarke et al.,

Figure 1.1:2 Lamin A post-translational processing of the C-

terminal tail domain to produce mature lamin a from a WT

lamin A nascent protein, or b mutant nascent lamin A

resulting from aberrant activation of a cryptic splice site in

exon 11. The ZMPSTE24 recognition site is absent in the

mutant protein

Page 22: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

8

1988; Sinensky et al., 1994). The CaaX box may also play a role in the formation

of higher-order lamin structures (Izumi et al., 2000).

The role of this cleavage-modification-cleavage is thought to aid localisation of

lamin A to the INM. It would seem that farnesylation is required for localisation

of prelamin A to the nuclear periphery, perhaps due to the hydrophobic nature

of the farnesyl and carboxymethyl groups (Hennekes and Nigg, 1994; Lutz et al.,

1992). Prelamin A may then be cleaved in situ (Lutz et al., 1992; Sasseville and

Raymond, 1995; Sinensky et al., 1994). Indeed, peripheral localisation of human

lamin C, which lacks its own temporal farnesylation signal, is dependent on the

re-location of lamin A (Vaughan et al., 2001). However, farnesylation is not

required in the mouse system. Mouse lamin A is able to locate to the periphery

in the absence of a farnesyl tag (Davies et al., 2010), and lamin C is also able to

integrate into the lamina without lamin A (Fong et al. 2006).

Not all lamin locates to the periphery, and this may be crucial for cell biology. A

sub-fraction of A-type lamins that remains in the nucleoplasm is purported to

form an intranuclear skeleton-like structure, which may aid recruitment of

transcription factories and DNA replication factors, tethering them to specific

nuclear regions (Dechat et al., 2008, 2007; Shumaker, 2003). The organisation of

intranuclear lamins may be mediated by LAP2α (Gant et al., 1999), and this

organisation results in a more motile and soluble structure than the lamin A

found at the periphery (Dahl et al., 2006). A fraction of B-type lamin is also

retained in the intranuclear region, and may too aid DNA replication (Moir et al.,

1994).

Page 23: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

9

1.1.3 Additional lamina components and lamin-interacting

partners

Numerous non-lamin proteins contribute to the functions of the lamina and, in

the cases where function has been elucidated, these interactions are known to

be important for organisation and positioning of the lamina, as well as

chromatin interactions with the lamina. Most interactions have been confirmed

through in vitro assays. Interactions are described below and summarised in

Figure 1.1:3.

1.1.3.1 The LINC complex

The nucleus is held in position in the cytoplasm via a complex of proteins

known as the linker of nucleoskeleton and cytoskeleton (LINC). The LINC

complex spans the INM and ONM, providing a physical bridge between the

nucleoplasm and the cytoskeleton. This complex interacts with and connects

elements of the lamina and the cytoskeleton, allowing mechanotransduction

Figure 1.1:3 Schematic of the nuclear membranes, including some of the known associated

factors (not to scale). Represented is the peripheral lamina, composed of a meshwork of A-

and B-type lamins and their associated factors as indicated in the legend. ER=endoplasmic

reticulum, PNS=perinuclear space, NPC=nuclear pore complex, INM=inner nuclear

membrane, ONM=outernuclear membrane.

Page 24: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

10

signalling between the two compartments, and absorbing mechanical strain

(Brosig et al., 2010; Chambliss et al., 2013; Lombardi et al., 2011). The complex

consists of a lamina-interacting, INM-associated protein, and an ONM-

associated factor capable of tethering to the cytoskeleton and binding within

the perinuclear space (PNS). In humans, the Sad1 and UNC84 domain-

containing (SUN) proteins SUN1 and SUN2 (encoded by the SUN1 and SUN2

genes respectively) form the basal INM unit for the LINC complex. Most SUN

proteins consist of a large N-terminal nucleoplasmic region, a central trans-

membrane domain and a coiled-coil domain, and a C-terminal SUN domain

which spans the PNS. The luminal C-terminus SUN domain is highly conserved

evolutionarily, from plants to yeast to mammals (Méjat and Misteli, 2010).

Homotrimerisation of SUN proteins is mediated by the SUN domain (Zhou et

al., 2012) and both the SUN and coiled-coil domains recruit and bind the ONM

LINC element by means of a Klarsicht/ANC-1/Syne-1 homology (KASH) domain

(Sosa et al., 2012). In mammals, the KASH-domain containing protein family is

composed of nesprins (nuclear envelope with spectrin repeats). The nesprin

family is large and extremely diverse, including isoforms ranging wildly in size

(from ~45kDa to ~1MDa), encoded by four genes (SYNE1-4). Although most

nesprins translocate to the ONM to form the LINC complex with SUN proteins,

specificity to the ONM or INM is dependent on the size of the nesprin and its

interaction partners. Small nesprins, including nesprin-1α, may be transported

through NPCs to the INM and interact directly with lamin A/C (Mislow et al.,

2002). Nesprin-1α targets muscle A-kinase anchoring protein (mAKAP) to the

nucleus of muscle cells (Pare et al., 2005), which may have importance in disease

development in the disorder Emery-Dreifuss muscular dystrophy (EDMD, see

section 1.1.4.1). Nesprin-1 and -2 mainly bind actin, while nesprin-4 associates

with microtubules and nesprin-3 is able to bind IFs and nuclear microtubules

indirectly through its own interacting partners (Ketema and Sonnenberg, 2011).

Page 25: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

11

1.1.3.2 LEM domain-containing proteins and BAF

The LEM domain is so-named due to its initial discovery in three proteins: LAP2,

Emerin and MAN1, all of which are able to bind lamins. Located towards the N-

terminus, the LEM domain consists of a motif that has evolved for specific

binding of barrier to autointegration factor (BAF) (Brachner and Foisner, 2011).

BAF is able to bind double stranded DNA in a non-specific manner (Zheng et al.,

2000), and is a key factor in mediating chromatin organisation through

coordination with the lamina.

Six LAP2 isoforms are encoded by alternative splicing of the LAP2 locus, and

most bind the lamina through lamin B1 (Foisner and Gerace, 1993). As well as a

LEM-domain, LAP2s contain a second, LEM-like, direct DNA binding motif (Cai

et al., 2001). LAP2α is the exception of the LAP2 group, as it does not contain a

transmembrane domain and so does not associate at the nuclear periphery, but

with intranuclear A-type lamins (Dechat et al., 2000). This interaction is required

for formation of the intranuclear lamin structure, and may coordinate chromatin

organisation within the nucleus (Dorner et al., 2007; Gant et al., 1999). As well as

interacting directly with A-type lamins, LAP2α also interacts with retinoblastoma

protein Rb, providing anchorage within the nucleoplasm that is essential for Rb

function, suggesting a role for lamins in cancer (Markiewicz et al., 2002), as well

as a potential route for delayed cell division observed in patients with lamin A

mutations (Dechat et al., 2007).

Emerin and MAN1 (encoded by genes EMD and MAN1, respectively) share

much homology and there is a large redundancy between the two proteins in

terms of their function in chromosome segregation at mitosis in C. elegans, (Liu

et al., 2003). Emerin and MAN1 are able to bind one another, as well as BAF

(Mansharamani and Wilson, 2005; Shimi et al., 2004). Emerin is also able to bind

lamin A (Lee et al., 2001), which is required for emerin localisation to the

nucleus (Vaughan et al., 2001). Emerin also stabilises the LINC complex through

Page 26: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

12

interactions with nesprin-1α and -2β (Mislow et al., 2002; Wheeler et al., 2007)

and SUN1/2 (Haque et al., 2010). Mutations in the EMD gene lead to the X-

linked form of EDMD (Bione et al., 1994).

BAF is a small protein (only 10kDa) that forms dimers that are able to

multimerise in vitro upon binding DNA (Zheng et al., 2000). As well as binding

DNA, BAF may be able to influence heterochromatin density (Segura-Totten et

al., 2002). BAF is able to bind LAPs, emerin and nesprins through the LEM

domain of each and this interaction is essential for proper nuclear re-assembly

of the lamina after mitosis, and for cell viability (Lee et al., 2001; Mansharamani

and Wilson, 2005; Shumaker et al., 2001).

1.1.3.3 Other binding partners

Large numbers of novel nuclear envelope transmembrane (NETs) and linker

proteins have been identified (Schirmer and Gerace, 2005), of which, most of

the best characterised are described above. However several other factors may

be pertinent to lamin biology. An actin-binding site has been identified and

confirmed in lamins, and lamin A contains an additional site that is able to

mediate F-actin bundling (Simon et al., 2010). NPC components NUP153 and

NUP88 have been shown to associate directly with lamin A through purified

pull-down methods (Al-Haboubi et al., 2011; Lussi et al., 2011), confirming an

early report of NPC association with the lamina (Aaronson and Blobel, 1975).

Overexpression of several NETs results in redistribution of chromatin in the

nucleus, as well as relocalisation of specific chromosomes (Zuleger et al., 2013).

This induces tissue-specific expression patterns, in accordance with findings that

the nuclear envelope proteome differs greatly between cell-types (Korfali et al.,

2012). Lamin A/C has been shown to bind the transcription factors

MAPK/MAK/MRK overlapping kinase (MOK2) (Dreuillet et al., 2002; Harper et al.,

2009) and sterol regulatory element binding protein 1 (SREBP1) (Lloyd et al.,

Page 27: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

13

2002). Mutations in lamin A resulting in familial partial lipodystrophy (FPLD, see

section 1.1.4.1) affect the affinity of lamin A to SREBP1, potentially affecting

adipocyte differentiation which SREBP1 is required for (Lloyd et al., 2002).

Reduction of lamin A or emerin compromises the differentiation potential of

muscle cells, associated with reduced levels of retinoblastoma protein (pRb),

Myosin D, desrin, M-cadherin and other factors involved in muscle

differentiation (Frock et al., 2006). In this study, transcript levels of myosin D and

desmin were found to be lowered in Lmna-/- myoblasts (Frock et al., 2006),

suggesting that lamin A could be mediating transcriptional activation, while

lamin A/C regulates pRb levels through inhibition of its proteasomal

degredation (Johnson et al., 2004). Although no direct binding has been

identified between lamins and the transcription factor megakaryoblastic

leukaemia 1 (MKL1), a mechanosensitive transcription factor important in

cardiac development, it’s normal nuclear distribution is dependent on the

presence of lamin A/C (Ho et al., 2013), reinforcing the importance of A-type

lamins in cardiac development.

Intranuclear A- and B-type lamins are able to mediate organisation of discreet

compartments containing RNA polymerase II and its associated splicing factors

in human and mammalian cells (Jagatheesan et al., 1999; Kumaran et al., 2002;

Spann et al., 2002). This association between lamins and splicing factors may be

cell-type dependent and required only during differentiation (Muralikrishna et

al., 2001). Lamins are also involved in DNA replication, and disruption of the

intranuclear lamin network affects the distribution of replication factors and

DNA synthesis efficiency (Spann et al., 1997).

The Lamin B receptor (LBR), aside from binding B-type lamins (Ye and Worman,

1994), is able to directly bind chromatin, anchoring it to the lamina

(Pyrpasopoulou et al., 1996), as well as bind heterochromatin proteins HP1-α

and HP1-γ (Ye and Worman, 1996; Ye et al., 1997). Interestingly, lamin A is able

Page 28: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

14

to bind the replication factor proliferating cell nuclear antigen PCNA, which may

have implications in DNA damage and cell cycle progression, as PCNA can be

relocalised in the presence of mislocalised, N-terminally truncated lamin A

(Spann et al., 1997). Lamins may interact with DNA in a more direct manner

through histones (Taniura et al., 1995), or even direct binding to duplex DNA

(Stierlé et al., 2003).

1.1.4 Lamin A dysfunction and laminopathies

The identification of mutations in LMNA has revealed a host of functions that

are reliant upon normal lamin A function. Such roles include chromatin

organisation and anchorage (Dechat et al., 2008), transcription (Shimi et al.

2008), RNA splicing (Kumaran et al., 2002), replication (Spann et al., 1997),

cellular and nuclear stability (Lombardi et al., 2011) and cell cycle progression

(Dechat et al., 2007). These diverse roles are mirrored by a wide variety of

disorders associated with mutations throughout the various domains of lamin A,

with symptoms that include muscular dystrophy and myopathy,

cardiomyopathy, lipodystrophy and progeria. Diseases caused by mutations in

genes encoding lamins and lamina-associated proteins are collectively termed

laminopathies. Mutations in lamin B are rarely reported, most likely due to the

requirement of a base-level of functionality for viability (Harborth et al., 2001).

Therefore the most commonly recorded lamin B mutations involve duplications

(Padiath and Fu, 2010), however mutations in LMNB2 have been implicated as

modifiers in acquired partial lipodystrophy (Hegele et al., 2006).

1.1.4.1 Non-progeroid laminopathies

Mutations occurring throughout lamin A result in several non-progeroid

disorders that tend to manifest as muscular dystrophies, but include dilated

cardiomyopthy (DCM), familial partial lipodystrophy (FPLD) and the neuropathy

Charcot-Marie-Tooth disorder (CMT).

Page 29: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

15

Muscle wasting disorders include Emery-Dreifuss muscular dystrophy (EDMD),

limb girdle muscular dystrophy subtype B1 (LGMDB1) and congenital muscular

dystrophy (CMD). EDMD is mainly caused by mutations in LMNA and EMD.

Symptoms include tendon contracture, muscular dystrophy and heart defects

(Emery, 1989). Mutations in EMD, which encodes emerin, cause the X-linked

form of the disease, and single mutations may result in broad phenotypic

variation (Hoeltzenbein et al., 1999). Rarer cases of EDMD result from mutations

in genes encoding nesprin-1 and -2 (Zhang et al., 2007) and putatively SUN1

and -2 (Meinke et al., 2014). LMNA mutations may be dominant or recessive and

two LMNA EDMD mutations result in the condition CMD, as well as different

amino acid changes at codon 249 (Figure 1.1:4).

CMD is caused by LMNA mutations affecting the rod domain and C-terminal

head. Muscle wasting may be accompanied by delayed development and joint

contractures, although there is a broad range of phenotypic variation (Quijano-

Roy et al., 2008). Symptoms include ventricular dilation and hypertrophy,

potentially leading to heart failure. Lamin A is mutated in >10% of DCM

individuals, and mutation may be inherited or sporadic. Mutations are located

along the length of lamin A (Figure 1.1:4). A genotype-phenotype analysis

revealed that individuals with lamin A mutations are prone to an earlier age of

onset and more severe symptoms (Taylor et al., 2003). Limb girdle muscular

dystrophy is also caused by mutations in several genes, however those

identified in LMNA cluster in the C-terminal tail domain of lamin A (Figure 1.1:4),

and may also result in EDMD (Muchir et al., 2000).

Familial partial lipodystrophy (FPLD), results from missense mutations in regions

(Figure 1.1:4). FPLD patients exhibit a normal appearance until after puberty,

when subcutaneous fat is then lost from the limbs and trunk, while

simultaneously accumulating in the face and neck. Other symptoms include

prominent musculature, altered skin pigmentation and diabetes (Köbberling

Page 30: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

16

and Dunnigan, 1986). Expression of residue R482 FPLD mutants holds no effect

on myocyte differentiation (Favreau et al., 2004), while adipocyte differentiation

is affected (Boguslavsky et al., 2006).

Charcot-Marie-Tooth disease is a genetically and symptomatically

heterogeneous collection of common neuropathic disorders with many

subtypes, a small proportion of which stem from a single homozygous mutation

at residue R298 in lamin A (De Sandre-Giovannoli et al., 2002), resulting in

subtype 2. Common symptoms include muscle weakness and wasting.

Generally, muscular dystrophy is associated with mutations which impact the

function and stability of the LINC complex, as highlighted by the gene

mutations in LINC components that cause EDMD. Mutations in lamin A that

result in muscular dystrophy and cardiomyopathy also result in impaired nuclear

response to mechanical stress (Zwerger et al., 2013). Emerin localisation is

affected by the presence of muscular dystrophy-associated LMNA mutants

Figure 1.1:4 Lamin A amino acid changes resulting in laminopathies. DCM=Dilated

cardiomyopathy, EDMD=Emery-Dreifuss muscular dystrophy, CMD= congenital muscular

dystrophy, FPLD=familial partial lipodystrophy, CMT2=Charcot-Marie-Tooth disorder type 2,

LGMDB1=limb girdle muscular dystrophy type B1, NT=N-terminus, CT=C-Terminus. V440M

and R584H from Hegele et al. (2000), R482W/Q/L from Shackleton et al. (2000). Adapted from

Scharner et al. (2010).

Page 31: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

17

(Manju et al., 2006), and mislocalisation of emerin is associated with impaired

function of MKL1 (Ho et al., 2013). Several of the mutation locations in LMNA

that associate with FPLD overlap the locations of atypical Hutchinson-Gilford

Progeria Syndrome (HGPS) and atypical Werner syndrome (WS). Lipodystrophy

is a common symptom of HGPS, indicating that these regions of lamin A as

specific to adipocyte function. This is most likely through disruption of the

interaction with SREBP1 which binds between lamin A residues 227 and 487,

where the majority of mutations in FPLD cases reside (Lloyd et al., 2002).

However, over-expression of WT lamin A affects adipocyte differentiation and

inhibits lipid synthesis and accumulation, again suggesting that aberrant

accumulation of even WT lamin A may be sufficient for disease (Boguslavsky et

al., 2006), and almost all laminopathies present with abnormal distribution of

adipose tissue.

1.1.5 Hutchinson-Gilford progeria syndrome

HGPS is an extremely rare condition affecting around one child in every eight

million (Pollex and Hegele, 2004). The vast majority of children with HGPS are

said to have classical HGPS and carry a single base de novo mutation (1824C>T)

in LMNA (Eriksson et al., 2003; Hennekam, 2006). Children suffering from this

classical form of the disease are born with a completely normal appearance but

within 1 to 2 years develop notable features including alopecia, loss of

subcutaneous fat and very slow growth. Further symptoms include hypo- and

hyper-pigmentation of the skin, sclerodermatous skin, arthritis and

osteoporosis. General frailty and short stature as well as distinctive, small facial

features, prominent veins due to subcutaneous fat loss and aged-looking skin

produce an overall appearance of old age. Due to the overlap with symptoms of

normal ageing, HGPS is categorised as a segmental premature ageing

syndrome. The average age of death is approximately 13 years and is almost

Page 32: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

18

invariably due to cardiac failure due to atherosclerosis or stroke (Al-Shali and

Hegele, 2004).

1.1.5.1 Lamin A processing and progerin expression in classical HGPS

In classical HGPS, the 1824C>T mutation activates a cryptic splice site in exon

11, consequently excluding the remaining 150bp of the exon (Eriksson et al.,

2003). The resultant 50 amino acid deletion includes the final ZMPSTE24

cleavage site required for release of the modified C-terminus (Figure 1.1:2b).

Activation of this site is “leaky”, in that some of the transcripts produced by the

mutant allele are WT. The WT allele produces WT transcripts at a normal rate,

leading to an overall ratio of 2:3 mutant to WT transcripts (Reddel and Weiss,

2004).

The omission of the ZMPSTE24 processing step resulting from the loss of its

recognition site produces a form of lamin A that lacks 50 amino acids but

retains farnesylation (Figure 1.1:2). This protein is termed progerin or LAΔ50

(Eriksson et al., 2003; Hennekam, 2006). Lamin C transcription is terminated in

exon 10 and as such is unaffected by the mutation (Figure 1.1:1). Cells that

express progerin exhibit several nuclear deformities, including blebbing of the

nuclear membrane, altered mechanosensitivity and dissociation from the

nuclear periphery and change in composition of heterochromatin (Goldman et

al., 2004).

The nuclei of HGPS cells display striking morphology, exhibiting lobulations,

blebs, folds and wrinkles (Figure 1.1:5) at high levels in cell populations,

particularly at later passages ex vivo (Scaffidi and Misteli, 2005). HGPS cells with

abnormal morphology within a sample population display higher levels of DNA

damage than wild-type or HGPS cells with a normal nuclear appearance,

indicating that alterations to the physical organisation of the lamina may have a

role in disease (Constantinescu et al., 2010). Interestingly, progerin is expressed

Page 33: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

19

at low levels in the cells of healthy individuals and may accumulate with age,

which is caused by sporadic activation of the cryptic splice site mutated in HGPS

(Scaffidi and Misteli, 2006). The presence of progerin in the cells of normally

aged individuals is associated with an increase in nuclear dysmorphia, as well as

other cellular symptoms common to HGPS (Scaffidi and Misteli, 2006), including

shortened telomeres and increased senescence (Cao et al., 2011). Evidence also

suggests that with increasing numbers of cellular divisions, progerin transcripts

are upregulated (Rodriguez et al., 2009). This indicates that progerin

progressively accumulates in the nucleus, even in normally aged individuals,

causing nuclear deformities, and that this process is accelerated in HGPS cells.

Lamin B organisation is also altered in HGPS cells (Goldman et al., 2004; Taimen

et al., 2009), and global levels are depleted (Scaffidi and Misteli, 2005) indicating

that lamin B and its interactors could also contribute to the HGPS phenotype.

1.1.5.2 Impact of progerin on nuclear lamin organisation

The presence of progerin results in an over-accumulation of A-type lamins at

the nuclear periphery. The lamina thickens in HGPS cells, and this effect worsens

with the age of cells in culture (Goldman et al., 2004). Progerin as well as WT

lamin is integrated into the lamina matrix more readily in HGPS cells, (Scaffidi

and Misteli, 2005). It is possible that accumulation of progerin at the lamina is

Figure 1.1:5 Examples of nuclei in a normal human foreskin fibroblast (control),

classical HGPS, late onset HGPS (T623S) and restrictive dermopathy (RD) fibroblasts

immunostained for lamin A/C. Images provided by the Shackleton laboratory and

reproduced with permission.

Page 34: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

20

due to aberrant localisation signalling from the farnesyl group, which instigates

morphological changes at the nuclear membrane (Capell et al., 2005). However,

though retention of the farnesyl group is sufficient to cause abnormal nuclear

morphology, several non-classical mutants which do not aberrantly retain C-

terminal farnesylation are also able to trigger nuclear dysmorphia (Huang et al.,

2008; Taimen et al., 2009).

The proclivity of progerin for integration into the lamina holds other

implications for the cell, as it results in reduced levels of progerin and lamin A

levels in the nucleoplasm (Scaffidi and Misteli, 2005). Intranuclear lamin A may

aggregate and coordinate the association of DNA damage response (DDR)

proteins and DNA in subnuclear compartments (Dechat et al., 2008; Shumaker,

2003). This depletion of lamin A from the nuclear space in HGPS cells (visible in

Figure 1.1:5) could therefore alter expression levels if transcription factor

recruitment is impaired. Alternatively, disrupted interactions at the periphery or

within the nuclear space that affect chromatin-associated factors may be

responsible for the alterations seen in heterochromatin observed in HGPS (see

below).

The stability of the lamina network is drastically altered in HGPS patients.

Fluorescence Recovery after Photobleaching (FRAP) assays have demonstrated a

progressive and severe decrease in A-type lamin mobility in HGPS nuclei with

cellular age (Dahl et al., 2006; Scaffidi and Misteli, 2005). This has consequences

for the mechanistic response of HGPS nuclei to external pressures. The HGPS

lamina loses elasticity in a progressive manner and has a reduced ability to

respond to mechanical stress (Verstraeten et al., 2008), as does the lamina of

cells ectopically expressing progerin (Dahl et al., 2006). This mechanistic effect

could itself be responsible for dissociation of chromatin and chromatin

associated factors from the lamina through steric hindrance (Scaffidi and

Misteli, 2005). It has also been suggested that increased cellular sensitivity to

Page 35: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

21

mechanical strain contributes to loss of vascular smooth muscle cells, and

therefore early death in HGPS patients caused by atherosclerosis (Verstraeten et

al., 2008).

1.1.5.3 Impact of progerin on chromatin

The locality of chromatin in the nucleus is important for, and indicative of

expression levels (Croft et al., 1999). Heterochromatin is thought to interact with

lamin A either directly or via LEM or other lamina-associated proteins (see

sections 1.1.3.2 and 1.1.3.3) and all lamins bind DNA in vitro (Shoeman and

Traub, 1990). Lamin A- and lamin B-associated chromatin is generally

heterochromatic and transcriptionally silent (Guelen et al., 2008; Kubben et al.,

2012). Dissociation of heterochromatin from the nuclear periphery is a key

marker of HGPS cells (Dechat et al., 2008; Goldman et al., 2004), and a

generalised increase in global decondensation has been observed (Csoka, 2004;

Shumaker et al., 2006). Trimethylation of histone 3, lysine residue 9 (H3K9me3),

a marker of constitutive chromatin, is reduced in HGPS cells and subsequently

chromatin interaction with HP1-α, a binding partner of LBR (Ye et al., 1997)

which binds through H3K9me3, is also reduced (Shumaker et al., 2006). In

contrast, trimethylation of histone 4, lysine 20 (H4K20me3), a facultative

chromatin marker, is upregulated in HGPS and progerin-expressing HeLa cells

(Shumaker et al., 2006). Progerin associates with almost 100 additional genes

when compared with WT lamin A, while losing interaction with only 8 upon

expression in MEFs (Kubben et al., 2012). As a consequence, the global

expression profile of HGPS cells is altered, with the greatest effects on

transcription factor and extracellular matrix protein expression (Csoka et al.,

2004). Heterochromatin alterations are observed before abnormal nuclear

morphology is observed (Shumaker et al., 2006), implying that lobulation of the

nuclear membrane cannot be directly linked to chromatin alterations. DNA

methylation is also affected in HGPS, altering expression of several genes

Page 36: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

22

including the inflammatory transcription factor nuclear factor kappa-light-

chain-enhancer of activated B cells NF-κB (Heyn et al., 2013).

A constitutively active DDR (Csoka, 2004), high incidence of double strand

breaks (DSBs) and large numbers of persistent DNA damage foci in HGPS and

other laminopathy and laminopathy-model cells are widely reported (Gonzalez-

Suarez et al., 2009; Li and Zou, 2005; Liu et al., 2008, 2006). Liu et al.

demonstrated a delayed response of p53 binding protein 1 53BP1, a key

mediator of DDR factors, to recruit to γ-H2AX foci (a marker for DSBs) in

Zmpste24-/- MEFs and HGPS cells following γ-irradiation, as well as activated

checkpoint protein CHK1 (Liu et al., 2005). This and other studies have recorded

the involvement of global DNA repair factors ataxia-telangiectasia mutated

(ATM) and ataxia telangiectasia and Rad3 related (ATR), in the DDR of HGPS

cells (see section 1.3.1), working through CHK1 and CHK2 to halt cells at the

G1/S phase and G2/M checkpoint (d’Adda di Fagagna et al., 2003; Liu et al.,

2006), presumably causing the slow cell cycle progression of HGPS cells. In

2008, Liu et al. suggested this could explain their finding that XPA (normally

exclusively a factor in nucleotide excision repair and unrelated to DSB repair) is

bound aberrantly to DSB ends (Liu et al., 2008). This is a potential mechanism

for DSB accumulation as XPA could be blocking binding of required factors and

contributing to the formation of permanent lesions. Lamins may also regulate

cycle through control of retinoblastoma protein (pRB) nuclear localisation and

proteasomal degradation (Johnson et al., 2004).

1.1.5.4 Franesyl transferase treatment

Farnesyl transferase inhibitors (FTIs) block the action of farnesyl transferases and

have been employed experimentally to lower farnesylated lamin A levels. FTIs

have proven to dramatically reduce lamin A accumulation at the nuclear

periphery and alleviate nuclear dysmorphia in HGPS cells ex vivo (Capell et al.,

2005; Constantinescu et al., 2010; Toth et al., 2005), in cells ectopically

Page 37: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

23

expressing lamin A mutants (Capell et al., 2005; Mallampalli et al., 2005) and in

mouse models (Loren G Fong et al., 2006; Yang et al., 2008, 2006, 2005). Several

other symptoms of HGPS nuclear dysfunction are also improved with FTIs,

including a return of chromatin localisation and the global gene expression

profile (Columbaro et al., 2005; Scaffidi and Misteli, 2005) and DSB repair

capacity (Constantinescu et al., 2010). Mice expressing a progerin allele

(LmnaHG/+) exhibit similar symptoms to humans with HGPS, including a normal

appearance at birth followed by growth deficiency, loss of subcutaneous fat,

bone weakening and reduced survival (Yang et al., 2006). Administering FTIs to

these mice and genetically identical MEFs not only improves nuclear

dysmorphia (Yang et al., 2005), but also improves the developmental

deficiencies described above (Yang et al., 2006), as well as improving survival

(Yang et al., 2008).

However, certain symptoms of HGPS are not addressed by FTIs, such as DNA

damage that has already accumulated (Constantinescu et al., 2010; Liu et al.,

2006), or mechanosensitivity, despite a correction of increased nuclear stiffness

(Houben et al., 2007). As increased senescence and/or apoptosis due to

mechanosensitivity in arterial cells could be the root of atherosclerosis in HGPS

patients (Verstraeten et al., 2008), the major cause of death in HGPS, FTIs may

not protect against early fatality.

1.1.6 Atypical HGPS and other progeroid laminopathies

Atypical HGPS conditions are so termed as they derive from mutations other

than the typical G608G, and have a tendency to deviate from the classical

phenotype, frequently displaying symptoms from other laminopathy syndromes

(Smallwood and Shackleton, 2010). ~80% of HGPS cases are a result of the

classical G608G mutation, while the remainder are caused by other, less well

classified mutations (Hennekam, 2006). Atypical mutations span the length of

LMNA and range from apparently simple missense mutations to alternative

Page 38: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

24

mechanisms of cryptic splice site activation (Figure 1.1:6). These atypical

mutations may result in a range of phenotypes present in classical HGPS, but

generally to a less severe extent, and in some cases symptoms resemble or

overlap with other laminopathy syndromes. A subset of individuals share many

symptoms of Mandibuloacral Dysplasia (MAD), which in many ways resembles a

milder form of HGPS (Hennekam, 2006).

MAD is a very rare disorder caused by mutations in various locations in the

LMNA gene (Figure 1.1:6), and results in mandibular and clavicular hypoplasia,

loss of subcutaneous fat, dappled skin pigmentation and progeroid features

(Simha et al., 2003). In a similar manner to atypical HGPS, manifestation of

symptoms varies between individuals (Ahmad et al., 2010; Bai et al., 2014; Garg

Figure 1.1:6 Lamin A amino acid changes resulting in progeroid disorders. Mutations are

limited to the first half of the rod domain and the C-terminal head domain. V169fs= frameshift

from codon 169 to stop codon (Scharner et al., 2010), G567del= intronic donor splice site

deletion IVS11+1G>A, resulting in deletion of exon 11 (Doh et al., 2009). G608G, although a

silent mutation, sporadically initiates splicing within exon 11, as does the T623S mutation. One

instance of G608G resulting in RD phenotype has been recorded (Navarro et al., 2004).

Adapted from Kudlow et al. 2007. HGPS mutants V607V and G567del from Moulson et al.

2007. T528M and M540T do not cause disease except when inherited together (Navarro et al.,

2004). HGPS=Hutchinson-Gilford Progeria Syndrome, A-WS=atypical Werner Syndrome,

MAD=Mandibuloacral dysplasia, RD=restrictive dermopathy, NT=N-terminus, CT=C-Terminus

Page 39: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

25

et al., 2005; Novelli et al., 2002; Simha et al., 2003). At the severe end of the

spectrum is another rare disorder, Restrictive Dermopathy (RD), which exhibits

several symptoms of HGPS and MAD, alongside several other severe

phenotypes.

RD is usually caused by homozygous or compound heterozygous ZMPSTE24

mutations that eliminate ZMPSTE24 function (Moulson et al., 2005; Navarro et

al., 2013, 2005). Due to the role of ZMPSTE24 in lamin A processing (see section

1.1.2.1), loss of function results in an absence of native lamin A expression,

which is replaced by prelamin A. Prelamin A is a partially processed form of

lamin A which retains the farnesyl modification. This results in devastating

consequences for the individual, evident by intrauterine growth retardation and

neonatal death. Symptoms include tight, sclerotic and easily broken skin,

particularly evident at joints, which are stiff and retracted. Shared symptoms

Figure 1.1:7 Reverse transcriptase

PCR results from T623S, HGPS and

control cells (U2OS) for WT and

mutant transcripts. Data was

generated in the Shackleton lab and

is reproduced with permission.

au=arbitrary units.

Table 1.1-1 Position and sequence of cryptic splice sites mutated in T623S and G608G LMNA,

compared to the consensus major-class splice activation sequence. Consensus bases are given

in bold. Mutant transcript sequences are given for bases 1863-1869 in T623S and bases 1818-

1824 in HGPS (G608G).

Page 40: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

26

with MAD and HGPS include prominent blood vessels, small facial bones,

clavicular hypoplasia and hair loss (Navarro et al., 2004). At the cellular level, RD

nuclei resemble those of HGPS patients, exhibiting blebbing and altered

chromatin organisation (Columbaro et al., 2010). ZMPSTE24 mutations may also

cause HGPS and MAD-like phenotypes (Agarwal et al., 2006, 2003; Denecke et

al., 2006; Shackleton et al., 2005), the milder symptoms of which are due to, and

whose severity negatively correlate with, residual ZMPSTE24 function

(Barrowman et al., 2012).

Two cases of RD resulting from mutations in LMNA have been reported. One

individual expressed a lamin A form lacking the 90 amino acids (termed lamin A

Δ90) encoded by exon 11 due to an intronic splice site mutation (Figure 1.1:6).

The other was genotyped as having the classical G608G mutation, suggesting

that genetic modifiers may affect the disease outcome (Navarro et al., 2004).

Although their conditions were extremely severe and resulted in very early

death, their symptoms were milder than classical RD, perhaps due to residual

expression of WT lamin A (Navarro et al., 2004).

An additional mutation, T623S, results from a single base change 1868C>G

resulting in deletion of exon 11 codons. This is a result of activation of a cryptic

splice site, different to that affected by the classical 1824C>T mutation, but

results in a similar transcript that lacks the ZMPSTE24 binding site, as well as 35

amino acids, and as such is termed lamin A Δ35 (Figure 1.1:6 and Table 1.1-1).

The T623S phenotype exhibits the symptoms of classical HGPS but at a slower

rate, and with a later age of onset (Fukuchi et al., 2004; Shalev et al., 2007). As

progressive loss of exon 11 in mutant transcripts correlates with an increase in

severity, this could demonstrate a functional importance of this region of lamin

A. However, it has been shown that over-expression of even wild-type lamin A

in human fibroblasts induces HGPS-like characteristics (Huang et al., 2008), and

that farnesylated lamin A accumulates at the nuclear periphery over time in

Page 41: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

27

HGPS fibroblasts, correlating with an increased nuclear membrane aberrations

and chromatin localization disruption (Goldman et al. 2004; McClintock et al.

2006).

The classical HGPS 1824C>T mutation adheres more strongly to the consensus

than the T623S 1868C>G mutation (Patel & Steitz 2003 and Table 3.1-1). As

such, the 1868 (T623S) mutation confers a splice site that is activated less

frequently than the classical 1824 mutation. Reverse transcriptase-PCR carried

out in the Shackleton laboratory demonstrates a 2.5-fold increase in progerin

transcripts compared to Δ35. This is in agreement with Western blot results

which demonstrate a higher proportion of progerin relative to WT lamin A when

compared to Δ35 and WT lamin A (Figure 3.1:2, p86). Also, two additional HGPS

mutations which cause elevated activation of the splice site used in classical

HGPS, V607V (1821G>A) and an intronic mutation (IVS11+1G>A), result in

higher levels of progerin expression and more severe phenotypes (Moulson et

al., 2007). It is therefore extremely likely that the delayed phenotype

manifestations in the atypical T623S form are a result of slower accumulation of

farnesylated lamin A at the nuclear periphery. It is possible that increased

expression of lamin A Δ90 could be a causative factor in the severity of its

accompanying RD-like syndrome, although this would require confirmation.

Werner Syndrome (WS) is a progeroid syndrome usually caused by mutations in

the WRN gene which encodes the helicase WRN (see section 1.2.3.6). However,

~20% of patients are referred to as atypical and have mutations in LMNA (Chen

et al., 2003; Doh et al., 2009). Mutations cluster in the first half of the rod

domain of lamin A (Figure 1.1:6). WS symptoms are variable between individuals

and do not become apparent until later in life. Symptoms include short stature,

premature hair greying and loss, cataracts, osteoporosis, atherosclerosis and

similar facial characteristics to HGPS patients (Huang et al., 2006; Kudlow et al.,

2007). Unlike HGPS, WS patients are susceptible to a variety of cancers (Goto et

Page 42: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

28

al., 1996). Cancer and cataracts are more representative of symptoms of natural

ageing, however individuals with LMNA mutations may not present with these

symptoms and onset is earlier (Chen et al., 2003), indicative of inherent

differences in disease mechanisms between disease caused by LMNA and WRN

mutations.

A number of genotype-phenotype correlations have been established. All

mutations that result in retention of the C-terminal cysteine residue (and

therefore, retain the farnesyl modification) result in the characteristic presence

of dysmorphic nuclei in patient cells (Bergo et al., 2002; Eriksson et al., 2003;

Mallampalli et al., 2005; Navarro et al., 2004). N-terminal point mutations do not

cause nuclear blebbing, and generally result in atypical symptoms (Smallwood

and Shackleton, 2010). Interestingly, farnesylation retention is not required for

the change in nuclear morphology as one N-terminal missense mutation (E578V

– see section 3.1.3) with abnormal nuclei shows (Fukuchi et al., 2004). Mutations

affecting residues that become buried within the immunoglobulin-like fold

located in the C-terminal globular region associate with skeletal muscle

phenotypes, while exposed residues associate with lipodystrophy (Scharner et

al., 2014).

1.2 Telomeres

1.2.1 Overview

Telomeres are highly conserved repetitive sequences located at the termini of

linear chromosomes. In humans, telomeres consist of a tandem array of the

hexamer TTAGGG and form a nucleoprotein complex with a set of proteins

known as shelterin. Shelterin is able to bind to telomeres and mediate the

formation of a t-loop, a structure in which the end of a telomere is sterically

hidden from DNA damage recognition factors. This prevents the cell from

Page 43: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

29

recognising the telomere terminus as a double strand break (DSB), which could

result in telomere-telomere fusion, exit from cell cycle and/or genomic

instability.

Due to the end replication problem, telomeres shorten with each cell division in

the absence of a telomere maintenance mechanism (TMM). When telomeres

reach a critical length senescence is initiated. The accumulation of senescent

cells is thought to be a key factor in the ageing phenotype. If senescence is

bypassed telomeres may continue to shorten until the cell reaches crisis. At this

stage, telomeres are so short that association of shelterin is compromised,

exposing the telomere to DNA damage recognition factors. Efforts by the cell to

erroneously repair this “break” lead to genomic instability. Mutations or

epigenetic changes that permit the cell to adopt a TMM allow the cell to

continue proliferating and this is a key step in carcinogenesis.

1.2.2 Telomere structure

Human telomeres consist of a long tract of canonical TTAGGG repeats, normally

extending between 5 and 15kb at the chromosome terminus (Samassekou et al.,

2010). Telomere length is highly variable between different cell types (de Lange

et al., 1990; Engelhardt et al., 1997), different telomere ends in a single cell

(Graakjaer et al., 2004; Lansdorp et al., 1996), and between alleles of sister

chromatids (Baird et al., 2003). Telomere length is partially inherited (Graakjaer

et al., 2006; Slagboom et al., 1994) with a bias towards paternal inheritance

(Nordfjäll et al., 2005), and may be determined by genetic factors (Baird et al.,

2006). Loci that may contribute to heritable telomere length have been

identified on chromosome 12 and the X chromosome (Nawrot et al., 2004;

Vasa-Nicotera et al., 2005). Opposing findings in mice (García-Cao et al., 2004),

one study in twins has established that the contribution of environmental and

epigenetic factors are minor in human telomere length determination

(Graakjaer et al., 2004).

Page 44: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

30

With advances in techniques that are capable of measuring single telomere

lengths, it has become apparent that individual telomeres show tendencies

towards individual characteristics. Short arm telomeres tend to be longer than

their long arm counterparts (Samassekou et al., 2009). The 17p telomere has a

strong tendency to be the shortest or one of the shortest telomeres (Britt-

Compton et al., 2006; Martens et al., 1998), while 3p, 4p and 5p are among the

longest (Samassekou et al., 2009). Unusual length dynamics are also apparent

at the telomeres of the sex chromosomes (Mayer et al., 2006; Perner et al.,

2003).

Adjacent to the 5-15kb long TTAGGG tract is a region of the telomere (Figure

1.2:1) which includes canonical TTAGGG repeats but mainly consists of

degenerate repeats, including TCAGGG, TGAGGG and TTGGG which may extend

for ~2kb (Allshire et al., 1989; Baird et al., 1995). Degenerate telomere

sequences may also be present within the main body of the telomere, which

may have consequences for telomeric stability (Mendez-Bermudez et al., 2009).

Directly adjacent to the telomere is a region of subtelomeric DNA. While some

Figure 1.2:1 Sequence composition of the telomere and subterminal regions of human

chromosomes. The telomere consists of ~2-20kb repeat tracts of the conserved TTAGGG

repeat and the 3’ overhang at the most distal (terminal) end of the chromosome.

Immediately adjacent to the telomere is the subtelomeric region, consisting of a patchwork

of degenerate and canonical telomere repeats.

Page 45: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

31

subtelomeres show little homology to others, such as 7q (Brown et al., 1990;

Riethman et al., 1993), historical duplications, reshuffling and exchange events

have resulted in a patchwork-like composition (Baird et al., 2000, 1995; Brown et

al., 1990; Cross et al., 1990; Martin-Gallardo et al., 1995; Monfouilloux et al.,

1998), and large amounts of homology exist across different chromosome ends

(>99.5% between duplicons) (Riethman et al., 2005). Because of this, and the

repetitive nature of these sequences, accurately defining subtelomeric regions is

a difficult process (Bailey et al., 2001). A schematic representation of

subterminal chromosome structure is provided in Figure 1.2:1.

The telomere terminus is marked by a 3’ G-rich overhang which is thought to

invade double stranded telomeric DNA to form a lariat termed the t-loop. A

short D-loop also results from the invasion of the 3’ end into the normal duplex

DNA (Griffith et al., 1999). This process is thought to be essential for telomere

function and is orchestrated by the components of the multi-protein complex

termed shelterin.

1.2.2.1 Shelterin components

The telomere-specific complex shelterin recognises and binds telomere

TTAGGG repeats to form a nucleoprotein complex, protecting telomere ends

from recognition by the DNA damage response (DDR), which could otherwise

recognise telomere termini as double strand breaks (DSBs). Shelterin is

composed of six proteins: TRF1, TRF2, RAP1, TIN2, TPP1, and POT1.

Telomere repeat-binding factors TRF1 and TRF2 are detectable along entire

stretches of telomeric repeats and are responsible for recruiting all other

shelterin components to telomeres (Palm and de Lange, 2008). Each contains a

C-terminal SANT/Myb domain that binds to the 7nt sequence TAGGGTT with

extremely high specificity (Court et al., 2005; Hanaoka et al., 2005). Both also

contain a TRF homology domain (TRFH) which mediates homodimerisation.

Page 46: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

32

Despite a large degree of homology between the two proteins they are unable

to directly interact (Broccoli et al., 1997). The TRFH also serves as a docking site

for several telomere binding factors (Chen et al., 2008). RAP1 binds TRF2,

increasing its affinity for telomeric sequence, and is able to bind DNA directly

(Arat and Griffith, 2012). TRF1-interacting nuclear factor 2, TIN2 acts as a

linchpin in shelterin, stabilising the complex through its simultaneous binding of

both TRF1 and TRF2 (Ye et al., 2004). TIN2 also binds Tripeptidyl Peptidase 1

TPP1 (Frescas and de Lange, 2014), through which protection of telomeres 1

(POT1) is recruited (Hockemeyer et al., 2007). POT1 recognises G-rich single

stranded telomeric DNA and is able to bind the overhang (Baumann and Cech,

2001), and the D-loop of the t-loop conformation (de Lange, 2005).

1.2.2.2 The 3’ overhang

The single-stranded G-rich overhang is conserved in most organisms

(Henderson and Blackburn, 1989). Electron microscope images of a lariat at the

terminus of telomeric DNA suggested that the overhang, through mediation by

TRF2, is able to invade double stranded telomeric sequence to form the t-loop

(Griffith et al., 1999). Recent innovations in microscopy have confirmed these

findings, and have excluded a role in t-loop formation for all shelterin

components except TRF2 (Doksani et al., 2013).

The overhang may range in size from ~12-300nt (Dai et al., 2010; Makarov et al.,

1997; McElligott and Wellinger, 1997; Stewart et al., 2003; Zhao et al., 2008) and

may be dependent on cell type and divisional age of the cell (Stewart et al.,

2003). The overhang is partly derived through the end-replication problem

(Levy et al., 1992), so called because DNA polymerase requires a primer in order

to initiate replication, and upon detachment of this primer there remains an

unreplicated section of DNA relative to at least the size of the primer

(Olovnikov, 1973). Therefore, the nascent strand created by lagging-stand

synthesis will be missing genomic sequence at the 5’ end. Semiconservative

Page 47: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

33

replication results in one daughter copy with an overhang in place (lagging

strand synthesis), and one blunt end (leading strand synthesis), which requires

processing to obtain an overhang. This processing is carried out on nascent

duplex telomeres derived from leading strand synthesis by the nuclease Apollo.

Apollo is recruited to telomeres through TRF2 and its nuclease activity is

regulated by POT1 (Wu et al., 2012). Exonuclease I (ExoI) may generate long

overhangs at both telomere ends, which are subsequently filled in by the CTC1,

STN1 and TEN1 (CST) complex (Wang et al., 2012; Wu et al., 2012). Leading

strand daughter duplex DNA shortens by ~30nt, while the lagging strand

shortens by ~100nt (Zhao et al., 2008). The final RNA primer is positioned ~70-

100nt from the ends during replication (Chow et al., 2012), which could account

for increased lagging-strand shortening, however, nuclease digestion would

seem to be carried out on both lagging- and leading-strand-derived telomeres

as the C-rich strand sequence shows a strong tendency (~80%) to end after the

first cysteine of the CCCAAT-5’ repeat (Sfeir et al., 2005). To add further

complexity, the overhang may in fact be extended through resection, then

shortened through fill-in in a cell cycle-dependent manner (Dai et al., 2010). This

could be regulated by differential association of telomere-associated proteins at

S/G2 (Verdun and Karlseder, 2006; Verdun et al., 2005). Ultimately this process

leads to the shortening of telomeres with every cell division.

1.2.3 Telomeres, DNA damage and ageing

Telomere shortening has been reproducibly observed in cultured primary cells

(Harley et al., 1990; Lansdorp et al., 1996), as well as in vivo (Allsopp et al., 1995;

Kajstura et al., 2000; Notaro et al., 1997). As continued shortening of

chromosomes would result in erosion of essential genetic code, telomeres

ensure the protection of the genome. Thus, upon shortening to a critical length,

a signalling pathway is set in motion that halts further cell division, as well as

telomere shortening. The result is senescence, a state in which the cell may no

Page 48: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

34

longer divide, and several biological and morphological changes occur in the

cell (Campisi, 2013). This process is a natural defence against catastrophic DNA

damage events that could result from fusion-breakage cycles (see section

1.2.3.5), and could ultimately lead to cancer. Tissues accumulate senescent cells

with age (Herbig et al., 2006; Lawless et al., 2010; Wang et al., 2009), and it is

likely that this is the cause of several functional changes which lead to tissue

dysfunction and disease associated with ageing (Artandi and DePinho, 2010;

Cawthon et al., 2003; Krtolica et al., 2001).

1.2.3.1 The mitotic clock and telomere-associated senescence

Limited replicative capacity of cultured cells was first observed by Hayflick in

1961 (Hayflick and Moorhead, 1961). Serving as a mitotic clock, the number of

times a cell divides ex vivo has a predictable limit, and this is often termed the

Hayflick limit after its discoverer. Telomeres shorten progressively with continual

cell division (Harley et al., 1990), and it is telomere length that dictates the

potential replicative capacity of a cell (Allsopp and Harley, 1995; Allsopp et al.,

1992). Telomeres shorten by ~30-160bp per cell division (Allsopp et al., 1992;

Baird et al., 2003; Counter et al., 1992; Harley et al., 1990; Levy et al., 1992; Vaziri

et al., 1993), until reaching a critical length. This process initiates cell cycle exit,

entering the cell into a non-dividing state where much of its expression profile

is altered, termed replicative senescence (Bodnar et al., 1998). Upon critical

shortening, elements of shelterin may dissociate from telomere ends, disrupting

the secondary structure of telomeres and activating a DDR-like response

involving many global DDR proteins (see section 1.2.3.5 and d’Adda di Fagagna

et al. 2003). Current understanding suggests that five telomeres or more must

shorten critically and exhibit a DDR (see below) before senescence is initiated

(Kaul et al., 2011). Genomic DNA damage may also induce an alternative

senescent state that has distinct downstream effectors from the senescence

pathway activated by shortened telomeres (Cesare et al., 2013).

Page 49: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

35

1.2.3.2 DNA damage response

Much of the DDR relies heavily on blanket signals provided by modification of

factors readily available in the nucleus. The first responder to double stranded

breaks is the MRN complex, consisting of Mre11, Rad50 and Nbs1. MRN binds

and processes damaged DNA ends (Carson et al., 2003; Moreno-Herrero et al.,

2005; Uziel et al., 2003). The key activator ataxia-telangiectasia mutated (ATM), a

serine/threonine kinase, is then recruited to double strand breaks, or in the case

of single strand breaks ataxia telangiectasia and Rad3 related (ATR), and

activates several DDR factors through phosphorylation. This includes

phosphorylation of serine residue 139 of histone variant H2AX (Burma et al.,

2001). This phosphorylated form is termed γ-H2AX, and spreads rapidly on

H2AX over hundreds of kilobases from the initial site of damage detection

(Rogakou et al., 1999). γ-H2AX is a marker of DNA damage and is key in the

recruitment of DNA repair effectors (Paull et al., 2000; Sharma et al., 2012).

As well as recruiting DNA repair proteins, ATM will also initiate a rapid halt to

cell cycle progression by phosphorylation of CHK1 and CHK2 (at residues serine

345, and threonine 68 respectively), which, now activated, inhibit with their

downstream effectors Cdc25A and Cdc25C phosphatases to bring about arrest

at G1/S or G2/M (Li and Zou, 2005). Upon completion of repair, cell cycle may

continue and the loss of phosphorylation of H2AX may play a role in resuming

the cell cycle (Downey and Durocher, 2006). p53, a powerful tumour suppressor,

is activated by ATM. It induces transcription of p21 which in turn may

upregulate p16INK4A. Should damage be irreparable, senescence is initiated

through this increased transcription of p21 and p16INK4A (Campisi and d’Adda

di Fagagna, 2007). Double-strand breaks may then be repaired by homologous

recombination (HR) or non-homologous end joining (NHEJ).

Page 50: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

36

1.2.3.3 Repair via homologous recombination

Double-strand breaks form as a result of exposure to reactive oxygen species

and ionizing radiation, and may be formed during replication due to fork

collapse. Whether the cell carries out HR or NHEJ is largely dependent on the

stage of the cell cycle, as well as the nature of the lesion (Symington and

Gautier, 2011). HR carries out fidelitous repair by using a sister-chromatid as a

template for recombination that is only available during S-phase (Saleh-Gohari

and Helleday, 2004), when replication fork collapse may occur. Current

knowledge of the HR repair system derives mostly from what has been learnt

Figure 1.2:2 DNA damage repair by homologous recombination a DNA damage is

induced and detected by DDR factors. 5’ resection is carried out and strand invasion is

mediated by RPA. Following DNA synthesis, HR repair may be resolved by one of the

three mechanisms shown in figures b-d b Integration of the remaining strand and

Holliday junction resolution resulting in crossover or non-crossover, and conservation of

genetic data c The extended strand is displaced and re-anneals to the remaining strand.

Processing and extension result in conservation of genetic data d A replication fork may

form and continue processively to the chromosome terminus. Any mutations will be

copied. Adapted from (Deriano and Roth, 2013)

Page 51: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

37

from the yeast homologous system. Breaks are usually not clean in that blunt

ends do not generally result and so processing of the ends of a break is

required before repair can be carried out. HR repair is dependent upon cyclin

dependent kinase 1 (CDK1), which is required for 5’-3’ resection of DNA ends at

the site of damage (Ira et al., 2004). This is mediated by the MRN complex, or in

yeast the MRX complex, and carried out by the exonucleases EXO1 and CtIP (Eid

et al., 2010; Nakada et al., 2004). The now exposed 3’ ssDNA is coated rapidly

with replication protein A (RPA), inhibiting secondary structure formation in the

ssDNA (Krejci et al., 2012). RPA is then replaced by Rad51, facilitated by

mediators such as breast cancer 2, early onset in humans (BRCA2) (Liu et al.,

2010). Rad51-ssDNA filaments then invade dsDNA to produce a D-loop, the

ssDNA of which is also incorporated into the Rad51 filament. Upon completion

of this stage, Rad51 may then dissociate from and expose the 3’ OH group

required to initiate DNA synthesis (Krejci et al., 2012). DNA synthesis and break

resolution may be carried out in one of three ways, as shown in Figure 1.2:2.

1.2.3.4 Repair via non-homologous end-joining

Much of what is understood about NHEJ derives from studying the

programmed rearrangement of antibody gene loci. Early in their development, T

and B lymphocytes carry out V(D)J recombination in the Variable Diverse and

Joining regions of the heavy and light antibody chain genes. Rearrangement of

these three regions provides a basis for antibody diversity. V(D)J recombination

is initiated by recombination activating gene proteins (RAGs), which induce

DSBs, and completed by classical-NHEJ. A second process, immunoglobulin

class switch recombination (CSR), takes place in mature B lymphocytes,

directing class changes of the antibody heavy chain. CSR also employs DSBs,

but resolves them with both classical- and atypical-NHEJ in a 53BP1-dependent

manner (Bothmer et al., 2010).

Page 52: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

38

In G0, G1 and early S phases of the cell cycle, HR is repressed and NHEJ is the

major effector of DSB repair. NHEJ does not require a homologous sister-

chromatid and instead will anneal broken ends to one another despite an

absence of sequence homology, or ligate sequences based on small sections of

homologous sequence (microhomologies), regardless of whether the two

sequences correspond to the same locus, which can result in deletions (Roth

and Wilson, 1986). NHEJ is also able to compensate for loss of HR in cells

deficient in key HR proteins (Takata et al., 1998). A major regulator of NHEJ

repair is 53BP1, which is able to direct repair to be carried out through one of

two routes (Bothmer et al., 2011, 2010): classical NHEJ (c-NHEJ) or alternative

NHEJ (a-NHEJ) (Figure 1.2:3).

In c-NHEJ, Ku, a heterodimer of Ku70 and Ku86, is first to bind DNA and

mediates binding of other NHEJ factors (Fattah et al., 2010). Ku binds and

maintains the proximity of the two ends of the break to one another, while also

recruiting DNA-dependent protein kinase catalytic subunit (DNA-PKcs), as well

as DNA ligase IV and its cofactors XRCC4 and XRCC4-like factor (XLF). DNA-PK

Figure 1.2:3 DNA damage repair by non-homologous end joining. Classical NHEJ (c-

NHEJ) results in minimal loss of genomic material, however alternative NHEJ (a-NHEJ)

relies upon alignment of microhomologous regions, potentially resulting in large

deletions, or translocations. Adapted from (Deriano and Roth, 2013)

Page 53: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

39

aids in complex recruitment and is also able to form a complex with the

exonuclease ARTEMIS, which may carry out DNA processing (Ma et al., 2002),

before the DNA ligase IV completes ligation on regions of microhomology

(Grawunder et al., 1997).

a-NHEJ is the least faithful mode of DSB repair, and frequently results in

translocations, deletions and insertions (McVey and Lee, 2008). The discovery of

a-NHEJ is relatively recent and as such its mechanisms are less well-defined

than other forms of repair. Although first identified as a backup system for c-

NHEJ (Kabotyanski et al., 1998; Liang and Jasin, 1996), a-NHEJ is now known to

function in the presence of a functional c-NHEJ pathway (Nussenzweig and

Nussenzweig, 2007). a-NHEJ relies upon the MRN complex, which may play a

direct role in end processing alongside the exonuclease CtIP (Deriano and Roth,

2013), but is carried out independently of Ku, which may be replaced by poly

(ADP-ribose) polymerase 1 (PARP1) (Haince et al., 2008). Ligation may be carried

out by DNA ligases I and III (Liang et al., 2008).

1.2.3.5 Telomere dysfunction induces senescence through a DNA

damage response pathway

Critical shortening or damage at telomeres may result in T-loop deconstruction,

loss of the overhang and shelterin dissociation, exposing the telomere (a

process termed uncapping) (d’Adda di Fagagna et al., 2003; de Lange, 2005;

Karlseder et al., 2002). This results in a DDR that involves the recruitment of

factors found at double-strand break sites. These aggregates have been termed

Telomere dysfunction induced foci (TIFs) (Takai et al., 2003). TIFs are

distinguishable from normal DSB sites by the presence of TRF1 (Takai et al.,

2003), although the particulars of downstream effectors are not well enough

understood to fully distinguish a telomere-specific response (Palm and de

Lange, 2008). Many proteins involved in the ordinary ATM directed response

co-localise in TIFs that are associated with senescent cells. These foci, which

Page 54: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

40

include γ-H2AX, 53BP1, the MRN complex and activated ATM, are thought to

induce telomere-associated senescence, causing cells to permanently and

irreversibly exit the cell cycle (d’Adda di Fagagna et al., 2003). TIFs may also

persist in senescent cells that accumulate with age in vivo (Scaffidi and Misteli,

2006; Sedelnikova et al., 2004). It has been shown that telomeric binding

proteins are not merely passively shielding telomere ends from detection by the

DDR apparatus but that TRF2 actively inhibits ATM activity, while POT1 may

inhibit ATR activity (Denchi and de Lange, 2007; Karlseder et al., 2004).

p16INK4A is not a contributor to establishing telomere-induced senescence

(Herbig et al., 2004), and CHK2 may not be activated despite ATM activation

(Cesare et al., 2013).

1.2.3.6 Werner syndrome

Werner Syndrome (WS) is a progeroid syndrome associated with deficiencies in

telomere biology. Classical-WS is an autosomal recessive disorder caused by

homozygous or compound heterozygous mutations in the RECQL2 gene.

RECQL2 encodes WRN, and WS mutations tend to result in truncated forms of

WRN which are incapable of relocating to the nucleus (Moser et al., 2000). WRN

is a RecQ helicase involved in several cellular processes, including DNA repair,

replication and recombination. As well as exhibiting symptoms of premature

ageing (see section 1.1.6), patients are susceptible to various forms of cancer

(Goto et al., 1996). WS patients also present with genomic instability, DNA

repair deficiencies, replication defects and attenuated apoptosis (Croteau et al.,

2014). At telomeres, the helicase activity of WRN acts in partnership with

exonucleases to unwind and dissociate the telomeric D-loop, mediated by TRF1

and TRF2 (Machwe et al., 2004; Opresko et al., 2004). WRN is required for

normal telomere lagging-strand synthesis (Crabbe et al., 2004), telomeric DNA

damage processing (Eller et al., 2006), and DNA damage regulation upon

telomeric dysfunction (Crabbe et al., 2007). The proliferative capacity of WS cells

Page 55: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

41

is reduced compared to normal cells, but may be restored through telomerase

expression (telomerase is able to extend telomeres - see section 1.2.4.1).

However, investigation of the rate of telomere shortening in WS has given

mixed results, with some cells exhibiting a normal or slightly elevated

shortening rate while others exhibited greatly accelerated shortening rates at

certain points in proliferative lifespan. Though this was likely as a result of clonal

expansions within the population, mean telomere length was extremely short in

WS cells at early passages (~6kb) (Baird et al., 2004), perhaps indicating a

developmental role rather than a persistent issue. Telomere length is also

shorter in vivo in WS than age matched controls in skin fibroblasts (Ishikawa et

al., 2011). Together, these results reinforce the importance of telomere length

and integrity in telomere-initiated senescence and ageing.

1.2.4 Telomere maintenance mechanisms and cancer

The genomic instability caused by telomere dysfunction may provide an entry

point to establishing immortalisation in cancer. Accumulation of mutations

leads to further genomic instability and any mutations conferring a growth

advantage will result in expansion of neoplastic cell types. A myriad of cellular

mechanisms exist to limit proliferation, including senescence induction upon

telomere dysfunction. As such, a key feature of cancer cells is their ability to

circumvent normal cell signalling responses that halt proliferation. In order to

propagate without limit, cancer cells must overcome the imposition of

senescence induced by telomere shortening. Through interruption of the p53

and pRb signalling pathways (Hara et al., 1991), cells may bypass senescence

and enter crisis – a state in which telomeres continue to erode (Counter et al.,

1992). This may result in cycles of telomere fusion, in which uncapped telomeres

are joined, followed by uneven breakage at cell division and causing extreme

chromosomal instability (Gisselsson et al., 2001; Lo et al., 2002; Maser et al.,

Page 56: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

42

2007). Within this population, cells may contain mutations which lead to the

activation of a telomere maintenance mechanism (TMM).

To this effect, cancer cells may take advantage of cellular mechanisms already

present in the genetic code, though transcriptionally restricted in most normal

cells. The most common TMM that is activated in cancer cells is telomerase, a

multi-subunit enzyme capable of extending telomeres through a reverse

transcription mechanism. Telomerase may be activated by epigenetic alterations

which activate transcription of the telomerase subunit telomerase reverse

transcriptase (TERT) (Sui et al., 2013). A second, less well-understood

mechanism, termed alternative lengthening of telomeres (ALT) is activated in a

minority of cancers, and seems to be dependent upon recombination-based

elongation of telomeres. The propensity of a particular cell type to initiate

telomerase expression or the ALT system in cancer may depend on inherent

differences in expression patterns (Lafferty-Whyte et al., 2009). Cell types of

mesenchymal origin are more predisposed to activating ALT, and common ALT

cancers include glioblastoma multiforme (Hakin-Smith et al., 2003), liposarcoma

(Costa et al., 2006), sarcoma and astrocytoma (Henson et al., 2005). Activation of

the ALT and telomerase pathways are not mutually exclusive and both may be

employed in human cancer cells (Cerone et al., 2001).

1.2.4.1 Telomerase

Not all cells are prone to telomere shortening. As telomeres are key to

maintaining genome integrity, certain human cell types that require telomere

length maintenance while continuing to divide, specifically germ cells and stem

cells (Kim et al., 1994), are able to lengthen telomeres through expression of

telomerase. However, in the remainder of normal cells, telomerase remains

inactive and cells whose telomeres reach a critical length will intiate, unless

bypassed, the senescence response.

Page 57: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

43

Telomerase is a multi-subunit holoenzyme, the core of which is composed of

the catalytic reverse transcriptase subunit hTERT in humans (Harrington et al.,

1997) and a telomerase RNA component hTR, which provides a template for

DNA synthesis (Feng et al., 1995). Telomerase also binds several auxiliary

factors, including the key component Dyskerin, which is required for stable

formation of the telomerase complex (Mitchell and Collins, 2000). hTERT is

highly conserved and binds telomeres through the shelterin component TPP1,

via its interaction with POT1 (Xin et al., 2007). hTR is a 451nt telomeric RNA

sequence which binds hTERT and serves as a template for its catalytic activity.

hTR may also aid the processivity of telomerase action (Nandakumar and Cech,

2013). In human cells expressing telomerase, telomere length remains

heterogeneous, but short telomeres and senescence are avoided, leading to the

theory that telomerase specifically targets short telomeres for lengthening

(Blackburn, 2001; Zhu et al., 1999). This has been confirmed through ectopic

expression of telomerase in various cell types, in which telomere length is

regulated to cell type-specific limits (Britt-Compton et al., 2009a). Telomerase

may extend the proliferative lifespan of a cell not only through telomere

elongation but also through blocking the DDR and apoptosis, and stimulating

proliferation (reviewed in Majerská et al. 2011).

Background levels of expression of hTR are present in most tissues, and

telomerase expression and subsequent telomere lengthening may be

reconstituted in human cells by induced expression of hTERT alone (Vaziri and

Benchimol, 1998), with no effect on genomic stability (Ouellette et al., 2000).

Due to the biological implications of maintaining telomere length, TERT

expression is tightly regulated by several factors related to oncogenesis (Kyo et

al., 2008). In contrast to humans, mice express telomerase in most somatic cell

types (Kipling, 1997), and is required for cell growth and survival (Lee et al.,

1998). Mouse telomere length is highly variable and telomeres can reach up to

Page 58: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

44

150kb (Kipling and Cooke, 1990). The representativeness of the mouse as a

model organism for telomere biology must therefore be carefully considered.

1.2.4.2 The ALT mechanism

The ALT pathway is activated in ~10-15% of cancers that do not express

telomerase (Heaphy et al., 2011). No single event or pathway has been

identified as the cause of the ALT pathway, making identification of ALT markers

difficult. Robust methods for identifying ALT are of interest due to the poor

prognosis associated with ALT in some cancer types (Cesare and Reddel, 2010),

although the prognosis of diffuse malignant peritoneal mesothelioma patients

exhibiting ALT is variable (Villa et al., 2008), possibly reflecting the

heterogeneous nature of ALT pathways. As such, ALT is identified by a collection

of characteristics as follows.

ALT telomeres are extremely heterogeneous in length, and can reach lengths

greater than 50kb (Bryan et al., 1997, 1995). Losses and gains may occur rapidly

(Murnane et al., 1994), most likely due to the high level of recombination at ALT

telomeres (see below). It is thought that these extreme lengths are not tolerated

by the cell, which initiates a trimming mechanism resulting in an abundance of

extrachromosomal linear and circular telomeric DNA in ALT cells, although the

mechanisms of this are not fully understood. t-circles are double-stranded

circular telomeric sequences, so called as it is thought they are formed by

resolution of t-loops (Cesare and Griffith, 2004; Wang et al., 2004). Although

abundant in ALT cells, t-circles are also found in telomerase positive cells

(Tomaska et al., 2009). More specific to ALT positive cells are c-circles composed

of a single strand C-rich circle, with some duplex portions containing the

complementary G-rich sequence (Henson et al., 2009). It has been proposed

that C-circles derive from G-strand specific degredation of t-circles. Less

common, but still present, are G-circles, the inverse manifestation of C-circles,

Page 59: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

45

and extremely large fragments of telomeric DNA containing secondary

structures, termed t-complex DNA (Nabetani and Ishikawa, 2009).

Promyelocytic leukaemia (PML) bodies are nuclear structures that contribute to

a number of nuclear functions, including tumour suppression (Bernardi and

Pandolfi, 2007). The presence of telomeric DNA in PML bodies is associated with

ALT and as such these structures are termed ALT-associated PML bodies (APBs).

APBs are identifiable by their association with telomeric DNA, as well as

telomere-associated proteins, including members of the shelterin complex and

DNA repair and recombination factors (Yeager et al., 1999). It would seem that a

subset of APBs are able to co-localise with telomeres, whose mobility is

increased compared to PML bodies, providing discreet compartments for ALT-

related recombination processes to take place (Jegou et al., 2009), although ALT

induced telomere lengthening may occur in their absence (Jeyapalan et al.,

2008). ALT cells are also more commonly prone to genomic instability than

telomerase positive cells (Lovejoy et al., 2012), and exhibit extraordinary

instability at the MS32 minisatellite locus (Jeyapalan et al., 2005).

High levels of sister-chromatid exchange (Londoño-Vallejo et al., 2004) and

rapid acquirement of telomere length polymorphisms (Murnane et al., 1994)

indicate that the ALT mechanism depends on recombination processes to

implement telomere elongation. Dunham et al. demonstrated via the insertion

of a neomycin resistance marker in telomeres and subtelomeric regions that

recombination occurs at telomeres while subtelomeres remain unaffected, and

that telomerase positive cells do not employ recombination at telomeres

(Dunham et al., 2000). Several models have been proposed as to how ALT cells

employ recombination-like processes in order to extend telomeres, including

sister-chromatid exchange and homologous recombination, as well as

mechanisms that use extrachromosomal linear or circular telomeric DNA as a

Page 60: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

46

template (reviewed in Cesare & Reddel 2010). Due to the heterogeneity of the

ALT phenotype, it is possible that several of these mechanisms may apply.

1.3 Links between telomere and lamin biology

Telomere length has been frequently linked to ageing and several reports have

shown a link between lamin A malfunction and disruption of normal telomere

biology (Allsopp et al., 1992; Decker et al., 2009; Gonzalez-Suarez et al., 2009;

Huang et al., 2008). Telomere dysfunction induces the DDR, which may result in

entry into senescence. Both the DDR and levels of senescence are elevated in

HGPS cells, and damage at telomeres is specifically enhanced (Benson et al.,

2010). Two studies so far have shown that classical HGPS cells have shorter

telomeres when compared to controls (Allsopp et al., 1992; Decker et al., 2009),

suggesting that increased cellular senescence in HGPS is attributable to

telomere-induced senescence. Rapid accumulation of TIFs in progerin-

expressing cells suggests that telomere damage signalling is occurring in cells

prior to critical shortening (Benson et al., 2010), thus senescence may be

induced by damaged telomeres that would otherwise continue to shorten

before initiating senescence.

1.3.1 DNA damage and lamin deficiencies

Ectopic expression of progerin in normal fibroblasts produces increased

damage signalling at telomeres – a fourfold increase in γ-H2AX association with

telomeres (Benson et al., 2010). These data mirror the accumulation of γ-H2AX

in senescing mesenchymal cells, which is coupled with aggregation of telomeres

with intranuclear lamin structures (Raz et al., 2008). The DDR is greatly

perturbed in HGPS cells. DNA damage accumulates and is slow to be repaired.

Patient cells display a deficiency in 53BP1 recruitment to sites of damage, and

lesions induced by ionising radiation (Liu et al., 2005). Similarly, 53BP1

Page 61: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

47

recruitment is slow in both unirradiated (Gonzalez-Suarez et al., 2009) and

irradiated Lmna-/- MEFs (Redwood et al., 2011), in which a reduction in 53BP1

levels is observed (Gonzalez-Suarez et al., 2011). Lmna-/- MEFs c- and a-NHEJ

deficiencies may be rescued through 53BP1 expression (Gonzalez-Suarez et al.,

2011; Redwood et al., 2011).

The importance of 53BP1 in NHEJ has been highlighted by its requirement for

V(D)J recombination. During the DDR, ATM phosphorylates, and thereby

activates 53BP1 which then protects and promotes DSB end-joining (Bothmer et

al., 2011; Rybanska-Spaeder et al., 2013) in a γ-H2AX-dependent manner

(Difilippantonio et al., 2008). 53BP1 also seems to play a key role in controlling

which mode of repair is carried out on a DNA break. 53BP1 preferentially

activates c-NHEJ in CSR (Bothmer et al., 2011) and co-operates with breast

cancer 1, early onset (BRCA1/Brca1) to activate NHEJ preferentially over HR in

mice (Bunting et al., 2010). 53BP1 plays a specific role in telomere damage

repair, being essential for NHEJ of dysfunctional telomeres. This process

increases telomere motility, is dependent on ATM, and requires a 53BP1-

H4K20me1 interaction for optimal repair (Dimitrova et al., 2008). An interesting

study on the cells of ataxia telangiectasia (AT) patients, who have defective ATM

(from whom the protein derives its name), revealed that telomeres are more

frequently associated with the lamina in AT cells than control cells (Smilenov et

al., 1999), indicating a connection between ATM, telomeres and the lamina

which could be explored.

It seems that rapid shortening events are not the cause of telomere attrition in

HGPS from data obtained in a telomere length study in lamin A stably

transfected cells (Huang et al., 2008), which suggests a steady but increased rate

of loss as measured by Q-PCR, however more data is required to confirm these

conclusions. Chromosome orientation fluorescence in situ hybridisation) has

shown no increase in sister-chromatid exchange (Gonzalez-Suarez et al., 2009),

Page 62: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

48

demonstrating no increase in recombination activity at telomeres in HGPS cells.

This suggests that the shortening process is replication-dependent or resultant

of a compensatory increase in division in cells in a population where a high

proportion of senescent cells put a strain on tissue maintenance.

1.3.2 Telomeres, heterochromatin, lamins and nuclear

localisation

Telomeres may associate with the nuclear lamina in humans (de Lange, 1992;

Ludérus et al., 1996), and a direct interaction between telomeres and lamina has

been confirmed via chromatin immunoprecipitation (ChIP) assay in mice

(Gonzalez-Suarez et al., 2009). This extensive study by Gonzalez-Suarez et al.

into the effects of lamin A deficiency on telomeres in Lmna-/- mice provides

convincing evidence for a critical interaction between the two with data

showing altered telomere localisation toward the nuclear periphery and an

increase in TIF formation. This study also identified a NHEJ repair deficiency in

Lmna-/- cells, which the authors attributed to reduced 53BP1 after establishing

no changes in the cellular levels of several DNA repair factors (Gonzalez-Suarez

et al., 2009).

Telomeres have a specific heterochromatin profile which is crucial in

maintaining telomere function (Michishita et al., 2008) and nuclear localisation

(Uhlírová et al., 2010). Both lamin A reduction and histone methylation affect

telomere localisation in MEFs (Uhlírová et al., 2010). Telomeric heterochromatin

contains high levels of H3K9 methylation and H4K20Me3, both of which are

greatly effected in HGPS cells (Columbaro et al., 2005; Dechat et al., 2008;

Scaffidi and Misteli, 2005; Shumaker et al., 2006). The lamin A mutant E145K cell

line shows an increase in telomere association with the lamina (Taimen et al.,

2009), and HGPS telomere mobility in the nucleus is reduced (De Vos et al.,

Page 63: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

49

2010). This suggests that telomere localisation may be altered in HGPS cells due

to epigenetic remodelling at telomeres.

Another overlap of telomere and lamin processes is the interaction of both

within the nucleoplasm with LAP2α during mitosis and nuclear assembly

(Dechat et al., 2004, 2000). LAP2α specifically binds intranuclear lamin A (Dechat

et al., 2000) and is thought to bind telomeres via the non-specific double

stranded DNA binding protein BAF (Dechat et al., 2004). This mediated and

temporal interaction could provide critical information as to the role of

intranuclear lamin A in genome organisation during mitosis, which in turn could

be investigated in laminopathy disorders. Loss of lamins also result in an

decreased association of telomeres with PML bodies in MEFs (Uhlírová et al.,

2010).

1.3.3 Telomere length is reduced by lamin A deficiencies

Two studies have recorded telomere length directly in HGPS cells. The first

identified a reduction in average telomere length in HGPS patient fibroblasts

versus age matched and parental controls (Allsopp et al., 1992). The second

observed shorter telomeres in patient cell lines versus controls, and an

increased variability of length versus controls (Decker et al., 2009). These

studies, have the inherent drawback of measuring telomere length at a single

time point. Telomere length varies greatly between individuals, and in various

tissues of the same individual. Allowing for the different periods of time that

cells were cultured ex vivo in the above experiments, a single measurement of

telomere length may not be representative, as telomeres shorten with every cell

division. Additionally, due to inter-individual differences, a measurement at one

time point that yields a short average telomere length may simply be

attributable to natural inter-individual variation. As such, control samples are

also difficult to select.

Page 64: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

50

An additional study overcame these barriers by ectopic expression of four

mutant forms of lamin A known to induce the HGPS cellular phenotype in

fibroblasts containing an excisable hTERT construct (Huang et al., 2008). Ectopic

expression of telomerase is known to inhibit telomere shortening as well as

lengthen telomeres in normally telomerase-negative, and can reverse the

proliferative deficiencies in HGPS cells (Benson et al., 2010; Kudlow et al., 2008;

Ouellette et al., 2000). Thus, hTERT expression allowed normalisation of

telomere length. The construct was then excised and lamin A mutants were then

expressed. Measurement of telomere length across several time points revealed

an accelerated telomere shortening rate in lamin A transfected cells versus

controls. Interestingly, WT lamin A over-expression also seemed to induce an

acceleration in telomere shortening.

Expression of telomerase can reverse proliferative deficiencies in HGPS cells and

may inhibit telomere shortening, as can be seen in various normal cell lines

(Benson et al., 2010; Kudlow et al., 2008; Ouellette et al., 2000). These studies

have shown that some of the effects of telomerase induction may be reversing

the p53, p21, p16 and pRB negative effects on cell cycle progression whose

expression is altered upon telomerase transduction, potentially via interference

with the altered mechanics of the telomere-lamina interaction. This coordinates

with findings that p53 suppression allows some recovery of replicative potential

(Kudlow et al., 2008) as does ATM/ATR suppression (Liu et al., 2006). Zmpste24,

p53 double knockout mice have also been bred and display increased lifespan

and weight gain (Varela et al., 2005). In terms of experimental design, it is

notable that a significant proportion of separate clonal expansions of HGPS cells

fail to immortalise upon transformation by telomerase (Wyllie et al., 2000).

Page 65: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

51

1.4 Objectives

The first issue to be address by this project is the unknown contribution of

HGPS molecular mechanisms to telomere shortening. Although short telomeres

have been observed in cell lines derived from classical HGPS patients, issues

regarding cell line derivation and the varied replicative history of cell lines

tested question the conclusiveness of these results. Additionally, these results

are not informative of any ongoing process that may affect telomere length. As

such, it was decided that telomere length should be recorded in HGPS cells over

the course of their proliferative lifespan. By recording the mean number of cell

divisions in an HGPS population it is possible to derive a mean telomere

shortening rate per cell division, which may be directly compared to control

lines. Thus the hypothesis that short telomeres previously observed in HGPS

cells are the result of an elevated telomere shortening rate, rather than sudden

loss of telomeric sequence, may be tested.

Considerable research has been carried out on classical HGPS cells, however,

due to the rarity of the condition, and the even scarcer prevalence of atypical

HGPS, considerably less is known about atypical HGPS mutants. It was therefore

decided that telomere shortening analysis would also be carried out in a

number of atypical HGPS cell lines, as well as an RD cell line for comparison.

Farnesylated forms of mutated lamin A are associated with nuclear membrane

deformity, however one atypical patient line with a C-terminal head domain

mutation that leaves lamin A processing unaffected (E578V) also exhibits some

nuclear membrane blebbing (Csoka 2004; Toth et al. 2005). Although the rescue

of nuclear membrane morphology may correspond with protection against the

acquisition of new DNA damage lesions (Constantinescu et al., 2010), the failure

to repair DNA damage upon treatment with FTIs suggests some separation

between these two consequences of farnesylated lamin A accumulation (Scaffidi

and Misteli, 2005)(Liu et al., 2006). Thus the E578V line was selected to test

Page 66: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

52

whether telomere biology is altered in the presence of aberrant nuclear

morphology, but not farnesylated lamin A.

Upon establishing telomere shortening rate, I aimed to extricate any underlying

factors that may contribute to any potential changes in telomere biology in

patient cells. Potential mechanisms were to be identified based on telomere

dynamics recorded by the shortening analysis. This was made possible by the

use of single telomere length analysis (STELA) for establishing length, which

allows a high resolution of the dynamics of single telomeres.

Telomere integrity and sufficient activation of senescence is crucial for avoiding

the development of cancer. HGPS patients do not typically develop cancer, and

this suggests that genomic integrity is not compromised in patients, despite

high levels of DNA damage. This implies that damage is either repaired more

slowly or results in premature passage into senescence. However, previously

published data suggest that A-type lamins impact telomere biology, including

telomere localisation within the nucleus. The mobility and localisation of

telomeres has shown to be altered in ALT cells. As such, it was hypothesised

that lamin biology may be altered in ALT cells, and experiments were designed

to observe differences between lamin:telomere interactions in telomerase

positive and ALT cells. This was first explored by comparison of cellular levels

and nuclear localisation of lamins and lamin-associated factors in ALT and

telomerase positive cells. As chromatin is altered in both laminopathy and ALT

cells, it was hypothesised that the altered levels and localisation of lamins

discovered via these methods would result in a disruption of the interaction

between chromatin and lamins, and this was explored using chromatin

immunoprecipitation (ChIP).

Page 67: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

53

Chapter 2 Materials and Methods

2.1 Cell culture

2.1.1 Subculturing cell lines

All cell lines were grown in appropriately supplemented media (see Table 2.1-1)

at 37°C in 5% CO2. Adherent cells were subcultured as follows: upon reaching

confluency of 80-100% (cell line dependent), media was aspirated off, cells were

washed in 1X phosphate buffered saline (PBS) before the addition of 0.25%

Trypsin unless otherwise stated in Table 2.1-1. Cells were returned to 37°C in

order to promote cell detachment, trypsin was inactivated with media, and cells

re-plated in fresh flasks in the following total volumes of media: 8ml in 25cm2

flask, 12ml in 75cm2 flask, 20ml in 175cm2 flask. Suspension cells were mixed

thoroughly via resuspension with a pipette in media before being split into

fresh flasks at dilutions between 1:2 and 1:32, in the same volume of fresh

media.

Tissue culture plastics were purchased from Greiner Bio-One, and media and

trypsin products from Invitrogen Gibco.

2.1.2 Population growth measurement

Cell counts were performed using a haemocytometer.

Cells were subcultured 1:2 (1:3 or 1:4 when necessary) at between 80 and 100%

confluencey and mean population doubling (PD) calculated using the equation:

𝑃𝐷 =[ ln(𝑐𝑜𝑢𝑛𝑡 𝑎𝑡 ℎ𝑎𝑟𝑣𝑒𝑠𝑡) − ln(𝑖𝑛𝑖𝑡𝑖𝑎𝑙 𝑐𝑒𝑙𝑙 𝑛𝑢𝑚𝑏𝑒𝑟) ]

ln 2

Counts were adjusted for viability, as assessed by the Trypan Blue assay.

Page 68: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

54

2.1.3 Trypan Blue staining

Trypan Blue is a stain that selectively stains dead cells or tissue, while uptake is

excluded from living cells, allowing an estimation of viability levels. 150l

Hyclone Trypan Blue Solution (Thermo Scientific) was added to 150l

resuspened cells in media. The solution was mixed well, left for 3 minutes then

loaded onto a haemocytometer. 300-400 cells were counted for each passage

and the proportion of dead (blue) cells recorded and excluded from cell counts

when calculating PD.

2.1.4 -galactosidase Senescence assay

β-galactosidase is present at higher concentrations in senescent cells and may

be used as a maker to identify senescence by the addition of X-gal in sub-

optimal conditions for enzymatic activity (see section 3.2.1). The assay was

carried out on each patient and control primary cell line at as many time-points

as possible. Cells were seeded onto 13mm HCl acid-treated glass coverslips in

24-well-plates between 24 and 72 hours before assay (2x104 cells per

coverslip). Cells were washed once in PBS, fixed for 3 minutes in 1X fixation

solution (2% formladehyde, 0.2% gluteraldehyde in 1X PBS), washed twice with

1X PBS then incubated in a staining solution for 16 hours at 37°C, protected

from light and without CO2 supplementation. Staining solution (90mM citric

acid (C6H8O7)/sodium phosphate (NaH2PO4) buffer pH5.98, 150mM NaCl, 2mM

MgCl2 5mM potassium ferricyanide K3[Fe(CN)6], 5mM potassium ferrocyanide

K4[Fe(CN)6], 400μg X-gal) was prepared as a fresh solution each time. Cells were

then washed in 1X PBS 3 times and fixed in 100% methanol for 30 seconds

before air-drying and mounting with Invitrogen ProLong Gold. -galactosidase

activity was recorded via manual counting on an Olympus BH-2 BHS

microscope. A minimum of 400 cells were counted and re-counted on each

coverslip, and the average percentage of β-galactosidase positive cells was

calculated.

Page 69: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

2.1.5 Cell lines

Name Immortalisaion

status Growth media

Telomere

maintenance Origin

G608G

(AG11498) Primary line

DMEM + 20% FBS +

1X pen/strep N/A

Skin fibroblast, 14 year old male. Heterozygous c. 1864C>G in LMNA

Purchased from Coriell Institute

E578V

(AG04110) Primary line

DMEM + 20% FBS +

1X pen/strep N/A

Skin fibroblast, 13 year old female. Heterozygous c. 1945A>T in LMNA

Purchased from Coriell Institute

RD Primary line DMEM + 20% FBS +

1X pen/strep N/A

Skin fibroblast, 1 day old, restricitive dermopathy German (neonatally fatal).

Homozygous null c. 1085_1086insT in ZMPSTE24

Gift from prof. Manfred Wehnert, Ernst Moritz Arndt University

T623S Primary line DMEM + 20% FBS +

1X pen/strep N/A

Skin fibroblast, 10 year old Bristish Caucasian female. Heterozygous c. 1868C>G

in LMNA

Received from Great Ormand Street Hospital

S143F Primary line DMEM + 20% FBS +

1X pen/strep N/A

Skin fibroblast, 6 year old German. Heterozygous c. 428C>T in LMNA

Gift from Prof. Manfred Wehnert, Ernst Moritz Arndt University

NHDF Primary line DMEM + 10% FBS +

1X pen/strep N/A

Normal Human Diploid Fibroblast. Skin fibroblast, healthy adult male.

Shackleton lab long-term stocks

155-BR Primary line DMEM + 15% FBS +

1% NEAA N/A Skin fibroblast, healthy 55 year old male

161-BR Primary line DMEM + 15% FBS +

1% NEAA N/A Skin fibroblast, healthy 49 year old male

HT1080 Tumour derived DMEM + 10% FBS Tel+ Fibrosarcoma, 35 year old male. p53 mutated, exhibits pseudodiploidy.

U20S Tumour derived DMEM + 10% FBS ALT Osteosarcoma, female, DSB repair defcient

WI-38

VA13/2RA

(WI38ALT)*

SV40 DMEM + 10% FBS ALT Fetal lung fibroblast, female. Werner’s –ve, DSB repair deficient

NT2D1 Tumour derived High glucose DMEM

+10% FBS Tel+ Pluripotent embryonic male lung, derived from metastasis of testis carcinoma

JFCF6T.5K post

crisis SV40

DMEM + 20% FBS

Trypsin LE ALT Jejunal fibroblast, cystic fibrosis

54

Page 70: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

JFCF.6T-1J/11E SV40 DMEM + 10% FBS ALT Jejunal fibroblast, cystic fibrosis

JFCF.6T-1J/11C SV40 DMEM + 10% FBS Tel+ Jejunal fibroblast, cystic fibrosis

IIIcf/a2 post

crisis Tumour derived RPMI + 20% FBS ALT Breast fibroblast, female with Li-Fraumeni. p53 null

HeLa Tumour derived DMEM + 10% FBS Tel+ Adenocarcinoma, 31 year old black female. HPV positive

Saos-2 Tumour derived DMEM + 15% FBS ALT Osteosarcoma, female.

LoVo Tumour derived DMEM + 10% FBS

Trypsin LE Tel+ Colon, male. MSH2 deficient

SW480 Tumour derived DMEM + 10% FBS

Trypsin LE Tel+ Adencarcinoma male. MSH2 deficient

SUSM-1 Chemical DMEM + 10% FBS ALT Embryonic liver fibroblast, DSB repair deficient

T24 Tumour derived RPMI + 10% FBS Tel+ Bladder carcinoma

WV SV40 DMEM + 10% FBS ALT Fibroblast, 45 year old male, Werner syndrome

Table 2.1-1 Summary of all information available for all cell lines used in this study. Abreviations: DMEM: Dulbecco’s Modified Eagle Medium (1X) with

GlutaMAX-I, 1g/L D-Glucose,sodium pyruvate (Invitrogen GIBCO #21885-025) was used for all cell lines except NT2D1, which was cultured in high glucose

(4.5g/L) DMEM without sodium pyruvate (Invitrogen GIBCO #61965-026); FCS: fetal calf serum; NEAA: non-essential amino acid (Invitrogen GIBCO); pen/strep:

penicillin/streptomycin, 1X=100U/100µg/ml (GE Healthcare PAA) * WI-38 VA13/2RA is referred to as WI38ALT herein.

55

Page 71: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

56

2.1.6 DNA/protein pellet preparation

Both DNA and whole cell extracts were derived from cell pellets collected in the

following manner. Cells were trysinised and pelleted at 1400 rpm for 5 minutes in a

Sorvall Legend RT benchtop centrifuge. Media was removed, cells resuspended in 1X

PBS and centrifugated 1400 rpm for 5 minutes. PBS was removed and the pellet

snap-frozen in dry ice or liquid nitrogen before storage at -80°C.

2.2 Genomic DNA extractions

2.2.1 DNA extractions

Small cell pellets (1x105-5x106 cells) were thawed quickly at room temperature and

resuspended in 50l of 1X SSC (see Appendix A). 50l lysis buffer (100mM Tris-HCl

pH7.5, 100mM NaCl, 10mM EDTA, 1% Sarkosyl) was added and lysates were

incubated at room temperature for 20 minutes with the addition of RNase (Sigma),

FC 10g/ml. Lysates were then incubated at 55°C for 6 hours with addition of

Proteinase K (Sigma), FC 100 g/ml. 100l phenol:chloroform:isoamyl alcohol

(25:24:1) was added to Qiagen MaxTract High density 1.5ml phase-lock tubes,

followed by lysates and mixed thoroughly. Tubes were centrifugated at 4000 rpm for

5 minutes, and the aqueous phase transferred to a clean tube. 20l (0.1X lysate

volume) 3M sodium acetate (Invitrogen) and 1l Invitrogen Pellet Paint were added

and mixed gently, and 100% ethanol poured gradually onto the surface of the

samples. Lysates were left at room temperature for 10 minutes to allow DNA

precipitation. Samples were gently inverted to spool the DNA pellet and 100%

ethanol removed. Pellets were then washed in 80% ethanol before removing ethanol

and drying at room temperature for 15 minutes.

For large pellets (>5x106cells), the above volumes were doubled or scaled up

appropriately. 2M sodium acetate was used and Pellet Paint was excluded from the

precipitation step. DNA pellets were dissolved in a volume of H2O appropriate to

Page 72: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

57

pellet size (20-100μl) and rotated overnight at 4°C to aid resolubilisation. Quantity

and quality of genomic DNA was confirmed using NanoDrop 1000 (Thermo

Scientific) and for STELA by electrophoresis of an aliquot of the sample on a 1%

agarose gel with 200ng high molecular weight marker.

2.2.2 Hirt DNA extractions

~5x106-1x107 cells were trypsinised and washed in PBS as in section 2.1.6. Pellets

were resuspended immediately after a PBS wash in 1ml Hirt buffer containing 0.6%

SDS and 10mM EDTA pH7.5, and incubated at room temperature for 20 minutes. 5M

NaCl was added to FC 1M and lysates were rotated overnight at 4˚C. The next day,

lysates were centrifugated at 18,000rpm at 4˚C. The supernatant was collected and

1ml phenol:chloroform:isoamyl alcohol (25:24:1) was added to Qiagen MaxTract High

density 1.5ml phase-lock tubes, followed by lysates and mixed thoroughly. Tubes

were centrifugated at 4000 rpm for 5 minutes, and the aqueous phase transferred to

a second MaxTract phase-lock tube and phenol:chloroform extraction repeated in

order to remove excess NaCl. 50l (0.05X lysate volume) 2M sodium acetate was

added and mixed gently, and 100% ethanol poured gradually onto the surface of the

samples. Lysates were left at room temperature for 10 minutes to allow DNA

precipitation. Samples were gently inverted to spool the DNA pellet and 100%

ethanol removed. Pellets were then washed in 80% ethanol before removing ethanol

and drying at room temperature for 15 minutes. DNA pellets were dissolved in a

100µl H2O rotated overnight at 4°C to aid resolubilisation, before restriction digests

and dot blotting (see section 2.8.2).

2.3 Single Telomere Length Analysis (STELA)

STELA is a PCR-based approach developed for this purpose and allows sizing of

individual telomere lengths of specific chromosome ends in a population. Details can

be found in section 3.1.1.

Page 73: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

58

EcoRI (NEB) restriction digests were carried out at 37°C for 2 hours to fragment

genomic DNA in reactions of 20l containing 40U enzyme and 1X NEB EcoRI

restriction enzyme buffer. Samples were then re-quantified using a NanoDrop 1000

spectrophotometer and diluted to 10ng/l before use in STELA PCR reactions.

STELA PCRs were carried using a method adapted from Baird et al. 2003. In a

specifically allocated PCR hood, digested genomic DNA was incubated with 0.9μM

Telorette 2 primer for 20 minutes prior to diluting in 1ng/l tRNA carrier solution to a

DNA concentration between 2.5ng-10ng/l. PCR reactions were set up as master

mixes before aliquoting into a 96-well plate. Water controls were included for each

primer set used for amplification. Each 10μl reaction contained 0.01M teltail primer,

0.3M subtelomere-specific primer, 0.9l 11.1X PCR buffer (see Appendix Table 1),

0.1l Taq (KAPA Biosystems): PWO (Genaxxon) (5:1, FC 0.5U and 0.05U respectively),

10ng tRNA and 1l DNA/tRNA/Telorette 2 mix (final DNA concentration of 250pg-

1ng). See Table 2.14-1 for primer sequences.

Initially, PCR was conducted on digested DNA samples at different concentrations to

optimise the number of bands produced by STELA so as not to over-crowd lanes or

leave lanes too sparse. Loading dye (0.1X TBE (see Appendix A), 6% glycerol,

0.1mg/ml bromophenol blue) was added to each well. Products were run overnight

at 120V on 40cm 0.8% HGT agarose gels in 0.5X TBE with 100ng Fermentas

GeneRulerTM 1Kb DNA ladder and 100ng Fermentas GeneRulerTM High Range DNA

ladder until the largest band of the marker ran 6cm from wells. Gels were imaged

using Gene Genius Bio Imaging System and GeneSnap software from SynGene to

confirm marker running and absence of smears, before trimming to 28-30cm in

length and Southern blotting (section 2.7).

2.3.1 PCR cycling conditions

STELA PCRs were carried out in Applied Biosystems Veriti 96-well thermal cyclers

under the following conditions:

Page 74: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

59

12q: 32 cycles of 20 seconds at 96°C, 40 seconds at 66°C, 10 minutes at 68°C. 1 cycle

of 10 minutes at 68°C.

XpYp: 32 cycles of 20 seconds at 96°C, 40 seconds at 64°C, 10 minutes at 68°C. 1

cycle of 10 minutes at 68°C.

17p: 32 cycles of 20 seconds at 96°C, 40 seconds at 65°C, 12 minutes at 68°C. 1 cycle

of 12 minutes at 68°C.

2.4 MS32 analysis

PCR was carried out on undigested genomic DNA. Each reaction contained 0.2μM

primers MS32B and MS32E, 1X PCR buffer (see Appendix A), 12.5mM Tris, 1ng/μl

high molecular weight salmon sperm DNA and 0.06ul of Taq (KAPA

Biosystems):cloned Pfu (Stratagene), (20:1 ratio, FC 0.3U and 0.015U respectively).

2.4.1 PCR cycling conditions

PCRs were carried out in Applied Biosystems Veriti 96-well thermal cyclers under the

following conditions: 32 cycles of 20 seconds at 96°C, 40 seconds at 62°C, 10 minutes

at 70°C. 1 cycle of 1 minute at 56°C, 10 minutes at 70°C

Products were size separated by electrophoresis overnight in a 20cm 1% agarose gel

before Southern Blotting (section 2.7).

2.5 DSN overhang assay

2.5.1 Digests

The Duplex Specific Nuclease (DSN) exhibits extremely specific preference for digest

of double-stranded DNA while leaving single stranded DNA intact (Shagin et al.,

2002). This allows near-complete digestion of double-stranded DNA but retention of

single stranded telomeric overhangs, which may then be quantified as described

Page 75: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

60

below. DSN assay was carried out as closely as possible to the method described by

Zhao et al. 2008.

One day prior to digests, genomic DNA was diluted to 350ng/μl and incubated at

37°C in a water bath overnight to aid complete solubilisation and accurate pipetting.

Digests were set up in pairs, and prior to DSN digest one sample was digested with

ExoI. ExoI degrades DNA in the 3’-5’ direction, digesting the 3’ overhang, allowing

this sample to be used as a control for background signal in the hybridisation

analysis. Experiments were set up as follows: DSN digest (Experiment): 5ug DNA,

1.7μl ExoI buffer (Fermentas), H2O to 17μl. DSN ExoI double digest (Control): 5μg

DNA, 10U ExoI enzyme (Fermentas), 1.7μl ExoI buffer (Fermentas), H2O to 17μl.

Digests were incubated for 1 hour at 37°C in a water bath before the addition of 0.2U

DSN enzyme and 2μl DSN 10X master buffer and a further incubation at 37°C in a

water bath, for 2 hours. Reactions were halted by the addition of 0.5μl 0.5M EDTA

pH8 and retained on ice until loaded onto an acrylamide gel.

Activity of DSN was previously confirmed as per manufacturer’s instructions by

carrying out restriction digest on sample plasmid DNA supplied with the enzyme.

DSN enzyme and all related buffers supplied by Evrogen.

2.5.2 Gel Electrophoresis

Polyacrylamide denaturing gel mix was prepared containing 6% 19:1 accugel

acrylamide 40% (National Diagnostics), 1 X TBE, 7.7M ultra-pure urea (Serva).

Gel mix was filtered via vacuum through 3MM Whatman paper and stored at 4°C,

protected from light.

Gels were cast in 16x16cm plates using the Protean II Xi system (Bio-Rad). Prior to

casting the gel, glass plates were cleaned with SDS solution, polished with IMS and

set up with 0.75mm spacers. To polymerise, 0.25μg/ml APS and 1ul/ml TEMED were

Page 76: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

61

added to the gel mix, which was then poured into the plate set-up and a 15-well

comb (Bio-Rad) inserted.

Once set and immediately prior to loading samples, wells were flushed with water

and excess urea and water removed with 3MM Whatman paper strips.

Meanwhile, samples were prepared by addition of half the sample volume of

formamide denaturing loading buffer (90% formamide, 0.01M EDTA pH8, 0.1μg/ml

w/v Xylene cyanol, with or without 0.1μg/ml w/v bromophenol blue) and heated at

65°C in a heat block for 5 minutes before loading into dry wells. Oligonucleotide

sequences for TTAGGG(n) markers are given in Table 2.5-1. Wells and Protean II Xi

cell were topped up with 1 X TBE running buffer and electrophoresis conducted for

approximately 80 minutes or until bromophenol blue had migrated two thirds down

the gel.

Name 5’>3’ sequence

Tel G3 GGGTTAGGGTTAGGGTTAGGG

Tel G4 TTAGGGTTAGGG

Tel G5 TTAGGGTTAGGGTTAGGGTTAGGGTTAGGG

Table 2.5-1 Oligonucleotides used as size markers in DSN assay. All oligonucleotides produced by

Sigma-Aldrich

2.5.3 Gel transfer

Two pieces of extra thick blotting paper (Bio-Rad) and a positively-charged nylon

membrane (Roche) were cut to size and pre-soaked in chilled 0.5X TBE for at least 20

minutes. The gel was placed onto the nylon membrane, on top of one piece of pre-

soaked (0.5X TBE) extra thick blotting paper (Bio-Rad), and covered with a second

piece of pre-soaked extra thick blotting paper before being placed into BioRad

Trans-Blot SD Semi-Dry transfer cell and air bubbles removed by applying an even,

rolling pressure in each direction across the sandwich. Transfer was carried out at

constant 400mA for 50 minutes. The membrane was then rinsed briefly in 0.5X TBE,

dried at 80°C for 15 minutes and cross linked (700kJ/cm2, CL-1000 UV cross-linker,

UVP).

Page 77: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

62

2.6 Chromatin Immunoprecipitation (ChIP)

ChIP buffers were prepared in advance and filter sterilised with 0.4μm syringe filter.

2.6.1 Cross-link and cell harvest

Cells to be harvested were cultured in 175cm2 flasks to ~90% confluency. In 10ml of

media, formaldehyde was added directly to adherent cells in tissue culture flasks at a

final concentration of 1% (v/v) and incubated for 10 minutes at room temperature.

125mM glycine was added to halt cross-linking and the flasks were incubated at

room temperature for 5 minutes. Flasks were then transferred onto ice. Cells were

first washed in 10ml ice cold 1X PBS then scraped in ice-cold 1X PBS and pelleted at

800rcf for 5 minutes at 4°C in a benchtop Sorvall Legend RT centrifuge. PBS was

removed and pellets snap-frozen and stored long-term at -80°C.

2.6.2 Bead preparation

One day prior to immunoprecipitation, 40μl IgG Dynabeads (Invitrogen) per ChIP

reaction were aliquotted into 1.5ml microcentrifuge screw-cap tubes and pelleted on

a magnetic rack (Dynamag-2, Invitrogen) and storage buffer was removed. Beads

were resuspended in ChIP-PBST wash buffer (1X PBS, 0.02% Tween 20) and rotated at

18rpm on a Stuart variable-speed rotator at 4°C overnight with the relevant amount

of antibody (see Table 2.16-1)

Beads were pelleted on a magnetic rack and washed once in ChIP PBST wash buffer

before adding 500μl ChIP dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM

EDTA pH 8.0, 16.7mM Tris-HCl pH 8.1, 150mM NaCl) with 1X protease inhibitor

cocktail (PIC) (Sigma Aldrich, P8340) and 75μl cell lysate (see preparation method

below).

Page 78: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

63

2.6.3 Cell lysis and chromatin preparation

Initially, ChIP was validated using a two-stage lysis to isolate nucleic DNA. This

involved a cell lysis step where ChIP pellets were resuspended to a concentration of

~5x107 cells/ml in chilled (4°C) ChIP Lysis Buffer (5mM PIPES, 20mM Tris-HCl pH 8.0,

85mM KCl, 0.5% IGEPAL CA-630 IP-40) with the addition of PIC, and incubated on ice

with regular vortexing to lyse cell membranes. Nuclei were then pelleted by

centrifugation at 4°C, at 800rcf for 4 minutes. The nuclei were resuspended at the

same dilution in chilled (4°C) Nuclear Lysis Buffer (50mM Tris-HCl pH8.0, 10mM EDTA

pH 8.0, 1% SDS) with PIC and retained on ice before sonication (see below). However,

it was found that cells did not lyse in ChIP Lysis Buffer, in accordance with a previous

report (Schoppee Bortz and Wamhoff, 2011), due to the level of protein cross-linking

at the cell membrane. It was also found that cell membranes did lyse in Nuclear Lysis

Buffer at room temperature, but that nuclear lysis was absent or minimal (Figure

2.6:1). Intact nuclei were, however, absent from lysates after sonication. As such the

lysis method was adapted and carried out as described below.

ChIP cell pellets were resuspended in Nuclear Lysis Buffer to a concentration of

~5x107 cells/ml and incubated at room temperature for 10-20 minutes (cell-line

dependent), while cell membrane lysis was monitored on an Olympus BH-2 BHS

microscope. During incubation, lysates were resuspended with a 0.5mm needle and

vortexed regularly to eradicate cell clumps and aid lysis. Nuclei were pelleted at

800rcf at 4°C for 5 minutes (Eppendorf 5415R microcentrifuge), resuspended in same

volume Nuclear Lysis Buffer and retained on ice. Lysates were sonicated in a

Diagenode standard Bioruptor on high power setting with the addition of 0.5cm of

ice as per the manufacturers instructions, for 6 cyles of 30 second on/30 seconds off

in 15ml Faclon tubes with a Bioruptor reflecting bar.

Page 79: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

64

In order to verify fragment size, 20μl lysate was reserved for gel confirmation (see

section 2.6.3.1). Lysates were then pre-cleared by centrifugation to aid removal of

background signal. Lysates were centrifugated at 13K rpm at 4°C for 10 minutes and

transferred to a new tube. This step was repeated once. 75μl lysate was then added

to the prepared bead:antibody complexes and incubated at room temperature for 1

hour with rotation (Stuart rotator SB3).

2.6.3.1 Gel confirmation of chromatin shearing

In order to confirm adequate shearing of chromatin to 200bp-1kb fragments, a small

aliquot of sonicated material was removed from each sonicated lysate and size

separated by gel electrophoresis. Lysates were first incubated overnight at 65°C with

rotation with the addition of 20μl IPure purification solution (see section 2.6.5).

Lysates were then purified using QIAquick PCR purification kit (Qiagen) as per the

maufacturer’s instructions and run on a 1.5% LE agarose gel with Low Molecular

Weight (LMW) ladder (NEB). Gels were imaged using Gene Genius Bio Imaging

System and GeneSnap software from SynGene.

2.6.4 Washes

Beads were washed with rotation (Stuart rotator SB3) at 4°C for 5 minutes in the

following chilled (4°C) buffers in the order of: Low Salt Immune Complex Wash Buffer

Figure 2.6:1 Brightfield images of HT1080 cells in lysis buffers. ChIP lysis buffer failed to lyse cell

membranes on ice or at room temperature. Nuclear lysis buffer was able to lyse cell membranes while

keeping nuclei intact a Cells in ChIP lysis buffer after 30 minutes incubation on ice. Cells remain intact and

clumped b Cells in ChIP lysis buffer after 30 minutes incubation at room temperature. Cells remain intact

and clumped c Free nuclei in nuclear lysis buffer after 15 minutes at room temperature. Cell membranes are

no longer intact and nuclei are retained.

Page 80: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

65

(0.1% SDS, 1% Triton X-100, 2nM EDTA pH 8.0, 20mM Tris-HCl pH 8.1, 150mM NaCl),

High Salt Immune Complex Wash Buffer (0.1% SDS, 1% Triton X-100, 2nM EDTA pH

8.0, 20mM Tris-HCl pH 8.1, 500mM NaCl) and Lithium Chloride Immmune Complex

Buffer (0.25M LiCl, 1% IGEPAL CA-630, 1% deoxycholic acid, 1mM EDTA pH 8.0,

10mM Tris HCl pH 8.1) in order to achieve high-stringency. These washes were

followed by two 5 minute washes in TE Wash Buffer (10mM Tris-HCl pH 8.0, 1nM

EDTA pH8.0) to remove excess salts.

2.6.5 Sample purification

DNA purification was carried out using the IPure kit (Diagenode) as per the

manufacturer’s instructions. Briefly, samples were incubated at 65°C with rotation

overnight in provided solution to elute and reverse cross-linking of DNA:protein

complexes. Dynabeads were removed and IPure magnetic beads added and

incubated for 1 hour at room temperature before washes in Dynabead wash buffers.

DNA was eluted in the elution buffer provided and used directly for dot blot and PCR

assays.

2.6.6 Semi-quantitative PCR

PCRs were set up as 20μl reactions as follows: 1X Taq Buffer (KAPA Biosystems),

0.5mM dNTPs (0.125mM of each), 0.3mM of each primer, 1U Taq. Primer sequences

may be found in Table 2.14-1. PCRs were carried out in Applied Biosystems Veriti 96-

well thermal cyclers under the following conditions: 1 cycle of 1 minute at 95°C. 32

cycles (30 cycles for D4Z4) of 15 seconds at 95°C, 15 seconds at 60°C, 30 seconds at

68°C (two-step PCR was carried out for GAPDH at 95°C and 68°C). 1 cycle of 1 minute

at 68°C.

2.6.7 Antibody verification

Antibody suitability for ChIP was confirmed by efficiency and specificity of the pull-

down of each protein by its respective antibody. Conjugated antibody:bead

Page 81: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

66

complexes were incubated with ChIP lysates for one hour, then washed briefly in

ChIP PBST wash buffer. Beads were then resuspended in mild elution buffer (1% SDS,

100mM NaHCO3) and rotated at room temperature for 2 minutes. Eluate was

retained and Laemelli buffer (see section 2.13.2) added to both the eluate (mild

elution) and the beads (harsh elution). Both mild and harsh elutions were carried out

in order to verify antibody binding to both the Dynabeads and the substrate. Most

protein:antibody interactions should be released under mild elution conditions,

allowing visualisation of the protein, while a harsh elution results in the release of the

antibody immunoglobulin chains. The samples were boiled for 10 minutes before

loading onto a SDS-PAGE gel. Western blotting was carried out as described in

section 2.13 and the antibody used for IP was also used as the primary to verify

specificity, except in the case of H3K9me3 (see section 6.2.1).

2.7 Southern Blotting

Gels were submerged and agitated in depurination solution (0.25M HCl) for 7

minutes. Gels were then rinsed in water and submerged in denaturation solution

(0.5M NaOH, 1M NaCl) for 20 minutes, changing the solution at 10 minutes, with

constant shaking. Gels were again rinsed before washing in neutralisation (0.5M Tris,

3M NaCl pH7.0) solution for 20 minutes (again changing solution once at 10

minutes). DNA was transferred onto 0.45μm Nylon Transfer Membrane (Osmonics),

by placing the gel onto 20X SSC wetted 3mm Whatman paper that was in contact

with a reservoir of 20X SSC. The membrane was placed on top, followed by two

sheets of Whatman previously soaked in 6X SSC. The blotting rig was topped with

paper towels and a weight and towels changed regularly for at least 5 hours to allow

consistent capillary action. At this point the rig was deconstructed and the

membrane was dried for 5 minutes at 80°C. DNA was cross-linked onto the

membrane by exposure to UV (short wave length 254nm, 700kJ/cm2) using the CL-

1000 UV crosslinker (UVP).

Page 82: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

67

2.8 DNA Dot blot preparation

2.8.1 Denaturing dot blots for ChIP analysis

ChIP samples were prepared in 96-well plates. The DNA was added to each well and

diluted in H2O to standardise samples to a final volume of 10μl. 5 volumes of

denaturing mix (0.5M NaOH, 2M NaCl, 25mM EDTA pH 8.0, with bromophenol blue)

per sample was added. Samples were resuspended 10 times, centrifugated briefly,

and incubated at room temperature for a minimum of 10 minutes. The samples were

then loaded onto a pre-soaked (H2O) 0.45μm Nylon Transfer Membrane (Osmonics),

cushioned with two sheets of pre-soaked 3MM Whatmann paper using a 96-well

Hybri-Dot manifold (Life Technologies), drawn through by vacuum. The DNA was

then washed twice with 120ul 2XSSC. Blots were dried for 10 minutes at 80°C and

DNA cross-linked onto membrane with UV (700kJ/cm2).

2.8.2 Native dot blots for overhang analysis

DNA samples were partially digested with AluI (NEB), with the addition of 1X

NEBuffer 4 at 37˚C for three hours. Half the sample was then digested with ExoI for

one hour at 37˚C. Both ExoI digested and undigested samples were prepared for

loading by direct addition of bromophenol blue and loaded onto 0.45μm Nylon

Transfer Membrane (Osmonics) as above. Two 150μl washes with 2X SSC were

carried out and blots were dried and cross-linked as above.

2.9 DNA radiolabelling

2.9.1 Random-primed labelling of DNA double stranded probes

15ng of PCR amplified probe (see section 2.9.2.1) or commercially bought ladder was

initially mixed with water and denatured by boiling for 5 minutes. 6l 5X OLB (see

Appendix Table A2), 12g bovine serum albumin (BSA), 7.5U DNA polymerase I large

Klenow fragment, 1.5l 32P -dCTP (0.555MBq) were added. Reactions were

Page 83: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

68

incubated at room temperature overnight and recovered with the addition 50l

Oligo Stop Solution (20mM NaCl, 20mM Tris-HCl pH 8.0, 2mM EDTA pH 8.0), 30l

high molecular weight salmon sperm DNA (30mg/ml), 25l 2M sodium acetate and

precipitating in 470l 100% ethanol. Pellets were then washed in 500μl 80% ethanol,

resuspended in 500μl H2O (1.5ml H2O for ladders) and incubated at 96°C for 5

minutes before adding to hybridisation tubes.

2.9.1.1 Telomere probe

PCR reactions for the telomere probe (Teloprobe) were set up in 40μl reactions

containing 1X PCR buffer (see Appendix A), 10μM Tel1 primer, 10M Tel2 primer,

26.6l H2O and 4U Taq polymerase (Kapa Biosystems) (see Table 2.14-1 for primer

sequences). PCR was carried out in Applied Biosystems Veriti 96-well thermal cyclers.

1ng of existing telomere probe was used as a template. PCR products were run on a

1% LE Agarose gel and products between 300bp and 12kb excised and purified using

QIAquick gel extraction kit (Qiagen).

Cycling conditions: 15-20 cycles of 40 seconds at 96°C, 40 seconds at 65°C, 5

minutes at 70°C.

2.9.2 Oligonucleotide probe end-labelling

Oligonucleotides (see Table 2.1-1 for sequences) were prepared as a master mix

containing 1X Kinase mix (70mM Tris-HCL pH7.0, 10mM MgCl2, 5mM spermidine

trichloride, 2mM dithiothreitol), 350U T4 polynucleotide kinase (NEB) and 0.15μl 32P

γ-dATP (55.5 kBq) per probe reaction. 9μl master mix was added to 0.14M

oligonucleotide per reaction. Reactions were incubated for 1 hour at 37°C before

overnight incubation at room temperature. Reactions were stopped by addition of

kinase stop solution (25mM EDTA pH 8.0, 0.1% SDS, 10μM ATP) at room

temperature.

Page 84: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

69

2.9.2.1 Oligonucleotides

Name 5’>3’ sequence

Tel G1 GTTAGGGTTAGGGTTAGG

TelC TAACCCTAACCCTAACCC

ssOc12 CCCTAACCCTAA

Table 2.9-1 Sequences of oligonucleotides used directly in end-labelling probe reactions for DSN overhang

assay. All oligonucleotides were generated by Sigma-Aldrich.

2.10 Hybridisation conditions

Radiolabelled probes were hybridised to DNA-bound membranes under the

following conditions:

2.10.1 STELA and MS32

Dried membranes were rinsed in water and placed between meshes in Hybaid

hybridisation tubes (Thermo). Membranes were pre-hybridised for 15 minutes prior

to hybridisation overnight with Teloprobe, LMW (NEB) and HMW (Thermo Scientific)

marker probes for STELA, and an MS32-specifc probe (derived in the Royle

laboratory) and 1kb marker (Thermo Scientific) for MS32 blots. Hybridisation and

pre-hybridisation were carried out in Modified Church Buffer (0.5M sodium

phosphate buffer, 1mM EDTA pH 8.0, 14% SDS) at 65°C. Membranes were then

washed in low stringency buffer (1XSSC, 0.1% SDS) in 15 minute intervals.

Membranes were then rinsed in water, blotted dry and mounted before exposure to

Amersham Phosphor Screens.

2.10.2 Native and denaturing dot blots

Pre-hybridisation was performed in TMAC hybridisation solution (3M TMAC, 0.6%

SDS, 1mM EDTA pH 8.0, 10mM Na3PO4, 5X Denhardt’s (see Appendix A), 4ug/ul S.

cerevisiae RNA (Sigma) at 58°C for 10 minutes. Hybridisation was carried out in TMAC

hybridisation solution for 5 hours or overnight at 48°C with the addition of random-

primed labelled probe. Washes were carried out in TMAC wash solution (3M TMAC,

Page 85: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

70

0.6% SDS, 1mM EDTA pH 8.0, 10mM Na3PO4), with three 5 minute washes at 48-49°C

and one final 15 minute wash at 50°C. Membranes were then rinsed in 3X SSC before

mounting and exposing to Amersham Phosphor Screen.

2.10.3 DSN overhang assay

Pre-hybridisation was performed in TMAC hybridisation solution (3M TMAC, 0.6%

SDS, 1mM EDTA pH 8.0, 10mM Na3PO4, 5X Denhardt’s (see Appendix A), 4ug/ul yeast

RNA) at 58°C for 10 minutes. Hybridisation was carried out overnight with the

addition of random-primed labelled telomeric oligonucleotide probes at 48°C for 18-

21mer probes, 25°C for the 12mer probe and end-labelled probes LMW marker

(NEB). Three washes were carried out at 48-50°C in TMAC wash solution (3M TMAC,

0.6% SDS, 1mM EDTA pH 8.0, 10mM Na3PO4) for 18-21mer probes, 28°C for 9-12mer

probes. Final 15 minute washes were at 52°C for 18-21mer probes, 30°C for 9-12mer

probes. Membranes were then rinsed in 3XSSC before mounting and exposing to

Amersham Phosphor Screen.

2.11 Analysis and quantification of hybridisation assays

2.11.1 STELA

Blots were exposed to Amersham Phosphor Screens and imaged using the Typhoon

9400 Variable Mode Imager (GE Healthcare). Image Quant Gel Imager 2D gel

software (GE Healthcare) analysis calculated lengths of individual bands from each

STELA according to size markers. Flanking sequence length was deducted before

further analysis using GraphPad Prism 6.0.

2.11.2 Dot Blots

Blots were exposed to Amersham Phosphor Screens and imaged using the Typhoon

9400 Variable Mode Imager (GE Healthcare). Gel Imager array software analysis

Page 86: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

71

provided intensity readings for each dot. These values were internally normalised to a

single dot from the input series before comparison.

2.11.3 DSN overhang assay

Standardising to a commercially available size marker, quantification was carried out

as previously described (Chai et al., 2005; Zhao et al., 2008). An example experiment

is shown for reference in Figure 2.11:1. After exposure to a Phosphor Imaging Screen,

the gel image was analysed using Image Quant array analysis software (GE

Healthcare). A column containing 100 boxes was overlaid on each of the

experimental and control lanes, as well as the marker lane, and numbered 0 to 100

(Figure 2.11:1a). Marker positions were manually assigned to a box position (to

1/10th of a box) and the size at the centre of each box was interpolated using

Graphpad Prism6 (Figure 2.11:1b).

Each box was then assigned this size and was plotted against the signal intensity as

recorded by Image Quant in each box for the control and experiment lanes (Figure

2.11:1c). The intensity of the control lane was subtracted from the intensity of the

experiment lane for each box to give an ODi value for each box (Figure 2.11:1c). As

proportionally more probe binds to larger fragments, each value was divided by the

size assigned to that value, giving an OD/Li, that is a relative value normalised to size.

Due to background at larger sizes, Zhao et al. suggested excluding larger fragments

above 400nt, and an upper limit of 500nt was initially assigned. As sizing became

impractical at the smallest sized fragments, a lower limit of 20nt was also assigned.

Results of this example are shown in Figure 2.11:1d.

Page 87: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

72

Control Experiment ODi

d

c

Figure 2.11:1 Quantification of DSN method a

Example gel image with and without the sizing

grid overlay (C=control, E=experiment), LMW

marker is shown with high and low contrast to

facilitate visualisation b Standard curve used for

sizing each grid-box c Control, experiment, and

experiment minus control results (ODi) d ODi/Li

results for signal between 50 and 500 nt. Line is

Gaussian fit (r2=0.8163, mean=49.30)

b

a

0

100

Page 88: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

73

2.12 Subtelomere genotyping

2.12.1 12q

Fragments from the 12q telomere-adjacent region were first amplified from genomic

DNA using the 12q sub-telomere specific primer pair 12qA and 12qB (see Table

2.14-1 for primer sequences). PCR reactions contained 0.9μl 1X PCR buffer (see

Appendix A), 3μM each primer, 2.5U Taq (KAPA Biosystems). PCRs were carried out in

Applied Biosystems Veriti 96-well thermal cyclers. These products were then sized

and quantified on a 1.5% gel before re-amplification in a secondary PCR containing

the same amounts of PCR reagents.

Cycling conditions: 1 cycle of 1 minute at 96°C. 32 cycles of 15 seconds at 96°C, 30

seconds at 61°C, 1 minute at 68°C. 1 cycle of 5 minutes at 68°C.

Restriction digests were carried out as described by Baird et al. (2000) with each of

the restriction enzymes illustrated in Figure 2.12:1. Restriction digests were incubated

for one hour with the appropriate 1X restriction enzyme buffer and at the

temperature indicated in Table 2.12-1. Restriction enzymes and buffers supplied by

NEB. Products were separated by gel electrophoresis in 1.5% LE agarose gels (BsmI

and KpnI) or 3.5% NuSieve agarose (AciI). In addition to the restriction analyses, a

larger fragment was amplified using primers KSRV2.D and 12qB under the same

Figure 2.12:1 Position of informative restriction sites and the 1440bp deletion at the 12q

subtelomere. Starting position of primers are also shown (bp)

Page 89: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

74

conditions to confirm the presence or absence of a large deletion reported to be

present in 6% of the population (Baird et al., 2000). All digests were first carried out

on CEPH DNA controls whose haplotypes were already established to confirm

experimental procedure.

2.12.2 Xp/Yp

PCR reactions for XpYp genotyping were set up as described for 12q, using the

primers Tsk8C and Tsk8G (Table 2.14-1).

Cycling conditions: 1 cycle of 1 minute at 96°C. 32 cycles of 40 seconds at 96°C, 50

seconds at 63°C, 1 minute at 68°C. 1 cycle of 5 minutes at 68°C.

Restriction digests were carried out at described by Baird et al. (1995) using the

appropriate 1X NEBuffer and incubation temperature indicated in Table 2.12-1 and

resolved on 3.5% NuSeive agarose gels. All digests were first carried out on CEPH

DNA controls whose haplotypes were already established to confirm experimental

procedure.

KpnI BsmI AciI DdeI AvaII TaqI MboII

NEBuffer 1 4 3 3 4 4 + BSA* 4

Temp. 37°C 65°C 37°C 37°C 37°C 65°C 37°C

Table 2.12-1 Respective NEBuffers and temperatures used for genotyping assay restriction digests.

*BSA added to FC 100μg/ml

Figure 2.12:2 Position of informative restriction sites at the XpYp subtelomere. Starting

position of primers are also shown (bp).

Page 90: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

75

2.13 Western Blotting

2.13.1 Gel preparation

SDS-PAGE gels were prepared by hand using Bio-Rad plates and manifolds (10 wells

per gel), and the percentage of acrylamide used was adapted for size of protein to be

analysed (Figure 2.13:1). Gels were prepared as follows: Separating gel: 6-15%

Acrylamide (Protogel (30%), 37.5:1 Acrylamide:Bisacrylamide, National Diagnostics),

0.21M Tris-HCl pH8.8, 0.1% SDS, 0.001% ammonium persulphate (APS); stacking gel:

4% Acrylamide, 0.24M Tris-HCl pH6.8, 0.1% SDS, 0.003% APS.

Tetramethylethylenediamine (TEMED) was added to a final concentration of 0.0008%

to polymerise gels.

2.13.2 Electrophoresis conditions and transfer

Lysates were diluted in Laemmlli loading buffer, FC: 63 mM Tris HCl, 10% Glycerol,

2% SDS, 0.0025% Bromophenol Blue, 2.5% β-mercaptoethanol. Typically 10ul of

Figure 2.13:1 Diagram indicating the percentage of acrylamide used in SDS-PAGE gels for

proteins within the range of sizes indicated. Sizes of proteins used in this study are indicated

after the protein name in brackets. α- and β-tubulin were electrophoresed on gels containing

7.5-12% acrylamide. Details of antibodies used may be found in Table 2.16-1

Page 91: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

76

sample was loaded per well with 5ul PageRuler Plus Prestained Protein Ladder

(Fermentas). Samples were boiled for 10 minutes for the first use, 2-5 minutes on

subsequent uses. Electrophoresis was carried out at 100V until loading dye passed

the stacker:running gel interface then at 150V until migration of the Protein Ladder

bands indicated adequate separation at the desired size. Amersham Hybond ECL

Nitrocellulose membrane (GE Healthcare) was pre-soaked for 10 minutes in 1X

transfer buffer (25mM Tris, 192mM glycine, 20% methanol), or in the case of 53BP1,

membrane was pre-soaked in transfer buffer excluding methanol and with the

addition of 0.01% SDS. Gels were removed from plates and soaked in transfer buffer

for a further 10 minutes. Sandwiches were constructed as follows: 3 layers of 3MM

Whatmann paper (soaked in 1X transfer buffer), Nitrocellulose membrane, SDS-PAGE

gel, 3 layers of soaked 3MM Whatmann paper. Transfer was carried out using the

BioRad Trans-Blot SD Semi-Dry transfer cell at constant ampage of 1mAmp/cm2.

Membranes were stained with Ponceau S solution (Sigma) to verify transfer before

air-drying. Membranes that were not to be used immediately were stored at 4°C.

2.13.3 Immunoblotting

Membranes were blocked in 5% Marvel skimmed milk diluted in 1X PBS with 0.1%

Tween 20 (PBST) for one hour with persistent agitation. The primary antibody was

diluted appropriately in 5% Marvel/PBST (see Table 2.16-1) and added to the

membrane for a minimum of 1 hour. After an initial rinse in PBST, membranes were

washed 3 times for 5 minutes in PBS/Tween. Secondary antibodies were diluted

1/10,000 in Marvel/PBST and added to the washed membrane for a minimum of 45

minutes. Washes were repeated and blots developed using Amersham ECL Prime

Western Blotting Detection Reagent (GE Healthcare) in accordance with

manufacturer’s instructions. Blots were immediately exposed to Kodak X-ray film and

developed.

Page 92: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

77

2.13.4 Quantification

Films were scanned in full colour and converted to 8-bit in ImageJ. ImageJ software

was used to derive a histogram of the intensity of each band and a relative intensity

value. Values of target proteins were normalised to the values of a control on the

same blot wherever possible. In order to accurately compare the normalised values

across different blots, initial values were first normalised to the total signal contained

in one blot for each protein immunoblotted.

2.14 Immunofluorescence

Cells were grown to a confluency of 50-70% (cell line-dependent) on 13mm glass

coverslips previously acid treated in 1M HCl and sterilised. Cells were then fixed in

cold 100% methanol (-20°C) and incubated at -20°C overnight.

Coverslips were washed 3 times in PBS and incubated for 1 hour in 3% BSA in PBS.

Coverslips were then incubated for 1 hour in 3% BSA-PBS containing primary

antibody at antibody-specific concentration (see Table 2.16-1). Coverslips were then

washed three times in PBS with a final wash with agitation for 5 minutes before

addition of secondary antibody at a dilution of 1/1000 in 3% BSA-PBS. Washes were

repeated as before and coverslips were mounted on microscope slides with Prolong

Gold mountant (Life Technologies). DNA was stained with the addition of DAPI to

either the secondary antibody or to Prolong Gold. Fluorescence detection screening

and analysis was carried out on the ScanR platform. Nuclear periphery was defined

by ± 3 pixels from the edge of the DAPI stain as detected by the software.

Page 93: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

78

2.15 Primers

Name 5’>3’ sequence

STELA

Telorette 2 TGCCCGTGCATCTGGCATCTAACCCT

Teltail TGCTCCGTGCATCTGGCATC

12qSTELA CGAAGCAGCATTCTCCTCAG

Xp/YpE2 TTGTCTCAGGGTCCTAGTG

17p6 GGCTGAACTATAGCCTCTGC

Semi-quantitative ChIP

D4Z4 F1 GACTGCCAGAAGTGCAACAA

D4Z4 R1 TGGCTCCACCTCATTACTCC

IGX1a F2 CGTGGATCATGGAGTAGGCT

IGX1a R2 ACTGGTGCTCTGTTCTGCCT

GAPDH F TACTAGCGGTTTTACGGGCG

GAPDH R TCGAAAGGAGGAGCAGAGAGCGA

MS32 analysis

MS32B AAGCTCTCCATTTCCAGTTTCT

MS32E CTTCCTCGTTCTCCTCAGCCCTAG

Subtelomere genotyping

12qA GGGACAGCATATTCTGGTTACC

12qB ATTTTCATTGCTGTCTTAGCACTGCAC

KSRV2.D ATCCTAGCAAAGCTGAGAACTCAG

12qArev CCCCAAAATATAATGGTAGGTAACC

TSK8C CAGGGACCGGGACAAATAGAC

TSK8G TCAGGGTCCTAGTGTGTCTGGA

Probe generation

Tel1 TTAGGGTTAGGGTTAGGG

Tel2 CTAACCCTAACCCTAACC

Table 2.14-1 Sequences of primers used in all PCRs

Page 94: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

79

2.16 Antibodies

Antibody Species Supplier Catalogue

number

Application/dilution

WB IF ChIP

Lamin A/C Goat Santa Cruz sc-6215 1/4000 - 6µg

Lamin A/C Rabbit Eric Schirmer - - 1/300 -

Lamin B Goat Santa Cruz sc-6217 1/150 1/150 -

SUN1 Rabbit Shackleton lab 2383-GE 1/500 - 8µg

SUN2 Rabbit Shackleton lab 2853 1/1000 - 8µg

53BP1 Rabbit Novus NB100-304 1/500 1/500 -

HP1-α Goat abcam ab77256 1/12,000 - 4µg

HP1-γ Rabbit abcam ab10480 1/200 1/200 4µg

H3K9me3 Rabbit Active Motif 3916 1/1000 - 4µg

H3K9me3 Rabbit Abcam ab8898 1/1000 - -

RAP1 Rabbit Santa Cruz sc-28197 1/500 - -

β-tubulin Rabbit abcam ab6046 1/4000 - -

α-tubulin Mouse Sigma T9026 1/4000 - -

Rabbit IgG Rabbit Millipore 12-370 - - 4µg

Table 2.16-1 Primary antibodies used in Western blotting (WB), immunofluorescence (IF) and

chromatin immunoprecipitation (ChIP). Dilutions used for WB and IF, and amount of antibody used in

each ChIP are given.

Antibody/

conjugate

Species

specificity

Host

species Supplier

Catalogue

number

Application/

dilution

WB IF

Anti IgG/HRP Rabbit Donkey GE

Healthcare

NA934 1/10,000 -

Anti IgG/HRP Mouse Sheep GE

Healthcare

NA931 1/10,000 -

Anti IgG/HRP Goat Donkey Santa Cruz sc-2020 1/10,000 -

Anti IgG/

AlexaFluor 488 Rabbit Donkey

Life

Technologies A-21206 - 1/1000

Anti IgG/

AlexaFluor 594 Goat Rabbit

Life

Technologies A-11080 - 1/1000

Table 2.16-2 Secondary antibodies used in Western blotting (WB) and immunofluorescence (IF).

IgG=immunoglobulin G, HRP=horseradish peroxidase.

Page 95: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

80

PART A • Telomere shortening in progeria cell lines

Chapter 3 Telomere shortening rate in progeroid

laminopathy cell lines established by STELA

3.1 Background

The relationship between ageing and telomeres, and the occurrence of

accelerated telomere shortening in non-laminopathy based progeria

syndromes, has prompted several studies aiming to identify telomere

abnormalities in HGPS cells and models. These studies have yielded promising,

if limited, results. Telomere length is shorter in HGPS patients compared to

controls (Allsopp et al., 1992; Decker et al., 2009) but as yet, no study has

directly investigated telomere shortening rate in patient cells. This is most likely

because of the small amounts of genetic material available due to the

difficulties involved in culturing HGPS cells.

The objective was therefore to establish telomere shortening rate in HGPS

dermal fibroblast cells. In addition to a classical HGPS line, telomere attrition

rate was also recorded in three other progeroid laminopathy fibroblast lines in

order to identify any correlation between altered telomere dynamics and the

nature and positioning of differing lamin A mutations. The lengths of two

telomeres (12q and XpYp) were measured at various time-points throughout

the proliferative lifespan of each cell line. In order to establish an accurate rate

of telomere shortening, the mean number of population doublings (PDs)

between each passage was counted. Telomere shortening rate was then

calculated relative to PD.

Page 96: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

81

3.1.1 Measuring telomere length: STELA

Several methods exist to record telomere length in cultured cells. The first to be

established, and the most commonly employed, is Terminal Restriction

Fragment (TRF) analysis (Moyzis et al., 1988). TRF was the first method used to

demonstrate telomere shortening in cultured fibroblasts (Harley et al., 1990).

The technique involves restriction digestion of relatively large amounts of

genomic DNA followed by gel electrophoresis and Southern blot hybridisation

to a telomeric probe. This identifies telomere fragments as a smear which may

be analysed to give a mean telomere length. Telomere lengths are extremely

heterogeneous, however, and large variability can be seen even within a single

cell type from a single individual (Harley et al., 1990; Moyzis et al., 1988; Weng

et al., 1997). Although a robust technique, this bulk measurement of telomeres

precludes the identification of any subtle population dynamics that may be

present. It is also not possible to accurately identify short telomeres that may be

responsible for senescence induction.

A similar problem arises with the use of Q-PCR. Due to the repetitive nature of

telomeric DNA, it is necessary to employ a specialised approach to allow PCR

amplification. The method described by Cawthon (2002) uses a primer pair with

enough homology to telomeric sequence to anneal to telomeres, but differing

from one another to such an extent as to avoid primer-dimer formation

(Cawthon, 2002). Q-PCR only allows quantification of telomeric sequence

relative to a control locus, rather than length, and results are not comparable

between separate PCR runs.

An alternative that allows for individual measurements of telomeres is

quantitative fluorescent in situ hybridisation (Q-FISH), which measures telomere

length on metaphase spreads (Egholm et al., 1993; Nielsen et al., 1991). By

employing a fluorescently labelled peptide nucleic acid (PNA) specific for

telomere repeats, a highly stable PNA:DNA hybridisation (Lansdorp et al., 1996)

Page 97: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

82

allows visualisation of all telomeres for quantification. Because Q-FISH identifies

single telomeres, individual chromosome dynamics have been revealed

(Martens et al., 1998), and the technique is ideal for documenting telomeric

aberrations (Hande et al., 1999). Q-FISH is a highly informative approach that

generally requires small amounts of starting material, however the starting cells

must have the potential to form metaphase spreads. Q-FISH quantification may

also be carried out through the use of fluorescence-activated cell sorting

(FACS). However, due to the high-throughput nature of this flow-FISH

technique, similar issues arise as with Q-PCR, in that relative telomeric

sequence, rather than length is measured. Although standard Q-FISH may be

highly informative, HGPS cells have an extremely high level of senescent and

slowly-dividing cells, excluding the possibility of harvesting metaphase spreads,

particularly at the end of proliferative lifespan.

As such, Single Telomere Length Analysis (STELA), a highly sensitive method

that is well-established in the Royle laboratory, was selected. STELA exploits

known and unique sequences from the subtelomeric regions of a limited

number of chromosome arms (2p, 11q, 12q, 17p and XpYp (Britt-Compton et

al., 2006) - so restricted due to the large sections of homology between

Figure 3.1:1 The distal end of the telomere. A STELA chromosome-specific primer anneals to a unique

subtelomeric sequence at a specific chromosome arm. Telorette 2 anneals in line with the 5’ strand and

is the template for the first round of PCR. This product contains the complementary sequence of the

second primer, Teltail. In subsequent PCR cycles, Teltail engages the template and initiates elongation.

Page 98: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

83

subtelomeres (Mefford and Trask, 2002). This allows telomere-specific

amplification. Figure 3.1:1 is a schematic representation of STELA-PCR. The

forward primer anneals within the subtelomeric region of the specified

chromosome arm. In lieu of a single reverse primer, a set of two partially

complementary primers are used. Initially, the first primer, Telorette, anneals to

the 3’overhang at its complementary 3’ end sequence. The 5’ end of the

Telorette contains a unique sequence, identical to a second primer, Teltail. At

the first round of PCR the G-rich 3’ end products are essentially tagged with this

sequence and in subsequent rounds of PCR the Teltail primer may initiate

elongation (Baird et al., 2003).

The products of STELA therefore represent individual telomeres from a

population. Gel electrophoresis of PCR products and Southern blot

hybridisation with a telomere-specific probe allows a direct comparison of PCR

products with size markers in order to establish length. The method may also be

used to amplify specific alleles in instances where single nucleotide

polymorphisms (SNPs) have been identified within regions of chromosome-

specific sequence, allowing allele-specific primer design.

STELA does have drawbacks and is a labour-intensive technique. As a PCR-

based approach, extremely large telomeres (>15-20kb) may not be as

effectively amplified. Although STELA requires only small amounts of DNA, the

DNA must be of extremely high quality to ensure retention of an undamaged 3’

overhang as it is the required substrate for the Telorette primer. Therefore the

use of columns and centrifugation is omitted from DNA extraction to avoid

mechanical force shearing, and to avoid co-precipitation of contaminants which

may affect PCR efficiency. The DNA pellet must therefore be visible and

manually isolated after precipitation. The minimum number of cells required for

a successful STELA DNA extraction was therefore established as ~5x105 cells

prior to growing patient cells. Despite this, STELA provides extremely high

Page 99: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

84

resolution, allowing visual identification of allelic subpopulations without the

use of allele-specific primers (Baird et al., 2003), and so has the potential to

distinguish subpopulation dynamics, including short telomeres that may be

responsible for senescence induction.

As such, STELA provides a sensitive and informative assay for the measurement

of telomere length from early and late proliferative stages, as well as insight into

individual telomere dynamics at the single cell level.

3.1.2 Telomere length in progeria cell lines

It has been previously reported that the telomeres of HGPS lines are shorter

than those of age-matched control lines (Allsopp et al., 1992). The first study

into HGPS telomere length, carried out in 1992 by Allsopp et al., employed TRF

analysis. This study gave the mean telomere length of five patients in

comparison with age matched controls. A similar comparative approach using

Q-FISH was carried out in 2009 (Decker et al., 2009) on three HGPS cell lines.

This study demonstrated that two out of three patient lines contained shorter

telomeres than normal controls. Although this study had the benefit of

identifying specific telomeres, the length analysis was only carried out at one

time point for all lines, and at greatly varying PDs (PD 17, 35 and 7). The HGPS

line that did not show significantly differently sized telomeres from controls

(AG11498) was analysed at PD 7. If the rate of telomere erosion is affected in

HGPS cells, longer telomeres at early sampling could represent the norm, and

the abnormally short telomeres recorded at later PDs in the other patient lines

may be a result of accelerated telomere erosion.

Because both these studies were carried out at only one time point, it is not

possible to determine whether short telomeres are the result of a

developmental setback, i.e. telomere length is initially short in these children, or

whether telomere shortening rate is increased in HGPS cells. Huang et al. (2008)

Page 100: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

85

developed a fibroblast line with an excisable hTERT construct, in order to

transfect mutant and wild-type lamin A forms and measure shortening rate in a

normalised background lacking the disadvantages of patient cells. Cells over-

expressing mutant forms of lamin A displayed an increased rate of shortening

as measured by average telomere length from Q-PCR. Although this approach

benefited from an isogenic starting point, ectopic expression of a transfected

mutant form of lamin A in the presence of endogenous lamin A resulted in

considerable over-expression which could affect the representativeness of the

results derived from this study.

3.1.3 Cell lines

A HGPS fibroblast line derived from a 14 year old male, with the classical

G608G, 1824C>T mutation was grown in culture. This line expresses progerin in

a comparable amount to WT LA (Figure 3.1:2), in accordance with previously

published data on this and several other classical HGPS cell lines (Rodriguez et

al., 2009). In order to elucidate relationships between lamin A mutations and

potential telomeric abnormalities, four more fibroblast lines derived from

patients with various progeria-associated laminopathies were also grown.

The first, T623S, is derived from a 10 year old female with a 1868C>G mutation

in LMNA that activates a cryptic splice site, as well as an amino acid change at

codon 623, although the amino acid change is unlikely to be causative as the

silent G608G results in a considerably more severe phenotype. The T623S

mutation results in similar processing problems as classical HGPS but is located

15 amino acids further along exon 11, resulting in a 35 amino acid deletion

(Δ35) which is located within the Δ50 deletion in RNA transcripts. As in classical

HGPS, the resultant protein remains farnesylated due to deletion of the

ZMPSTE24 recognition site, but an additional 15 amino acids are retained (see

section 1.1.6 and Figure 3.1:2).

Page 101: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

86

The T623S patient began to manifest minor symptoms at 6 years of age (Shalev

et al., 2007) and a similarly late onset was observed in a previous 1868C>G case.

This previous male patient lived to 45 years of age, by which time, after gradual

deterioration, his symptoms were identical to those of classical HGPS (Fukuchi

et al., 2004). These differences in onset may be due to some function conserved

by the 15 amino acids missing in classical HGPS, or due to lower expression of

the T623S mutant isoform (section 1.1.6).

The retention of the pre-lamin A farnesyl group has been implicated as a major

contributor to the pathology of classical HGPS (Capell et al. 2005; Toth et al.

2005; Fong et al. 2006; Constantinescu et al. 2010). In order to compare a non-

farnesylated mutant, a line derived from a biopsy of a 13 year old female with a

heterozygous 1945A>T missense mutation at the beginning of exon 11 was also

Figure 3.1:2 Position and resultant protein expression of LMNA mutations used in this

study a Western blot of lamin A/C in patient and control lines. HH is a normal human

fibroblast cell line. Arrows indicate fully processed lamin A and C as well as incompletely

processed forms found in the HGPS, T623S and RD lines b schematic of lamin A indicating

position of amino acid changes in the cell lines used in this study. Regions encoding the

N-terminal (NT) and C-terminal (CT) domains are indicated.

a

b

Page 102: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

87

examined. This mutation results in a single amino acid change of glutamic acid

to valine at position 578 and as such the line is referred to as E578V. Aberrantly

spliced farnesylated products do not accumulate (Csoka, 2004; Toth et al., 2005),

however, an increased proportion of E578V cells have misshapen nuclei

compared to normal lines (Csoka 2004; Toth et al. 2005). FTI treatment reduces

nuclear deformity and peripheral lamin A (Toth et al., 2005), supporting lamin A

accumulation in the nuclear matrix as a key contributor to the HGPS phenotype.

This patient exhibits relatively mild symptoms of progeria and atypical Werner’s

syndrome.

A second non-farnesylated line, S143F was selected and the growth rate and

senescence information was recorded. However, the limited number of PDs this

line completed in culture before reaching senescence precluded it from the

STELA analysis. The S143F line is derived from a 6 year old patient with early

onset myopathy who developed progeria symptoms at 5 years of age (Kirschner

et al., 2005). The heterozygous mutation is located in the second exon (428C>T)

and does not result in aberrant splicing (Figure 3.1:2 and Kirschner et al. 2005).

Although not apparent in Western blots carried out here (Figure 3.1:2), pre-

lamin A may be present at low amounts in this line due to an interference with

lamin A processing (Capanni et al., 2005).

A Restrictive Dermopathy (RD) line was also included in the analyses. RD is a

genodermatosis caused by mutations in the ZMPSTE24 gene (see section 1.1.6).

Mutations are homozygous or compound heterozygous and usually completely

eradicate ZMPSTE24 proteolytic activity (Moulson et al., 2005; Navarro et al.,

2013, 2004). The posttranslational processing carried out by ZMPSTE24 is

essential for the formation of mature lamin A and as such, RD cells produce only

pre-lamin A (Figure 3.1:2a). RD is categorised with other laminopathies for this

reason, and is commonly studied as a more severe manifestation of HGPS due

to overlapping symptoms. Two children with LMNA mutations, including one

Page 103: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

88

with the classical G608G mutation, have been diagnosed with RD (Navarro et

al., 2004).

RD was first described in 1986, characterised by an extremely severe cellular and

physiological phenotype and neonatal death (Witt et al., 1986). After pregnancy

issues and premature birth, babies exhibit growth retardation, tight, thin skin

that is easily eroded and limited joint manoeuvrability. It is possible that the

differential symptoms of RD that contribute to the severity of the disorder arise

due to the complete absence of mature lamin A or a deficiency in ZMPSTE24

activities not related to lamin A processing. The cell line used in this study was

derived from a patient carrying a homozygous 1085-1086insT in the ZMPSTE24

gene. This mutation is the most common in RD (Moulson et al., 2005; Navarro et

al., 2005) and results in a premature transcriptional stop and no enzyme

functionality.

Three normal skin fibroblast lines derived from healthy individuals were used as

controls: Normal Human Diploid Fibroblast (NHDF), 155-BR and 161-BR, see

Table 2.1-1).

3.2 Results

3.2.1 Growth rates and senescence levels

Cell lines were grown under ambient O2, 5% CO2 and with high humidity. All the

patient cell lines were grown in DMEM media supplemented with 20% FCS and

100U/μg penicillin/streptomycin per ml media. The NHDF line was cultured in

10% FCS-supplemented DMEM, with 100U/μg penicillin/streptomycin per ml

media. The 151-BR and 161-BR line were grown without antibiotics, with 15%

FCS supplementation in DMEM, and 1% non-essential amino acids were added

to the 155-BR medium. Patient cells generally grew very slowly (in some cases

less than 1 PD every 5 days at early PDs) and splits were limited to 1:2. Faster

Page 104: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

89

growing cell lines which divided roughly once every 24 hours, such as NHDF

and E578V, were split 1:2-1:4. Cell counting was employed to establish

population doublings (PDs). All cell lines were cultured from the lowest available

PD from lab stocks and were grown to senescence where possible (i.e. cell

number did not significantly increase after one week and/or senescence was

recorded at >95%). Trypan Blue staining was employed to establish viability

levels of all cell lines. Non-viable cells were counted and then discounted from

all calculations.

Patient cells exhibited a correlation between replicative capacity and phenotypic

severity (Figure 3.2:1a). All cell lines maintained a level of 95-100% viability

throughout their proliferative lifespan, excluding 155-BR and HGPS which

displayed a sudden drop in viability at their final passage (80.5% and 84.5%

respectively), accounting for the variability in viability in these lines (Figure

3.2:1b). These data suggest that LMNA mutations do not have an effect on

overall cell viability. Cell lines derived from patients with the most severe

phenotypes tended to grow more slowly throughout their cultured lifespan than

Figure 3.2:1 Final PD and viability of each cell line a Final PD reached by each cell line

cultured, excluding NHDF, which did not reach senescence. The HGPS value given is the

average of two separate rounds of subculturing b Mean viability of patient and control lines

which were carried forward to the STELA analysis, derived from Trypan Blue assays carried out

throughout the proliferative lifespan of all cell lines.

Page 105: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

90

cells derived from patients with milder symptoms (Figure 3.2:2). The normal cell

lines that were passaged alongside the patient lines showed varying rates of

growth. The NHDF line was abandoned prior to its reaching senescence due to

contamination. However, senescence would be anticipated to halt proliferation

at around 60 PDs (based on previous work in the lab as well as Suzuki et al.

2012), and at 35PDs the line still retained a largely actively dividing population

of ~80% (Figure 3.2:3). Unexpectedly, 161-BR reached senescence at only 15.5

PDs (Figure 3.2:3b&c). As 161-BR is derived from a healthy 49 year old male, it is

likely that at the time this cell line went into culture, the number of PDs

recorded was greatly underestimated, perhaps as a result of the initial

derivation of the line or erroneous record-keeping. The 155-BR also fell short of

its anticipated growth potential, reaching only ~30PDs before reaching

senescence. The HGPS, RD and T623S cell lines failed to divide beyond around

20 PDs, in accordance with previous reports (Benson et al., 2010; Goldman et al.,

2004; Liu et al., 2006; Shumaker et al., 2006). The S143F cell line (mild progeria)

grew to 35PDs, while E578V (very mild progeria symptoms), continued to divide

to around 50 PDs, which is within the range of the expected lifespan of a normal

fibroblast line (Hayflick, 1965).

Throughout the cultured lifespan of the patient and control cell lines, β-

galactosidase senescence assay was carried out. This method exploits the

specific increased abundance of β-galactosidase associated with an increased

lysosomal function present only in senescent cells. Upon the addition of X-gal

substrate in sub-optimal conditions (pH6 rather than pH4), the excessive

amount of β-galactosidase present in senescent cells is able to cleave X-gal,

which appear blue. Presenescent and quiescent cells remain unstained (Dimri et

al., 1995).

In all cell lines, an increase in β-galactosidase staining correlated with a

decrease in rate of division (Figure 3.2:2 and Figure 3.2:3). As expected, the

Page 106: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

91

HGPS line displayed a high proportion of senescent cells. Even while growth

rate remained steady, the HGPS population had a consistently higher

proportion of β-galactosidase positive cells, with ~40% of cells senescent

between the first PD analysed and ~PD 20 (Figure 3.2:2a). Despite the severity

of RD, up to 89% of the cell population were not senescent in earlier passages

(Figure 3.2:2b). However, this level drops to ~60% at around PD 13 – a similar

time-point to the starting PD for HGPS. As the RD cell line was available at an

earlier PD than HGPS, and the first recordable senescence data for the HGPS

line was at roughly PD13, it is possible that younger HGPS cells may also have

have a lower initial proportion of senescent cells.

Figure 3.2:2 Patient cell lines were cultured

from the lowest PD available to senescence

except in the case of E578V which was

initially cultured to around 80% senescence,

then another frozen stock was recovered

and grown to complete senescence.

Growth experiment was repeated in full for

HGPS. The second experiment is indicated

on for both E578V (e) and HGPS (a) by a

dotted line.

Page 107: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

92

The other patient lines exhibited low initial numbers of senescent cells (~20%),

similar to levels in the normal lines NHDF and 155-BR. The high proportion of β-

galactosidase positive cells present in 161-BR confirm the impression that 161-

BR was in fact much closer to senescence than was recorded prior to the cells

being thawed. Due to the small number of pellets that it was possible to harvest

from 161-BR over less than five PDs, it was excluded from STELA analysis. The

155-BR line grew unusually slowly in culture for almost 15 PDs before the

proportion of senescent cells began to increase (Figure 3.2:3). Although it is

possible that the divisional “age” of the cell line was also underestimated at the

point of thawing, the senescence data, in combination with the number of PDs

completed by 155-BR before senescence levels elevated, indicate that the slow

growth rate, and potentially the early entry into senescence, genuinely reflect

the biology of this line. This implies a potential similarity to the laminopathy

lines in terms of growth capacity (discussed in section 3.3.3).

β-galactosidase assay is not without pitfalls. The required pH of exactly 6.0

proves to be difficult to confirm due to the limitations of equipment accuracy.

Figure 3.2:3 Control cell lines were

cultured from the lowest PD available

from lab stocks to the point of

senescence (i.e. cell number did not

significantly increase after one week

and/or senescence was recorded at

>95%), except in the NHDF line which

was grown to ~35 PDs only.

Page 108: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

93

Also, differences between cell lines may affect the optimal pH for observing

only senescence-associated activity. As such, data accrued from cell passaging

experiments is slightly erratic in places and if it is assumed that β-galactosidase

activity decreases in a linear or exponential manner, from the data acquired it

can be inferred that the error rate is as much as ±10%. However, the β-

galactosidase data collected does demonstrate observable trends for all cell

lines.

3.2.2 Telomere shortening in laminopathy and control

fibroblasts

STELA was carried out on DNA derived from samples taken at various time-

points for four patient lines and two normal lines. Representative Southern blots

for each cell line are shown in Appendix B.1. Each gel was run with a control

DNA (extracted at one time point from a single cell pellet of HT1080 cells) in

order to establish technical reproducibility of PCR and size separation. HT1080

bands were sized on each gel and the distributions were statistically tested in

order to confirm no significant difference before pooling data collected from

separate gels for each cell line. Data for all HT1080 bands is given in Appendix

B.2. STELA primers are positioned at a set distance from the beginning of the

telomere and so the flanking sequence was subtracted from sized bands prior

to regression analysis using Graphpad Prism6 to establish a shortening rate at

each end. A reduction in the median length of telomeres over time was

observed in all but the NHDF line.

3.2.2.1 NHDF

The Normal Human Diploid Fibroblast cell line, NHDF, is derived from a healthy

adult male. Problems in tissue culture precluded growing this line to

senescence; however cell pellets were collected for over twenty PDs and used

for STELA analysis. Five Southern blots were prepared on NHDF STELA products

Page 109: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

94

and PCR efficiency and consistent gel running conditions were confirmed by

ANOVA of the HT1080 control (Appendix B.2). The telomeres of NHDF cells

have an extremely large length distribution at 12q and XpYp. The median values

for each time point analysed did not decrease regularly as would be expected

(Figure 3.2:4a, Table 3.2-1). This caused difficulties in deriving a significant

shortening rate at the 12q telomere, which regression analysis assigned a rate

Figure 3.2:4 Telomere length analysis on the NHDF

cell line a 12q STELA results after subtraction of

544bp b Regression analysis on 12q STELA data.

Shortening rate is 16.32±14.77bp/PD (p=

0.2579). Dotted line indicates 95% confidence

band c Regression analysis on 10th and 90th

percentiles, shortening rates -50.18±5.006bp/PD

(p<0.0001) and -7.42±16.66bp/PD (p=0.6576)

respectively. Dotted line indicates 95% confidence

band d XpYp STELA results after subtraction of

406bp e Regression analysis on XpYp STELA data.

excluding PD 28.0. Shortening rate is -90.98

±18.35bp/PD (p<0.0001). Dotted line indicates

95% confidence band.

Page 110: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

95

of -16.94±14.96bp/PD (p=0.2579).

A distribution histogram identifies two distinct peaks within the 12q STELA data

(Figure 3.2:5a). This clustering implies the presence of one large and one short

subpopulation, with some telomeres of intermediate length. Allelic length

differences between maternal and paternal chromosomes are a possible source

of this population anomaly. Unfortunately the NHDF line was genotyped to be

homozygous at the 12q sub-telomere (Appendix C.1), precluding the use of

allele-specific STELA to isolate a single subpopulation and alleviate noise. In

order to eliminate the possibility of a contaminating concomitant cell

population in the NHDF population, a minisatellite analysis was carried out. The

MS32 minisatellite is a highly variable region, and amplification of a single

individual’s genome results in one or two products only, indicating

Figure 3.2:5 Smoothed histogram of NHDF 12q STELA data shown a in entirety and b expanded to

highlight short telomere length distribution (500-5000bp). Excluding PD6.4, the proportion of

shorter telomeres decreases over time. Relative frequency in PD 28.0 is especially low for short

telomeres, suggesting a loss of cells with short telomeres at this PD. Bin width set at 500bp.

Table 3.2-1 Statistics for NHDF cell line telomere length data, all values given in bp.

Page 111: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

96

homozygosity or heterozygosity at the locus. The analysis was carried out on all

cell lines, which all proved to have either one or two alleles at MS32, confirming

no admixture of cell lines in any of the cultures, including NHDF. Results are

shown in Appendix D.

In the absence of any compounding non-native subpopululation, this data

highlights two foreseen concerns regarding the use of STELA analysis, and likely

any technique used for determination of telomere length of the same

population over time. Firstly, as with any PCR-based method, there remains a

limit to the size of fragments that may be amplified, thus imposing a ceiling on

the upper limit of recordable telomere size. It is not possible to circumvent this

outcome by limiting the use of STELA data to later passages as large telomeres

that may be present in the population would begin to phase into the PCR

product pool. This could also mask any change in telomere dynamics

throughout the proliferative lifespan of cell lines. Also, when considering the

patient lines, exclusion of time-points from the analysis would be unfeasible

due to the limited proliferation of these cells.

Secondly, cells with short telomeres may be lost from the population prior to

PCR. With serial passaging, and even at early PDs when the vast majority of cells

are actively dividing and a large proportion of cells have long telomeres, a

continually increasing proportion of cells will enter senescence due to telomere

shortening (Martin-Ruiz et al., 2004). Senescent cells have many biological and

morphological differences to actively dividing cells (Campisi, 2013), including

their ability to re-adhere after trypsinisation. If this is the case, the result would

be a loss of a proportion of senescent cells at each passage. If the proportion of

cells activating senescence remains low, relatively steady gain and loss of

senescent cells from the population could explain the steady-state levels of

senescence observed initially in cell lines, or indeed the peaks and troughs

around ~80%, rather than the expected continually increasing proportion if

Page 112: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

97

senescent cells were not lost. As cells with short telomeres at one end are more

likely to be cells that have doubled more than the average number of times in

this population, it is likely that telomeres on other chromosomes are shorter in

these cells, and that many of these cells are senescent. Cells with short

telomeres may therefore be lost from the population in a biased manner,

resulting in the telomeres of these cells to be disproportionally lost from the

pool of telomeres in the STELA reactions.

Both these factors appear to have manifested in the 12q data for NHDF. The

maximum length at the final three time-points confers a decrease in length,

with the largest telomere to be amplified being 14904bp at PD 18.7 (Table

3.2-1). No fragments amplified above this size are present in the products of the

first two time points. Distribution analysis reveals a trend towards an increase in

the proportion of long telomeres, suggesting that additional long telomeres are

being added into the recorded population (Figure 3.2:5).

This suggests that the largest telomeres are failing to be amplified by STELA at

PD 6.4 and 12.3 (Figure 3.2:4a). Conversely, the minimum length recorded at

each PD is smaller as the population ages, with the exception of the final time

point (PD 28.0). The proportion of telomeres that are below 4kb decreases at

later PDs (Figure 3.2:5). STELA is able to detect even the smallest fragments, and

so the likelihood of amplification failure of short telomeres is extremely low. As

telomere length is not anticipated to increase in primary cell lines, this reduction

in smaller PCR products supports the loss of senescent cells from the

population. These factors will result in an observed reduction in telomere length

at early PDs and the number of shorter telomeres at later PDs, and therefore a

tendency to underestimate telomere shortening rate.

Taking these factors into account, sensitivity analysis was carried out by

regression analysis of the 10th and 90th percentiles and one-factor-at-a-time

(OFAT) style exclusion of time points with uncertainty. By isolating the 10th and

Table 3.2-2

Page 113: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

98

90th percentiles, the shortest and longest telomeres are selected for analysis,

which gives significantly different shortening rates of -50.18 ± 5.006bp/PD

(p<0.0001) and -7.416 ± 16.66bp/PD (p=0.6576) respectively. An extremely

significant p value for the shortest telomeres indicates that this rate is the more

representative as a linear rate of change is anticipated, particularly in the

exponential growth phase (see section 3.3.2). Removing PD 28.0 from the 10th

percentile data due to the presumed loss of small ends, gives a shortening rate

of -54.41 ± 5.957 (p<0.0001). This rate, though not significantly different from

the rate including PD 28.0 (p=0.588), is likely the more representative. Next,

time points were excluded OFAT from the 90th percentile data. None of the

regression lines derived from this method provided a significant p value. This

may be mainly attributable to the loss of longer telomeres at earlier PDs (see

section 3.3.1)

The distribution of XpYp telomere lengths appears to have been within the

upper limit of STELA amplification, with possible drop out of the longest ends at

PD 6.4 (Figure 3.2:4d&e). A shortening rate of -99.37±16.77bp/PD was

calculated from all data (p<0.0001), while exclusion of PD 6.4 unexpectedly led

to a reduction in rate to -81.65 ± 23.65bp/PD, as well as a drop in significance

(p=0.0006). Due to the small number of points collected for PD 28.0 (n=35),

this time-point was also excluded from the complete dataset resulting in an

insignificant change in rate to -90.98 ±18.35 bp/PD (p<0.0001).

3.2.2.2 155-BR

A second normal fibroblast line derived from a healthy 55 year old male, 155-

BR, was passaged to senescence but grew notably slowly for a normal line

(Figure 3.2:3b), and at a similar rate to patient lines. STELA reactions and

Southern hybridisations for 12q were carried out by both Kim Fitzpatrick (KF)

and Rachel Turner (RT) as indicated in Figure 3.2:6b across seven gels. Sizing

was carried out by RT. HT1080 distribution confirmed no significant difference

Page 114: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

99

between those STELAs carried out by KF and those by RT (p=0.9780) and so

data was pooled to give a resultant shortening rate of -55.96±15.51bp/PD

(p=0.0003, Figure 3.2:6b). 12q STELA results demonstrated a large telomere size

distribution, including very small molecules. As with NHDF, the proportion of

shorter telomeres is reduced over time (Figure 3.2:7). Repeating the regression

analysis excluding PD 28.9 increases the shortening rate to -79.72±18.14bp/PD

(p<0.0001).

At XpYp, a faster shortening rate of -111.4 ± 21.06bp/PD (p<0.0001) was

Figure 3.2:6 Telomere length analysis on the

155-BR cell line a 12q STELA results after

subtraction of 544bp b PCR products

generated by RT and KF c Regression analysis

on 12q STELA data excluding PD 28.0.

Shortening rate is -79.72±18.14bp/PD

(p<0.0001). Dotted line indicates 95%

confidence band d XpYp STELA results

(generated by RT) after subtraction of 406bp e

Regression analysis on XpYp STELA data

excluding PD 16. Shortening rate is -

194.3±29.28bp/PD, (p<0.0001). Dotted line

indicates 95% confidence band.

Page 115: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

100

recorded. Results at PD 16.0 have a lower median and distribution than at the

next time point. Again, at this early PD it is quite possible that the longest

telomeres would not be within the amplification range of STELA. The

distribution at this time point also seems to have shifted in that the main body

of telomere lengths is shorter than would be expected. This may be indicative of

a shift in PCR efficiency, as, due to the presence of long telomeres, polymerase

may be preferentially elongating shorter telomeres in a compensatory manner.

This time point was excluded in sensitivity analysis and the resultant shortening

rate of -194.3±29.28bp/PD, (p<0.0001) (Figure 3.2:6d) is significantly higher

than at 12q (p=0.0037), even with the exclusion of PD 28.9 from the 12q

analysis (p=0.0152). Differences in erosion rate at different ends have previously

been recorded (Mendez-Bermudez and Royle, 2011). However, the XpYp

shortening rate obtained is faster than even the fastest shortening rates

previously recorded in normal fibroblasts (Baird et al., 2003; Vaziri et al., 1993).

It is possible that this line has a faster telomere shortening rate than would be

expected, and the similarity between growth rates of 155-BR versus patient lines

may have implications for interpreting the patient results (see discussion).

Figure 3.2:7 Histogram of telomere length distributions for 155-BR at 12q shown a in

entirety and b expanded to highlight short telomere length distribution (500-3000bp). Bin

width set at 500bp.

Page 116: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

101

3.2.2.3 HGPS

Due to the extremely slow growth rate of HGPS cells, and their rapid decline to

senescence (Figure 3.2:2a), it was only possible to collect a limited number of

cell pellets. These pellets were also very low in cell number (e.g. 2-6 x 105 per

25cm2 flask) and upon reaching around 20PDs even lower numbers of cells were

collected. As with all cell lines approaching senescence, this was in part due to

the increased size of senescent cells, but also the impaired ability of senescent

cells to adhere to tissue culture plastics after passaging. In the case of HGPS, the

window for collecting cells was very narrow (Figure 3.2:2), and only a limited

number of pellets contained enough cells to extract a visible DNA pellet.

As a consequence, STELA was performed on four pellets taken less than three

PDs apart. This proved to be too short a window to derive an accurate

shortening rate and regression analysis failed to produce a statistically

significant slope in the case of 12q (-165.1±145.9bp/PD, p=0.2583). To

eliminate noise, regression was attempted on the 10th and 90th percentiles,

yielding the respective significant rates of -240.7±105.6bp/PD (p=0.0264) and -

540.9±172.0bp/PD (p=0.0026). As these rates are not significantly different from

one another (p=0.1389), it is possible to derive a pooled rate of -390.7bp/PD.

However, as the range of these rates is large due to the inaccuracies of

performing a regression analysis across such a short timeframe, these results

are only indicative of an increased rate of telomere shortening. Notably,

extremely short telomeres are present (<400bp), and telomeres are highly

variable in length in accordance with previous findings using Q-FISH (Decker et

al., 2009).

XpYp shortening rate was also not significantly different from zero (-

215.8±119.1bp/PD, p=0.0704). Analysis of the 10th and 90th percentiles was

attempted and the longer ends provided a significant and rapid shortening rate

of -393.8±170.7bp/PD (p=0.0252). The 10th percentile regression did not yield a

Page 117: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

102

significant result (-56.25±169.0bp/PD, p=0.7406), most likely due to varying

amounts of smaller products present in the STELAs.

Figure 3.2:8 Telomere length analysis on the HGPS cell line a 12q STELA results after subtraction

of 544bp b Regression analysis on 12q STELA data. Shortening rate is -165.1±145.9bp/PD,

p=0.2583. Dotted line indicates 95% confidence band c Regression lines for 10th and 90th

percentile telomeres. Shortening rates -240.7±105.6bp/PD (p=0.0264) and -540.9±172.0bp/PD

(p=0.0026) respectively d XpYp STELA results after subtraction of 406bp e Regression analysis on

XpYp STELA data. Shortening rate is -194.3±29.28bp/PD (p=0.0704). Dotted line indicates 95%

confidence band f Regression lines for 10th and 90th percentile telomeres. Shortening rates -

56.25±169.0bp/PD (p=0.7406) and -393.8±170.7bp/PD (p=0.0252) respectively.

Page 118: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

103

Excluding a single outlier from the final time-point, the range of HGPS 12q

telomere length changes from 15269-849.9 to 11841-413.6. Taking the median

at these time points gives a rate change of -504.6bp/PD. Identical treatment at

XpYp gives a rate of -245.8bp/PD (range change: 12675-1073bp/PD to 11074-

3040bp/PD, excluding outlier at PD7.7). Again, these rates are indicative of an

increased telomere shortening rate in HGPS cells.

3.2.2.4 RD

The RD cell line was available from PD 6 and as such it was possible to extract

DNA from pellets taken across a longer time-frame than the HGPS line, and to

expand the cells so that at senescence a sizeable pellet could be acquired,

despite the severity of the effects of the mutation. As such, telomeres spanning

12.8 PDs were sized and a significant shortening rate (p<0.0001) was observed

at both 12q (-210.5±24.67bp/PD) and XpYp (-183.5±34.40bp/PD). STELA

reactions and Southern blots for XpYp were carried out by Kim Fitzpatrick. Due

to apparent loss of large telomeres at PD7.5, as indicated by the median and

distribution, regression analysis was repeated for XpYp excluding PD 7.5

resulting in a rate of -221.8±49.13bp/PD (p<0.0001).

For further insight into shortening rates at different chromosome ends in a

severe phenotype, STELA was also carried out on RD at 17p. STELA results

yielded a shortening rate of -196.8 ± 20.28 (p<0.0001). Products derived from

the DNA sample taken at PD 7.5 appear to have failed to amplify efficiently,

most likely due to the large distance between the annealing site of the 17p

STELA primer available and the start of the telomere (3078bp). Excluding this

data point results in a shortening rate of -223.8±26.85bp/PD (p<0.0001). All

three ends demonstrated a shortening rate considerably faster than that

previously recorded in normal cells, and are not significantly different from one

another (p=0.9515).

Page 119: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

104

Figure 3.2:9 Telomere length analysis on the RD cell line a 12q STELA results after subtraction

of 544bp b Regression analysis on 12q STELA data. Shortening rate is -210.5±24.67bp/PD

(p<0.0001). Dotted line indicates 95% confidence band c XpYp STELA results after subtraction

of 406bp. PCR products and Southern blots generated by KF for XpYp telomere d Regression

analysis on XpYp STELA data excluding PD7.5. Shortening rate is -221.8±49.13bp/PD

(p<0.0001). Dotted line indicates 95% confidence band e 17p STELA results after subtraction of

3078bp f Regression analysis on 17p STELA data excluding PD7.5. Shortening rate is -

223.8±26.85bp/PD (p<0.0001). Dotted line indicates 95% confidence band.

Page 120: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

105

Interestingly, a distinct subpopulation with longer telomeres is visible at 12q.

This individual has inherited the same 12q allele from each parent (Appendix

C.1), and so it is not possible to state whether this subpopulation is due to an

allelic difference. In this instance, this subpopulation does not appear to be

interfering with the regression analysis as it has been amplified efficiently at

every time point.

3.2.2.5 T623S

The T623S phenotype is considered a milder form of classical HGPS, with the

same symptoms but later onset and slower disease progression. In culture,

T623S cells did not continue to PDs much beyond HGPS cells (approximately 22

PDs vs 20), however, cells of low PD were available for this line and STELA was

carried out at six time-points across 13.3 PDs. 12q STELA PCRs and Southern

blots were generated by both RT and KF.

Kruskall-Wallis analysis of HT1080 data at 12q demonstrated significant

differences between two sets of gels, each set containing three out of six time

points (gels 1,2 and 3 carried out by KF, gels 4 and 5 carried out by RT). Tukey’s

multiple comparisons test revealed significant differences were only present

between gel 1 and gel 4, and gel 2 and gel 4. Differences in HT1080 sets were

due to increased efficiency of large fragment amplification in the PCR reactions

carried out by RT (Appendix B.2). As larger fragments are not absent from the

T623S PCRs carried out by KF, both datasets were considered for analysis.

Initially, the two datasets were analysed separately to give similar shortening

rates of 219.2±27.27bp/PD (KF, p<0.0001) and 191.9±21.34bp/PD (RT,

p<0.0001), and comparison of the slopes revealed no significant difference

(p=0.4236). As such the data was considered as a whole to give the largest

insight across the longest timespan and repeating the regression analysis with

all data pooled gives a shortening rate of -213.7±16.75bp/PD (p<0.0001) at

12q.

Page 121: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

106

In the STELA data for XpYp it is apparent that a subpopulation of very long

telomeres is present in these cells (almost 20kb at PD 15.6). Due to their length,

this subpopulation is only amplifiable by STELA at later PDs, once shortening

has reduced their length. As these points are only present in the final three PDs

(although one long end appears to have been amplified at PD 10.5), it is not

possible to easily ascertain distinct groupings using 90th percentile data. XpYp

data was sensitive to these bands, as removing outliers by ROUT analysis in

Prism changed the shortening rate from -85.33±40.48bp/PD to -

171.7±38.38bp/PD (p<0.0001). It should be noted that apparent outliers at PD

Figure 3.2:10 Telomere length analysis on the T623S cell line a 12q STELA results after

subtraction of 544bp b Regression analysis on 12q STELA data excluding PD 21.3 (see main

text). Shortening rate is 225.3.±18.48bp/PD (p<0.0001). Dotted line indicates 95% confidence

band c XpYp STELA results after subtraction of 406bp, yellow dots indicate outliers identified by

ROUT analysis (Q=1%) d Regression analysis on XpYp STELA data excluding outliers. Shortening

rate is 171.7±38.38bp/PD (p<0.0001). Dotted line indicates 95% confidence band

Page 122: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

107

15.6 were not identified by the ROUT analysis with the parameters used, and

may have some bearing on the regression analysis, though the central position

of this PD in the dataset would most likely effect the elevation and y-intercept

of the slope, rather than the gradient itself. XpYp failed to amplify at PD 21.3,

indicating contamination or quality issues with this sample. As such, PD 21.3

was also excluded from the 12q dataset to give a rate of -225.3±18.48bp/PD.

3.2.2.6 E578V

The E578V line is derived from a patient with a comparatively very mild

phenotype. During cell culture, these cells behaved similarly to the NHDF line,

doubling approximately every 24 hours, and reached a high number of PDs

before reaching senescence. However, median telomere length was quite short

at even low PDs (4508bp median length at PD23.4 for 12q versus 6719bp and

6405bp at equivalent time points for 155-BR and NHDF), though this could be

attributable to normal inter-individual differences. Shortening at 12q displayed

a normal rate of –53.92±4.54bp/PD, whereas XpYp exhibited a faster attrition of

-120.9±10.76bp/PD, though this is potentially still within normal rates.

Unusually, the difference between 12q and XpYp was very large despite quite

well defined distributions at each time point in both cases resulting in very small

ranges and tight confidence bands in regression analyses. Repeating the

regression analysis after removal of the first time point resulted in slightly faster

shortening rates, as would be expected, with no significant difference from the

rates derived prior to sensitivity analysis. 12q gave a rate of -61.69 ±

6.768bp/PD, while XpYp increased to -155.8 ± 15.48bp/PD.

A summary of all shortening rates may be found in Table 3.3-3 (p124).

Page 123: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

108

3.3 Discussion

Statistically significant shortening rates were acquired for all cell lines except

NHDF at 12q and HGPS at both 12q and XpYp. Due to population dynamics, the

sensitivity of several cell line shortening rates was tested to establish the effects

of senescent cell detachment and inefficient amplification of large telomeres

and extremely significant rates of loss (p<0.0001) were obtained at all ends

except HGPS at XpYp. Trends suggest that telomere shortening rate is

accelerated in the cell lines derived from patients with more severe phenotypes.

Figure 3.2:11 Telomere length analysis on the E578V cell line a 12q STELA results after subtraction of

544bp b Regression analysis on 12q STELA data. Shortening rate is -53.92±4.538bp/PD (p<0.0001).

Dotted line indicates 95% confidence band c XpYp STELA results after subtraction of 406bp d

Regression analysis on XpYp STELA data. Shortening rate is -120.9±10.76bp/PD (p<0.0001). Dotted

line indicates 95% confidence band.

Page 124: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

109

3.3.1 Exploring the effects of population dynamics and long

molecule amplification efficiency on shortening rate

The increasing proportion of senescent cells in the population should result in a

biased and continual loss of those cells with shorter telomeres from the

population. With increased divisions, the proportion of cells containing shorter

telomeres should also increase. It appeared from the data collected that a

proportion of these cells was being lost, in some cases increasing the median

telomere length recorded by STELA at later PDs, impacting the overall observed

shortening rate. This apparent loss of cells also has implications for the accuracy

of PD counts. As the PD calculation is made based on the number of cells

counted when cells are plated out, if cells are lost after this time, and before the

next count, additional divisions by the dividing population would be

contributing to the PD count but go undetected. This would result in the

observed PD to be an underestimation of the actual mean PD rate. In extreme

instances, as was the case with HGPS, senescence levels were so high at late PDs

that after passaging, less cells were counted at the final time-point than in the

penultimate, resulting in a negative effect on cumulative PD at the final time-

point (Figure 3.2:2a). If growth rate is recorded more slowly than is the case,

faster telomere erosion could be seen as an artefact of this process. In other

words, if shortening rate is presumed constant, and the number of PDs between

time-points is underestimated, this would result in an observation of the same

amount of telomere shortening occurring over fewer PDs, giving an increase in

the observed shortening rate relative to PD. As the number of senescent cells

increases over time, the greater the loss of cells, potentially leading to a

continually increasing observed rate of shortening.

Two main factors indicated the loss of senescent cells from cell populations,

firstly, a reduction in the proportion of shorter telomere ends in length analysis

over time. Secondly, as a consistent gain of senescent cells would be anticipated

Page 125: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

110

over time, the relatively steady proportion of senescent cells (~20%) observed in

exponentially growing populations indicates that some senescent cells must be

detaching after replating. At this stage in proliferation it could be anticipated

that the same proportion of cells are entering senescence with each passage,

however, the magnitude of this proportion, as well as the proportion of

senescent cells detaching, cannot be inferred and would need to be determined

empirically, and may vary between cell lines.

Potentially, cell counting at the point of passaging could be followed by

counting cells that detach and become free in the media. It would be essential

to determine the proportion of senescent cells in parallel, as well as distinguish

between senescent and dead detached cells. It would be provident to use

multiple markers for senescence, as some markers are more reliable than others

and combinations of markers have been found to be highly informative (Lawless

et al., 2010). It would also need to be determined how quickly the viability of

detached senescent cells is affected, so as not to affect counts by

misrepresenting the proportion of senescent versus dead cells. FACS would

seem an ideal platform for distinguishing live from dead cells, however this

experiment would potentially have to be carried out on a huge scale in order to

provide the cell numbers required for FACS. However, if this issue could be

addressed, it would be possible to utilise DAPI or propidium iodide staining to

identify non-viable cells with sub-G1 DNA content, while simultaneously

identifying a senescence marker, such as ki67 negativity combined with high γ-

H2AX levels (Lawless et al., 2010). Once senescence levels in the population, and

the number of detached senescent cells was established, it would be possible to

derive a rate at which senescent cells detach.

By monitoring senescence levels in the patient and control lines, it is possible to

interpret population dynamics to some extent. The β-galactosidase assay was

carried out after re-plating cells, meaning a proportion of senescent cells could

Page 126: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

111

be lost prior to β-galactosidase assay, potentially causing the irregularities

observed in some cell line senescence data, and complicating interpretation of

the data. The discontinued contribution of senescent cells to the dividing

population further complicates accurately assigning the mean population

doubling value. PD was calculated using the following formula: [log10(final

count)- log10(initial count)]/log102. Senescent cells that have re-adhered are

included in both the initial and final count, although as time passes, a larger and

larger proportion of the cell population is not contributing to division

(senescent cells). If half the cell population is doubling normally, and half is

senescent, a count of 1 PD count would represent a mean of 2 PDs carried out

by the dividing population. As the proportion of senescent cells increases

beyond 50%, this effect would be amplified and cell populations will contain an

Figure 3.3:1 Illustration of population dynamics in an ageing cell line. Each column

represents observations each time the population is passaged, counted, and assayed for

β–galactosidase activity. Yellow background indicates cells that are included in cell

counts, grey indicates cells that are lost prior to counting. The percentage of senescent

cells, according to counting, is given at the bottom of the figure. Initially, cells with the

longest telomeres (top left), while included in counts, may not contribute to STELA

results. This does not affect the accuracy of PDs, but has implications for interpreting

STELA data. Over time, more cells enter senescence, ~50% of which are lost and not

counted. Dividing cells continue to divide, compensating for cells lost from counts. These

cells should also continue to enter senescence at the same rate, which may influence the

proportion of senescent cells.

Page 127: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

112

increasing proportion of cells that are misrepresented by the observed

cumulative PD. This is the likely cause of previous reports of observed increases

in telomere shortening rate, rather than a genuine increase. In order to

compensate for the discontinued contribution of senescent cells to the dividing

population, the senescent cell count may not be simply excluded from cell

counts, as with dead cells, because of their perpetuation in the population. To

rectify this, the difference between the proportion of senescent cells from one

passage to the next could be deducted at the following passage as this new

senescent population is not contributing to doubling, however the inaccuracies

of the β-galactosidase assay preclude this. As such, accounting for these

population dynamics when establishing PD in a primary cell population would

require a mathematical model to be devised which takes into account the

presence of senescent cells and their contribution to cell counts, but their

inability to divide. A basis for this model is illustrated in Figure 3.3:1.

In order to further dissect the effects of losing cells, and therefore a proportion

of telomeres, on the accuracy of telomere shortening rate, a set of example data

was created. A bimodal population is represented, with one subpopulation

consisting of a broad distribution of short to medium-length telomeres, and the

other a smaller subpopulation composed of long telomeres (Figure 3.3:2). For

the purposes of observing the effect on regression analysis, short telomeres

were allowed to fall below zero base pairs in the intial example data. In this

example, the telomeres of this population range from 400-12,000bp and

shorten at a rate of 100bp/PD, and a window of 15PDs is observed. In a scenario

where all telomere lengths are measurable, regression analysis in Graphpad

Prism 6 calculates the telomere shortening rate of -100bp/PD as significant,

with an error of ±43.99 (Example A1, Figure 3.3:2a). Taking into account that

telomeres cannot shorten perpetually, a lower threshold of 600bp was applied,

and any example telomeres shortening beyond this were set at 600bp (Example

A2, Figure 3.3:2b). This had a small but observable effect on the shortening rate,

Page 128: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

113

Figure 3.3:2 Example dataset A a Example data containing two subpopulations. This data

was manipulated in figures b-f in order to predict the effects of excluding various

subpopulations, see main text and Table 3.3-1 for more detail g regression comparison of

each scenario from figures b-f

Page 129: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

114

reducing it to -89.48±42.83 bp/PD. This rate remained significantly different

from zero. To compare the effect of the loss of long telomeres due to

amplification failure, an upper threshold was set at 10kb. Ends exceeding this

length were removed from the dataset, causing a dramatic shift in shortening

rate to -22.89±37.40 bp/PD (Example A3, Figure 3.3:2c). Combining these

scenarios results in a shortening rate of -34.13±38.94 bp/PD (Example A4,

Figure 3.3:2d). In order to reflect the loss of short telomeres from the

population, as was evident from the STELA data, roughly 50% of the “senescent”

population (the 600bp telomeres) was removed. This combination of loss of

long and short telomeres resulted in the least accurate shortening rate

(4.435±37.49 bp/PD, Example A5, Figure 3.3:2e). As the results from Examples 2

and 3 suggested that the effect of losing longer ends has more serious

consequences for the accuracy of the regression analysis, a final scenario was

explored. Conditions were repeated as for Example A5 (i.e. telomeres will not

shorten below 600bp and approximately 50% of these telomeres are lost at

each time point), but all long ends were included (Example A6, Figure 3.3:2f).

This resulted in a shortening rate of -60.90±43.24 bp/PD. A comparison of

regression lines may be found in Figure 3.3:2g, and a summary in Table 3.3-1.

This analysis points to the loss of long telomeres as having the greatest impact

on shortening rate accuracy. In order to exclude the possibility that this was due

to the bimodal distribution being skewed, with fewer long telomeres, the

process was repeated with another theoretical population, containing a small

subpopulation of short telomeres and a large population of medium-length to

long telomeres with the same shortening rate (Figure 3.3:3a, Example 1). This

new distribution has the same range and the same error in shortening rate

(±43.99bp/PD). Using the same parameters as above, the “senescent”

population was assigned to telomeres under 600bp, and ~50% of these were

removed from the analysis at each time point. This results in only a slight drop

in accuracy when compared to the equivalent Example A6 (-55.51±41.52bp/PD).

Page 130: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

115

Figure 3.3:3 Example dataset B a example data containing two subpopulatipns, skewed towards

longer telomere. This data was manipulated in figures b to predict the effects of excluding

senescent cells from a population and in c to predict the effects of excluding long telomeres,

reflecting a loss of PCR efficiency of longer molecules d regression comparison of a-c

Table 3.3-1 Summary table of the exclusions from the example data, including shortening rate

and p values as derived from Graphpad Prism 6.

Page 131: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

116

As would be expected, removing long telomeres (>10kb) from this population

has a slightly less deleterious effect on the regression analysis than its

equivalent Example A3 (-46.96±43.10bp/PD).

These analyses confirm that a loss of senescent cells from the population, as

well as PCR amplification failure of long telomeres, will affect the telomere

shortening rate derived from STELA data. While a failure to amplify larger

telomeres remains a PCR-specific issue, any technique used to establish

telomere shortening rate will be affected by the loss of senescent cells. As

anticipated, all the scenarios tested lead to a reduction in telomere shortening

rate, suggesting that any published rates to date may be underestimated. It

would seem that the non-amplification of long telomeres has a greater

influence on shortening rate than the effects caused by senescent cell loss.

Deviation from the actual shortening rate, and the significance of regression

analysis are more greatly affected when long telomeres drop-out, as is slope

elevation. This can be seen by direct comparison of Examples A5 and A6. This

effect would seem to be amplified by the distance of the longest telomeres

from the median and the main distribution of lengths, as a dataset with a long

subpopulation is affected to a greater degree than a dataset that contains long

telomeres within the main distribution (Example A3 versus Example B3). A

subpopulation of short telomeres does not affect regression analysis to the

same extent when the effects of senescence (i.e. telomeres do not shorten

below a set length) are taken into account (Example B2 versus A6), and this may

be due to the retention of a proportion of the telomeres. Still, the presence of

isolated long and short subpopulations had the most impact on shortening rate,

as was highlighted by the XpYp data for T623S. Extreme care should therefore

be taken when interpreting any dataset that contains an isolated short or long

subpopulation. Excluding T623S at XpYp, none of the cell lines appeared to

contain subpopulations that may have been susceptible to the effect described

Page 132: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

117

above. Therefore, in this study, the effect of losing senescent cells may not have

resulted in a large impact on shortening rate, however, the effect of senescent

cell detachment on PD accuracy could be affecting the accuracy of shortening

rate to an unknown degree. However, with the exception of the NHDF line

which did not approach senescence, all cell lines experienced an increase in the

proportion of senescent cells, which could minimise this effect. Additionally, any

effect on shortening rate would become more apparent towards the end of the

cell lines’ proliferative lifespan. This possibility is explored below.

3.3.2 Telomere shortening rate over lifespan

A drop-off in telomere length towards the end of replicative lifespan has been

previously recorded in fibroblasts (Britt-Compton et al., 2009b, 2006). However,

if, towards the end of proliferative lifespan, PD rate is recorded as slower than is

the case, this increase in shortening rate will be observed in error. In order to

identify changes in shortening rate over the lifespan of each cell line, the

median values of each time-point were plotted (Figure 3.3:4a-f), and the rate of

change between each median value was calculated (Table 3.3-2). Telomere

length and cell division rate were also plotted over time in order to observe any

interactions between these two factors (Figure 3.3:4g-l). Figure 3.3:5 is provided

as a reference for senescence levels at the time points where DNA was

harvested for STELA.

The 155-BR line has a particularly low r2 value when a regression line is derived

from the median values for both 12q and XpYp (Table 3.3-2). At several time

points, the median varies greatly from the trend, causing difficulty interpreting

rates of change, and as such does not provide reliable information. The NHDF

line conforms well to a linear model, with high r2 values at XpYp and at 12q

when using the 10th percentile data. Taking the change in rate between each

time-point shows a steady rate that fluctuates marginally around the rate

established by regression of the raw data. The NHDF data was collected while

Page 133: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

118

the cell line was still in its exponential growth phase, and as NHDF maintained a

steady level of senescent cells (Figure 3.2:3a), the rate of telomere loss would

not be expected to be affected by counting inaccuracies caused by senescent

cell loss. This consistent growth rate is confirmed by the high r2 value derived by

regression analysis of growth rate over time (r2=0.9990 (Figure 3.3:4g)). The

slight decrease in rate between the final two time-points at XpYp could

therefore be due to experimental fluctuation.

The patient lines show varying degrees of linearity. Due to the short period of

time between points for HGPS it is not feasible to analyse the intervening rates.

Slow initial rates of shortening in most cell lines are most likely attributed to

amplification failure of the longest telomeres at the first time-point. This ceiling

on amplification limited the detection of telomeres that exceed approximately

15kb for 12q and between 18-20kb for XpYp. The T623S data demonstrates this

at the first two rates for XpYp, and the first rate for 12q. After this time, the rates

do not vary greatly. Disregarding the first value for both telomere ends of

E578V, a similar linearity is observed, however the shortening rate appears to

decrease between the final time-points. Conversely, the RD shortening rate

increases at the final rate-change at all three ends tested, suggesting that the

efficiency of senescent cell re-adherence may be poorer in RD cells, consistent

with the severe nature of the cellular phenotype. Comparing the time difference

between the final three time-points for RD shows a marked decrease in

doubling rate. 32 days passed between the final 3.8 PDs, in contrast to the

previous 5.1 PDs which took place over only 14 days. High levels of senescent

cells at this final stage could be skewing counting and creating an artificial

observed increase in shortening rate as has been discussed. As the shortening

rate is more constant when plotted against time rather than PD (r2 values for

regression on final 3 time points increase from: 12q=0.9658, XpYp= 0.9002,

17p= 0.9649 to: 12q=0.9856, XpYp=0.998, 17p=0.9861), doubling time could

directly impact the rate of telomere shortening (see next section).

Page 134: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

119

Figure 3.3:4

Shortening rates of

each cell line over

time. Values given

for NHDF 12q are

the 10th percentile

data. Graphs a-f

show the median

telomere length

(derived from

complete datasets

prior to sensitivity

testing) at each

time-point, plotted

against PD. Dotted

lines indicate range.

Graphs g-l show

median (derived as

above) telomere

length and PD

plotted against

time (days). Dotted

lines indicate range.

Page 135: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

120

Figure 3.3:5 Growth rates as determined by cell counting and senescence levels as

determined by β-galactosidase assay. Time points where genomic DNA was derived and

used as a template in STELA reactions are indicated by pink dots for ease of interpretation.

Table 3.3-2 Rate of shortening and r2 values for shortening rate of each cell line, as derived from a

regression based on the median and IQR of telomere length at each time-point. Slope between

median time-points gives exact shortening rates between median lengths at each sequential time-

point. Figures or slopes are negative unless otherwise shown. Sensitivity corrected shortening rates

derived in section 3.2.2 are also given (final column). *10th percentile data

Page 136: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

121

It has been shown that the rate of telomere erosion is independent of the

length of the telomere (Baird et al., 2003). However, at 12q in the RD line, a

subpopulation of long telomeres is distinguishable, roughly correlating with the

75th percentile, and appears to be shortening more quickly than the main body

of telomeres (Figure 3.3:6). This was confirmed by normalising the data of each

time point according to its median, so that they may be overlaid on a histogram

plot (Figure 3.3:7a&b). This effect could be due to allelic differences between

the two subpopulations and so regression analysis of the shortening rate of all

cell lines was carried out.

Median telomere length of each line, at each telomere, was plotted against the

sensitivity-corrected shortening rate. As shortening rate for HGPS XpYp was not

reliable, this data was excluded from this analysis. Regression analysis reveals a

statistically significant, if minor, effect of telomere length on shortening rate

(p=0.0079 (Figure 3.3:7c)). Direct comparison of XpYp and 12q shows a non-

Figure 3.3:6 Differential shortening of

subpopulations in RD at 12q a Regression

analysis of the upper quartile versus the

remaining data points. Shortening rates are

shown on the figure. Slopes are significantly

different (p=0.0059). UQ=upper quartile b

Smoothed histogram of 12q data, the

median of each time point has been set to

zero to allow comparison c Blow-up of the

longer telomere subpopulation. Over time

this subpopulation peak shifts towards the

median, indicating a faster rate of telomere

shortening.

Page 137: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

122

significant trend for XpYp to shorten more rapidly, though if longer telomere

lengths result in faster shortening, this may be explained by the longer median

telomere lengths in this group (Figure 3.3:7d). A similar difference was observed

in the comparison of patient and control cells (Figure 3.3:7e), suggesting that

12q and/or patient telomeres are less susceptible to this length-dependent rate

change. If long telomeres shorten more quickly this would cause a gradual

decrease over time in the rate of shortening and could explain the gradual

decrease in rate observed at the final point in the E578V line. If longer

telomeres are in fact shortening more rapidly, this suggests that there is an

inherit mechanism by which telomeres shorten gradually and processively at

different rates. This mechanism may be the source of natural variation in

telomere erosion rates, and potentially the differences between control and

patient rates.

Figure 3.3:7 Regression analyses of

shortening rates again median telomere

length a Regression analysis of median

telomere length over shortening rate of each

telomere analysed, excluding HGPS XpYp

(r2=0.5224, p=0.0079) b Comparison of

shortening rates versus median telomere

length regression between 12q (r2= 0.7748,

p=0.0488) and XpYp (r2=0.7659, p=0.0224) c

Comparison of shortening rates between

patient (r2=0.9731, p=0.0136) and control

lines (r2=0.5900, p=0.0260)

Page 138: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

123

3.3.3 Cell line differences in telomere shortening rate

A summary of the shortening rates at 12q and XpYp for all cell lines tested is

given in Table 3.3-3a and Figure 3.3:8. Comparison of all shortening rates was

carried out in Graphpad Prism6 and the resultant p values are given in Table

3.3-3b-e. Initial observations show considerably less variation in telomere

shortening rate between cell lines at XyYp than 12q. The shortening rates of

both control lines were significantly slower than T623S and RD at 12q (Table

3.3-3) and the data was not sensitive, having significantly faster shortening rates

at 12q both before and after sensitivity testing. Previous reports for fibroblast

shortening rate record a loss of between ~30-85bp/PD (Allsopp et al., 1992;

Counter et al., 1992; Harley et al., 1990; Levy et al., 1992), with some studies

reporting up to ~160bp/PD (Baird et al., 2003; Vaziri et al., 1993). The RD rate (-

210.5 ±24.67 bp/PD), T623S (-225.3 ± 18.48) and provisional HGPS (-390.7

bp/PD) shortening rates all exceed normal rates.

The shortening rate data for HGPS, although not significant in itself, was found

to be significantly different from the NHDF control line and the E578V line, but

not RD or T623S at 12q after sensitivity testing (Table 3.3-3). At 12q, the E578V

line did not differ significantly from the normal cell lines, but did differ from the

other patient lines significantly in all cases except HGPS prior to sensitivity

analysis. Sensitivity-corrected testing of XpYp reveals a significant difference

between the E578V shortening rate and NHDF. However, this rate (-155.8 ±

15.48 bp/PD) is still within normal parameters. This similarity to normal lines

reflects the milder nature of the symptoms accompanying E578V and its normal

rate of cell division in culture, and could indicate that any effect on telomere

shortening due to lamin A mutation is either minimal or absent in this line.

The failure to record a significant rate of telomere shortening in the HGPS line

was due to the short window in which samples were available. Crucially, the

aberrantly high shortening rates recorded for the RD line are observed between

Page 139: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

124

Table 3.3-3 a Shortening rates at 12q and XpYp fpr each cell line, before and after sensitivity analysis.

The sex of each individual is indicated as Male, Female or Unknown in brackets after the cell line name.

Non-significant p values are given in red *10th percentile data ** Pooled from 10th and 90th percentile

data (p=0.0264 and p=0.0026 respectively) *** 90th percentile data. Letters in brackets after cell lines

indicate Male or Female b-e p values from regression comparisons carried out pair-wise across all cell

lines. Values are shown for 12q b before and c after sensitivity testing and XpYp d before and e after

sensitivity testing

Page 140: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

125

PD 11.4 and 16.5 for XpYp and 17p, and from PD 7.5 at 12q (Figure 3.3:4d). At

these lower PDs, the cell population is dividing rapidly and senescence levels

are relatively low (Figure 3.3:5f), which would minimise the effects of cell loss on

artificially increasing the observed telomere shortening rate, thus confirming a

genuine accelerated telomere shortening rate in this line. Accelerated

shortening is also observed at early PDs in the T623S line while senescence

levels are minimal in. This is apparent at 12q, while confirmation of this pattern

at XpYp is made difficult by the “drop-in” subpopulation with long telomeres

(Figure 3.3:4e and Figure 3.3:5d). These rates from early time points are consistent

with the hypothesis that an inherent mechanism of increased telomere attrition

Figure 3.3:8 Comparison of telomere shortening rates in patient and control cell lines,

with error. Shortening rates of patient and control cell lines at 12q a before and b after

sensitivity testing (HGPS: circles indicate the rate of shortening of the 90th and 10th

percentiles and error bars here indicate the combined ranges of the 90th and 10th

percentile rates) and at XpYp c before and d after sensitivity testing.

Page 141: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

126

at replication in these lines. As the T623S cells are derived from a patient with a

disorder comparable to classical HGPS, except for slower onset and disease

progression rate, the similarity of the molecular mechanisms behind both

disorders, data from other studies, and the sensitivity tested HGPS data at 12q,

it is likely that the rate of HGPS shortening is comparable to or faster than

T623S and RD.

At XpYp, in a comparison of severe phenotypes and NHDF, a significant

difference was only observed between the NHDF line and RD line (Table 3.3-3c),

despite faster shortening rates than would expected in normal fibroblasts in

T623S (-171.7 ± 38.38 bp/PD), and implied by sensitivity-corrected HGPS data (-

393.8±170.7 bp/PD). After sensitivity testing, the T623S shortening rate was not

significantly different from the NHDF rate of -90.98 ± 18.35 bp/PD despite no

overlap in the ranges of each line (p=0.05582).

The most unexpected result is that of 155-BR at XpYp, which exhibits a

shortening rate faster than would be expected of a normal fibroblast line

(194.3±29.28bp/PD) between PDs 18.4 and 28.9. This unusually elevated erosion

rate could be attributable to loss of senescent cells, and perhaps the longer

telomeres at XpYp in comparison to 12q contribute to this increase.

Alternatively, viewing the similarities between the 155-BR line and the severe-

phenotype patient cell growth rates, it is possible that long division times may

exact an effect on telomere length. Previous work has shown that slowly

dividing clonally-derived fibroblast populations have exhibited accelerated

shortening rates compared to their more quickly dividing counterparts, and this

was attributed to an acceleration of telomere shortening of cells approaching

senescence (Britt-Compton et al., 2006). However, as the XpYp shortening rate

was relatively stable at earlier PDs when senescence levels were low, this

suggests that slowly growing populations may in fact incur accelerated

telomere shortening. It would be possible to determine if this is the case by

Page 142: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

127

imposing growth restrictions on a normal line and comparing shortening rate to

a control. This could be achieved by chemical inhibition, however it would be

crucial not to expose the cells to any toxic substance that may affect the

integrity of the telomere. Although potentially toxic to a certain degree, media

starvation could provide a low-impact alternative to chemical arrests.

The sex-related nature of the XpYp causes other complications when

interpreting the XpYp shortening rates. X-inactivation in females results in an

increase of telomere shortening rate at the inactivated X (Xi) (Surrallés et al.,

1999). This will produce a direct effect on telomere length distribution in

females, in this data set E778V and T623S, and could explain the faster

shortening rate of the E578V at XpYp versus 12q, however, the T623S

shortening rate at XpYp is slower than at 12q, though not significantly

(p=0.5734). However, significant increases in shortening rate at XpYp versus 12q

may be observed for both NHDF and 155-BR, despite both being male. X-

inactivation is marked by condensed chromatin across the Xi. Although the X

chromosome is not relocated within the nuclei of HGPS cells (Mehta et al.,

2011), patients lose histone 3 trimethylation on lysine residue 27 (H3K27me3), a

marker for facultative chromatin on the Xi (Shumaker et al., 2006). This may

Figure 3.3:9 Last recorded 12q telomere length for each line at a 12q and b XpYp. PD at

final measurement: NHDF=28.0 (not senescent), 155-BR=28.9 E578V=46.5, T623S=22.1,

HGPS=20.3, RD=20.3

Page 143: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

128

accelerate or influence the rate of shortening of Xi in patient cells in an

unpredictable manner, and the 12q data could be a more reliable source of

information than the XpYp data. Expression of sex-related genes could also be

altered in patient cells.

Finally, the last recordable median telomere length in each cell line was

compared for 12q and XpYp (Figure 3.3:9). At both ends there is a trend for cell

lines with the most severe phenotypes to have a higher median length at the

point of senescence. However, the range of telomere lengths of the RD line at

senescence included very short telomeres. The distribution of telomere length

in RD is also very similar to that of control line 155-BR at both ends. Given this,

and the natural variation in telomere length distribution in humans, it is possible

that this reflects natural variation not associated with a senescence-induction

mechanism.

3.3.4 Potential sources for increased telomere shortening in

laminopathy patient cells

Because the 155-BR cell line did not behave as a normal cell line, using NHDF as

a controlled comparison, and in comparison to previously published telomere

shortening rates relative to PD (Allsopp et al., 1992; Baird et al., 2003; Counter et

al., 1992; Harley et al., 1990; Levy et al., 1992; Vaziri et al., 1993), it would appear

that the T623S and RD cell lines are shortening at an accelerated rate. This is

especially clear at 12q, which is more representative of the autosomal genome.

It is most likely that telomeres of the HGPS line are also shortening more

rapidly. The patient cell lines demonstrate a correlation between increased

senescence and short telomeres confirming that patient fibroblasts have a

shorter lifespan than normal cells as a direct consequence of accelerated

telomere shortening. This is in agreement with a previous study in which

Page 144: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

129

expression of the telomerase subunit TERT abolished senescence in HGPS and

progerin-expressing cells (Benson et al., 2010).

The major commonality between the T623S, HGPS and RD lines is the

accumulation of farnesylated forms of lamin A. Farnesylated lamin A aggregates

at the periphery, forming a thickened lamina and dysmorphic nuclear

membrane. Farnesylated lamin A is also associated with slow cell cycle

progression, DNA damage accumulation and signalling and chromatin

organisation disruption. Therefore, increased telomere shortening may be an

effect of farnesylation. Given that E578V exhibits limited nuclear blebbing in the

absence of farnesylated lamin A (Csoka, 2004; Toth et al., 2005), and a relatively

high rate of shortening at XpYp, it is possible that accumulation of lamin A at

the nuclear periphery and/or the resultant physical aberrations could be

influencing telomere shortening. However, as shortening rates at both 12q and

XpYp were both within the normal range of telomere shortening, it would seem

that retention of the farnesyl group is necessary for accelerated telomere

shortening.

DNA damage levels are highly elevated in HGPS and other laminopathy and

laminopathy-model cells (Csoka, 2004; Gonzalez-Suarez et al., 2009; Li and Zou,

2005; Liu et al., 2008, 2006), and current understanding could attribute this to

accelerated proteasomal degradation of DNA damage factor 53BP1 (Gonzalez-

Suarez et al., 2011). The repercussive slow response of 53BP1 to recruit to y-

H2AX foci provides one explanation for the slow cell cycle progression of HGPS

cells (Liu et al., 2005). Additionally, the PCNA replication factor is re-localised

upon truncation of lamin A (Spann et al., 1997), increasing the likelihood of fork

stalls, and increased fork-stalling has been recorded in Lmna-/- MEFs (Singh et

al., 2013). In the presence of progerin, telomeres exhibit increased γ-H2AX

damage-signalling (Decker et al., 2009). If damage is concentrated at telomeres,

failure to repair that damage could result in loss of telomeric repeats. However,

Page 145: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

130

one would expect this damage to be distributed randomly throughout

telomeric sequences. DNA breaks and subsequent loss of DNA would be

initiated at various points along the telomere, resulting in telomeres of a range

of sizes and potentially telomere fusions and other aberrations. STELA amplifies

telomeres of all sizes under a certain range, and sudden large deletions are

readily detectable (Benson et al., 2010). None of the patient lines show signs of

such jumps in product size on STELA blots, suggesting that sudden loss is not a

cause of increased shortening. It is possible that some forms of damage could

result in the loss of the 3’-overhang, the substrate required for STELA PCR.

Consequently, STELA may not be able to identify all damaged telomeres.

Using Q-FISH, Benson et al. (2010) found chromosomal aberrations at higher

levels at telomeres in HGPS cells compared to controls, consisting of a wide

variety of abnormalities, including sister telomere fusions and losses, telomere

doublets, chromosomal breaks and extra-chromosomal telomeres observed at

only 3.5% of HGPS chromosomes (approximately three anomalies per cell)

versus 0.8% of controls (approximately one anomaly per cell) in accordance with

findings in Lmna-/- MEFs, despite an absence of sister-chromatid exchange

(Gonzalez-Suarez et al., 2009). This marginal difference suggests that although

repair of telomeric lesions is impeded in HGPS cells, it is unlikely to impact

telomere shortening rate to the degree observed here. It would seem that cells

are undergoing a continually increased loss of telomeric DNA. Additionally, a

previous study using STELA has identified a frequency of large-scale or

catastrophic deletion events of ~3% in normal cells (Baird et al., 2003),

suggesting that STELA may in fact be a more sensitive analysis for the

identification of lesions. Although it remains possible that a specific route of

DNA damage is activated in HGPS cells that nullifies the effectiveness of STELA,

the variety of aberrations recorded by Benson et al. suggest that this is not the

case.

Page 146: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

131

HGPS cells exhibited a large distribution of telomere lengths, in accordance with

previous findings (Benson et al., 2010), and it is likely that a proportion of the

shorter telomeres are the result of catastrophic events. In normal cells, short

telomeres that have suffered major deletions do not perpetuate in the

population, as their numbers do not accumulate over time (Baird et al., 2003). In

the case of the HGPS results, it would be difficult to separate this fraction of

small telomeres from the senescent population, even if a longer divisional

history could be examined, and so it is not possible to identify whether these

telomeres are lost from the population, or contributing to accelerated

senescence. By observing the smallest telomeres of the remaining patient cell

lines, and the distribution of lengths at each time point, neither appreciably

alters in any way over time other than has been already described. This further

supports the notion that, while catastrophic events may occur at higher rates in

patient cells, the effect of this is minimal in terms of a long-term effect on

telomere length.

There are three possible scenarios for the fate of a telomere that has been

damaged. Firstly, damage may be repaired, and resultant shorter telomeres that

acquire or retain an overhang would remain detectable by STELA, but were not

observed. Damage resulting in loss of overhangs is not detectable through this

method, but if overhangs are left unrepaired, this may initiate a second

scenario, in which damage at telomeres or anywhere in the genome cannot be

repaired and apoptosis is initated, however a reduction in viability was not

observed in the patient lines according to Trypan Blue analysis. An undetectable

effect on viability during proliferation could be attributed to only a small

number of cells exhibiting fatal damage, and as such probably would not

impact telomere shortening rate to any detectable degree. Thirdly, prolonged

DNA damage signalling would activate entry into damage-induced senescence.

Although this may not be accurately inferred from the data available, the

consistent distribution of telomere lengths throughout the STELA analyses,

Page 147: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

132

most visible in instances where discreet subpopulations exist, as well as the size

and distribution of telomere at senescence do not suggest a strong tendency to

enter premature cell arrest. Although a combination of these minor effects may

potentially be creating the observed effect on shortening rate, another

mechanism is more likely at work which elevates the rate of continual telomere

shortening in patient cells. An ongoing, increased erosion fits well with the

STELA data, and it is possible that this may be a result of slowed cell cycle

progression (see previous section).

3.3.5 Summary

Five progeroid laminopathy fibroblast lines were grown in culture to

senescence, four of which were carried forward for a telomere shortening rate

analysis using data collected from STELA and cell division rates as derived from

cell counts. In parallel, three control fibroblast lines were cultured, and telomere

shortening analysis was carried out on two of these lines, only one of which

reached senescence. This cell line (155-BR) grew slowly, resembling the

laminopathy lines, although one patient line (E578V) grew at the same rate as

the second normal line (NHDF), which grew at a normal rate (~1PD/day). E578V

exhibited a normal shortening rate of ~60bp/PD at 12q but a borderline rate of

~155bp/PD at XpYp, perhaps indicative of the mild nature of symptoms

associated with the E578V mutation, but could be within normal inter-individual

variance in shortening rate. The RD cell line exhibited a significantly elevated

rate of telomere shortening at both XpYp and 12q compared to controls. T623S

also had a significantly elevated shortening rate at 12q compared to controls,

however, despite the rapid rate of shortening at XpYp (~170bp/PD), failed to

reach significance compared to controls. A very limited number of PDs was

analysed for the HGPS line, causing difficulty in deriving a shortening rate,

however results implied an elevated rate, similar to that of T623S and RD.

Page 148: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

133

Due to the chromosomal specificity and high resolution of STELA, it was

possible to perform in-depth analyses of the changing telomere population in a

dividing primary cell line. Loss of senescent cells from the population was

confirmed and found to effect the accuracy of regression analysis to derive

telomere shortening rate by affecting the representativeness of PD counts. PCR

amplification failure of extremely long telomeres could have a major effect on

shortening rate accuracy if long telomere subpopulations drop in during the

course of shortening rate experiments. These two factors have several

implications for deriving an accurate telomere shortening rate, and for any

time-course experiments carried out in primary lines that rely upon accurate PD

counts.

In the datasets presented here, only T623S and NHDF seemed dramatically

affected by amplification failure at XpYp and 12q respectively. Also, the effect of

senescent cell loss proved to be minimal, implying that loss of senescent cells,

while an issue, did not appear to have had a major effect on the calculated

shortening rates.

Page 149: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

134

Chapter 4 Exploring mechanisms behind accelerated

telomeres shortening in laminopathy patient cells

4.1 Background

The STELA data demonstrates an accelerated telomere shortening rate in

patient lines containing mutations that result in lamin A forms that retain the

farnesyl modification. As none of the patient lines appeared to exhibit sudden

loss of large sections of telomeric DNA, it is most likely that the acceleration is

caused by a processive and continual increased loss of telomere repeats.

Therefore, lagging strand synthesis and post-replication end-processing are

potential sources of increased telomere loss. Control of telomere overhang

processing is heavily regulated by the cell cycle both in yeast (Frank et al., 2006;

Vodenicharov and Wellinger, 2006) and humans (Dai et al., 2010). Dai et al.

(2010) discovered that human overhangs increase in size during the cell cycle,

peaking at late S and early G2 phases, when, they concluded, the 5’ strand is

extended to reduce overhang length. If telomere end processing is cell cycle

dependent, it is possible that extended stays at the various checkpoints could

result in abnormally-processed telomere overhangs. As the length of the 5’ end

accompanying the 3’ overhang dictates the maximum replicable length of one

of the two daughter strands of any replicated telomere, overhang length drives

telomere shortening in a proportional manner (Huffman et al., 2000). If

processing is occurring in an uncontrolled manner and forming longer

overhangs in patient cells, the shorter length of the 5’ strand will drive

accelerated telomere attrition.

Sfier and de Lange found that in the absence of 53BP1, overhangs were ten

times longer in MEF cells (Sfeir and de Lange, 2012). Upon serial culturing of

53BP1 null MEFs, Q-FISH revealed a 20-30% reduction in telomere length after

Page 150: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

135

only five days. 53BP1 is present at lower levels in lamin null MEFs (Gonzalez-

Suarez et al., 2009) and is slow to be recruited to sites of DNA damage in HGPS

fibroblasts, as well as Zmpste24-/- MEFs (Liu et al., 2005). Mutated lamin A is able

to up-regulate CTSL activity (a major endosomal/lysosomal pathway

component), which results in 53BP1 export from the nucleus and its subsequent

proteosomal degradation (Gonzalez-Suarez et al., 2011). If the absence of 53BP1

results in dramatically longer overhangs, it is quite feasible that the altered

biology of 53BP1 in patient cells could lead to increased overhang length, and

the accompanying telomere shortening acceleration.

Although several approaches have been published describing quantification of

telomere overhangs, most of these techniques are extremely labour-intensive

and require specialist equipment. As the telomeric overhang makes up a

miniscule fraction of total genomic DNA, quantification is extremely difficult. So

limiting is the amount of DNA that is to be measured, many approaches aim

only to establish a relative volume measurement of overhang DNA. That is, the

amount of telomeric overhang DNA may be quantified relative to the amount of

telomeric or genomic DNA, but the overhang lengths are unknown. The

methods of in-gel hybridisation, in which terminal restriction fragments are run

on a native gel and hybridised to a telomeric probe in order to detect single

stranded telomeric DNA (Dionne and Wellinger, 1996; McElligott and Wellinger,

1997), and the G-tail telomere hybridization protection assay (HPA) which

anneals chemiluminescent probes in native conditions to the overhang for

direct quantification (Tahara et al., 2005), offer such results. HPA was developed

as a high-throughput alternative to the overhang protection assay (OPA). In

OPA, overhangs are coated in UP1 and gp32 proteins which protect the

overhangs from restriction digest by DNaseI, before proteinase K treatment and

native gel analysis and size quantification (Chai et al., 2005). In a similar

approach, oligonucleotides may be ligated to the overhang in the telomere

nucleotide ligation assay (T-OLA) (Cimino-Reale et al., 2001). The method

Page 151: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

136

chosen to carry forward for analysis of overhangs in patient cells, the duplex-

specific nuclease (DSN) method, is less reliant on stringency and specialist

reagents than these methods. The method exploits the nuclease activity of the

DSN enzyme, derived from the Red King crab. This enzyme shows high

specificity for double-stranded DNA, and extremely low activity on single

stranded DNA (Shagin et al., 2002). Upon complete digestion with DSN,

remaining double-stranded products have been reported to be of 6 nucleotides

or less, (Shagin et al., 2002). The undigested single-stranded DNA may be run

on a native gel and probed for G-rich repeats.

As a negative control for the DSN digest, an ExoI digest is set up in parallel. The

3’->5’ activity of ExoI should fully digest telomeric overhangs. Products are run

on a denaturing polyacrylamide gel, Southern blotted and detected by

radiolabelled CCCTAA(n) probe specific for the G-rich telomere repeats. Products

in the experimental sample appear as a smear which may be normalised to the

control lane. The experimental process is outlined in Figure 4.1:1, and the

quantification method is detailed in Materials and Methods section 2.11.3.

Figure 4.1:1 DSN digests are carried out on genomic DNA derived from cultured cells a The

experimental sample is digested with DSN alone, leaving the 3’ overhang intact b Control

sample is incubated with ExoI prior to DSN digestion. Exonuclease activity of ExoI digests 3’

overhang, releasing deoxyribonucleoside 5'-monophosphates c Experimental and control

sample are run side-by-side with a size marker on a denaturing polyacrylamide gel, Southern

blotted, and hybridised with radiolabelled C-rich probe (see section 2.9.1.1).

Page 152: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

137

4.2 Results

4.2.1 Initial observations of DSN method

In order to establish the method and optimise conditions, the experiment was

carried out on control DNA from HT1080 cell-line stocks. Parallel experimental

and control digests were run side-by-side. Smears were observed in the

experimental lane, and absent from the control lane when probing with a C-rich

labelled oligo (Figure 4.2:1a). Preliminary experiments also showed no signal

when probed with a G-rich oligo (Figure 4.2:1c), indicating that single-stranded

Figure 4.2:1 Preliminary results from DSN overhang assay. A DSN digest and a DSN + ExoI

digest were carried out on HT1080 DNA (5µg/reaction), run on a polyacrylamide denaturing gel

and transferred to a positively charged nylon membrane. Marker sizes are indicated in nt on the

left-hand side of the figure. The membrane was probed with a CCCTAA(n) radioactively end-

labelled oligonucleotide (TelC, see Table 2.9-1) and exposed to a Phosphor Imaging screen (a).

The probe was then removed and the membrane re-exposed to confirm probe removla (b).

Finally, the membrane was re-hybridised to a TTAGGG(n) end-labelled oligonucleotide

oligonucleotide (G1, see Table 2.9-1) to demonstrate overhang specificity (c).

Page 153: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

138

DNA in the experimental digest consisted of ExoI-sensitive single stranded DNA

composed of G-rich 3’overhang.

Standardising to a commercially available size marker, quantification was carried

out as previously described (Chai et al., 2005; Zhao et al., 2008) (see section

2.11.3). However, several problems with quantification arose. As the smallest

markers of commercially available ladders are either above 20nt or are so small

as to result in undetectable signal after hybridisation, extrapolation of the low

end of the size range was necessary. As such, a method for quantification of

small molecules was devised to ensure accurate sizing of the smallest fragments

(see below).

4.2.2 Troubleshooting

It was necessary to develop low molecular markers beyond the range of

commercially available ladders and so several oligos consisting of differing

numbers of G-rich telomere repeats were generated. The possibility of deriving

a ssDNA ladder in the lab was explored but oligonucleotides containing G-rich

telomere sequence were chosen to reflect the size and migration of overhang

fragments (sequence rich in cytosine runs considerably faster than guanine-rich

sequence in polyacrylamide (Frank and Köster, 1979)), and following a personal

communication with Professor Woody Wright. However, initial electrophoresis

of these oligonucleotides resulted in unexpected sizing relative to the LMW

marker. In order to correct this, high-performance liquid chromatography

(HPLC) purified oligonucleotides were purchased, however the results were

similar (Figure 4.2:2), and the smallest oligonucleotide (G4, 12nt) did not bind

the radiolabelled probe efficiently enough to emit a detectable signal.

A more fundamental issue arose at the gel electrophoresis stage. It was

apparent from the loading buffer dye front that the migration of the digested

samples was retarded in comparison to the size markers (Figure 4.2:3a&b). This

Page 154: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

139

caused major problems with quantification as the relative migration of markers

appeared to vary with each gel electrophoresis, and it was clear that the sizes

derived were not representative. In order to identify and resolve the cause of

the migration problems, several additives were supplemented to a commercial

marker, including inactivated enzymes, glycerol and BSA equivalent to that

added to digest reactions (Figure 4.2:3d). SDS was added to one digest in an

attempt to correct the migration issues, however this was extremely disruptive

of migration. It was apparent from agarose gel electrophoresis that small

molecules were abundant in DSN digests (Figure 4.2:3c). In order to confirm

whether these small fragments were the source of the migration issues, λ and Φ

markers were added to digested and undigested genomic DNA (Figure 4.2:3e).

When markers were added to undigested DNA, the marker bands migrated

unhindered through the gel, however, upon digestion with DSN, migration was

seriously affected. Ethanol precipitation was carried out on one digest prior to

the addition of marker in order to identify whether small fragments and/or

mononucleotides were the cause of the migration problems, as well as a

proteinase K treatment to determine whether the protein content of the digests

Figure 4.2:2 HPLC purified

oligonucleotides electrophoresed under

DSN overhang conditions. Three

oligonucleotides of various sizes

(G3=21nt, G4=12nt, G5=30nt, see Table

2.5-1) were size separated alongside a

low molecular weight marker as

indicated. LMW marker sizes are

indicated in nt at the far left of the

image. Size of each oligonucleotide

marker was estimated through

extrapolation of the LMW marker data,

via the method described in Section

2.11.3. In order to effectively detect the

small-sized oligonucleotides, a C-rich

12mer (ssOc12 - Table 2.9-1) was

radiolabelled and used in the

hybridisation after optimisation.

Page 155: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

140

Figure 4.2:3 Results of DSN overhang optimisation experiments a DSN assay carried out on

HT108 DNA with control (DSN+ExoI) and experiment (DSN alone) reactions. Dotted blue line

indicates the migration of the bromophenol blue dye front b Photograph of an acrylamide gel

during electrophoresis. Lanes contain: 1: LMW ladder, enzyme buffers and glycerol, 2= HT1080

control digest, 3= HT1080 experiment digest, 4= H2O, 5= KK control digest, 6=KK experiment

digest c 1% agarose gel containing genomic DNA from HT1080 and KK cell lines. DNA was

digested with DSN and and with an without ExoI treatment d LMW ladder resolved on a

denaturing polyacrylamide gel with the following additives: 1= none, 2 = heat inactivated

enzymes, 3 = control digest products, 4 =BSA, 5 = control digest products + SDS. (e) e λ and

Φ markers were mixed with genomic DNA and digested with DSN. Ethanol precipitation (EtOH)

and proteinase K (Prot. K) treatment were carried out as indicated.

Page 156: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

141

was an issue. As can be seen in Figure 4.2:3e, the addition of ethanol

precipitated DNA allowed similar migration and expected size resolution to the

addition of undigested DNA, while proteinase K treatment had no effect.

After completing this process of elimination, it was apparent that the digest

process itself was the source of the migration issues. If average overhang length

is considered to be 65nt long (Zhao et al., 2008), a conservative estimate for the

amount of overhang DNA in a 5µg sample would be ~2pg. Assuming that the

completed 5µg digests consisted of almost entirely of ≤6nt fragments, it

appeared that overloading of the lanes at this size range interfered with

electrophoresis of the very small quantity of overhang products. As current DNA

clean-up methods inescapably result in sample loss, and given the closeness in

size between the digest bi-products and smaller overhangs, a clean-up method

that would discriminate between the two could not be devised.

In order to tailor the DSN assay to the use of patient DNA, digests were

attempted using lower amounts of DNA. Signal from hybridisations of digests

carried out on less than 5µg DNA was imperceptible. Ultimately, it was not

possible to resolve the problems highlighted above, and so it was concluded

that this technique is not suitable for use with the smaller amounts of DNA

available from patient cells lines.

4.2.3 Native dot blots

As the amount of genomic DNA available from patient cells was a major limiting

factor, development of a volume-based method to quantify telomeric single-

stranded overhangs was attempted. Although this precluded direct analysis of

overhang length, it was apparent from the shortcomings of the DSN method

that length analysis would not be possible with the amounts of DNA available

from patient cells. Native dot-blotting was selected as a simple and direct

method for relative overhang quantification. In order to minimise any pipetting

Page 157: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

142

Figure 4.2:4 Results of native dot blot optimisation experiments a T7 digestion timecourse

carried out on U2OS cell DNA and hybridised to the radiolabelled probes indicated b

Quantification of (a) c Example dot blot showing relative signal before (1) and after (2) ExoI

digest from U2OS DNA. Samples were divided into two and loaded onto two separate

membranes before probing each with either a C-rich or G-rich telomere-specific probe

(contrast levels shown for TelC- and G3-probed membranes not are not equivalent) d As (c)

but with Hirt extracted DNA e Quantified C-rich probe signal from three independent

replicas (rep 1-3), Hirt extracted DNA was left undigested (1) or ExoI digested (2). Error bars

represent SEM from three dots, values normalised to undigested DNA. au=arbitrary units

Page 158: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

143

error and normalise the amount of DNA loaded onto membranes, all samples

were partially digested with AluI prior to further digestion and loading.

In order to test the sensitivity of this method, a time-course was carried out on

U2OS genomic DNA with T7 exonuclease. T7 is able to resect the 5’ strand of

dsDNA, effectively extending overhangs. Digest products were split across two

dot blot membranes and then hybridised with either a C-rich telomere probe,

TelC, in order to detect the G-rich overhang, or a G-rich probe, G3 as a negative

control (see Table 2.9-1 for probe sequences). The relative signal from

hybridisation with the TelC probe increased over time but unexpectedly

hybridisation with the G-rich probe resulted in a quantifiable signal which was

affected by T7 digestion in a similar proportion to the C-rich probe results

(Figure 4.2:4a).

Once acquired, DNA samples were split in half. One half was treated with ExoI to

remove overhangs while the other half was left undigested. ExoI digested and

undigested DNA was then loaded side-by-side in triplicate onto a membrane

under native conditions (see section 2.8.2). Membranes were hybridised to G-

and C-rich probes (Figure 4.2:3b), and the amount of overhang signal

(undigested sample) could then be quantified relative to the signal from the

control sample (ExoI digested). Digestion of DNA with ExoI should eliminate

overhang signal, however C-rich probe was still bound to dots (Figure 4.2:3b).

This signal, and the signal from G-rich probe may have been attributable to

large amounts of single stranded telomeric DNA present in ALT cells (Nabetani

and Ishikawa, 2009). To counter this effect, modified Hirt extractions were

carried out on DNA samples. The Hirt extraction procedure (Hirt, 1967) is

normally employed in order to isolate extrachromosomal DNA, such as

plasmids, from mammalian cells. A centrifugation step is carried out which

results in two fractions – genomic DNA in the pellet and extrachromosomal

Page 159: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

144

DNA in the supernatant. In this case, the supernatant was discarded and the

pellet resolubilised in order to purify chromosomal DNA (see section 2.2.2).

Experiments were repeated with Hirt extracted DNA, and although background

signal improved, signal was still detectable with the C-rich probe on ExoI

digested DNA still, and on G-probed DNA. This was also the case for telomerase

positive cell lines (data not shown), which do not contain extra-chromosomal

telomeric sequences (Cesare and Griffith, 2004). Large telomere molecules are

observed when hybridising with G- and C-rich probes using the DSN method,

and this is attributed by the method authors to an unknown fraction of DSN-

inaccessible single stranded DNA (Zhao et al., 2008). Either Hirt extraction was

not able to separate this fraction from chromosomal DNA, perhaps due to its

size, or the stringency of this method is not adequate.

Several identical experiments (with Hirt extraction) were carried out in order to

determine whether this background signal from extrachromosomal telomeric

DNA in the ALT U2Os cell line caused issues for quantification (Figure 4.2:3d).

Results showed some variation in signal from one experiment to another. The

signal from ExoI digests was considered as baseline background, and a relative

value for each experiment was derived by a ratio of digested:undigested DNA.

In the three experiments shown in Figure 4.2:3d, the resultant ratios were as

follows: replica 1 = 0.358, replica 2 = 0.531, replica 3 = 0.432. Due to this

variation it was decided to abandon this method for quantifying telomere

single-stranded overhangs.

4.3 Discussion

4.3.1 Alternative methods for overhang analysis

As neither method above provided consistent or robust results, and given the

limited amount of genomic material, it is unlikely that any of the well-defined

Page 160: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

145

techniques that measure overhang directly would be suitable for measurement

of overhangs in DNA from patient cell lines, in particular those that measure

length, rather than relative measurements.

Cellular levels of POT1 are relative to the amount of single-stranded telomeric

overhang (Loayza and De Lange, 2003). Immunofluorescence using a POT1

primary antibody was evaluated as a method for quantification of overhangs.

However, the quality of currently available commercial antibodies was not high

enough to derive quantifiable data. An alternative application of POT1 antibody

would be as an immunoblotting method by quantification of POT1 in lysates

derived from patient cells. Again, quantification was not possible due to the

quality of the available antibody.

Although every attempt was made to carry out the analysis directly on patient

DNA, as the quantity of DNA holds a crucial barrier to accurate quantification it

would be more feasible to express mutant lamin A forms ectopically in

immortalised cells and harvest large quantities of DNA, and It would be

necessary to inactivate telomerase in order to eliminate extension of overhangs.

Alternatively, several other possible causes for accelerated shortening were

considered and could be explored. These are discussed and some preliminary

data presented in section 7.1.1.

Page 161: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

146

PART B • The nuclear lamina and telomere biology in

ALT and Telomerase positive cancer cells

Chapter 5 Differential expression of lamins and

related proteins in ALT and telomerase positive cell

lines

5.1 Background

An essential stage in cancer cell development is the acquisition of a TMM in

order to escape senescence and continue division. Around 85% of cancers carry

out telomere extension via activation of telomerase (Shay and Bacchetti, 1997).

The remainder of cancers activate an Alternative Lengthening of Telomeres

(ALT) mechanism (Bryan et al., 1997), which utilises recombination-based

processes to maintain telomeres (Dunham et al., 2000; Varley et al., 2002). As

mutations in A-type lamins have been shown to influence telomere

maintenance in Chapter 3, A-type lamin function may differ within the context

of differential maintenance mechanisms.

Several aspects of nuclear organisation and chromatin status are affected in

both lamins-deficient and ALT cells. HGPS cells display a global reduction of

condensed heterochromatin, including the constitutive heterochromatin

markers found commonly at telomeres (Csoka, 2004; Shumaker et al., 2006),.

Loss of telomere chromatin condensation also occurs in ALT cells (Episkopou et

al., 2014), coupled with increased motility of shortened and de-protected

telomeres (Jegou et al., 2009). This increased motility is dependent on 53BP1

(Dimitrova et al., 2008), whose nuclear abundance is modulated by the presence

of A type lamins (Gonzalez-Suarez et al., 2011). Therefore, levels of lamin A

Page 162: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

147

could be crucial for 53BP1 activity in ALT cells, and its associated effect on

telomere motility.

As well as their role in 53BP1 regulation, lamins may play a more direct role in

the mobility of nuclear factors. Telomeres are relocated towards the nuclear

periphery in senescent cells and this change in localisation and increased

aggregation of telomeres is associated with increasing lamina deformity with

continuing cell divisions (Raz et al., 2008, 2006). Lmna-/- MEFs also display a

redistribution of telomeres towards the periphery (Gonzalez-Suarez et al., 2009;

Uhlírová et al., 2010). Telomeric DNA-containing ALT-associated PML bodies

(APBs) are an important marker of ALT activity (see section 1.2.4.2). Although

evidence suggests that APBs are resident to recombinatorial processes of ALT

(Draskovic et al., 2009), their role remains mysterious as ALT processes may

occur in their absence (Jeyapalan et al., 2008). PML bodies associate with the

nucleoskeleton (Stuurman et al., 1990) and move more rapidly and freely in

lamin A deficient cells, aggregating and localising to the nuclear periphery more

frequently (Stixová et al., 2012). APBs may also accumulate outside the nuclear

membrane following rupture in laminopathy cells (Houben et al., 2013).

Absence of lamin A in MEFs reduces telomere association with PML bodies

(Uhlírová et al., 2010), suggesting that an increase in lamin A could aid APB

formation. As PML bodies are associated with intranuclear lamins, and a subset

of ALT-associated PML bodies are able to mobilise and co-localise with

telomeres (Jegou et al., 2009), altered lamin biology in ALT cells could be

contributing to APB:telomere interactions.

Previously, no investigation into the status of lamins in ALT cells has been

carried out. As such, investigation began with the quantification of lamins, a

selection of lamina associated factors, and potential linkers between the lamina

and chromatin in a panel of ALT and telomerase positive cell lines (summarised

in Table 5.1-1). Three lines with a common derivation but differing telomere

Page 163: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

148

maintenance status were included (JFCF.6T.5K, JFCF.6T-1J/11C and JFCF.6T/11E,

hereafter referred to as 5K, 11C and 11E, respectively) in order to observe

potential differences between ALT and telomerase lines in the context of as

close an isogenic background as possible.

ALT Telomerase positive (Tel+)

Cell line Origin/derivation Cell line Origin

WI38ALT Lung fibroblast (foetal) JFCF.6T-

1J/11C

CF jejunal fibroblast,

transformed

SUSM-1 Liver fibroblast (embryonic),

transformed

HT1080 Fibrosacrcoma

JFCF6T.5K CF jejunal fibroblast (post

crisis), transformed

HeLa Cervical adenocarcinoma

JFCJ.6T-

1J/11E

CF jejunal fibroblast,

transformed

LoVo HNPCC

WV Werner syndrome fibroblast,

transformed

SW480 Colonic adenocarcinoma

Saos Osteosarcoma T24 Bladder carcinoma

U2OS Osteosarcoma NT2D1 testis carcinoma metastasis

(embryonic)

IIIcf/2a Li-Fraumeni breast

fibrosarcoma

- -

5.2 Results

5.2.1 Protein quantification by Western blot

Western blotting was employed to establish endogenous levels of lamins and

related proteins in ALT and telomerase cells. Initially, the telomerase positive

line NT2D1 was included in the cell line panel. However, this line displayed

Table 5.1-1 ALT and telomerase positive cell lines used in this chapter. Further information, including

culturing conditions may be found in Table 2.1-1. CF=cystic fibrosis, HNPCC=Hereditary non-

polyposis colon cancer

Page 164: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

149

unusual characteristics, such as absence of A-type lamins and SUN proteins.

Statistically, protein levels differed significantly from the other telomerase

positive lines for all but 53BP1, SUN1 and SUN2 content, despite the apparent

absence of SUN from NT2D1 (Appendix E). As A-type lamins have key roles in

differentiation and development, these differences were most likely attributable

to the cell line’s derivation from pluripotent embryonic cells, and as such NT2D1

was excluded from the final panel.

Lamins A and C were quantified using an antibody raised against an N-terminal

orientated epitope capable of identifying both splice forms. Lamin A was

present in telomerase positive cells at almost three times the amount in ALT

cells (p<0.0001), and lamin C was also in significantly greater abundance in

Figure 5.2:1 Representative Western blot of each protein and cell line in the panel. Whole cell

lysates were derived from cell pellets of each line by the addition of Laemmli buffer. Lysates

were size separated on SDS-PAGE gels containing the appropriate percentage of acrylamide

with Page Ruler Plus protein marker (see section 2.13). Control immunoblotting was carried

out on the same membrane wherever possible. Bands were normalised within each blot in

order to allow comparison across blots. ALT (A) or telomerase positive (T) status is indicated at

the bottom of the figure.

Page 165: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

150

telomerase positive cells, but at only a 1.4-fold increase (p=0.0016). Lamin B was

significantly elevated in telomerase positive cells (p=0.0007), by a factor of 1.2

(Figure 5.2:1 and Figure 5.2:2a-c). Loss of A-type lamins has been shown to

activate the degradation pathway of 53BP1 in MEFs (Gonzalez-Suarez et al.,

2011). To assay whether levels of 53BP1 were affected by the differential levels

of A-type lamins in ALT and telomerase positive cells, 53BP1 levels were also

quantified. However, 53BP1 levels were not significantly different between the

two sets of cell lines, nor did lower levels of lamin A in individual cell lines

Figure 5.2:2 Total protein content as quantified by Western blot in ALT and telomerase

positive cell lines, normalised to β-tubulin. Error bars show SEM from three independent

biological replicas. p values are derived from t tests on ALT versus telomerase positive cells.

au=arbitrary units a Lamin A levels are significantly higher in telomerase positive cells

(p<0.0001) b Lamin C levels are significantly higher in telomerase positive cells (p=0.0016)

c Lamin B1 levels are significantly higher in telomerase positive cells (p=0.0007) d 53BP1

levels are not significantly different between ALT and telomerase positive cells.

Page 166: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

151

correlate with lower 53BP1 levels (compare Figure 5.2:2a&d). SUSM-1, a DSB

repair deficient line derived from embryonic tissue, showed extremely low levels

of 53BP1, likely contributing to its DNA repair status.

The HP1 protein family was identified as a candidate linker for lamina and

Figure 5.2:3 Total protein content as

quantified by Western blot in ALT and

telomerase positive cell lines, normalised

to either α-tubulin or β-tubulin as

indicated in brackets in legend. Error bars

show SEM from three independent

biological replicas. p values are derived

from t tests on ALT versus telomerase

positive cells. au=arbitrary untis a HP1-α

results (β) b HP1-γ results (β) c SUN1

results (α) d SUN2 results (β) e RAP1

results (α)

Page 167: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

152

chromatin interactions (see sections 1.1.3.3 & 1.1.5.3). HP1-α is capable of

binding LBR (Ye et al., 1997), pre-lamin A and LAP2α (Lattanzi et al., 2007). HP1-

α levels are reduced in HGPS cells (Scaffidi and Misteli, 2005), while APB

formation is dependent on the presence of HP1 proteins (Jiang et al., 2009).

Therefore levels of HP1-α were investigated but were not significantly different

between ALT and telomerase lines (Figure 5.2:3a). Subsequently, HP1-γ was

considered due to its role in telomere maintenance (Canudas et al., 2011) and

were found to be almost two-fold higher in telomerase positive cells (p<0.0001,

Figure 5.2:3b).

SUN proteins are integral membrane proteins associated with the lamina, and

SUN1 is required for chromatin detachment from the nuclear membrane in

mitosis (Patel et al., 2014; Turgay et al., 2014). SUN1 also plays a role in

tethering telomeres to the nuclear membrane, aiding post-mitotic re-assembly

(Crabbe et al., 2012), and so may have a role in telomere anchorage in ALT.

SUN1 levels were investigated and proved to be present in telomerase cells at

almost 4.5 times the level in ALT cells (p<0.0001, Figure 5.2:3c). SUN2 was also

examined and no significant differences were found between SUN2 levels in ALT

and Telomerase positive cells. Interestingly, SUN2 levels were so low in the

telomerase positive lines SW480 and LoVo as to be virtually absent (Figure 5.2:1

and Figure 5.2:3d). As both these lines are derived from colon tissue, this is

most likely indicative of an underlying biology specific to this cell type. Upon

exclusion of these cell lines from the SUN2 data, differences remain non-

significant (p=0.5891). As SUN1 may link chromatin to the lamina via RAP1

(Crabbe et al., 2012), levels of RAP1 were quantified and found to be more than

twice as abundant in telomerase positive cells (Figure 5.2:3e).

Page 168: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

153

5.2.2 Protein quantification and identification of lamin

localisation by immunofluorescence microscopy

In order to confirm the Western findings, and verify any differences in protein

localisation between ALT and telomerase positive cell lines,

immunofluorescence (IF) microscopy was carried out on three representative

ALT and three representative telomerase positive lines, including 11E (ALT) and

11C (Tel+). Lamin A/C, lamin B, 53BP1 and HP1-γ were chosen for further

investigation by IF. The Olympus Scan R platform was used to gather the IF

data. Several hundred cells were scanned for each protein in each cell line, and

several parameters were recorded, including the total signal intensity of each

Figure 5.2:4 Comparison of Western blot and IF total protein results.

IF values are normalised internally to allow comparison to Western

data. t tests were carried out to compare ALT and Tel+ cell line total

protein results for IF and did not reveal significant differences. Error

bars indicate error from two biologically independent repeats for IF,

and three independent repeats for Westerns. au=arbitrary units.

Page 169: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

154

protein per nucleus, and the size-normalised signal per nucleus, derived by

normalising to DAPI signal. DAPI staining was also used to select for single

nuclei, and exclude nuclei displaying extreme abnormalities, such as very low

circularity.

In order to compare total protein as assayed by Western blot and IF, the total

signal intensity values provided by the Scan R software were internally

normalised to sit within the range of the Western total protein values (arbritrary

units). As lamin A and lamin C were both quantified using a single antibody, it

was not possible to discriminate between the two when analysing the IF data,

and so total lamin A and C Western values were combined prior to comparison.

The results for IF and Westerns differed for relative total protein content (Figure

5.2:4), most likely due to the differential availability of epitopes to the primary

antibodies between the two techniques. IF total signal of each protein was not

found to differ significantly between ALT and telomerase cells (see below),

despite close parity between the total protein results for IF and Western results

for lamin B and HP1-γ (Figure 5.2:4b&d).

This could be due to the automated counting system, as optimal parameters for

nucleus recognition is not possible between different cell lines. This may result

in cells with certain morphologies to be discounted from the data pool. For

example, the relative size of nuclei did not differ significantly between ALT and

telomerase cells (data not shown). ALT cell nuclei were observed to be more

variable in size through manual screening, potentially due to increased amounts

of DNA resulting from extrachromosomal telomeric DNA. This implies that large

nuclei were not included in the ALT data, although every care was taken to allow

inclusion of these cells. Circularity also did not differ significantly between

telomerase and ALT cells (data not shown) despite the presence of considerable

numbers of misshapen nuclei observed by manual screening particularly in

WI38ALT and U2OS cells (Figure 5.2:5-Figure 5.2:8). However, the screening did

Page 170: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

155

DA

PI

FIT

C:la

min

A/C

M

erg

e

HT1080 WI38ALT HeLa JFCF/11E JFCF/11C U2OS

ALT Tel+

Figure 5.2:5 IF results for lamin A/C a Microscope images showing DAPI and lamin A/C

immunostaining as indicated b Mean total intensity of lamin A/C in each cell line c Mean

size-normalised intensity, defined as the total amount of lamin A/C relative to DAPI

staining in each cell, as derived by Scan R software d Mean total intensity of lamin A/C

located at the nuclear periphery e Mean ratio of intranuclear lamin A/C relative to

peripheral lamin A/C. Error bars represent error from two independent biological replicas.

au=arbitrary units

Page 171: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

156

DA

PI

TxR

ed

:lam

in B

M

erg

e

WI38ALT HeLa JFCF/11E JFCF/11C U2OS

ALT Tel+

HT1080

Figure 5.2:6 IF results for lamin B a Microscope images showing DAPI and lamin B

immunostaining as indicated b Mean total intensity of lamin B in each cell line c Mean

size-normalised intensity, defined as the total amount of lamin B relative to DAPI staining

in each cell, as derived by Scan R software d Mean total intensity of lamin B located at the

nuclear periphery e Mean ratio of intranuclear lamin B relative to peripheral lamin B. Error

bars represent error from two independent biological replicas. au=arbitrary units

Page 172: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

157

reveal differences between lamin localisation in ALT and telomerase cells.

Nuclear levels of Lamin A/C did not differ between ALT and telomerase positive

cells when measured according to total lamin A/C (Figure 5.2:5b), or relative to

size as indicated by DAPI staining (Figure 5.2:5c). In order to determine whether

lamin A/C localisation at the nuclear periphery varied between ALT and

telomerase cell lines, Scan R was programmed to identify the nuclear perimeter

according to the edge of the DAPI staining and quantify the amount of lamin

within 3 pixels either side of this boundary. Total peripheral lamin A/C was not

significantly different between ALT and telomerase positive cells (Figure 5.2:5d),

however intranuclear levels of Lamin A/C were significantly higher relative to

peripheral levels in ALT cells (p=0.0164, Figure 5.2:5e), although this effect was

minimal (1.1 fold higher).

These measurements were repeated for lamin B (Figure 5.2:6). Again, nuclear

levels of lamin B were not significantly different between ALT and telomerase

positive cells (Figure 5.2:6b&c), although total lamin B was almost significantly

higher in ALT cells (p=0.0532), counter-intuitive to the Western data.

Examination of total peripheral lamin B again revealed no significant difference

(p=0.2704, Figure 5.2:6d), while the proportion of intranuclear lamin B was 1.4

times higher in ALT cells (p=0.0087, Figure 5.2:6e).

Due to the extremely large size of 53BP1 (>250kDa), a specialised transfer

procedure was developed for Western blotting, however, variability between

replicas was relatively high. Due to this level of uncertainty, IF was partly carried

out in order to verify the Western findings. 53BP1 results correlated with

Western findings, in that no significant difference was found between the total

53BP1 in ALT and telomerase cells (Figure 5.2:7b&c), although IF also exhibited

more variation between replicas than the rest of the proteins examined in the

panel. Automatic foci counting was carried out on 53BP1 immunostained cells

Page 173: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

158

DA

PI

FIT

C:5

3B

P1

Merg

e

HT1080 WI38ALT HeLa JFCF/11E JFCF/11C U2OS

ALT Tel+

Figure 5.2:7 IF results for 53BP1 a Microscope images showing DAPI and 53BP1

immunostaining as indicated. Green channel has been enhanced post-capture in order to

aid visualisation b Mean total intensity of 53BP1 in each cell line c Mean size-normalised

intensity, defined as the total amount of 53BP1 relative to DAPI staining in each cell, as

derived by Scan R software d Percentage of cells with zero, 1-5 or >5 53BP1 nuclear foci as

detected by Scan R in ALT and telomerase positive cells.

Page 174: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

159

and foci counts were found to differ despite no overall difference in 53BP1

levels. Telomerase positive cells were three times more likely to contain no

53BP1 damage foci than ALT cells (p=0.0230), and ALT cells were more than

twice as likely to contain more than five foci (p=0.0059, Figure 5.2:7d.). This is in

accordance with elevated damage levels and rearrangements in ALT cells. The

comparable amounts of 53BP1 between ALT and telomerase cells may have

DA

PI

FIT

C:H

P1-γ

M

erg

e

HT1080 WI38AL

T

HeLa JFCF/11

E

JFCF/11C U2O

S

ALT Tel+

Figure 5.2:8 IF results for HP1-γ a Microscope images showing DAPI and HP1-γ

immunostaining as indicated b Mean total intensity of HP1-γ in each cell line c Mean size-

normalised intensity, defined as the total amount of HP1-γ relative to DAPI staining in each

cell, as derived by Scan R software.

Page 175: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

160

been due to a higher quantity of diffuse 53BP1 in telomerase positive cells

(Figure 5.2:7a). Finally, although total HP1-γ levels were not found to be

significantly different between ALT and telomerase positive cells, which may

have been due to highly variable amounts of HP1-γ, primarily in the telomerase

positive cells, size-normalised values were approximately 1.5 times higher in

ALT cells (p=0.0379, Figure 5.2:8).

5.3 Discussion

Several differences in lamina-associated factors were identified between ALT

and telomerase positive cells through protein quantification and localisation

assays. Lamins A, B and C and SUN1 were found in higher abundance in

telomerase positive cells, compared to ALT cells, as well as HP1-γ and RAP1,

candidate linkers for chromatin:lamina interactions. The panel of ALT and

telomerase positive cells that were investigated with Western blots included

three cell lines derived from a single individual which have adopted different

telomere maintenance mechanisms during establishment in culture. As these

three lines share the same origin, differences observed between them are more

likely to reflect differential mechanistic properties between the ALT and

telomerase maintenance programs, rather than inter-individual differences.

Some divergence will have occurred in the derivation of these lines, as may be

seen in the occasional variation between 11E and 5K despite both being ALT,

although this may be as a result of differing ALT pathways (alternative modes of

ALT-based recombination have been reviewed by Cesare & Reddel 2010).

Direct comparison of the JFCF.6T cell Western results supports most of the

findings from the broader panel (Figure 5.3:1). Levels of A-type lamins were

found at higher concentration in the telomerase positive 11C line, while no

difference was found between the ALT and telomerase positive JFCF.6T lines for

lamin B. As lamin B levels were found to be extremely consistent in telomerase

Page 176: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

161

cell lines (Figure 5.2:2c), this is possibly due to the greater variability in the 11E

replicas. The results are sensitive to an outlier in the 11E data-set and exclusion

of this point gives a significant difference between the ALT and telomerase cells

(p=0.032). Excluding this outlier, all proteins examined followed the trend of

increased protein levels in the telomerase positive line, although the differences

for SUN1 and RAP1 failed to reach significance (Figure 5.3:1). Interestingly,

SUN2 and HP1-α levels were found to be significantly lower in the ALT lines 11E

and 5K lines, compared to 11C, whereas no significant difference was identified

in the full panel. 11E and 5K differed significantly in their SUN2 content, despite

both being ALT positive (p=0.0310), and levels were highly variable between

both the different ALT lines and telomerase lines in the full panel. This supports

the initial finding from the complete panel that there is no relationship between

SUN2 levels and TMM. No such difference existed between 11E and 5K for HP1-

Figure 5.3:1 Comparison of JFCF.6T lines. 5K/11E=ALT, 11C=Telomerase positive. The mean

of three biological replicas (represented by triangles) is shown. Error bars indicate SEM.

Significance is denoted above each set, derived from unpaired t-tests.

Page 177: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

162

α (p=0.6184), putting the panel analysis for HP1-α into question. In a similar

fashion to SUN2, HP1-α levels were dramatically lower in the colon-derived

SW480 and LoVo telomerase positive lines. Exclusion of these lines from the

analysis results in a signifcant difference in HP1-α levels (p=0.0125), marking it

as potential candidate for involvement in ALT and telomerase differential

activity.

Lamin A, B1 and C were found at significantly higher levels in telomerase

positive cells compared to ALT cells according to the complete panel data,

although this finding was not repeated with the immunofluorescence data. The

reduced and denatured conditions of Western blotting allow for greater

accessibility to antibodies, and is therefore a more reliable reference for total

protein content. Also, IF signal may not be linear in relation to the amount of

protein in the cell due to steric hindrance in this respect. This discrepancy

alludes to the existence of a subset of lamins in telomerase positive cells that is

less accessible, and could be explored via solubility assays. At least one epitope

of lamin A located between residues 171 and 246 is accessible to an antibody

characterised by Jagatheesan et al. (1999) on intranuclear lamin A, but not the

peripheral fraction (Jagatheesan et al., 1999). Again, this supports the

hypothesis that intranuclear lamin A forms alternate structures compared to

peripheral lamin A, and may reflect the alternate functions of the two lamin A

pools (Dechat et al., 2010). If intranuclear lamin A plays a role in APB formation

or localisation, this may explain how a decrease in lamin A that would be

anticipated to inhibit APB formation (Uhlírová et al., 2010) is present in ALT cells.

The recorded data from the IF screen for area and circularity did not reflect the

observed variable size or increase in nuclear deformity of ALT cells, and it is

possible that large and/or misshapen nuclei may have been excluded from the

screen. If this is the case, and the Western data is the most representative, it is

possible that the amount of lamin could remain constant, while becoming more

Page 178: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

163

diffuse within enlarging ALT nuclei. As lamins quantitation was relative to

tubulins, in this scenario tubulin levels would need to increase to compensate

for the increased nuclear size. This correlation could be identified via the use of

fluorescence-activated cell sorting, which would be able to size each nucleus

according to DNA content, while quantifying lamin signal.

ALT cells that were included in the IF screen contained a lower proportion of

lamin A, B1 and C at the nuclear periphery compared to telomerase positive

cells. Increased relative levels of lamin A at the nuclear periphery is a marker of

HGPS, resulting from the increased integration of lamins into the nuclear matrix,

and is associated with a reduction of lamin A mobility (Dahl et al., 2006; Scaffidi

and Misteli, 2005). In normal cells, lamins are continually produced and

integrated into the lamina (Gerace et al., 1984). Two distinct fractions of lamin A

are distinguishable through their differential motility – approximately 15% of

lamin A is able to move freely and rapidly throughout the nucleoplasm, while

the remaining fraction, as well as periphery lamin A is more stable, indicating a

higher order of lamin structure (Dahl et al., 2006). On the other hand, the

presence of progerin eliminates almost all mobility of both WT lamin A and

progerin, resulting in the sequestration of both at the nuclear periphery, and the

typical thickened lamina observed in HGPS (Dahl et al., 2006; Goldman et al.,

2004; Scaffidi and Misteli, 2005). In FRET assays, fluorescence recovery is absent

from the lamina after normal levels of lamin A have recovered within the

nucleoplasm even in WT cells (Dahl et al., 2006), underlining the stable and

immobile nature of lamin A that is integrated into the lamina. Additionally, once

integrated, mature lamin A does not return to the intranuclear space, but

reforms solely at the periphery after mitosis (Goldman et al., 1992), further

supporting the existence of alternative structure formations with differential

solubility and localisation properties.

Page 179: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

164

These data support the existence of a less soluble and proportionally larger

fraction of peripheral A-type lamins in telomerase cells compared with ALT cells.

The capacity of progerin to impair nuclear mobility, including telomere

movement (De Vos et al., 2010) suggests that alterations in lamina organisation

may directly influence telomere mobility. Indeed, loss of lamin A results in an

increase in telomere mobility in fibroblasts (De Vos et al., 2010). Given that

telomere motility is abnormally elevated in ALT cells (Jegou et al., 2009), it is

most likely that the properties of lamin A in telomerase cells reflect those of

normal cells, while in ALT cells higher order organisation of A-type lamins is

limited in order to maintain a more motile nuclear environment, conducive to

the ALT mechanism. The differential motility of lamins in ALT, telomerase

positive and normal cells could be established through the application of FRAP,

however introduction of exogenous, fluorescently-tagged lamin A into ALT cells

could result in a reversal of increased nuclear motility, as this phenotype is

could be due to the reduced amount of lamin A in these cells, as lamin

organisation is concentration dependent (Guo et al., 2014).

53BP1 promotes increased telomere mobility (Dimitrova et al., 2008), however

levels were not found to be affected in ALT cells. In the absence of progerin,

lamin A deficiency results in an increase in telomere motility in fibroblasts (De

Vos et al., 2010) despite the reduction in 53BP1 that this would be expected to

incur (Gonzalez-Suarez et al., 2011). It is therefore possible that the observed

reduction in lamin A/C is adequate to facilitate nuclear motility without

activating 53BP1 degradation, or that 53BP1 function is altered in a way that

cannot be observed through quantification alone, which may be the case in

HGPS patient cells (see preliminary data in section 7.1.1.1).

A-type lamins are capable of stabilising DNA repair foci in the nucleus (Mahen

et al., 2013). Positional immobilisation of chromatin in the nucleus restricts DSB

repair, while increased mobility results in increased genomic instability through

Page 180: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

165

induction of inappropriate exchanges (Chiolo et al., 2011; Soutoglou et al.,

2007), which would benefit ALT cells. Depletion of macromolecular lamin

structures responsible for increased nuclear stability could contribute to

increased mobility of telomere ends to promote recombination in ALT cells.

Loss of lamin A results in decondensation of heterochromatin in MEFs

(Gonzalez-Suarez et al., 2009), and decondensation of telomeres enables

enhanced telomere elongation through recombination in mouse cells (Benetti

et al., 2007; Blasco, 2007; García-Cao et al., 2004). The observed reduction in

HP1-γ is consistent with this mode of action. If loss of A-type lamins contributes

to the loss of heterochromatin in ALT cells, this presents another potential

mechanism by which reduced nuclear lamin could facilitate the ALT telomere

maintenance program.

Page 181: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

166

Chapter 6 Interactions between chromatin and

lamins in ALT and telomerase positive cells

6.1 Background

As A-type lamins are at a lower level in ALT cells and this may be contributing

to increased telomere motility, it is possible that direct or indirect interactions

between the lamina and chromatin would be altered in ALT cells. Differential

distribution of peripheral lamins and intranuclear lamins suggests a

conformational change in higher order lamin A structure, which in turn could

affect steric interactions with DNA binding factors. Lamin A is able to engage

chromatin through γ-H2AX in order to stabilise DNA damage repair foci

(Mahen et al., 2013), and binds and associates with several factors that bind

DNA directly such as BAF, LBR (Dechat et al., 2004), lamin B (Dechat et al., 2004;

Martin et al., 2010), HP1-α (Lattanzi et al., 2007) and LAP2α (Dechat et al.,

2004; Lattanzi et al., 2007) as well as indirectly, for example LBR may bind to

chromatin through HP1-α (Ye et al., 1997). Although many of these interactions

are transiently involved in mitosis, differences in nuclear matrix components

were clearly detectable in asynchronous cells (see Chapter 5), suggesting a role

for the lamina in telomere organisation outside of the mitotic cycle.

Chromatin immunoprecipitation (ChIP) was therefore carried out on ALT and

telomerase positive asynchronous populations in order to identify differential

telomere binding to A-type lamins. These experiments were carried out prior to

the publication of a study identifying SUN1 as a linker between the lamina and

telomeres via its binding to RAP1 (Crabbe et al., 2012). As SUN proteins had

already been identified as chromatin interactor candidates, ChIP was attempted

with both SUN1 and SUN2 antibodies but no telomeric DNA was pulled down

(data not shown). As such, chromatin immunoprecipitation (ChIP) was carried

Page 182: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

167

out using a lamin A/C antibody with the aim of co-immunoprecipitating

interacting partners of lamin A that were responsible for telomere binding,

along with any bound telomeric DNA. As HP1 proteins were also promising

candidates identified in the quantification stage, ChIP was also performed using

HP1-α and HP1-γ antibodies.

6.2 Results

6.2.1 Optimisation and verification of ChIP

To investigate a direct or indirect interaction between A-type lamins and

telomeric chromatin, ChIP was undertaken on a selection of ALT and telomerase

positive cell lines. The goal of ChIP is to isolate genomic DNA sequences

associated with proteins of interest. In order to do so, proteins and chromatin

must first be cross linked with the use of formaldehyde before shearing the

chromatin to a suitable size, in this case via sonication. Fragmented chromatin is

then immunoprecipitated with an antibody specific to the protein of interest,

purified and quantified. Large numbers of cells are generally required, although

several relatively new techniques are available which require lower amounts of

starting material (Acevedo et al., 2007; Adli and Bernstein, 2011; Dahl and

Collas, 2008; Gilfillan et al., 2012).

Due to the complexity of the ChIP technique, several stages required

optimisation and verification. Firstly, choice of antibody is imperative to the

success of IP. A polyclonal Lamin A/C antibody previously used in ChIP

experiments in MEFs (Gonzalez-Suarez et al., 2009) was chosen as no previous

work had published the use of an A-type lamin antibody in ChIP of human

chromatin. To confirm specificity of the IP, a “ChIP-validated” H3K9me3

antibody was selected as an experimental control for ChIP due to its common

application in this role and its well established association with

Page 183: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

168

heterochromatin. A normal rabbit IgG was used as a negative control. ChIP was

also attempted with commercially available “ChIP-validated” antibodies against

HP1-α and HP1-γ.

Figure 6.2:1 a Immunoblot for H3K9me3 ChIP

validation using two lots of ab8898 (lot number

indicated at top of blot) showing differential IP

efficiency. E1 indicates a mild elution, E2 is a harsh

elution (see section 2.6.7) b H3K9me3 ChIP

validation using Active Motif H3K9me3 antibody

3916, Immunoblotted with Active Motif 3916 and

ab8898 c Immunoblot validating HP1-α and –γ

antibodies for use in ChIP d Immunoblot

validating lamin A/C sc-6215 antibody for use in

ChIP e ChIP chromatin after sonication (see

section 2.6.3.1). IP=immunoprecipitation, IB=

immunoblotted protein

a b

c d

e

Page 184: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

169

The specificity of all antibodies was tested via IP under ChIP conditions (see

section 2.6.7) and Western blotting (Figure 6.2:1). Repeated rounds of

verification on the same “ChIP-verified” H3K9me3 antibody purchased from the

same company at different times revealed a high degree of batch-to-batch

variation in IP efficiency (Figure 6.2:1a). Ultimately, to avoid this variation, only

one batch of each antibody was used for full-scale ChIP after verification. The

H3K9me3 antibody subsequently purchased from Active Motif and used in ChIP

was specific for H3K9me3 in IP but was not suitable for Western. This IP was

therefore verified by use of the previous H3K9me3 antibody purchased from

Abcam (Figure 6.2:1b). Verification via this method gave specific and consistent

results for the remaining antibodies used (Figure 6.2:1c&d).

Another essential step in ChIP optimisation is the shearing treatment of

chromatin to ensure acquisition of fragments of a suitable size for capture

during the IP steps. A standard size range for input chromatin is 400-600bp

(Schoppee Bortz and Wamhoff, 2011) allowing for at least two nucleosomes

(usually spaced 147bp apart (Richmond and Davey, 2003)) to be bound to

individual fragments. However, when investigating indirect chromatin

interactions, increasing the size of this range may aid in IP of target sequence

due to increased inefficiency (Prof Rob Klose - personal communication).

Shearing efficiency was initially optimised for each cell line and then median

size of chromatin fragments was confirmed for input chromatin for each replica

(Figure 6.2:1c&d). Lysis was also optimised after it was found that cells were not

lysed in standard ChIP lysis buffers. As a result, nuclei isolation took place in

buffer containing SDS at room temperature (see section 2.6.3).

The length of time that lysates and antibody-bead conjugates were incubated

was optimised in parallel with the amount of antibody for optimal noise to

signal ratio. As small amounts of short DNA fragments are isolated by ChIP, a

commercially available kit specifically developed for ChIP purification was used.

Page 185: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

170

The IPure kit (Diagenode) exploits the negative charge of the DNA backbone

and uses magnetic beads for purification in order to minimise DNA loss. ChIP

output of telomeric sequence was quantified by dot blot and the specificity of

ChIP verified by PCR. Primer sets were chosen for regions known to be

composed of open or closed chromatin to verify heterochromatin specificity for

H3K9me3, HP1-α and HP1-γ. The tandem repeat locus D4Z4 has previously

been reported to associate with lamin A, and previously published primer

sequences were used as a positive control for lamin A interactions (Ottaviani et

al., 2010).

6.2.2 A-type lamins do not associate differently with telomeric

chromatin in ALT versus telomerase positive cells

The results for each replica of ChIP are shown in Figure 6.2:2, Figure 6.2:3 and

Figure 6.2:4. The GAPDH promoter primer set was used as an indicator of open

chromatin (designed by Jon Williams, Royle laboratory), due to its constituitive

expression. An unidentified, non-specific small product was sporadically

amplified with this primer set (see Figures 6.2:2-3). The IgX primers (designed by

Abcam) anneal at a gene desert, and were used to indicate and confirm closed

chromatin. D4Z4 is a tandem repeat and therefore also represents closed

chromatin. The results of the semi-quantitative PCR assays for IgX suggest that

the H3K9me3 ChIP was highly inefficient as considerable binding of this marker

for closed chromatin would be expected at this gene desert. Despite the PCR

analysis being only semi-quantitative, the dot blots carried out confirmed low

efficiency as low levels of telomeric DNA was also pulled down. Initial

optimisations of ChIP were able to IP telomeric DNA and the IgX region at a

level that far exceeded the final results, while pulling down minimal amounts of

open chromatin as confirmed by the GAPDH promoter PCRs. However these

results could not be reproduced. It is possible that the optimised conditions

arrived at in order to quantify lamin A/C interactions became sub-optimal for

Page 186: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

171

Figure 6.2:2 First ChIP replica a Results of semi-quantitative PCR on ChIP samples

with the primer sets indicated. Products were run on a 2.5% NuSieve agarose gel b

H2O control samples for semi-quantitative PCR c Diagram illustrating sample

locations in each section of d d Dot blot of ChIP samples including input

references. 10μl of a 50μl reaction was loaded for each ChIP.

HT1080 HeLa SUSM-1 WI38-ALT

11E 11C 5K IgG

Page 187: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

172

Figure 6.2:3 Second ChIP replica a Results of semi-quantitative PCR on ChIP

samples with the primer sets indicated. Products were run on a 2.5% NuSieve

agarose gel b H2O control samples for semi-quantitative PCR c Diagram illustrating

sample locations in each section of d d Dot blot of ChIP samples including input

references. 10μl of a 50μl reaction was loaded for each ChIP.

HT1080 SUSM-1 HeLa WI38-ALT

11E 11C 5K

Page 188: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

173

Figure 6.2:4 Third ChIP replica a & b Results of semi-quantitative PCR on ChIP

samples with the primer sets indicated.. Products were run on a 2.5% NuSieve

agarose gel c H2O control samples for semi-quantitative PCR d Diagram illustrating

sample locations in e e Dot blot of ChIP samples including input references. 10μl of

a 50μl reaction was loaded for each ChIP, except in the case of HeLa where all

quantities were doubled.

HT1080 SUSM-1 WI38-ALT

11E 11C 5K

HeLa

Page 189: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

174

direct interactions with chromatin. Due to the ChIP inefficiency of HP1-γ at low

antibody concentrations that would allow for the use of a single batch of

antibody, this IP was discontinued, although this initial ChIP indicated elevated

association of HP1-γ with telomeres in HeLa and HT1080 cells (Figure 6.2:5e).

The final ChIP data for telomeric IP achieved from each antibody is given in

duplicate only in Figure 6.2:5 as the third replica showed high levels of

background in IgG ChIPs in both the PCRs and dot blots, as well as an apparent

absence of IP in the JFCF.6T lines and HT1080 (Figure 6.2:4b&e). In accordance

with published data comparing ALT and telomerase lines derived from a

Figure 6.2:5 a ChIP results for H3K9me3,

error bars indicate SEM from replicas 1 and 2

b Western blot result for H3K9me3 using

antibody ab8898. Error bars indicate SEM

from three independent biological replicas c

ChIP results for lamin A/C, error bars indicate

SEM from replicas 1 and 2 d ChIP results for

HP1-α, error bars indicate SEM from replicas

1 and 2 e ChIP results from replica 1 for

HP1-γ

Page 190: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

175

common source (Episkopou et al., 2014), H3K9me3 has a higher association

with telomeres in telomerase positive cell lines than in ALT lines (p=0.008,

Figure 6.2:5a). In order to allow comparison with total H3K9me3, Western

blotting was carried out on each line (Figure 6.2:5b). This did not reveal a

significant difference between a selection of ALT and telomerase positive cell

lines from the panel. No significant difference was observed between ALT and

telomerase cells for lamin A and HP1-α ChIPs (Figure 6.2:5c&d).

6.2.3 Discussion

Although the efficiency of ChIP was low, results for H3K9me3 were in

agreement with previously published ChIP data (Episkopou et al., 2014).

Telomeres were found to be associated with H3K9me3 at higher levels in

telomerase positive cells compared with the ALT lines, despite no difference in

global H3K9 trimethylation levels. No significant difference was found between

the JFCF ALT and telomerase lines (p=0.1333).

Episkpou et al. found that ALT telomere chromatin is packaged more loosely at

telomeres, that is, a greater distance between nucleosomes was observed. The

proportion of H3K9 residues that were trimethylated remained constant,

regardless of telomerase activation status and the reduced abundance of

histones at ALT telomeres. Also, heterochromatin status was found to be

unchanged at ALT centromeres, and miccrococcal nuclease access to genomic

chromatin was unaffected, making it very unlikely that nucleosome

redistribution or H3K9 methylation status would be significantly altered in the

rest of the genome.

It is quite possible that telomere histone content is relatively constant between

ALT and telomerase positive nuclei, and that nucleosomes are merely spread

more diffusely across the additional telomeric DNA present in ALT cells. A

relative increase in telomeric content in ALT versus telomerase cells is apparent

Page 191: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

176

in the signal differences in the dot blots shown in Figures 6.2:2-4, in agreement

with previously published Q-PCR findings (Lau et al., 2013). The ALT cell lines

reproducibly exhibited up to ten times the amount of telomeric DNA in input

dilutions despite similar cell numbers in each sample. Indeed, to maintain a

comparable range, double loading was eventually used on the HeLa ChIP

extracts (Figure 6.2:4e).

Total H3K9me3 levels were not reported in the study by Episkopou et al., but

together these data suggest that the number of H3 histones at telomeres

remains unaltered upon a cell adopting an ALT pathway, as does the

trimethylation status of the H3K9 residue. This implies that distribution of

nucleosomes is affected only at telomeres, which become elongated in an

unregulated fashion in ALT, and the level of histone binding does not

compensate for this additional length. This is consistent with the suggestion

that shelterin binding may be reduced at ALT telomeres, and with evidence of

differential composition of binding factors at ALT telomeres (Conomos et al.,

2013). Alterations in ALT telomere structure result in the loss of association of

several known telomere-binding proteins, as well as ALT-specific binding of

orphan receptors to telomeres (Déjardin and Kingston, 2009). As longer

telomeres in telomerase positive cells have a higher density of H3K9me3

(Arnoult et al., 2012; Déjardin and Kingston, 2009), the process by which

telomere repeats are gained must dictate whether or not additional nucleosome

units are formed, and could therefore be processively linked to the action of

telomerase.

As no previous investigation has been carried out into differential association of

HP1-α and –γ at telomeres in ALT and telomerase cells it is not possible to

compare these data to any published work. The initial results from HP1-γ ChIP

correlate with increased telomeric binding to heterochromatin proteins in

telomerase positive cells, however this increase aligns with an increased

Page 192: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

177

presence of HP1-γ in telomerase cells (although 11C is not in accordance with

this trend (Figure 6.2:6d)). HP1-γ binds chromatin through the H3K9me3 residue

(Lachner et al., 2001), and so reduced H3K9me3 binding to ALT telomeres could

be anticipated to affect HP1-γ binding relative to telomeric content in this

manner. Hp1-γ levels were found to be almost two-fold higher in telomerase

positive versus ALT cells from Western analysis (section 5.2.1). Given that global

levels of H3K9me3 do not differ between the two TMM types, this could

indicate an additional role for HP1-γ besides chromatin compaction in

telomerase positive cells.

Figure 6.2:6 Comparison of Western and ChIP results. ChIP results have been

internally normalised to facilitate comparison. a H3K9me3 b lamin A/C c HP1-α

d HP1-γ

Page 193: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

178

There was no observable trend between levels of HP1-α and levels of bound

telomeric DNA, and increased levels of HP1-α in telomerase positive cells did

not reflect an increase in HP1-α association with telomeres (Figure 6.2:6c). This

difference may be due to the differential roles of HP-1γ and HP1-α at

heterochromatin. While HP1-α is primarily associated with centromeric

chromatin (Frydrychova et al., 2008; Hayakawa et al., 2003; Perrini et al., 2004),

HP1-γ may locate to both heterochromatin and euchromatin (Luke and Lingner,

2009; Schoeftner and Blasco, 2010).

TERRAs are Telomeric repeat-containing RNA transcribed at telomeres and may

have a role in epigenetic remodelling of telomeres and telomerase expression

(Minc et al., 2000). Arnoult et al. (2012) found HP1-α expression is required to

repress TERRA expression, while TERRA expression is elevated in ALT cells

(Episkopou et al., 2014). Lower expression of HP1-α in ALT cells could therefore

contribute to TERRA up-regulation. Reduced methylation of H3K9 and other

histone residues associated with closed chromatin also results in up-regulated

TERRA expression (Arnoult et al., 2012), which is again consistent with the

findings here. The authors of these studies suggest that the increased level of

TERRA expression in ALT cells is conducive to homologous recombination, as

was found to be the case in yeast (Balk et al., 2013). Increased levels and

association of HP1-γ could also play a role in TERRA expression as, in co-

ordination with H3K9me3, HP1-γ is able to induce transcription (Vakoc et al.,

2005).

In the time since these experiments were carried out, one group has attempted

to carry out ChIP on human telomeric DNA with lamin A, and also experienced

low chromatin recovery (Crabbe et al., 2012). During validation and

optimisation, ChIP runs using the lamin A/C antibody pulled down considerably

more telomeric chromatin (>8% input) than in the final data presented. This

drop in efficiency reflects the difficulties of ChIP, which are mentioned by the

Page 194: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

179

authors of both the Crabbe et al. and Episkopou et al. studies, and these

problems are enhanced when attempting IP with a non-direct chromatin

interactor such as lamin A. The study by Crabbe et al. moved away from

traditional ChIP, employing a more typical protein IP method after protein-

chromatin formaldehyde cross-linking, by assaying protein rather than

chromatin IP levels. Under these conditions, the complex structures and

interactions at the nuclear membrane would most likely result in the majority of

protein IP to be as a result of protein-protein interactions, As such, co-IPs would

be a comparable approach, and further experiments utilising lamin A antibodies

could be attempted in ALT and telomerase positive cells. As the variation in A-

Type lamin levels could reflect an altered mobility in the ALT nucleus, it is

possible that interactions between telomeric chromatin (and genomic

chromatin in general) and lamins or their chromatin linkers are not altered, and

are merely more motile due to increased intranuclear lamin mobility.

Page 195: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

180

Chapter 7 Final Discussion

7.1 Accelerated telomere shortening in patient cells

expressing farnesylated lamin A forms

The first aim of this project was to establish whether the short telomeres

previously observed in HGPS cell lines are a result of an increase in the erosion

rate of telomeres. The STELA results presented in chapter three indicated an

elevated telomere shortening rate in patient cell lines with mutations resulting

in retention of C-terminal farnesylation in lamin A products. Closer analysis of

STELA results indicated clear differences in shortening rates between the 12q

and XpYp telomeres in some cell lines. The ranges of shortening rates at each

telomere did not overlap for the cell lines NHDF, 155-BR and E578V, and a

marginal overlap of 3bp/PD was observed at T623S. This may have been

attributable to unusual telomere dynamics related to senescence or X-

inactivation at XpYp (Martens et al., 2000; Surrallés et al., 1999).

As STELA provides a high-resolution profile of single telomere dynamics, an in-

depth analysis of telomere dynamics was carried out to identify any issues that

may arise when deriving shortening rate relative to population doublings. This

highlighted several problematic factors regarding the acquisition of an accurate

mean PD rate. Although the proportion of senescent cells increases over time,

potentially causing an increase of cells lost from cell counts, models indicated

that this may have a minimal effect on telomere shortening rate relative to PD.

It could be proposed that the deficiencies of HGPS cells, which may result in

loss of the 3’ overhang and therefore not be recordable by STELA, could be

causing an increase in cell death which could reduce the proliferative capacity of

the population. However, this was not reflected in a cell viability assay.

Page 196: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

181

Models also revealed that amplification failure of subpopulations of extremely

long telomeres (>15-20kb), may have an impact on deriving an accurate

shortening rate, and that both this and the loss of senescent cells will lead to a

tendency to underestimate telomere shortening rate. By understanding these

subpopulation dynamics, it is possible to account for PCR efficiency issues

through exclusion of any subpopulations that drop in through the analysis,

although these should only arise in instances where telomeres in some cells are

extremely long. It was not possible to identify through the data accumulated

whether increased numbers of cells enter senescence due to telomere damage

signalling arising from damage rather than critical shortening, although very

short telomeres (<400bp) were observed in the HGPS cell line.

Another issue that could not be addressed through modelling based on STELA

results is the effect of accumulating numbers of senescent cells on the

representativeness of the mean PD measurement. High levels of senescent cells,

or even a subpopulation that is slow growing due to high levels of DNA

damage, will result in an overall delay in the time it takes for a population to

complete a mean doubling. That is, cells that are actively dividing will double

more times than is evident in the mean PD measurement. Although the purpose

of calculating a mean PD measurement is to account for variations within the

population, the increasing proportion of mitotically static senescent cells which

are not contributing to division but remain in the population, will continually

skew the mean PD towards a lower value than represents the dividing

population in which telomeres are shortening. The resulting underestimation of

PD number results in an overestimation of telomere shortening relative to PD.

The analysis of shortening rates in the cell lines T623S and RD over time, which

were available at early enough PDs to afford telomere length measurements at

time points with high and low levels of senescence, indicated that telomere

shortening rate is elevated in these cell lines, and does not vary greatly between

early and late time point measurements. As an increase in senescent cells would

Page 197: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

182

be expected to increase the shortening rate, this implies that this effect is either

minor enough to not impact the telomere shortening rate relative to PD, or it is

cancelled out by the anticipated observed reduction in shortening rate due to

senescent cell accumulation described above. In any future studies on primary

cell lines, it would be essential to observe telomere shortening rate from as

early a PD as possible. Ideally, clonal populations with less telomere length

variability should be derived, and it is unfortunate that the limited doubling

capacity of HGPS and other laminopathy cell lines precludes the option of

isolating clonal cultures with longer telomeres before senescence levels

interfere.

7.1.1 Future work

Efforts were made to isolate the cause of accelerated telomere shortening in

patient cells. Methods to measure the length of the 3’ overhang were

attempted but failed to yield reliable results (see Chapter 4). Other potential

pathways resulting in accelerated telomere shortening in patient cell lines were

also considered, and several experimental options are out-lined below.

7.1.1.1 The role of 53BP1

53BP1 affects overhang length (Sfeir and de Lange, 2012), but also has key roles

in DNA damage repair. Telomeres in HGPS cells suffer increased DNA damage

compared to the rest of the genome (Benson et al., 2010), while 53BP1 is

recruited more slowly to sites of DNA damage in HGPS cells (Liu et al., 2005),

and is degraded in the absence of lamin A in MEFs (Gonzalez-Suarez et al.,

2011). Therefore, 53BP1 could play other roles in telomere integrity, and rescue

of 53BP1 levels in patient cells may result in elimination of increased erosion.

Low levels of 53BP1 in BRCA1 deficient breast tumour cells may be restored by

treatment with vitamin D via its interruption of CTSL activity (Gonzalez-Suarez et

al., 2011; Grotsky et al., 2013). In order to test the feasibility of vitamin D

Page 198: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

183

treatment in patient cells, attempts were made to down-regulate activation of

CTSL in U2OS cells with vitamin D, with an ultimate goal of repeating the

experiment in patient and control cells for as many PDs as possible in order to

derive a telomere shortening rate. U2OS cells were exposed continually to

vitamin D for several days before CTSL and 53BP1 levels were assayed by

Western blot analysis. Unfortunately, preliminary results were unreliable (data

not shown). As increased levels of 53BP1 could have consequences for the cell,

and CTSL alone is not sufficient to eliminate 53BP1 degradation (Gonzalez-

Suarez et al., 2011), it is possible that up-regulation of 53BP1 is tightly regulated

through another pathway. It would therefore be necessary to repeat these

experiments in 53BP1-deficient cells, ideally in a patient line or BRCA1-deficient

line.

Direct quantification of 53BP1 in laminopathy cell lines has not been published

previously. Preliminary results gained from Western blots of patient cell lysates

indicated a trend for 53BP1 levels to decrease with the number of cell divisions

in normal and patient cell lines. Unexpectedly, relative levels of 53BP1 in HGPS

(and RD) were higher than controls and cell lines from patients with milder

phenotypes at final passages (Figure 7.1:1). This is somewhat at odds with the

observation that 53BP1 is slow to recruit to sites of DNA damage in patient

cells, and the low levels of 53BP1 in Lmna-/- mouse models, although the

complete absence of lamin A in these mice could account for the discrepancy. It

is possible that the constant state of DDR activation, and the potential continual

degradation of 53BP1 in HGPS and RD cells could result in 53BP1 upregulation,

compensating for proteasomal action, while still effecting the efficient

recruitment of 53BP1.

Toxicity of vitamin D has been shown to be tolerable for use in human patients,

however, vitamin D may cause a reduction in proliferation rate of cells (Chen et

al., 2000). If slow growth rate is a factor in the accruement of telomere loss, and

Page 199: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

184

vitamin D did have a negative effect on cell growth rate, appropriate controls

would need to be considered. Culturing the cells under normoxic conditions is

one potential measure that may compensate for a reduction in growth rate and

a side-by-side comparison against cells grown under standard hyperoxic

conditions could reveal a time-dependent mechanism by which telomeres

shorten. If 53BP1 were found to play a role in telomere shortening, overhang

length would ultimately still need to be confirmed as 53BP1 is involved in a

broad range of events and processes that could impact telomere biology (see

section 1.3.1).

Figure 7.1:1 53BP1 levels in control and

laminopathy cell lines a Whole cell lysates were

derived from cell pellets harvested at various

PDs and immunoblotted against a 53BP1

antibody. Due to the size and number of

modifications present on 53BP1, several bands

are visible for each sample, of which all were

quantified b Due to the large size of 53BP1

(250kDa) it was necessary to run a control

(GAPDH) on a separate gel c 53BP1 signal was

normalised to GAPDH. au=arbritary units.

Page 200: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

185

7.1.1.2 Cohesion

Shortened telomeres have been observed in dyskeratosis congentia (DC)

patients with mutations in the Shelterin component TIN2 (Walne et al., 2008).

Overexpression of TIN2 with DC mutations results in telomere shortening in

tumour cells (Yang et al., 2011). TIN2 is also required for sister-telomere

cohesion (Canudas and Smith, 2009) through the recruitment of HP1-α and –γ

(Canudas et al., 2011). Mutations in the region of the HP1 binding motif in TIN2

disrupt this interaction and result in telomere shortening in association with a

loss of cohesion (Canudas et al., 2011).

HP1-α is reduced in HGPS cells, and preliminary data suggests a trend towards

a reduction in HP1-γ levels in cells with age, and in HGPS cells (Figure 7.1:2). If

HP1 proteins are required for cohesion, laminopathy cells may suffer a drop in

sister telomere cohesion, and resultant effects on telomere length. Interestingly,

Canudas et al. (2011) found that in the presence of mutated TIN2, lymphoblasts

proved less susceptible to cohesion loss. Lamin A is found at considerably low

concentration in lymphoblastoid lines (Figure 7.1:3) in accordance with previous

reports of low A-type lamin expression (Guilly et al., 1987; Rober et al., 1990).

Figure 7.1:2 HP1-γ levels in control and laminopathy cell lines a Whole cell lysates were

derived from cell pellets harvested at various PDs and immunoblotted against a 53BP1

antibody b Signal was normalised to GAPDH signal (run on the same gel) used for

quantification in Figure 7.1:1. au=arbitrary units

Page 201: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

186

Additionally, telomere length in lymphocytes of HGPS patients is not reduced

relative to controls, in contrast to telomere length in fibroblasts from the same

patients (Decker et al., 2009).

If HP1 is reduced by the presence of mutant lamin A, sister-telomeres in HGPS

cells may not be able to maintain normal levels of cohesion, which could result

in accelerated telomere shortening through a similar mechanism to DC. Utilising

the techniques from the Canudas et al. study, it would be possible to record

sister telomere cohesion in patient cell lines via FISH on specific chromosome

ends in combination with a telomere-specific PNA probe in interphase cells. As

such, BACs containing 12q and 16p terminal sequences were purchased in order

to produce telomere-specific probes, and these are currently in development in

the Royle laboratory. As lymphoblastoid cells contain very low levels of lamin A,

lymphoblasts could be used as an ideal control to identify whether farnesylated

and mutated lamin A forms or if over-accumulation of A-type lamins alone are

the major contributors to HGPS cellular phenotype, without the need for

ectopic overexpression. A role for HP1 could be verified by recording HP1 levels

in lymphoblasts.

Figure 7.1:3 Western blot of whole cell lysates derived from lymphoblastoid lines KK, KUK

and GM1229 and various fibroblast-derived lines. Lysates were immunoblotted against a

lamin A/C antibody (LA) Loading control GAPDH is shown for comparison. Short=short

exposure, long=long exposure

Page 202: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

187

7.2 Altered lamin state in ALT cells may enable the ALT

phenotype

Investigation of lamin levels and localisation in ALT and telomerase positive

cancer cells revealed an association between lower levels of lamins and less

lamin integration into the nuclear matrix in ALT cells. Previous reports would

indicate that the mobility of nuclear factors and telomeres in ALT cells is

elevated (Jegou et al., 2009), and that this plays a role in the biology of ALT

telomere maintenance. The organisation of chromatin within the nucleus is

highly dependent on lamina-associated factors (Zuleger et al., 2013).

Intranuclear lamins mediate and stabilise DNA and repair factors (Mahen et al.,

2013), and are required for efficient replication (Spann et al., 1997). The data

presented here suggest further evidence for a physical connection between

intranuclear lamins, telomeres and associated factors required for repair and

replication, as well as alternative conformation of macromolecular structures

within the nucleus compared to the periphery, although an intranuclear network

remains to be reproducibly visualised to a satisfactory level.

7.2.1 Future work

The immunofluorescence results are indicative of a shift in the stability of the

secondary structure of peripheral and intranuclear lamins in ALT cells.

Visualisation of altered lamin mobility in ALT cells with the use of FRAP would

require the introduction of fluorescently-labelled lamin proteins. Unfortunately,

a contributing factor to the altered lamin organisation in ALT cells is likely due

to the reduced amount of lamins recorded in these cells, as the subnuclear

organisation of lamins is concentration dependent (Guo et al., 2014). To avoid

interfering with lamin levels, solubility assays may be carried out instead, using

telomerase positive and ALT cells without any alteration in expression. This

would confirm whether the lamina is adopting a less complex and insoluble

Page 203: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

188

structure in ALT versus telomerase positive cells. In order to observe whether

this change in nuclear environment is able to affect telomere dynamics, it would

be necessary to record telomere movements in live cells through the

introduction of a fluorescently labelled telomere tag.

Once this is established, it may be possible to make use of the concentration-

dependent nature of lamin organisation. Lamin B has been shown to have a role

in post-mitotic organisation of chromatin and is required for chromatin

compartmentalisation (Dechat et al., 2004; Martin et al., 2010). Although lamin B

is not dependent on lamin A for its organisation within the nucleus (Shimi et al.

2008), lamin B may be excluded from blebs and lobulations in HGPS cells that

still contain lamin A (Goldman et al., 2004) and recent findings suggest that

coordination of lamina organisation is dictated by the total concentration of

lamins, and more specifically lamin A (Guo et al., 2014). It is therefore highly

likely that re-introduction of lamin A could result in redistribution of both A-

and B-type lamins, and an increased formation of higher-order complexes. This

could potentially restore local telomere stabilisation, at least in part, and

subsequently may reduce the heterogeneity of ALT telomere length through

repression of recombination. Live microscope imaging recording telomere

movement could be undertaken before and after ectopic introduction of A-type

lamins or progerin (which also promotes aggregation and stabilisation of

lamins) into ALT cells. Various aspects of ALT biology in these cells could then

also be undertaken. In order to determine the effects of lamins on

recombination, co-FISH could be carried out and rates of sister-chromatid

exchange compared before and after increased lamin expression. In order to

explore the putative link between lamins and APB organisation, the dynamics

and association of APBs and telomeres could be recorded by recreating live cell

imaging experiments carried out by Jegou et al. (2009). Stable expression and

extended tissue culture of ALT cells would provide the opportunity to carry out

STELA to confirm whether telomere length and distribution are effected by the

Page 204: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

189

gain of lamins. Also, alteration of chromatin mobility could impact instability

levels at the MS32 minisatellite locus, which could be assayed as described in

section 2.4.

SUN1 has roles in mitosis, as does lamin B, and specifically binds telomeres

during post-mitotic assembly (Crabbe et al., 2012). It is possible that nuclear re-

assembly is different in ALT and telomerase cells, as the comparatively increased

amount of SUN1 in telomerase positive cells could provide increased telomere

anchorage through increased RAP1 at re-assembly, in manner undetectable by

ChIP in unsynchronised cells (Crabbe et al., 2012). Nuclear disassembly could

also be affected by SUN1 levels (Patel et al., 2014; Turgay et al., 2014). As these

interactions are cell cycle-dependent, cell synchronisation prior to quantification

by Western blot as in section 5.2.1 could reveal more dramatic differences

between the cell lines. As was suggested in section 6.2.3, protein co-IP may be a

more robust method for identification of differential levels of lamin A or SUN1

interaction with chromatin components, as well as with one another in ALT and

telomerase positive cells. Cell synchronisation and sampling at different points

in the cell cycle prior to IP may provide more insight into these interactions.

7.3 Concluding remarks

In the case of HGPS and related laminopathies, the existence of a

telomere:lamin connection has great implications for the telomere biology of

laminopathy cells. The retention of the farnesyl group at the C-terminus of

some mutated forms of lamin A associates with an increase in telomere

shortening rate, and could therefore contribute to the elevated levels of

senescent cells found in patient tissues and the accelerated ageing phenotype.

In these cell lines lamin A is integrated more readily into the nuclear matrix,

greatly depleting the intranuclear fraction. This could be related to a sliding

scale whereby ALT cells represent a highly mobile internal nuclear structure in

Page 205: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

190

which telomeres move rapidly and may be recruited to sites of recombination

and replication. Telomerase positive cells represent an intermediate state,

retaining some lamins within the nucleus which may perform key nuclear

functions, while progeroid laminopathy lines that retain farnesylation, in which

reorganisation of lamins inhibits the motility of intranuclear lamin structures (as

well as the amount of intranuclear lamin), exhibit replication and repair

deficiencies. It is likely that the telomerase positive lamin state represents

something similar to a normal cell line, and further experiments into telomere

motility in cancer cells would require the inclusion of normal cell lines to verify

this. Results of chromatin immunoprecipitation suggest that these alterations in

lamina composition do not impede the degree to which lamins interact with

chromatin and it is the physical relocation and flexibility of lamins integrated

into the intranuclear lamin network versus the periphery which dictate

chromatin organisation and motility, and therefore relative frequency of

telomere co-localisation with replication, repair and recombination factors.

In conclusion, nuclear lamina composition is linked to the telomere

maintenance mechanism adopted by a cell line. The reduction in A-type lamins

in ALT cells could have multiple knock-on effects including increased mobility,

and subsequently increased recombination events of ALT telomeres. The

reduced presence of lamins and their associated factors may be necessary to

facilitate telomere exchange in ALT cells by allowing freer movement and

recombination of telomeres, while in progeroid laminopathies telomere motility

is likely impeded, diminishing the ability of the cell to repair DNA damage at

telomeres. A series of experiments investigating telomere motility could confirm

this theory. Interestingly, although lamin A deficiency in mice results in a

reduction in 53BP1 levels, preliminary data suggest this is not the case in HGPS

and RD cells. Also, reduced lamin A/C in ALT cells does not correlate with

reduced 53BP1. This could support a major role for telomere organisation and

Page 206: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

191

motility in the telomere biology of laminopathy cells that is not dependent on

53BP1.

Page 207: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

192

Appendices

Appendix A Materials and Methods supplemental

PCR buffer: Prepared as 11.1X mixture by Dr Nicola Royle and diluted to 1X in

PCR reactions

Reagent 11.1X

concentration

1X

concentration

Tris HCl pH 8.8 500mM 45mM

Ammonium Sulphate 122mM 11mM

MgCl2 50mM 4.5mM

β-mercaptoethanol 74mM 6.7mM

EDTA pH 8.0 49µM 4.4µM

dATP 11.1mM 1mM

dCTP 11.1mM 1mM

dGTP 11.1mM 1mM

dTTP 11.1mM 1mM

BSA 1.25mg/ml 113µg/ml

Denhardt’s solution: Prepared a as a 50X stock and diluted 1 in 10 for a final

5X concentration in hybridisation solution.

FC: 0.5% Ficoll 400, 0.5% BSA, 0.5% polyvinylpyrrolidone (all w/v)

SSC: Taken from Departmental 20X stock solution and diluted as indicated in

Materials and Methods section.

20X: 3M NaCl, 0.3M Na3C6H5O7

TBE: Taken from Departmental 10X/5X stock and diluted to 0.5-1X as indicated

in Materials and Methods section.

10X: 0.89M Tris, 0.89M Boric acid, 20mM EDTA

Page 208: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

193

Oligonucleotide labelling buffer (OLB)

OLB was prepared as a 6X in the department by preparation of 3 separate

solutions which were then combined:

Solution A (10µl):

1.25ml 2M Tris HCL pH 8.0, 50µl 5M Magnesium Chloride, 700µl Elga water

(double deionised/distilled), 36µl β-mercaptoethanol

10µl each (100mM stock): dATP, dGTP, dTTP

Solution B (25µl):

2M Hepes pH 6.6

Solution C (15µl):

Pd(N)6 Random Hexamer Sodium Salt (Fisher/Fermentas #SO142). Provided as

a 120µl stock containing 24ug of Hexamer.

Page 209: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

194

Appendix B STELA

B.1 STELA Southern blots

Representative images of STELA hybridisations for each telomere assayed for each cell

line. Population doubling (PD) is indicated at the top of each gel image, as well as the

HT1080 technical control, and H2O PCR controls. All PCRs, gel electrophoresis and

Southern hybridisations carried out by Rachel Turner unless otherwise stated.

Page 210: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

195

RD XpYp PCR, gel electrophoresis and Southern hybridisations carried out by Kimberly

Fitzpatrick (KF).

Page 211: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

196

T623S 12q PCR, gel electrophoresis and Southern hybridisations carried out by both

Rachel Turner (RT) and Kim Fitzpatrick (KF).

Page 212: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

197

B.2 HT1080 control data

Page 213: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

198

Quantified sizes of HT1080 products from STELAs for each gel carried out in the STELA

analysis.

Page 214: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

199

Appendix C Confirmation of cell line sub-telomere

genotypes

C.1 12q genotyping

Due to the presence of sub-populations in the STELA data, it was theorised that

these populations could derive from two parental alleles. Given the difficulties

interpreting particularly the NHDF and HGPS 12q data, allele-specific STELA

could be employed to isolate a single subpoplulation for analysis, reducing the

data distribution. Genotyping was undertaken as described in (Baird et al.,

2000).

Firstly, PCR was conducted to identify the presence or absence of a ~2.2kb

deletion in each of the sample DNAs. PCR was then conducted on a small

region of the 12q sub-telomere containing SNPs within each of three restriction

sites that are fully informative for 12q subtelomere genotype, as well as a 29

base insertion/deletion (see table C.1-1 and section 2.12.1). Results of the

restriction digest are shown in Figure C.1:1 and Table C.1-2.

Table C.1-1 12q subtelomere genotypes and relative positions of informative restriction sites.

*(Baird et al., 2000)

Page 215: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

200

Table C.1-2 SNP typing and haplotypes from results in Figure C.1:1

Figure C.1:1

12q genotyping a PCR products from the

deletion assay run on a 1% agarose gel.

133101 and 134602 are CEPH DNAs

previously characterised as negative and

positive, respectively, for the 2.2kb

deletion b & c PCR products were

digested with indicated restriction enzyme

and run on a 3% NuSieve agarose gel

Page 216: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

201

C.2 XpYp genotyping

Genotyping was carried out on XpYp for all patient and control cells to further

characterise the cell lines. In a similar approach to 12q, a region of the XpYp

sub-telomere was amplified and restriction analysis carried out as described in

(Baird et al., 1995)

Details of XpYp sub-telomere haplotypes as defined by informative SNPs within

restriction sites (Table C.2-1 and section 2.12.2). Results from the restriction

analysis are shown in Figure C.2:1 and summarised in Table C.2-2.

Table C.2-1 XpYp subtelomere genotypes and relative positions of informative restriction sites

*(Baird et al., 1995)

Figure C.2:1 XpYp genotyping. PCR products were digested with indicated restriction enzyme

and run on a 3% NuSieve agarose gel

Page 217: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

202

Table C.2-2 SNP typing and haplotypes from results in Figure C.2:1

Appendix D Confirmation of cell line purity with MS32

analysis

Results of MS32 screening. PCR products were resolved in acrylamide gels before

Southern blotting and hybridisation with MS32-specific probe.

T623S 155-BR E578V NHDF HT1080 RD HGPS

Page 218: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

203

Appendix E NT2D1 exclusion

Results from Western blotting and quantification of telomerase cell lines for each of the

proteins indicated. NT2D1 was separated from the telomere groups and t tests

between NT2D1 values and the remaining telomerase lines were carried out. * indicates

level of significance, ns=not significant, au=arbitrary units.

Page 219: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

204

Appendix F ChIP

Figures F1-3: Intensity measurements taken from dot blot input signals from

each replica. Four dilutions were loaded onto the membrane in duplicate for

each ChIP. Intercept was set at zero and the slope derived from line of best fit

was used to normalise the percentage input for each ChIP. au=arbitrary units

Figure F1

First ChIP replica

Figure F2

Second ChIP replica

Page 220: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

205

Table F-1 slope gradients and r2 values derived from input dilution series’ in Figures F1-3.

Figure F3

Third ChIP replica

Page 221: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

206

References

Aaronson, R.P., Blobel, G., 1975. Isolation of nuclear pore complexes in

association with a lamina. Proc. Natl. Acad. Sci. U. S. A. 72, 1007–1011.

doi:10.1073/pnas.72.3.1007

Acevedo, L., Leonardo Iniguez, a., Holster, H., Zhang, X., Green, R., Farnham, P.,

2007. Genome-scale ChIP-chip analysis using 10,000 human cells.

Biotechniques 43, 791–797. doi:10.2144/000112625

Adli, M., Bernstein, B.E., 2011. Whole-genome chromatin profiling from limited

numbers of cells using nano-ChIP-seq. Nat. Protoc. 6, 1656–68.

doi:10.1038/nprot.2011.402

Agarwal, A.K., Fryns, J.P., Auchus, R.J., Garg, A., 2003. Zinc metalloproteinase

ZMPSTE24, is mutated in mandibuloacral dysplasia. Hum. Mol. Genet. 12,

1995–2001. doi:10.1093/hmg/ddg213

Agarwal, A.K., Zhou, X.J., Hall, R.K., Nicholls, K., Bankier, A., Van Esch, H., Fryns, J.-

P., Garg, A., 2006. Focal segmental glomerulosclerosis in patients with

mandibuloacral dysplasia owing to ZMPSTE24 deficiency. J. Investig. Med.

54, 208–213. doi:10.2310/6650.2006.05068

Ahmad, Z., Zackai, E., Medne, L., Garg, A., 2010. Early onset mandibuloacral

dysplasia due to compound heterozygous mutations in ZMPSTE24. Am. J.

Med. Genet. A 152A, 2703–10. doi:10.1002/ajmg.a.33664

Al-Haboubi, T., Shumaker, D.K., Köser, J., Wehnert, M., Fahrenkrog, B., 2011.

Distinct association of the nuclear pore protein Nup153 with A- and B-type

lamins. Nucleus 2, 500–9. doi:10.4161/nucl.2.5.17913

Allshire, R.C., Dempster, M., Hastie, N.D., 1989. Human telomeres contain at

least three types of G-rich repeat distributed non-randomly. Nucleic Acids

Res. 17, 4611–4627. doi:10.1093/nar/gkn942

Allsopp, R., Chang, E., Kashefi-Aazam, M., Rogaev, E., Piatyszek, M.A., Shay, J.W.,

Harley, C.B., 1995. Telomere shortening is associated with cell division in

vitro and in vivo. Exp. Cell Res. 220, 194–200.

doi:http://dx.doi.org/10.1006/excr.1995.1306

Allsopp, R.C., Harley, C.B., 1995. Evidence for a critical telomere length in

senescent human fibroblasts. Exp. Cell Res. doi:10.1006/excr.1995.1213

Page 222: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

207

Allsopp, R.C., Vaziri, H., Patterson, C., Goldstein, S., Younglai, E. V, Futcher, a B.,

Greider, C.W., Harley, C.B., 1992. Telomere length predicts replicative

capacity of human fibroblasts. Proc. Natl. Acad. Sci. U. S. A. 89, 10114–8.

Al-Shali, K.Z., Hegele, R.A., 2004. Laminopathies and atherosclerosis. Arterioscler.

Thromb. Vasc. Biol. doi:10.1161/01.ATV.0000136392.59656.8b

Arat, N.Ö., Griffith, J.D., 2012. Human Rap1 interacts directly with telomeric DNA

and regulates TRF2 localization at the telomere. J. Biol. Chem. 287, 41583–

41594. doi:10.1074/jbc.M112.415984

Arnoult, N., Van Beneden, A., Decottignies, A., 2012. Telomere length regulates

TERRA levels through increased trimethylation of telomeric H3K9 and

HP1α. Nat. Struct. Mol. Biol. 19, 948–956. doi:10.1038/nsmb.2364

Artandi, S.E., DePinho, R.A., 2010. Telomeres and telomerase in cancer.

Carcinogenesis 31, 9–18. doi:10.1093/carcin/bgp268

Bai, S., Lozada, A., Jones, M.C., Dietz, H.C., Dempsey, M., Das, S., 2014.

Mandibuloacral Dysplasia Caused by LMNA Mutations and Uniparental

Disomy. Case Rep. Genet. 2014, 508231. doi:10.1155/2014/508231

Bailey, J.A., Yavor, A.M., Massa, H.F., Trask, B.J., Eichler, E.E., 2001. Segmental

duplications: Organization and impact within the current human genome

project assembly. Genome Res. 11, 1005–1017. doi:10.1101/gr.GR-1871R

Baird, D.M., Britt-Compton, B., Rowson, J., Amso, N.N., Gregory, L., Kipling, D.,

2006. Telomere instability in the male germline. Hum. Mol. Genet. 15, 45–

51. doi:10.1093/hmg/ddi424

Baird, D.M., Coleman, J., Rosser, Z.H., Royle, N.J., 2000. High levels of sequence

polymorphism and linkage disequilibrium at the telomere of 12q:

implications for telomere biology and human evolution. Am. J. Hum. Genet.

66, 235–250. doi:10.1086/302721

Baird, D.M., Davis, T., Rowson, J., Jones, C.J., Kipling, D., 2004. Normal telomere

erosion rates at the single cell level in Werner syndrome fibroblast cells.

Hum. Mol. Genet. 13, 1515–24. doi:10.1093/hmg/ddh159

Baird, D.M., Jeffreys, A.J., Royle, N.J., 1995. Mechanisms underlying telomere

repeat turnover, revealed by hypervariable variant repeat distribution

patterns in the human Xp/Yp telomere. EMBO J. 14, 5433–5443.

Page 223: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

208

Baird, D.M., Rowson, J., Wynford-Thomas, D., Kipling, D., 2003. Extensive allelic

variation and ultrashort telomeres in senescent human cells. Nat. Genet. 33,

203–7. doi:10.1038/ng1084

Balk, B., Maicher, A., Dees, M., Klermund, J., Luke-Glaser, S., Bender, K., Luke, B.,

2013. Telomeric RNA-DNA hybrids affect telomere-length dynamics and

senescence. Nat. Struct. Mol. Biol. 20, 1199–205. doi:10.1038/nsmb.2662

Barrowman, J., Wiley, P.A., Hudon-Miller, S.E., Hrycyna, C.A., Michaelis, S., 2012.

Human ZMPSTE24 disease mutations: Residual proteolytic activity

correlates with disease severity. Hum. Mol. Genet. 21, 4084–4093.

doi:10.1093/hmg/dds233

Baumann, P., Cech, T.R., 2001. Pot1, the putative telomere end-binding protein

in fission yeast and humans. Science (80-. ). 292, 1171–1175.

doi:10.1126/science.1060036

Beck, L.A., Hosick, T.J., Sinensky, M., 1990. Isoprenylation is required for the

processing of the lamin A precursor. J. Cell Biol. 110, 1489–1499.

doi:10.1083/jcb.110.5.1489

Benetti, R., Gonzalo, S., Jaco, I., Schotta, G., Klatt, P., Jenuwein, T., Blasco, M.A.,

2007. Suv4-20h deficiency results in telomere elongation and derepression

of telomere recombination. J. Cell Biol. 178, 925–36.

doi:10.1083/jcb.200703081

Ben-Harush, K., Wiesel, N., Frenkiel-Krispin, D., Moeller, D., Soreq, E., Aebi, U.,

Herrmann, H., Gruenbaum, Y., Medalia, O., 2009. The Supramolecular

Organization of the C. elegans Nuclear Lamin Filament. J. Mol. Biol. 386,

1392–1402. doi:10.1016/j.jmb.2008.12.024

Benson, E.K., Lee, S.W., Aaronson, S.A., 2010. Role of progerin-induced telomere

dysfunction in HGPS premature cellular senescence. J. Cell Sci. 123, 2605–

12. doi:10.1242/jcs.067306

Bergo, M.O., Gavino, B., Ross, J., Schmidt, W.K., Hong, C., Kendall, L. V, Mohr, A.,

Meta, M., Genant, H., Jiang, Y., Wisner, E.R., Van Bruggen, N., Carano, R. a D.,

Michaelis, S., Griffey, S.M., Young, S.G., 2002. Zmpste24 deficiency in mice

causes spontaneous bone fractures, muscle weakness, and a prelamin A

processing defect. Proc. Natl. Acad. Sci. U. S. A. 99, 13049–54.

doi:10.1073/pnas.192460799

Page 224: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

209

Bernardi, R., Pandolfi, P.P., 2007. Structure, dynamics and functions of

promyelocytic leukaemia nuclear bodies. Nat. Rev. Mol. Cell Biol. 8, 1006–

1016. doi:10.1038/nrm2277

Bione, S., Maestrini, E., Rivella, S., Mancini, M., Regis, S., Romeo, G., Toniolo, D.,

1994. Identification of a novel X-linked gene responsible for Emery-Dreifuss

muscular dystrophy. Nat. Genet. 8, 323–327. doi:10.1038/ng1294-323

Blackburn, E.H., 2001. Switching and signaling at the telomere. Cell 106, 661–

673. doi:10.1016/S0092-8674(01)00492-5

Blasco, M.A., 2007. The epigenetic regulation of mammalian telomeres. Nat. Rev.

Genet. 8, 299–309. doi:10.1038/nrg2047

Bodnar, a. G., Ouellette, M., Frolkis, M., Holt, S.E., Chiu, C.-P., Morin, G.B., Harley,

C.B., Shay, J.W., Lichtsteiner, S., Wright†, W.E., 1998. Extension of Life-Span

by Introduction of Telomerase into Normal Human Cells. Science (80-. ).

279, 349–352. doi:10.1126/science.279.5349.349

Boguslavsky, R.L., Stewart, C.L., Worman, H.J., 2006. Nuclear lamin A inhibits

adipocyte differentiation: Implications for Dunnigan-type familial partial

lipodystrophy. Hum. Mol. Genet. 15, 653–663. doi:10.1093/hmg/ddi480

Bossie, C.A., Sanders, M.M., 1993. A cDNA from Drosophila melanogaster

encodes a lamin C-like intermediate filament protein. J. Cell Sci. 104, 1263–

1272.

Bothmer, A., Robbiani, D.F., Di Virgilio, M., Bunting, S.F., Klein, I.A., Feldhahn, N.,

Barlow, J., Chen, H.T., Bosque, D., Callen, E., Nussenzweig, A., Nussenzweig,

M.C., 2011. Regulation of DNA End Joining, Resection, and Immunoglobulin

Class Switch Recombination by 53BP1. Mol. Cell 42, 319–329.

doi:10.1016/j.molcel.2011.03.019

Bothmer, A., Robbiani, D.F., Feldhahn, N., Gazumyan, A., Nussenzweig, A.,

Nussenzweig, M.C., 2010. 53BP1 regulates DNA resection and the choice

between classical and alternative end joining during class switch

recombination. J. Exp. Med. 207, 855–865. doi:10.1084/jem.20100244

Brachner, A., Foisner, R., 2011. Evolvement of LEM proteins as chromatin tethers

at the nuclear periphery. Biochem. Soc. Trans. 39, 1735–1741.

doi:10.1042/BST20110724

Page 225: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

210

Britt-Compton, B., Capper, R., Rowson, J., Baird, D.M., 2009a. Short telomeres are

preferentially elongated by telomerase in human cells. FEBS Lett. 583,

3076–3080. doi:10.1016/j.febslet.2009.08.029

Britt-Compton, B., Rowson, J., Locke, M., Mackenzie, I., Kipling, D., Baird, D.M.,

2006. Structural stability and chromosome-specific telomere length is

governed by cis-acting determinants in humans. Hum. Mol. Genet. 15, 725–

33. doi:10.1093/hmg/ddi486

Britt-Compton, B., Wyllie, F., Rowson, J., Capper, R., Jones, R.E., Baird, D.M.,

2009b. Telomere dynamics during replicative senescence are not directly

modulated by conditions of oxidative stress in IMR90 fibroblast cells.

Biogerontology 10, 683–693. doi:10.1007/s10522-009-9216-4

Broccoli, D., Smogorzewska, A., Chong, L., de Lange, T., 1997. Human telomeres

contain two distinct Myb-related proteins, TRF1 and TRF2. Nat. Genet. 17,

231–235. doi:10.1038/ng1097-231

Broers, J.L. V, Peeters, E.A.G., Kuijpers, H.J.H., Endert, J., Bouten, C.V.C., Oomens,

C.W.J., Baaijens, F.P.T., Ramaekers, F.C.S., 2004. Decreased mechanical

stiffness in LMNA-/- cells is caused by defective nucleo-cytoskeletal

integrity: Implications for the development of laminopathies. Hum. Mol.

Genet. 13, 2567–2580. doi:10.1093/hmg/ddh295

Broers, J.L. V, Ramaekers, F.C.S., 2004. Dynamics of nuclear lamina assembly and

disassembly. Symp. Soc. Exp. Biol. 177–192.

Brosig, M., Ferralli, J., Gelman, L., Chiquet, M., Chiquet-Ehrismann, R., 2010.

Interfering with the connection between the nucleus and the cytoskeleton

affects nuclear rotation, mechanotransduction and myogenesis. Int. J.

Biochem. Cell Biol. 42, 1717–1728. doi:10.1016/j.biocel.2010.07.001

Brown, W.R.A., MacKinnon, P.J., Villasante, A., Spurr, N., Buckle, V.J., Dobson,

M.J., 1990. Structure and polymorphism of human telomere-associated

DNA. Cell 63, 119–132. doi:10.1016/0092-8674(90)90293-N

Bryan, T.M., Englezou, A., Dalla-Pozza, L., Dunham, M.A., Reddel, R.R., 1997.

Evidence for an alternative mechanism for maintaining telomere length in

human tumors and tumor-derived cell lines. Nat. Med. 3, 1271–1274.

doi:10.1038/nm1197-1271

Bryan, T.M., Englezou, A., Gupta, J., Bacchetti, S., Reddel, R.R., 1995. Telomere

elongation in immortal human cells without detectable telomerase activity.

EMBO J. 14, 4240–4248.

Page 226: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

211

Bunting, S.F., Callén, E., Wong, N., Chen, H.T., Polato, F., Gunn, A., Bothmer, A.,

Feldhahn, N., Fernandez-Capetillo, O., Cao, L., Xu, X., Deng, C.X., Finkel, T.,

Nussenzweig, M., Stark, J.M., Nussenzweig, A., 2010. 53BP1 inhibits

homologous recombination in brca1-deficient cells by blocking resection of

DNA breaks. Cell 141, 243–254. doi:10.1016/j.cell.2010.03.012

Burma, S., Chen, B.P., Murphy, M., Kurimasa, A., Chen, D.J., 2001. ATM

Phosphorylates Histone H2AX in Response to DNA Double-strand Breaks. J.

Biol. Chem. 276, 42462–7. doi:10.1074/jbc.C100466200

Cai, M., Huang, Y., Ghirlando, R., Wilson, K.L., Craigie, R., Clore, G.M., 2001.

Solution structure of the constant region of nuclear envelope protein LAP2

reveals two LEM-domain structures: One binds BAF and the other binds

DNA. EMBO J. 20, 4399–4407. doi:10.1093/emboj/20.16.4399

Campisi, J., 2013. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75,

685–705. doi:10.1146/annurev-physiol-030212-183653

Campisi, J., d’Adda di Fagagna, F., 2007. Cellular senescence: when bad things

happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–40.

doi:10.1038/nrm2233

Canudas, S., Houghtaling, B.R., Bhanot, M., Sasa, G., Savage, S.A., Bertuch, A.A.,

Smith, S., 2011. A role for heterochromatin protein 1gamma at human

telomeres. Genes Dev. 25, 1807–1819. doi:10.1101/gad.17325211

Canudas, S., Smith, S., 2009. Differential regulation of telomere and centromere

cohesion by the Scc3 homologues SA1 and SA2, respectively, in human

cells. J. Cell Biol. 187, 165–173. doi:10.1083/jcb.200903096

Cao, K., Blair, C.D., Faddah, D.A., Kieckhaefer, J.E., Olive, M., Erdos, M.R., Nabel,

E.G., Collins, F.S., 2011. Progerin and telomere dysfunction collaborate to

trigger cellular senescence in normal human fibroblasts 121, 2833–44.

doi:10.1172/JCI43578.associated

Capanni, C., Mattioli, E., Columbaro, M., Lucarelli, E., Parnaik, V.K., Novelli, G.,

Wehnert, M., Cenni, V., Maraldi, N.M., Squarzoni, S., Lattanzi, G., 2005.

Altered pre-lamin A processing is a common mechanism leading to

lipodystrophy. Hum. Mol. Genet. 14, 1489–1502. doi:10.1093/hmg/ddi158

Capell, B.C., Erdos, M.R., Madigan, J.P., Fiordalisi, J.J., Varga, R., Conneely, K.N.,

Gordon, L.B., Der, C.J., Cox, A.D., Collins, F.S., 2005. Inhibiting farnesylation

of progerin prevents the characteristic nuclear blebbing of Hutchinson-

Page 227: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

212

Gilford progeria syndrome. Proc. Natl. Acad. Sci. U. S. A. 102, 12879–84.

doi:10.1073/pnas.0506001102

Carson, C.T., Schwartz, R.A., Stracker, T.H., Lilley, C.E., Lee, D. V., Weitzman, M.D.,

2003. The Mre11 complex is required for ATM activation and the G2/M

checkpoint. EMBO J. 22, 6610–6620. doi:10.1093/emboj/cdg630

Casey, P.J., Seabra, M.C., 1996. Protein prenyltransferases. J. Biol. Chem. 271,

5289–5292. doi:10.1074/jbc.271.10.5289

Cawthon, R.M., 2002. Telomere measurement by quantitative PCR. Nucleic Acids

Res. 30, e47. doi:10.1093/nar/30.10.e47

Cawthon, R.M., Smith, K.R., O’Brien, E., Sivatchenko, A., Kerber, R.A., 2003.

Association between telomere length in blood and mortality in people

aged 60 years or older. Lancet 361, 393–395. doi:10.1016/S0140-

6736(03)12384-7

Cerone, M.A., Londono-Vallejo, J.A., Bacchetti, S., 2001. Telomere maintenance

by telomerase and by recombination can coexist in human cells. Hum. Mol.

Genet. 10, 1945–1952.

Cesare, A., Hayashi, M., Crabbe, L., Karlseder, J., 2013. The Telomere

deprotection response is functionally distinct from the Genomic DNA

damage response. Mol. Cell 51, 141–155. doi:10.1016/j.molcel.2013.06.006

Cesare, A.J., Griffith, J.D., 2004. Telomeric DNA in ALT cells is characterized by

free telomeric circles and heterogeneous t-loops. Mol. Cell. Biol. 24, 9948–

9957. doi:10.1128/MCB.24.22.9948-9957.2004

Cesare, A.J., Reddel, R.R., 2010. Alternative lengthening of telomeres: models,

mechanisms and implications. Nat. Rev. Genet. 11, 319–30.

doi:10.1038/nrg2763

Chai, W., Shay, J.W., Wright, W.E., 2005. Human telomeres maintain their

overhang length at senescence. Mol. Cell. Biol. 25, 2158–2168.

doi:10.1128/MCB.25.6.2158-2168.2005

Chambliss, A.B., Khatau, S.B., Erdenberger, N., Robinson, D.K., Hodzic, D.,

Longmore, G.D., Wirtz, D., 2013. The LINC-anchored actin cap connects the

extracellular milieu to the nucleus for ultrafast mechanotransduction. Sci.

Rep. 3, 1087. doi:10.1038/srep01087

Chen, L., Lee, L., Kudlow, B.A., Dos Santos, H.G., Sletvold, O., Shafeghati, Y.,

Botha, E.G., Garg, A., Hanson, N.B., Martin, G.M., Mian, I.S., Kennedy, B.K.,

Page 228: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

213

Oshima, J., 2003. LMNA mutations in atypical Werner’s syndrome. Lancet

362, 440–445. doi:10.1016/S0140-6736(03)14069-X

Chen, T.C., Schwartz, G.G., Burnstein, K.L., Lokeshwar, B.L., Holick, M.F., 2000. The

in vitro evaluation of 25-hydroxyvitamin D3 and 19-nor-1alpha,25-

dihydroxyvitamin D2 as therapeutic agents for prostate cancer. Clin. Cancer

Res. 6, 901–908.

Chen, Y., Yang, Y., van Overbeek, M., Donigian, J.R., Baciu, P., de Lange, T., Lei,

M., 2008. A shared docking motif in TRF1 and TRF2 used for differential

recruitment of telomeric proteins. Science (80-. ). 319, 1092–1096.

doi:10.1126/science.1151804

Chiolo, I., Minoda, A., Colmenares, S.U., Polyzos, A., Costes, S. V., Karpen, G.H.,

2011. Double-strand breaks in heterochromatin move outside of a dynamic

HP1a domain to complete recombinational repair. Cell 144, 732–744.

doi:10.1016/j.cell.2011.02.012

Chow, T.T., Zhao, Y., Mak, S.S., Shay, J.W., Wright, W.E., 2012. Early and late steps

in telomere overhang processing in normal human cells: the position of the

final RNA primer drives telomere shortening. Genes Dev. 26, 1167–1178.

doi:10.1101/gad.187211.112

Cimino-Reale, G., Pascale, E., Battiloro, E., Starace, G., Verna, R., D’Ambrosio, E.,

2001. The length of telomeric G-rich strand 3’-overhang measured by

oligonucleotide ligation assay. Nucleic Acids Res. 29, E35.

Clarke, S., Vogel, J.P., Deschenes, R.J., Stock, J., 1988. Posttranslational

modification of the Ha-ras oncogene protein: evidence for a third class of

protein carboxyl methyltransferases. Proc. Natl. Acad. Sci. U. S. A. 85, 4643–

4647. doi:10.1073/pnas.85.13.4643

Columbaro, M., Capanni, C., Mattioli, E., Novelli, G., Parnaik, V.K., Squarzoni, S.,

Maraldi, N.M., Lattanzi, G., 2005. Rescue of heterochromatin organization in

Hutchinson-Gilford progeria by drug treatment. Cell. Mol. life Sci. 62, 2669–

78. doi:10.1007/s00018-005-5318-6

Columbaro, M., Mattioli, E., Schena, E., Capanni, C., Cenni, V., Levy, N., Navarro,

C.L., Del Coco, R., Squarzoni, S., Camozzi, D., Hutchison, C.J., Wehnert, M.,

Lattanzi, G., 2010. Prelamin A processing and functional effects in restrictive

dermopathy. Cell Cycle 9, 4766–4768. doi:10.4161/cc.9.23.14210

Page 229: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

214

Conomos, D., Pickett, H. a, Reddel, R.R., 2013. Alternative lengthening of

telomeres: remodeling the telomere architecture. Front. Oncol. 3, 27.

doi:10.3389/fonc.2013.00027

Constantinescu, D., Csoka, A.B., Navara, C.S., Schatten, G.P., 2010. Defective DSB

repair correlates with abnormal nuclear morphology and is improved with

FTI treatment in Hutchinson-Gilford progeria syndrome fibroblasts. Exp.

Cell Res. 316, 2747–59. doi:10.1016/j.yexcr.2010.05.015

Constantinescu, D., Gray, H.L., Sammak, P.J., Schatten, G.P., Csoka, A.B., 2006.

Lamin A/C expression is a marker of mouse and human embryonic stem

cell differentiation. Stem Cells 24, 177–85. doi:10.1634/stemcells.2004-0159

Corrigan, D.P., Kuszczak, D., Rusinol, A.E., Thewke, D.P., Hrycyna, C. a, Michaelis,

S., Sinensky, M.S., 2005. Prelamin A endoproteolytic processing in vitro by

recombinant Zmpste24. Biochem. J. 387, 129–38. doi:10.1042/BJ20041359

Costa, A., Daidone, M.G., Daprai, L., Villa, R., Cantù, S., Pilotti, S., Mariani, L.,

Gronchi, A., Henson, J.D., Reddel, R.R., Zaffaroni, N., 2006. Telomere

maintenance mechanisms in liposarcomas: Association with histologic

subtypes and disease progression. Cancer Res. 66, 8918–8924.

doi:10.1158/0008-5472.CAN-06-0273

Coulombe, P.A., Chan, Y.M., Albers, K., Fuchs, E., 1990. Deletions in epidermal

keratins leading to alterations in filament organization in vivo and in

intermediate filament assembly in vitro. J. Cell Biol. 111, 3049–3064.

doi:10.1083/jcb.111.6.3049

Counter, C.M., Avilion, A.A., LeFeuvre, C.E., Stewart, N.G., Greider, C.W., Harley,

C.B., Bacchetti, S., 1992. Telomere shortening associated with chromosome

instability is arrested in immortal cells which express telomerase activity.

EMBO J. 11, 1921–1929.

Court, R., Chapman, L., Fairall, L., Rhodes, D., 2005. How the human telomeric

proteins TRF1 and TRF2 recognize telomeric DNA: a view from high-

resolution crystal structures. EMBO Rep. 6, 39–45.

doi:10.1038/sj.embor.7400314

Crabbe, L., Cesare, A.J., Kasuboski, J.M., Fitzpatrick, J.A.J., Karlseder, J., 2012.

Human Telomeres Are Tethered to the Nuclear Envelope during Postmitotic

Nuclear Assembly. Cell Rep. 2, 1521–1529. doi:10.1016/j.celrep.2012.11.019

Crabbe, L., Jauch, A., Naeger, C.M., Holtgreve-Grez, H., Karlseder, J., 2007.

Telomere dysfunction as a cause of genomic instability in Werner

Page 230: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

215

syndrome. Proc. Natl. Acad. Sci. U. S. A. 104, 2205–10.

doi:10.1073/pnas.0609410104

Crabbe, L., Verdun, R.E., Haggblom, C.I., Karlseder, J., 2004. Defective telomere

lagging strand synthesis in cells lacking WRN helicase activity. Science (80-.

). 306, 1951–1953. doi:10.1126/science.1103619

Croft, J. a, Bridger, J.M., Boyle, S., Perry, P., Teague, P., Bickmore, W. a, 1999.

Differences in the localization and morphology of chromosomes in the

human nucleus. J. Cell Biol. 145, 1119–31.

Cross, S., Lindsey, J., Fantes, J., McKay, S., McGill, N., Cooke, H., 1990. The

structure of a subterminal repeated sequence present on many human

chromosomes. Nucleic Acids Res. 18, 6649–6657.

Croteau, D.L., Popuri, V., Opresko, P.L., Bohr, V. a, 2014. Human RecQ helicases

in DNA repair, recombination, and replication. Annu. Rev. Biochem. 83,

519–52. doi:10.1146/annurev-biochem-060713-035428

Csoka, a B., 2004. Novel lamin A/C gene (LMNA) mutations in atypical progeroid

syndromes. J. Med. Genet. 41, 304–308. doi:10.1136/jmg.2003.015651

Csoka, A.B., English, S.B., Simkevich, C.P., Ginzinger, D.G., Butte, A.J., Schatten,

G.P., Rothman, F.G., Sedivy, J.M., 2004. Genome-scale expression profiling

of Hutchinson-Gilford progeria syndrome reveals widespread

transcriptional misregulation leading to mesodermal/mesenchymal defects

and accelerated atherosclerosis. Aging Cell 3, 235–43. doi:10.1111/j.1474-

9728.2004.00105.x

d’Adda di Fagagna, F., Reaper, P.M., Clay-Farrace, L., Fiegler, H., Carr, P., Von

Zglinicki, T., Saretzki, G., Carter, N.P., Jackson, S.P., 2003. A DNA damage

checkpoint response in telomere-initiated senescence. Nature 426, 194–8.

doi:10.1038/nature02118

Dahl, J.A., Collas, P., 2008. A rapid micro chromatin immunoprecipitation assay

(microChIP). Nat. Protoc. 3, 1032–45. doi:10.1038/nprot.2008.68

Dahl, K.N., Scaffidi, P., Islam, M.F., Yodh, A.G., Wilson, K.L., Misteli, T., 2006.

Distinct structural and mechanical properties of the nuclear lamina in

Hutchinson-Gilford progeria syndrome. Proc. Natl. Acad. Sci. U. S. A. 103,

10271–6. doi:10.1073/pnas.0601058103

Dai, Q., Choy, E., Chiu, V., Romano, J., Slivka, S.R., Steitz, S.A., Michaelis, S.,

Philips, M.R., 1998. Mammalian prenylcysteine carboxyl methyltransferase is

Page 231: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

216

in the endoplasmic reticulum. J. Biol. Chem. 273, 15030–15034.

doi:10.1074/jbc.273.24.15030

Dai, X., Huang, C., Bhusari, A., Sampathi, S., Schubert, K., Chai, W., 2010.

Molecular steps of G-overhang generation at human telomeres and its

function in chromosome end protection. EMBO J. 29, 2788–2801.

doi:10.1038/emboj.2010.156

Davies, B.S.J., Barnes, R.H., Tu, Y., Ren, S., Andres, D.A., Peter Spielmann, H.,

Lammerding, J., Wang, Y., Young, S.G., Fong, L.G., 2010. An accumulation of

non-farnesylated prelamin A causes cardiomyopathy but not progeria.

Hum. Mol. Genet. 19, 2682–2694. doi:10.1093/hmg/ddq158

De Lange, T., 2005. Shelterin: the protein complex that shapes and safeguards

human telomeres. Genes Dev. 19, 2100–10. doi:10.1101/gad.1346005

De Lange, T., 1992. Human telomeres are attached to the nuclear matrix. EMBO

J. 11, 717–724. doi:10.1038/ng1097-231

De Lange, T., Shiue, L., Myers, R.M., Cox, D.R., Naylor, S.L., Killery, A.M., Varmus,

H.E., 1990. Structure and variability of human chromosome ends. Mol. Cell.

Biol. 10, 518–527. doi:10.1128/MCB.10.2.518.Updated

De Sandre-Giovannoli, A., Chaouch, M., Kozlov, S., Vallat, J.-M., Tazir, M.,

Kassouri, N., Szepetowski, P., Hammadouche, T., Vandenberghe, A., Stewart,

C.L., Grid, D., Lévy, N., 2002. Homozygous defects in LMNA, encoding lamin

A/C nuclear-envelope proteins, cause autosomal recessive axonal

neuropathy in human (Charcot-Marie-Tooth disorder type 2) and mouse.

Am. J. Hum. Genet. 70, 726–736. doi:10.1086/339274

De Vos, W.H., Houben, F., Hoebe, R. a, Hennekam, R., van Engelen, B., Manders,

E.M.M., Ramaekers, F.C.S., Broers, J.L. V, Van Oostveldt, P., 2010. Increased

plasticity of the nuclear envelope and hypermobility of telomeres due to

the loss of A-type lamins. Biochim. Biophys. Acta 1800, 448–58.

doi:10.1016/j.bbagen.2010.01.002

Dechat, T., Gajewski, A., Korbei, B., Gerlich, D., Daigle, N., Haraguchi, T.,

Furukawa, K., Ellenberg, J., Foisner, R., 2004. LAP2alpha and BAF transiently

localize to telomeres and specific regions on chromatin during nuclear

assembly. J. Cell Sci. 117, 6117–28. doi:10.1242/jcs.01529

Dechat, T., Gesson, K., Foisner, R., 2010. Lamina-independent lamins in the

nuclear interior serve important functions. Cold Spring Harb. Symp. Quant.

Biol. 75, 533–543. doi:10.1101/sqb.2010.75.018

Page 232: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

217

Dechat, T., Korbei, B., Vaughan, O. a, Vlcek, S., Hutchison, C.J., Foisner, R., 2000.

Lamina-associated polypeptide 2alpha binds intranuclear A-type lamins. J.

Cell Sci. 113, 3473–84.

Dechat, T., Pfleghaar, K., Sengupta, K., Shimi, T., Shumaker, D.K., Solimando, L.,

Goldman, R.D., 2008. Nuclear lamins: major factors in the structural

organization and function of the nucleus and chromatin. Genes Dev. 22,

832–53. doi:10.1101/gad.1652708

Dechat, T., Shimi, T., Adam, S. a, Rusinol, A.E., Andres, D. a, Spielmann, H.P.,

Sinensky, M.S., Goldman, R.D., 2007. Alterations in mitosis and cell cycle

progression caused by a mutant lamin A known to accelerate human aging.

Proc. Natl. Acad. Sci. U. S. A. 104, 4955–60. doi:10.1073/pnas.0700854104

Decker, M.L., Chavez, E., Vulto, I., Lansdorp, P.M., 2009. Telomere length in

Hutchinson-Gilford progeria syndrome. Mech. Ageing Dev. 130, 377–83.

doi:10.1016/j.mad.2009.03.001

Déjardin, J., Kingston, R.E., 2009. Purification of Proteins Associated with Specific

Genomic Loci. Cell 136, 175–186. doi:10.1016/j.cell.2008.11.045

Denchi, E.L., de Lange, T., 2007. Protection of telomeres through independent

control of ATM and ATR by TRF2 and POT1. Nature 448, 1068–71.

doi:10.1038/nature06065

Denecke, J., Brune, T., Feldhaus, T., Robenek, H., Kranz, C., Auchus, R.J., Agarwal,

A.K., Marquardt, T., 2006. A homozygous ZMPSTE24 null mutation in

combination with a heterozygous mutation in the LMNA gene causes

Hutchinson-Gilford progeria syndrome (HGPS): Insights into the

pathophysiology of HGPS. Hum. Mutat. 27, 524–531.

doi:10.1002/humu.20315

Deriano, L., Roth, D.B., 2013. Modernizing the nonhomologous end-joining

repertoire: alternative and classical NHEJ share the stage. Annu. Rev. Genet.

47, 433–55. doi:10.1146/annurev-genet-110711-155540

Difilippantonio, S., Gapud, E., Wong, N., Huang, C.-Y., Mahowald, G., Chen, H.T.,

Kruhlak, M.J., Callen, E., Livak, F., Nussenzweig, M.C., Sleckman, B.P.,

Nussenzweig, A., 2008. 53BP1 facilitates long-range DNA end-joining

during V(D)J recombination. Nature 456, 529–533. doi:10.1038/nature07476

Dimitrova, N., Chen, Y.-C.M., Spector, D.L., de Lange, T., 2008. 53BP1 promotes

non-homologous end joining of telomeres by increasing chromatin

mobility. Nature 456, 524–528. doi:10.1038/nature07433

Page 233: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

218

Dimri, G.P., Lee, X., Basile, G., Acosta, M., Scott, G., Roskelley, C., Medrano, E.E.,

Linskens, M., Rubelj, I., Pereira-Smith, O., 1995. A biomarker that identifies

senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad.

Sci. U. S. A. 92, 9363–9367. doi:10.1073/pnas.92.20.9363

Dionne, I., Wellinger, R.J., 1996. Cell cycle-regulated generation of single-

stranded G-rich DNA in the absence of telomerase. Proc. Natl. Acad. Sci. U.

S. A. 93, 13902–13907. doi:10.1073/pnas.93.24.13902

Doh, Y.J., Kim, H.K., Jung, E.D., Choi, S.H., Kim, J.G., Kim, B.W., Lee, I.K., 2009.

Novel LMNA gene mutation in a patient with atypical Werner’s syndrome.

Korean J. Intern. Med. 24, 68–72. doi:10.3904/kjim.2009.24.1.68

Doksani, Y., Wu, J.Y., De Lange, T., Zhuang, X., 2013. Super-resolution

fluorescence imaging of telomeres reveals TRF2-dependent T-loop

formation. Cell 155, 345–56. doi:10.1016/j.cell.2013.09.048

Dorner, D., Gotzmann, J., Foisner, R., 2007. Nucleoplasmic lamins and their

interaction partners, LAP2alpha, Rb, and BAF, in transcriptional regulation.

FEBS J. 274, 1362–1373. doi:10.1111/j.1742-4658.2007.05695.x

Downey, M., Durocher, D., 2006. y-H2AX as a checkpoint maintenance signal.

Cell Cycle. doi:10.4161/cc.5.13.2899

Draskovic, I., Arnoult, N., Steiner, V., Bacchetti, S., Lomonte, P., Londoño-Vallejo,

A., 2009. Probing PML body function in ALT cells reveals spatiotemporal

requirements for telomere recombination. Proc. Natl. Acad. Sci. U. S. A. 106,

15726–15731. doi:10.1073/pnas.0907689106

Dreuillet, C., Tillit, J., Kress, M., Ernoult-Lange, M., 2002. In vivo and in vitro

interaction between human transcription factor MOK2 and nuclear lamin

A/C. Nucleic Acids Res. 30, 4634–4642.

Dunham, M.A., Neumann, A.A., Fasching, C.L., Reddel, R.R., 2000. Telomere

maintenance by recombination in human cells. Nat. Genet. 26, 447–450.

doi:10.1038/82586

Egholm, M., Buchardt, O., Christensen, L., Behrens, C., Freier, S.M., Driver, D.A.,

Berg, R.H., Kim, S.K., Norden, B., Nielsen, P.E., 1993. PNA hybridizes to

complementary oligonucleotides obeying the Watson-Crick hydrogen-

bonding rules. Nature 365, 566–568. doi:10.1038/365566a0

Eid, W., Steger, M., El-Shemerly, M., Ferretti, L.P., Peña-Diaz, J., König, C.,

Valtorta, E., Sartori, A.A., Ferrari, S., 2010. DNA end resection by CtIP and

Page 234: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

219

exonuclease 1 prevents genomic instability. EMBO Rep. 11, 962–968.

doi:10.1038/embor.2010.157

Eller, M.S., Liao, X., Liu, S., Hanna, K., Bäckvall, H., Opresko, P.L., Bohr, V.A.,

Gilchrest, B.A., 2006. A role for WRN in telomere-based DNA damage

responses. Proc. Natl. Acad. Sci. U. S. A. 103, 15073–15078.

doi:10.1073/pnas.0607332103

Emery, a E., 1989. Emery-Dreifuss syndrome. J. Med. Genet. 26, 637–641.

doi:10.1136/jmg.26.10.637

Engelhardt, M., Kumar, R., Albanell, J., Pettengell, R., Han, W., Moore, M.A., 1997.

Telomerase regulation, cell cycle, and telomere stability in primitive

hematopoietic cells. Blood 90, 182–193.

Episkopou, H., Draskovic, I., Van Beneden, A., Tilman, G., Mattiussi, M., Gobin, M.,

Arnoult, N., Londoño-Vallejo, A., Decottignies, A., 2014. Alternative

Lengthening of Telomeres is characterized by reduced compaction of

telomeric chromatin. Nucleic Acids Res. 42, 4391–4405.

doi:10.1093/nar/gku114

Erber, A., Riemer, D., Hofemeister, H., Bovenschulte, M., Stick, R., Panopoulou, G.,

Lehrach, H., Weber, K., 1999. Characterization of the Hydra lamin and its

gene: A molecular phylogeny of metazoan lamins. J. Mol. Evol. 49, 260–271.

doi:10.1007/PL00006548

Eriksson, M., Brown, W.T., Gordon, L.B., Glynn, M.W., Singer, J., Scott, L., Erdos,

M.R., Robbins, C.M., Moses, T.Y., Berglund, P., Dutra, A., Pak, E., Durkin, S.,

Csoka, A.B., Boehnke, M., Glover, T.W., Collins, F.S., 2003. Recurrent de novo

point mutations in lamin A cause Hutchinson-Gilford progeria syndrome.

Nature 423, 293–8. doi:10.1038/nature01629

Farnsworth, C.C., Wolda, S.L., Gelb, M.H., Glomset, J.A., 1989. Human lamin B

contains a farnesylated cysteine residue. J. Biol. Chem. 264, 20422–20429.

Fattah, F., Lee, E.H.H., Weisensel, N., Wang, Y., Lichter, N., Hendrickson, E.A.,

2010. Ku regulates the non-homologous end joining pathway choice of

DNA double-strand break repair in human somatic cells. PLoS Genet. 6,

1000855. doi:10.1371/journal.pgen.1000855

Favreau, C., Higuet, D., Courvalin, J.-C., Buendia, B., 2004. Expression of a mutant

lamin A that causes Emery-Dreifuss muscular dystrophy inhibits in vitro

differentiation of C2C12 myoblasts. Mol. Cell. Biol. 24, 1481–1492.

doi:10.1128/MCB.24.4.1481-1492.2004

Page 235: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

220

Feng, J., Funk, W.D., Wang, S.S., Weinrich, S.L., Avilion, A.A., Chiu, C.P., Adams,

R.R., Chang, E., Allsopp, R.C., Yu, J., et al., 1995. The RNA component of

human telomerase. Science (80-. ). 269, 1236–1241.

doi:10.1126/science.7544491

Foisner, R., Gerace, L., 1993. Integral membrane proteins of the nuclear envelope

interact with lamins and chromosomes, and binding is modulated by

mitotic phosphorylation. Cell 73, 1267–1279. doi:10.1016/0092-

8674(93)90355-T

Fong, L.G., Frost, D., Meta, M., Qiao, X., Yang, S.H., Coffinier, C., Young, S.G.,

2006. A protein farnesyltransferase inhibitor ameliorates disease in a mouse

model of progeria. Science (80-. ). 311, 1621–3.

doi:10.1126/science.1124875

Fong, L.G., Ng, J.K., Lammerding, J., Vickers, T.A., Meta, M., Coté, N., Gavino, B.,

Qiao, X., Chang, S.Y., Young, S.R., Yang, S.H., Stewart, C.L., Lee, R.T., Bennett,

C.F., Bergo, M.O., Young, S.G., 2006. Prelamin A and lamin A appear to be

dispensable in the nuclear lamina. J. Clin. Invest. 116, 743–752.

doi:10.1172/JCI27125

Foster, H.A., Stokes, P., Forsey, K., Leese, H.J., Bridger, J.M., 2007. Lamins A and C

are present in the nuclei of early porcine embryos, with lamin A being

distributed in large intranuclear foci. Chromosom. Res. 15, 163–174.

doi:10.1007/s10577-006-1088-8

Frank, C.J., Hyde, M., Greider, C.W., 2006. Regulation of Telomere Elongation by

the Cyclin-Dependent Kinase CDK1. Mol. Cell 24, 423–432.

doi:10.1016/j.molcel.2006.10.020

Frank, R., Köster, H., 1979. DNA chain length markers and the influence of base

composition on electrophoretic mobility of oligodeoxyribonucleotides in

plyacrylamide-gels. Nucleic Acids Res. 6, 2069–2087.

doi:10.1093/nar/6.6.2069

Frescas, D., de Lange, T., 2014. Binding of TPP1 to TIN2 is required for POT1a,b-

mediated telomere protection. J. Biol. Chem. 289, 24180–7.

doi:10.1074/jbc.M114.592592

Frock, R.L., Kudlow, B.A., Evans, A.M., Jameson, S.A., Hauschka, S.D., Kennedy,

B.K., 2006. Lamin A/C and emerin are critical for skeletal muscle satellite cell

differentiation. Genes Dev. 20, 486–500. doi:10.1101/gad.1364906

Page 236: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

221

Frydrychova, R.C., Mason, J.M., Archer, T.K., 2008. HP1 is distributed within

distinct chromatin domains at drosophila telomeres. Genetics 180, 121–131.

doi:10.1534/genetics.108.090647

Fukuchi, K., Katsuya, T., Sugimoto, K., 2004. LMNA mutation in a 45 year old

Japanese subject with Hutchinson-Gilford progeria syndrome. J. Med.

Genet. 41, 67. doi:10.1136/jmg.2003.014688

Furukawa, K., Hotta, Y., 1993. cDNA cloning of a germ cell specific lamin B3 from

mouse spermatocytes and analysis of its function by ectopic expression in

somatic cells. EMBO J. 12, 97–106.

Furukawa, K., Inagaki, H., Hotta, Y., 1994. Identification and cloning of an mRNA

coding for a germ cell-specific A-type lamin in mice. Exp. Cell Res. 212,

426–430. doi:10.1006/excr.1994.1164

Gant, T.M., Harris, C.A., Wilson, K.L., 1999. Roles of LAP2 proteins in nuclear

assembly and DNA replication: Truncated LAP2β proteins alter lamina

assembly, envelope formation, nuclear size, and DNA replication efficiency

in Xenopus laevis extracts. J. Cell Biol. 144, 1083–1096.

doi:10.1083/jcb.144.6.1083

García-Cao, M., O’Sullivan, R., Peters, A.H.F.M., Jenuwein, T., Blasco, M.A., 2004.

Epigenetic regulation of telomere length in mammalian cells by the

Suv39h1 and Suv39h2 histone methyltransferases. Nat. Genet. 36, 94–9.

doi:10.1038/ng1278

Garg, A., Cogulu, O., Ozkinay, F., Onay, H., Agarwal, A.K., 2005. A novel

homozygous Ala529Val LMNA mutation in Turkish patients with

mandibuloacral dysplasia. J. Clin. Endocrinol. Metab. 90, 5259–5264.

doi:10.1210/jc.2004-2560

Gerace, L., Blobel, G., 1980. The nuclear envelope lamina is reversibly

depolymerized during mitosis. Cell 19, 277–287. doi:10.1016/0092-

8674(80)90409-2

Gerace, L., Comeau, C., Benson, M., 1984. Organization and modulation of

nuclear lamina structure. J. Cell Sci. Suppl. 1, 137–160.

Gilfillan, G.D., Hughes, T., Sheng, Y., Hjorthaug, H.S., Straub, T., Gervin, K., Harris,

J.R., Undlien, D.E., Lyle, R., 2012. Limitations and possibilities of low cell

number ChIP-seq. BMC Genomics 13, 645. doi:10.1186/1471-2164-13-645

Page 237: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

222

Gisselsson, D., Jonson, T., Petersén, A., Strömbeck, B., Dal Cin, P., Höglund, M.,

Mitelman, F., Mertens, F., Mandahl, N., 2001. Telomere dysfunction triggers

extensive DNA fragmentation and evolution of complex chromosome

abnormalities in human malignant tumors. Proc. Natl. Acad. Sci. U. S. A. 98,

12683–12688. doi:10.1073/pnas.211357798

Goldman, a E., Moir, R.D., Montag-Lowy, M., Stewart, M., Goldman, R.D., 1992.

Pathway of incorporation of microinjected lamin A into the nuclear

envelope. J. Cell Biol. 119, 725–35.

Goldman, A.E., Maul, G., Steinert, P.M., Yang, H.Y., Goldman, R.D., 1986. Keratin-

like proteins that coisolate with intermediate filaments of BHK-21 cells are

nuclear lamins. Proc. Natl. Acad. Sci. U. S. A. 83, 3839–3843.

doi:10.1073/pnas.83.11.3839

Goldman, R.D., Shumaker, D.K., Erdos, M.R., Eriksson, M., Goldman, A.E., Gordon,

L.B., Gruenbaum, Y., Khuon, S., Mendez, M., Varga, R., Collins, F.S., 2004.

Accumulation of mutant lamin A causes progressive changes in nuclear

architecture in Hutchinson-Gilford progeria syndrome. Proc. Natl. Acad. Sci.

U. S. A. 101, 8963–8. doi:10.1073/pnas.0402943101

Gonzalez-Suarez, I., Redwood, A.B., Grotsky, D. a, Neumann, M. a, Cheng, E.H.-Y.,

Stewart, C.L., Dusso, A., Gonzalo, S., 2011. A new pathway that regulates

53BP1 stability implicates Cathepsin L and vitamin D in DNA repair. EMBO J.

30, 3383–96. doi:10.1038/emboj.2011.225

Gonzalez-Suarez, I., Redwood, A.B., Perkins, S.M., Vermolen, B., Lichtensztejin, D.,

Grotsky, D. a, Morgado-Palacin, L., Gapud, E.J., Sleckman, B.P., Sullivan, T.,

Sage, J., Stewart, C.L., Mai, S., Gonzalo, S., 2009. Novel roles for A-type

lamins in telomere biology and the DNA damage response pathway. EMBO

J. 28, 2414–27. doi:10.1038/emboj.2009.196

Goto, M., Miller, R.W., Ishikawa, Y., Sugano, H., 1996. Excess of rare cancers in

Werner syndrome (adult progeria). Cancer Epidemiol. Biomarkers Prev. 5,

239–246.

Graakjaer, J., Der-Sarkissian, H., Schmitz, A., Bayer, J., Thomas, G., Kolvraa, S.,

Londoño-Vallejo, J.A., 2006. Allele-specific relative telomere lengths are

inherited. Hum. Genet. 119, 344–350. doi:10.1007/s00439-006-0137-x

Graakjaer, J., Pascoe, L., Der-Sarkissian, H., Thomas, G., Kolvraa, S., Christensen,

K., Londoño-Vallejo, J.-A., 2004. The relative lengths of individual telomeres

are defined in the zygote and strictly maintained during life. Aging Cell 3,

97–102. doi:10.1111/j.1474-9728.2004.00093.x

Page 238: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

223

Grawunder, U., Wilm, M., Wu, X., Kulesza, P., Wilson, T.E., Mann, M., Lieber, M.R.,

1997. Activity of DNA ligase IV stimulated by complex formation with

XRCC4 protein in mammalian cells. Nature 388, 492–495.

doi:10.1038/41358

Griffith, J.D., Comeau, L., Rosenfield, S., Stansel, R.M., Bianchi, A., Moss, H., de

Lange, T., 1999. Mammalian telomeres end in a large duplex loop. Cell 97,

503–14.

Grotsky, D. a, Gonzalez-Suarez, I., Novell, A., Neumann, M. a, Yaddanapudi, S.C.,

Croke, M., Martinez-Alonso, M., Redwood, A.B., Ortega-Martinez, S., Feng,

Z., Lerma, E., Ramon y Cajal, T., Zhang, J., Matias-Guiu, X., Dusso, A.,

Gonzalo, S., 2013. BRCA1 loss activates cathepsin L-mediated degradation

of 53BP1 in breast cancer cells. J. Cell Biol. 200, 187–202.

doi:10.1083/jcb.201204053

Gruenbaum, Y., Landesman, Y., Drees, B., Bare, J.W., Saumweber, H., Paddy, M.R.,

Sedat, J.W., Smith, D.E., Benton, B.M., Fisher, P.A., 1988. Drosophila nuclear

lamin precursor Dm0 is translated from either of two developmentally

regulated mRNA species apparently encoded by a single gene. J. Cell Biol.

106, 585–596. doi:10.1083/jcb.106.3.585

Guelen, L., Pagie, L., Brasset, E., Meuleman, W., Faza, M.B., Talhout, W., Eussen,

B.H., de Klein, A., Wessels, L., de Laat, W., van Steensel, B., 2008. Domain

organization of human chromosomes revealed by mapping of nuclear

lamina interactions. Nature 453, 948–951. doi:10.1038/nature06947

Guilly, M.N., Bensussan, A., Bourge, J.F., Bornens, M., Courvalin, J.C., 1987. A

human T lymphoblastic cell line lacks lamins A and C. EMBO J. 6, 3795–

3799.

Guo, Y., Kim, Y., Shimi, T., Goldman, R.D., Zheng, Y., 2014. Concentration-

dependent lamin assembly and its roles in the localization of other nuclear

proteins. Mol. Biol. Cell 25, 1287–97. doi:10.1091/mbc.E13-11-0644

Haince, J.F., McDonald, D., Rodrigue, A., Déry, U., Masson, J.Y., Hendzel, M.J.,

Poirier, G.G., 2008. PARP1-dependent kinetics of recruitment of MRE11 and

NBS1 proteins to multiple DNA damage sites. J. Biol. Chem. 283, 1197–

1208. doi:10.1074/jbc.M706734200

Hakin-Smith, V., Jellinek, D.A., Levy, D., Carroll, T., Teo, M., Timperley, W.R.,

McKay, M.J., Reddel, R.R., Royds, J.A., 2003. Alternative lengthening of

telomeres and survival in patients with glioblastoma multiforme. Lancet

361, 836–838. doi:10.1016/S0140-6736(03)12681-5

Page 239: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

224

Hall, V.J., Cooney, M.A., Shanahan, P., Tecirlioglu, R.T., Ruddock, N.T., French, A.J.,

2005. Nuclear lamin antigen and messenger RNA expression in bovine in

vitro produced and nuclear transfer embryos. Mol. Reprod. Dev. 72, 471–

482. doi:10.1002/mrd.20381

Hanaoka, S., Nagadoi, A., Nishimura, Y., 2005. Comparison between TRF2 and

TRF1 of their telomeric DNA-bound structures and DNA-binding activities.

Protein Sci. 14, 119–130. doi:10.1110/ps.04983705

Hande, M.P., Samper, E., Lansdorp, P., Blasco, M.A., 1999. Telomere length

dynamics and chromosomal instability in cells derived from telomerase null

mice. J. Cell Biol. 144, 589–601. doi:10.1083/jcb.144.4.589

Haque, F., Mazzeo, D., Patel, J.T., Smallwood, D.T., Ellis, J.A., Shanahan, C.M.,

Shackleton, S., 2010. Mammalian SUN protein interaction networks at the

inner nuclear membrane and their role in laminopathy disease processes. J.

Biol. Chem. 285, 3487–3498. doi:10.1074/jbc.M109.071910

Hara, E., Tsurui, H., Shinozaki, A., Nakada, S., Oda, K., 1991. Cooperative effect of

antisense-Rb and antisense-p53 oligomers on the extension of life span in

human diploid fibroblasts, TIG-1. Biochem. Biophys. Res. Commun. 179,

528–534. doi:10.1016/0006-291X(91)91403-Y

Harborth, J., Elbashir, S.M., Bechert, K., Tuschl, T., Weber, K., 2001. Identification

of essential genes in cultured mammalian cells using small interfering

RNAs. J. Cell Sci. 114, 4557–4565.

Harley, C.B., Futcher, A.B., Greider, C.W., 1990. Telomeres shorten during ageing

of human fibroblasts. Nature 345, 458–460. doi:10.1038/345458a0

Harper, M., Tillit, J., Kress, M., Ernoult-Lange, M., 2009. Phosphorylation-

dependent binding of human transcription factor MOK2 to lamin A/C. FEBS

J. 276, 3137–3147. doi:10.1111/j.1742-4658.2009.07032.x

Harrington, L., Zhou, W., McPhail, T., Oulton, R., Yeung, D.S.K., Mar, V., Bass, M.B.,

Robinson, M.O., 1997. Human telomerase contains evolutionarily conserved

catalytic and structural subunits. Genes Dev. 11, 3109–3115.

doi:10.1101/gad.11.23.3109

Hatzfeld, M., Weber, K., 1991. Modulation of keratin intermediate filament

assembly by single amino acid exchanges in the consensus sequence at the

C-terminal end of the rod domain. J. Cell Sci. 99, 351–362.

Page 240: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

225

Hayakawa, T., Haraguchi, T., Masumoto, H., Hiraoka, Y., 2003. Cell cycle behavior

of human HP1 subtypes: distinct molecular domains of HP1 are required for

their centromeric localization during interphase and metaphase. J. Cell Sci.

116, 3327–3338. doi:10.1242/jcs.00635

Hayflick, L., 1965. The limited in vitro lifetime of human diploid cell strains. Exp.

Cell Res. 37, 614–636. doi:10.1016/0014-4827(65)90211-9

Hayflick, L., Moorhead, P.S., 1961. The serial cultivation of human diploid cell

strains. Exp. Cell Res. 25, 585–621.

Heaphy, C.M., Subhawong, A.P., Hong, S.M., Goggins, M.G., Montgomery, E.A.,

Gabrielson, E., Netto, G.J., Epstein, J.I., Lotan, T.L., Westra, W.H., Shih, I.M.,

Iacobuzio-Donahue, C.A., Maitra, A., Li, Q.K., Eberhart, C.G., Taube, J.M.,

Rakheja, D., Kurman, R.J., Wu, T.C., Roden, R.B., Argani, P., De Marzo, A.M.,

Terracciano, L., Torbenson, M., Meeker, A.K., 2011. Prevalence of the

alternative lengthening of telomeres telomere maintenance mechanism in

human cancer subtypes. Am. J. Pathol. 179, 1608–1615.

doi:10.1016/j.ajpath.2011.06.018

Hegele, R.A., Cao, H., Anderson, C.M., Hramiak, I.M., 2000. Heterogeneity of

nuclear lamin A mutations in dunnigan-type familial partial lipodystrophy. J.

Clin. Endocrinol. Metab. 85, 3431–3435. doi:10.1210/jc.85.9.3431

Hegele, R.A., Cao, H., Liu, D.M., Costain, G.A., Charlton-Menys, V., Rodger, N.W.,

Durrington, P.N., 2006. Sequencing of the reannotated LMNB2 gene reveals

novel mutations in patients with acquired partial lipodystrophy. Am. J. Hum.

Genet. 79, 383–389. doi:10.1086/505885

Henderson, E.R., Blackburn, E.H., 1989. An overhanging 3’ terminus is a

conserved feature of telomeres. Mol. Cell. Biol. 9, 345–348.

doi:10.1128/MCB.9.1.345.Updated

Hennekam, R.C.M., 2006. Hutchinson – Gilford Progeria Syndrome : Review of

the Phenotype. Am. J. Med. Genet. 140, 2603 – 2624. doi:10.1002/ajmg.a

Hennekes, H., Nigg, E.A., 1994. The role of isoprenylation in membrane

attachment of nuclear lamins. A single point mutation prevents proteolytic

cleavage of the lamin A precursor and confers membrane binding

properties. J. Cell Sci. 107, 1019–29.

Henson, J.D., Cao, Y., Huschtscha, L.I., Chang, A.C., Au, A.Y.M., Pickett, H.A.,

Reddel, R.R., 2009. DNA C-circles are specific and quantifiable markers of

Page 241: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

226

alternative-lengthening-of-telomeres activity. Nat. Biotechnol. 27, 1181–

1185. doi:10.1038/nbt.1587

Henson, J.D., Hannay, J.A., McCarthy, S.W., Royds, J.A., Yeager, T.R., Robinson,

R.A., Wharton, S.B., Jellinek, D.A., Arbuckle, S.M., Yoo, J., Robinson, B.G.,

Learoyd, D.L., Stalley, P.D., Bonar, S.F., Yu, D., Pollock, R.E., Reddel, R.R.,

2005. A robust assay for alternative lengthening of telomeres in tumors

shows the significance of alternative lengthening of telomeres in sarcomas

and astrocytomas. Clin. Cancer Res. 11, 217–225. doi:11/1/217 [pii]

Herbig, U., Ferreira, M., Condel, L., Carey, D., Sedivy, J.M., 2006. Cellular

senescence in aging primates. Science (80-. ). 311, 1257.

doi:10.1126/science.1122446

Herbig, U., Jobling, W.A., Chen, B.P.C., Chen, D.J., Sedivy, J.M., 2004. Telomere

shortening triggers senescence of human cells through a pathway involving

ATM, p53, and p21(CIP1), but not p16(INK4a). Mol. Cell 14, 501–513.

Herrmann, H., Strelkov, S. V., Burkhard, P., Aebi, U., 2009. Intermediate filaments:

Primary determinants of cell architecture and plasticity. J. Clin. Invest.

doi:10.1172/JCI38214

Heyn, H., Moran, S., Esteller, M., 2013. Aberrant DNA methylation profiles in the

premature aging disorders Hutchinson-Gilford Progeria and Werner

Syndrome. Epigenetics 8, 28–33. doi:10.4161/epi.23366

Hirt, B., 1967. Selective extraction of polyoma DNA from infected mouse cell

cultures. J. Mol. Biol. 26, 365–369. doi:10.1016/0022-2836(67)90307-5

Ho, C.Y., Jaalouk, D.E., Vartiainen, M.K., Lammerding, J., 2013. Lamin A/C and

emerin regulate MKL1-SRF activity by modulating actin dynamics. Nature

497, 507–11. doi:10.1038/nature12105

Hockemeyer, D., Palm, W., Else, T., Daniels, J.-P., Takai, K.K., Ye, J.Z.-S., Keegan,

C.E., de Lange, T., Hammer, G.D., 2007. Telomere protection by mammalian

Pot1 requires interaction with Tpp1. Nat. Struct. Mol. Biol. 14, 754–761.

doi:10.1038/nsmb1270

Hoeltzenbein, M., Karow, T., Zeller, J.A., Warzok, R., Wulff, K., Zschiesche, M.,

Herrmann, F.H., Grosse-Heitmeyer, W., Wehnert, M.S., 1999. Severe clinical

expression in X-linked Emery-Dreifuss muscular dystrophy. Neuromuscul.

Disord. 9, 166–170. doi:10.1016/S0960-8966(98)00120-5

Page 242: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

227

Holtz, D., Tanaka, R.A., Hartwig, J., McKeon, F., 1989. The CaaX motif of lamin A

functions in conjunction with the nuclear localization signal to target

assembly to the nuclear envelope. Cell 59, 969–977. doi:10.1016/0092-

8674(89)90753-8

Houben, F., De Vos, W.H., Krapels, I.P.C., Coorens, M., Kierkels, G.J.J., Kamps,

M.A.F., Verstraeten, V.L.R.M., Marcelis, C.L.M., Van Den Wijngaard, A.,

Ramaekers, F.C.S., Broers, J.L. V, 2013. Cytoplasmic localization of PML

particles in laminopathies. Histochem. Cell Biol. 139, 119–134.

doi:10.1007/s00418-012-1005-5

Houben, F., Ramaekers, F.C.S., Snoeckx, L.H.E.H., Broers, J.L. V, 2007. Role of

nuclear lamina-cytoskeleton interactions in the maintenance of cellular

strength. Biochim. Biophys. Acta 1773, 675–86.

doi:10.1016/j.bbamcr.2006.09.018

Huang, S., Lee, L., Hanson, N.B., Lenaerts, C., Hoehn, H., Poot, M., Rubin, C.D.,

Chen, D.F., Yang, C.C., Juch, H., Dorn, T., Spiegel, R., Oral, E.A., Abid, M.,

Battisti, C., Lucci-Cordisco, E., Neri, G., Steed, E.H., Kidd, A., Isley, W.,

Showalter, D., Vittone, J.L., Konstantinow, A., Ring, J., Meyer, P., Wenger, S.L.,

Von Herbay, A., Wollina, U., Schuelke, M., Huizenga, C.R., Leistritz, D.F.,

Martin, G.M., Mian, I.S., Oshima, J., 2006. The spectrum of WRN mutations

in Werner syndrome patients. Hum. Mutat. 27, 558–567.

doi:10.1002/humu.20337

Huang, S., Risques, R.A.R.A., Martin, G.M., Rabinovitch, P.S., Oshima, J., 2008.

Accelerated telomere shortening and replicative senescence in human

fibroblasts overexpressing mutant and wild-type lamin A. Exp. Cell Res. 314,

82–91. doi:10.1016/j.yexcr.2007.08.004

Huffman, K.E., Levene, S.D., Tesmer, V.M., Shay, J.W., Wright, W.E., 2000.

Telomere shortening is proportional to the size of the G-rich telomeric 3’-

overhang. J. Biol. Chem. 275, 19719–19722. doi:10.1074/jbc.M002843200

Ira, G., Pellicioli, A., Balijja, A., Wang, X., Fiorani, S., Carotenuto, W., Liberi, G.,

Bressan, D., Wan, L., Hollingsworth, N.M., Haber, J.E., Foiani, M., 2004. DNA

end resection, homologous recombination and DNA damage checkpoint

activation require CDK1. Nature 431, 1011–1017. doi:10.1038/nature02964

Ishikawa, N., Nakamura, K.I., Izumiyama-Shimomura, N., Aida, J., Ishii, A., Goto,

M., Ishikawa, Y., Asaka, R., Matsuura, M., Hatamochi, A., Kuroiwa, M.,

Takubo, K., 2011. Accelerated in vivo epidermal telomere loss in Werner

syndrome. Aging (Albany. NY). 3, 417–429. doi:100315 [pii]

Page 243: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

228

Izumi, M., Vaughan, O.A., Hutchison, C.J., Gilbert, D.M., 2000. Head and/or CaaX

domain deletions of lamin proteins disrupt preformed lamin A and C but

not lamin B structure in mammalian cells. Mol. Biol. Cell 11, 4323–4337.

Jagatheesan, G., Thanumalayan, S., Muralikrishna, B., Rangaraj, N., Karande, A.A.,

Parnaik, V.K., 1999. Colocalization of intranuclear lamin foci with RNA

splicing factors. J. Cell Sci. 112 ( Pt 2, 4651–4661.

Jegou, T., Chung, I., Heuvelman, G., Wachsmuth, M., Görisch, S.M., Greulich-

Bode, K.M., Boukamp, P., Lichter, P., Rippe, K., 2009. Dynamics of telomeres

and promyelocytic leukemia nuclear bodies in a telomerase-negative

human cell line. Mol. Biol. Cell 20, 2070–82. doi:10.1091/mbc.E08-02-0108

Jeyapalan, J.N., Mendez-Bermudez, A., Zaffaroni, N., Dubrova, Y.E., Royle, N.J.,

2008. Evidence for alternative lengthening of telomeres in liposarcomas in

the absence of ALT-associated PML bodies. Int. J. Cancer 122, 2414–2421.

doi:10.1002/ijc.23412

Jeyapalan, J.N., Varley, H., Foxon, J.L., Pollock, R.E., Jeffreys, A.J., Henson, J.D.,

Reddel, R.R., Royle, N.J., 2005. Activation of the ALT pathway for telomere

maintenance can affect other sequences in the human genome. Hum. Mol.

Genet. 14, 1785–1794. doi:10.1093/hmg/ddi185

Jiang, W.Q., Zhong, Z.H., Nguyen, A., Henson, J.D., Toouli, C.D., Braithwaite, A.W.,

Reddel, R.R., 2009. Induction of alternative lengthening of telomeres-

associated PML bodies by p53/p21 requires HP1 proteins. J. Cell Biol. 185,

797–810. doi:10.1083/jcb.200810084

Johnson, B.R., Nitta, R.T., Frock, R.L., Mounkes, L., Barbie, D. a, Stewart, C.L.,

Harlow, E., Kennedy, B.K., 2004. A-type lamins regulate retinoblastoma

protein function by promoting subnuclear localization and preventing

proteasomal degradation. Proc. Natl. Acad. Sci. U. S. A. 101, 9677–82.

doi:10.1073/pnas.0403250101

Kabotyanski, E.B., Gomelsky, L., Han, J.O., Stamato, T.D., Roth, D.B., 1998.

Double-strand break repair in Ku86- and XRCC4-deficient cells. Nucleic

Acids Res. 26, 5333–5342. doi:10.1093/nar/26.23.5333

Kajstura, J., Pertoldi, B., Leri, A., Beltrami, C.A., Deptala, A., Darzynkiewicz, Z.,

Anversa, P., 2000. Telomere shortening is an in vivo marker of myocyte

replication and aging. Am. J. Pathol. 156, 813–819. doi:10.1016/S0002-

9440(10)64949-8

Page 244: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

229

Karlseder, J., Hoke, K., Mirzoeva, O.K., Bakkenist, C., Kastan, M.B., Petrini, J.H.J., de

Lange, T., 2004. The telomeric protein TRF2 binds the ATM kinase and can

inhibit the ATM-dependent DNA damage response. PLoS Biol. 2, 240.

doi:10.1371/journal.pbio.0020240

Karlseder, J., Smogorzewska, A., de Lange, T., 2002. Senescence induced by

altered telomere state, not telomere loss. Science (80-. ). 295, 2446–9.

doi:10.1126/science.1069523

Kaul, Z., Cesare, A.J., Huschtscha, L.I., Neumann, A.A., Reddel, R.R., 2011. Five

dysfunctional telomeres predict onset of senescence in human cells. EMBO

Rep. doi:10.1038/embor.2011.227

Ketema, M., Sonnenberg, A., 2011. Nesprin-3: a versatile connector between the

nucleus and the cytoskeleton. Biochem. Soc. Trans.

doi:10.1042/BST20110669

Kim, N.W., Piatyszek, M. a, Prowse, K.R., Harley, C.B., West, M.D., Ho, P.L.,

Coviello, G.M., Wright, W.E., Weinrich, S.L., Shay, J.W., 1994. Specific

association of human telomerase activity with immortal cells and cancer.

Science (80-. ). 266, 2011–2015. doi:10.1126/science.7605428

Kipling, D., 1997. Telomere structure and telomerase expression during mouse

development and tumorigenesis. Eur. J. Cancer 33, 792–800.

doi:10.1016/S0959-8049(97)00060-9

Kipling, D., Cooke, H.J., 1990. Hypervariable ultra-long telomeres in mice. Nature

347, 400–402. doi:10.1038/347400a0

Kirschner, J., Brune, T., Wehnert, M., Denecke, J., Wasner, C., Feuer, A.,

Marquardt, T., Ketelsen, U.-P.P., Wieacker, P., Bönnemann, C.G.,

Korinthenberg, R., 2005. p.S143F mutation in lamin A/C: a new phenotype

combining myopathy and progeria. Ann. Neurol. 57, 148–51.

doi:10.1002/ana.20359

Köbberling, J., Dunnigan, M.G., 1986. Familial partial lipodystrophy: two types of

an X linked dominant syndrome, lethal in the hemizygous state. J. Med.

Genet. 23, 120–127. doi:10.1136/jmg.23.2.120

Korfali, N., Wilkie, G.S., Swanson, S.K., Srsen, V., de Las Heras, J., Batrakou, D.G.,

Malik, P., Zuleger, N., Kerr, A.R.W., Florens, L., Schirmer, E.C., 2012. The

nuclear envelope proteome differs notably between tissues. Nucleus 3,

552–64. doi:10.4161/nucl.22257

Page 245: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

230

Krejci, L., Altmannova, V., Spirek, M., Zhao, X., 2012. Homologous recombination

and its regulation. Nucleic Acids Res. doi:10.1093/nar/gks270

Krtolica, A., Parrinello, S., Lockett, S., Desprez, P.Y., Campisi, J., 2001. Senescent

fibroblasts promote epithelial cell growth and tumorigenesis: a link

between cancer and aging. Proc. Natl. Acad. Sci. U. S. A. 98, 12072–12077.

doi:10.1073/pnas.211053698

Kubben, N., Adriaens, M., Meuleman, W., Voncken, J.W., Steensel, B. Van, Misteli,

T., 2012. Mapping of lamin A- and progerin-interacting genome regions.

Chromosoma 121, 447–464. doi:10.1007/s00412-012-0376-7

Kudlow, B. a, Kennedy, B.K., Monnat, R.J., 2007. Werner and Hutchinson-Gilford

progeria syndromes: mechanistic basis of human progeroid diseases. Nat.

Rev. Mol. Cell Biol. 8, 394–404. doi:10.1038/nrm2161

Kudlow, B.A., Stanfel, M.N., Burtner, C.R., Johnston, E.D., Kennedy, B.K., 2008.

Suppression of Proliferative Defects Associated with Processing-defective

Lamin A Mutants by hTERT or Inactivation of p53. Mol. Biol. Cell 19, 5238 –

5248. doi:10.1091/mbc.E08

Kumaran, R.I., Muralikrishna, B., Parnaik, V.K., 2002. Lamin A/C speckles mediate

spatial organization of splicing factor compartments and RNA polymerase

II transcription. J. Cell Biol. 159, 783–793. doi:10.1083/jcb.200204149

Kyo, S., Takakura, M., Fujiwara, T., Inoue, M., 2008. Understanding and exploiting

hTERT promoter regulation for diagnosis and treatment of human cancers.

Cancer Sci. doi:10.1111/j.1349-7006.2008.00878.x

Lachner, M., O’Carroll, D., Rea, S., Mechtler, K., Jenuwein, T., 2001. Methylation of

histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410,

116–120. doi:10.1038/35065132

Lafferty-Whyte, K., Cairney, C.J., Will, M.B., Serakinci, N., Daidone, M.-G.,

Zaffaroni, N., Bilsland, A., Keith, W.N., 2009. A gene expression signature

classifying telomerase and ALT immortalization reveals an hTERT regulatory

network and suggests a mesenchymal stem cell origin for ALT. Oncogene

28, 3765–3774. doi:10.1038/onc.2009.238

Lammerding, J., Fong, L.G., Ji, J.Y., Reue, K., Stewart, C.L., Young, S.G., Lee, R.T.,

2006. Lamins A and C but not lamin B1 regulate nuclear mechanics. J. Biol.

Chem. 281, 25768–25780. doi:10.1074/jbc.M513511200

Page 246: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

231

Lammerding, J., Schulze, P.C., Takahashi, T., Kozlov, S., Sullivan, T., Kamm, R.D.,

Stewart, C.L., Lee, R.T., 2004. Lamin A/C deficiency causes defective nuclear

mechanics and mechanotransduction. J. Clin. Invest. 113, 370–378.

doi:10.1172/JCI200419670

Lansdorp, P.M., Verwoerd, N.P., Van De Rijke, F.M., Dragowska, V., Little, M.T.,

Dirks, R.W., Raap, A.K., Tanke, H.J., 1996. Heterogeneity in telomere length

of human chromosomes. Hum. Mol. Genet. 5, 685–691.

doi:10.1093/hmg/5.5.685

Lattanzi, G., Columbaro, M., Mattioli, E., Cenni, V., Camozzi, D., Wehnert, M.,

Santi, S., Riccio, M., Del Coco, R., Maraldi, N.M., Squarzoni, S., Foisner, R.,

Capanni, C., 2007. Pre-lamin A processing is linked to heterochromatin

organization. J. Cell. Biochem. 102, 1149–1159. doi:10.1002/jcb.21467

Lau, L.M.S., Dagg, R.A., Henson, J.D., Au, A.Y.M., Royds, J.A., Reddel, R.R., 2013.

Detection of alternative lengthening of telomeres by telomere quantitative

PCR. Nucleic Acids Res. 41. doi:10.1093/nar/gks781

Lawless, C., Wang, C., Jurk, D., Merz, A., Zglinicki, T. von, Passos, J.F., 2010.

Quantitative assessment of markers for cell senescence. Exp. Gerontol. 45,

772–778. doi:10.1016/j.exger.2010.01.018

Lee, H.W., Blasco, M.A., Gottlieb, G.J., Horner, J.W., Greider, C.W., DePinho, R.A.,

1998. Essential role of mouse telomerase in highly proliferative organs.

Nature 392, 569–574. doi:10.1038/33345

Lee, K.K., Gruenbaum, Y., Spann, P., Liu, J., Wilson, K.L., 2000. C. elegans nuclear

envelope proteins emerin, MAN1, lamin, and nucleoporins reveal unique

timing of nuclear envelope breakdown during mitosis. Mol. Biol. Cell 11,

3089–99.

Lee, K.K., Haraguchi, T., Lee, R.S., Koujin, T., Hiraoka, Y., Wilson, K.L., 2001.

Distinct functional domains in emerin bind lamin A and DNA-bridging

protein BAF. J. Cell Sci. 114, 4567–4573.

Lehner, C.F., Stick, R., Eppenberger, H.M., Migg, E.A., 1987. Differential

expression of nuclear lamin proteins during chicken development. J. Cell

Biol. 105, 577–587. doi:10.1083/jcb.105.1.577

Levy, M.Z., Allsopp, R.C., Futcher, A.B., Greider, C.W., Harley, C.B., 1992. Telomere

end-replication problem and cell aging. J. Mol. Biol. 225, 951–60.

Page 247: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

232

Li, L., Zou, L., 2005. Sensing, signaling, and responding to DNA damage:

organization of the checkpoint pathways in mammalian cells. J. Cell.

Biochem. 94, 298–306. doi:10.1002/jcb.20355

Liang, F., Jasin, M., 1996. Ku80-deficient cells exhibit excess degradation of

extrachromosomal DNA. J. Biol. Chem. 271, 14405–14411.

doi:10.1074/jbc.271.24.14405

Liang, L., Deng, L., Nguyen, S.C., Zhao, X., Maulion, C.D., Shao, C., Tischfield, J.A.,

2008. Human DNA ligases I and III, but not ligase IV, are required for

microhomology-mediated end joining of DNA double-strand breaks.

Nucleic Acids Res. 36, 3297–3310. doi:10.1093/nar/gkn184

Liu, B., Wang, J., Chan, K.M., Tjia, W.M., Deng, W., Guan, X., Huang, J., Li, K.M.,

Chau, P.Y., Chen, D.J., Pei, D., Pendas, A.M., Cadiñanos, J., López-Otín, C.,

Tse, H.F., Hutchison, C., Chen, J., Cao, Y., Cheah, K.S.E., Tryggvason, K., Zhou,

Z., 2005. Genomic instability in laminopathy-based premature aging. Nat.

Med. 11, 780–5. doi:10.1038/nm1266

Liu, J., Doty, T., Gibson, B., Heyer, W.-D., 2010. Human BRCA2 protein promotes

RAD51 filament formation on RPA-covered single-stranded DNA. Nat.

Struct. Mol. Biol. 17, 1260–1262. doi:10.1038/nsmb.1904

Liu, J., Lee, K.K., Segura-Totten, M., Neufeld, E., Wilson, K.L., Gruenbaum, Y.,

2003. MAN1 and emerin have overlapping function(s) essential for

chromosome segregation and cell division in Caenorhabditis elegans. Proc.

Natl. Acad. Sci. U. S. A. 100, 4598–4603. doi:10.1073/pnas.0730821100

Liu, J., Rolef Ben-Shahar, T., Riemer, D., Treinin, M., Spann, P., Weber, K., Fire, A.,

Gruenbaum, Y., 2000. Essential roles for Caenorhabditis elegans lamin gene

in nuclear organization, cell cycle progression, and spatial organization of

nuclear pore complexes. Mol. Biol. Cell 11, 3937–3947.

doi:10.1091/mbc.11.11.3937

Liu, Y., Rusinol, A., Sinensky, M., Wang, Y., Zou, Y., 2006. DNA damage responses

in progeroid syndromes arise from defective maturation of prelamin A. J.

Cell Sci. 119, 4644–9. doi:10.1242/jcs.03263

Liu, Y., Wang, Y., Rusinol, A.E., Sinensky, M.S., Liu, J., Shell, S.M., Zou, Y., 2008.

Involvement of xeroderma pigmentosum group A (XPA) in progeria arising

from defective maturation of prelamin A. FASEB J. 22, 603–11.

doi:10.1096/fj.07-8598com

Page 248: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

233

Lloyd, D.J., Trembath, R.C., Shackleton, S., 2002. A novel interaction between

lamin A and SREBP1: implications for partial lipodystrophy and other

laminopathies. Hum. Mol. Genet. 11, 769–777. doi:10.1093/hmg/11.7.769

Lo, A.W.I., Sabatier, L., Fouladi, B., Pottier, G., Ricoul, M., Murnane, J.P., 2002.

DNA Amplification by Breakage/Fusion/Bridge Cycles Initiated by

Spontaneous Telomere Loss in a Human Cancer Cell Line. Neoplasia 4, 531–

538. doi:10.1038/sj.neo.7900267

Loayza, D., De Lange, T., 2003. POT1 as a terminal transducer of TRF1 telomere

length control. Nature 423, 1013–1018. doi:10.1038/nature01688

Loewinger, L., Mckeon, F., 1988. Mutations in the nuclear lamin proteins

resulting in their aberrant assembly in the cytoplasm. EMBO J. 7, 2301–

2309.

Lombardi, M.L., Jaalouk, D.E., Shanahan, C.M., Burke, B., Roux, K.J., Lammerding,

J., 2011. The interaction between nesprins and sun proteins at the nuclear

envelope is critical for force transmission between the nucleus and

cytoskeleton. J. Biol. Chem. 286, 26743–53. doi:10.1074/jbc.M111.233700

Londoño-Vallejo, J.A., Der-Sarkissian, H., Cazes, L., Bacchetti, S., Reddel, R.R.,

2004. Alternative Lengthening of Telomeres Is Characterized by High Rates

of Telomeric Exchange. Cancer Res. 64, 2324–2327. doi:10.1158/0008-

5472.CAN-03-4035

Lovejoy, C.A., Li, W., Reisenweber, S., Thongthip, S., Bruno, J., de Lange, T., De, S.,

Petrini, J.H.J., Sung, P.A., Jasin, M., Rosenbluh, J., Zwang, Y., Weir, B.A.,

Hatton, C., Ivanova, E., Macconaill, L., Hanna, M., Hahn, W.C., Lue, N.F.,

Reddel, R.R., Jiao, Y., Kinzler, K., Vogelstein, B., Papadopoulos, N., Meeker,

A.K., 2012. Loss of ATRX, genome instability, and an altered DNA damage

response are hallmarks of the alternative lengthening of Telomeres

pathway. PLoS Genet. 8, 1002772. doi:10.1371/journal.pgen.1002772

Ludérus, M.E.E., Van Steensel, B., Chong, L., Sibon, O.C.M., Cremers, F.F.M., De

Lange, T., 1996. Structure, subnuclear distribution, and nuclear matrix

association of the mammalian telomeric complex. J. Cell Biol. 135, 867–881.

doi:10.1083/jcb.135.4.867

Luke, B., Lingner, J., 2009. TERRA: telomeric repeat-containing RNA. EMBO J. 28,

2503–2510. doi:10.1038/emboj.2009.166

Page 249: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

234

Lussi, Y.C., Hügi, I., Laurell, E., Kutay, U., Fahrenkrog, B., 2011. The nucleoporin

Nup88 is interacting with nuclear lamin A. Mol. Biol. Cell 22, 1080–1090.

doi:10.1091/mbc.E10-05-0463

Lutz, R.J., Trujillo, M.A., Denham, K.S., Wenger, L., Sinensky, M., 1992.

Nucleoplasmic localization of prelamin A: implications for prenylation-

dependent lamin A assembly into the nuclear lamina. Proc. Natl. Acad. Sci.

U. S. A. 89, 3000–4.

Ma, Y., Pannicke, U., Schwarz, K., Lieber, M.R., 2002. Hairpin opening and

overhang processing by an Artemis/DNA-dependent protein kinase

complex in nonhomologous end joining and V(D)J recombination. Cell 108,

781–794. doi:10.1016/S0092-8674(02)00671-2

Machiels, B.M., Zorenc, A.H.G., Endert, J.M., Kuijpers, H.J.H., Van Eys, G.J.J.M.,

Ramaekers, F.C.S., Broers, J.L. V, 1996. An alternative splicing product of the

lamin A/C gene lacks exon 10. J. Biol. Chem. 271, 9249–9253.

doi:10.1074/jbc.271.16.9249

Machwe, A., Xiao, L., Orren, D.K., 2004. TRF2 recruits the Werner syndrome

(WRN) exonuclease for processing of telomeric DNA. Oncogene 23, 149–

156. doi:10.1038/sj.onc.1206906

Mahen, R., Hattori, H., Lee, M., Sharma, P., Jeyasekharan, A.D., Venkitaraman,

A.R., 2013. A-Type Lamins Maintain the Positional Stability of DNA Damage

Repair Foci in Mammalian Nuclei. PLoS One 8.

doi:10.1371/journal.pone.0061893

Majerská, J., Sýkorová, E., Fajkus, J., 2011. Non-telomeric activities of telomerase.

Mol. Biosyst. 7, 1013–1023. doi:10.1039/c0mb00268b

Makarov, V.L., Hirose, Y., Langmore, J.P., 1997. Long G tails at both ends of

human chromosomes suggest a C strand degradation mechanism for

telomere shortening. Cell 88, 657–666. doi:10.1016/S0092-8674(00)81908-X

Mallampalli, M.P., Huyer, G., Bendale, P., Gelb, M.H., Michaelis, S., 2005.

Inhibiting farnesylation reverses the nuclear morphology defect in a HeLa

cell model for Hutchinson-Gilford progeria syndrome. Proc. Natl. Acad. Sci.

U. S. A. 102, 14416–21. doi:10.1073/pnas.0503712102

Manju, K., Muralikrishna, B., Parnaik, V.K., 2006. Expression of disease-causing

lamin A mutants impairs the formation of DNA repair foci. J. Cell Sci. 119,

2704–2714. doi:10.1242/jcs.03009

Page 250: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

235

Mansharamani, M., Wilson, K.L., 2005. Direct binding of nuclear membrane

protein MAN1 to emerin in vitro and two modes of binding to barrier-to-

autointegration factor. J. Biol. Chem. 280, 13863–13870.

doi:10.1074/jbc.M413020200

Markiewicz, E., Dechat, T., Foisner, R., Quinlan, R.A., Hutchison, C.J., 2002. Lamin

A / C Binding Protein LAP2alpha is Required for Nuclear Anchorage of

Retinoblastoma Protein. Mol. Biol. Cell 13, 4401– 4413.

doi:10.1091/mbc.E02

Martens, U.M., Chavez, E.A., Poon, S.S., Schmoor, C., Lansdorp, P.M., 2000.

Accumulation of short telomeres in human fibroblasts prior to replicative

senescence. Exp. Cell Res. 256, 291–299. doi:10.1006/excr.2000.4823

Martens, U.M., Zijlmans, J.M., Poon, S.S., Dragowska, W., Yui, J., Chavez, E.A.,

Ward, R.K., Lansdorp, P.M., 1998. Short telomeres on human chromosome

17p. Nat. Genet. 18, 76–80. doi:10.1038/ng0198-76

Martin, C., Chen, S., Jackson, D.A., 2010. Inheriting nuclear organization: Can

nuclear lamins impart spatial memory during post-mitotic nuclear

assembly? Chromosom. Res. 18, 525–541. doi:10.1007/s10577-010-9137-8

Martin-Gallardo, A., Lamerdin, J., Sopapan, P., Friedman, C., Fertitta, A.L., Garcia,

E., Carrano, A., Negorev, D., Macina, R.A., Trask, B.J., 1995. Molecular

analysis of a novel subtelomeric repeat with polymorphic chromosomal

distribution. Cytogenet. Cell Genet. 71, 289–295. doi:10.1159/000134129

Martin-Ruiz, C., Saretzki, G., Petrie, J., Ladhoff, J., Jeyapalan, J., Wei, W., Sedivy, J.,

Von Zglinicki, T., 2004. Stochastic Variation in Telomere Shortening Rate

Causes Heterogeneity of Human Fibroblast Replicative Life Span. J. Biol.

Chem. 279, 17826–17833. doi:10.1074/jbc.M311980200

Maser, R.S., Choudhury, B., Campbell, P.J., Feng, B., Wong, K.-K., Protopopov, A.,

O’Neil, J., Gutierrez, A., Ivanova, E., Perna, I., Lin, E., Mani, V., Jiang, S.,

McNamara, K., Zaghlul, S., Edkins, S., Stevens, C., Brennan, C., Martin, E.S.,

Wiedemeyer, R., Kabbarah, O., Nogueira, C., Histen, G., Aster, J., Mansour,

M., Duke, V., Foroni, L., Fielding, A.K., Goldstone, A.H., Rowe, J.M., Wang,

Y.A., Look, A.T., Stratton, M.R., Chin, L., Futreal, P.A., DePinho, R.A., 2007.

Chromosomally unstable mouse tumours have genomic alterations similar

to diverse human cancers. Nature 447, 966–971. doi:10.1038/nature05886

Maske, C.P., Hollinshead, M.S., Higbee, N.C., Bergo, M.O., Young, S.G., Vaux, D.J.,

2003. A carboxyl-terminal interaction of lamin B1 is dependent on the

Page 251: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

236

CAAX endoprotease Rce1 and carboxymethylation. J. Cell Biol. 162, 1223–

1232. doi:10.1083/jcb.200303113

Mayer, S., Brüderlein, S., Perner, S., Waibel, I., Holdenried, A., Ciloglu, N., Hasel,

C., Mattfeldt, T., Nielsen, K. V., Möller, P., 2006. Sex-specific telomere length

profiles and age-dependent erosion dynamics of individual chromosome

arms in humans. Cytogenet. Genome Res. 112, 194–201.

doi:10.1159/000089870

McClintock, D., Gordon, L.B., Djabali, K., 2006. Hutchinson-Gilford progeria

mutant lamin A primarily targets human vascular cells as detected by an

anti-Lamin A G608G antibody. Proc. Natl. Acad. Sci. U. S. A. 103, 2154–2159.

doi:10.1073/pnas.0511133103

McElligott, R., Wellinger, R.J., 1997. The terminal DNA structure of mammalian

chromosomes. EMBO J. 16, 3705–14. doi:10.1093/emboj/16.12.3705

McVey, M., Lee, S.E., 2008. MMEJ repair of double-strand breaks (director’s cut):

deleted sequences and alternative endings. Trends Genet.

doi:10.1016/j.tig.2008.08.007

Mefford, H.C., Trask, B.J., 2002. The complex structure and dynamic evolution of

human subtelomeres. Nat. Rev. Genet. 3, 91–102. doi:10.1038/nrg727

Mehta, I.S., Eskiw, C.H., Arican, H.D., Kill, I.R., Bridger, J.M., 2011.

Farnesyltransferase inhibitor treatment restores chromosome territory

positions and active chromosome dynamics in Hutchinson-Gilford progeria

syndrome cells. Genome Biol. doi:10.1186/gb-2011-12-8-r74

Meinke, P., Mattioli, E., Haque, F., Antoku, S., Columbaro, M., Straatman, K.R.,

Worman, H.J., Gundersen, G.G., Lattanzi, G., Wehnert, M., Shackleton, S.,

2014. Muscular dystrophy-associated SUN1 and SUN2 variants disrupt

nuclear-cytoskeletal connections and myonuclear organization. PLoS Genet.

10, e1004605. doi:10.1371/journal.pgen.1004605

Méjat, A., Misteli, T., 2010. LINC complexes in health and disease. Nucleus 1, 40–

52.

Mendez-Bermudez, A., Hills, M., Pickett, H.A., Phan, A.T., Mergny, J.-L., Riou, J.-F.,

Royle, N.J., 2009. Human telomeres that contain (CTAGGG)n repeats show

replication dependent instability in somatic cells and the male germline.

Nucleic Acids Res. 37, 6225–38. doi:10.1093/nar/gkp629

Page 252: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

237

Mendez-Bermudez, A., Royle, N.J., 2011. Deficiency in DNA mismatch repair

increases the rate of telomere shortening in normal human cells. Hum.

Mutat. 32, 939–946. doi:10.1002/humu.21522

Michishita, E., McCord, R. a, Berber, E., Kioi, M., Padilla-Nash, H., Damian, M.,

Cheung, P., Kusumoto, R., Kawahara, T.L.A., Barrett, J.C., Chang, H.Y., Bohr,

V.A., Ried, T., Gozani, O., Chua, K.F., 2008. SIRT6 is a histone H3 lysine 9

deacetylase that modulates telomeric chromatin. Nature 452, 492–6.

doi:10.1038/nature06736

Minc, E., Courvalin, J.C., Buendia, B., 2000. HP1gamma associates with

euchromatin and heterochromatin in mammalian nuclei and chromosomes.

Cytogenet. Cell Genet. 90, 279–284. doi:10.1159/000056789

Mislow, J.M.K., Holaska, J.M., Kim, M.S., Lee, K.K., Segura-Totten, M., Wilson, K.L.,

McNally, E.M., 2002. Nesprin-1α self-associates and binds directly to emerin

and lamin A in vitro. FEBS Lett. 525, 135–140. doi:10.1016/S0014-

5793(02)03105-8

Mitchell, J.R., Collins, K., 2000. Human telomerase activation requires two

independent interactions between telomerase RNA and telomerase reverse

transcriptase. Mol. Cell 6, 361–371. doi:10.1016/S1097-2765(00)00036-8

Moir, R.D., Montag-Lowy, M., Goldman, R.D., 1994. Dynamic properties of

nuclear lamins: Lamin B is associated with sites of DNA replication. J. Cell

Biol. 125, 1201–1212. doi:10.1083/jcb.125.6.1201

Monfouilloux, S., Avet-Loiseau, H., Amarger, V., Balazs, I., Pourcel, C., Vergnaud,

G., 1998. Recent human-specific spreading of a subtelomeric domain.

Genomics 51, 165–176. doi:10.1006/geno.1998.5358

Moreno-Herrero, F., de Jager, M., Dekker, N.H., Kanaar, R., Wyman, C., Dekker,

C., 2005. Mesoscale conformational changes in the DNA-repair complex

Rad50/Mre11/Nbs1 upon binding DNA. Nature 437, 440–443.

doi:10.1038/nature03927

Moser, M.J., Kamath-Loeb, A.S., Jacob, J.E., Bennett, S.E., Oshima, J., Monnat, R.J.,

2000. WRN helicase expression in Werner syndrome cell lines. Nucleic Acids

Res. 28, 648–654. doi:gkd149 [pii]

Moulson, C.L., Fong, L.G., Gardner, J.M., Farber, E.A., Go, G., Passariello, A.,

Grange, D.K., Young, S.G., Miner, J.H., 2007. Increased Progerin Expression

Associated With Unusual LMNA Mutations Causes Severe Progeroid

Syndromes. Hum. Mutat. 28, 882–889. doi:10.1002/humu

Page 253: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

238

Moulson, C.L., Go, G., Gardner, J.M., Van Der Wal, A.C., Smitt, J.H.S., Van Hagen,

J.M., Miner, J.H., 2005. Homozygous and compound heterozygous

mutations in ZMPSTE24 cause the laminopathy restrictive dermopathy. J.

Invest. Dermatol. 125, 913–919. doi:10.1111/j.0022-202X.2005.23846.x

Moyzis, R.K., Buckingham, J.M., Cram, L.S., Dani, M., Deaven, L.L., Jones, M.D.,

Meyne, J., Ratliff, R.L., Wu, J.R., 1988. A highly conserved repetitive DNA

sequence, (TTAGGG)n, present at the telomeres of human chromosomes.

Proc. Natl. Acad. Sci. U. S. A. 85, 6622–6626. doi:10.1073/pnas.85.18.6622

Muchir, A., Bonne, G., van der Kooi, A.J., van Meegen, M., Baas, F., Bolhuis, P.A.,

de Visser, M., Schwartz, K., 2000. Identification of mutations in the gene

encoding lamins A/C in autosomal dominant limb girdle muscular

dystrophy with atrioventricular conduction disturbances (LGMD1B). Hum.

Mol. Genet. 9, 1453–1459. doi:ddd158 [pii]

Muralikrishna, B., Dhawan, J., Rangaraj, N., Parnaik, V.K., 2001. Distinct changes

in intranuclear lamin A/C organization during myoblast differentiation. J.

Cell Sci. 114, 4001–4011.

Murnane, J.P., Sabatier, L., Marder, B.A., Morgan, W.F., 1994. Telomere dynamics

in an immortal human cell line. EMBO J. 13, 4953–4962.

Nabetani, A., Ishikawa, F., 2009. Unusual telomeric DNAs in human telomerase-

negative immortalized cells. Mol. Cell. Biol. 29, 703–713.

doi:10.1128/MCB.00603-08

Nakada, D., Hirano, Y., Sugimoto, K., 2004. Requirement of the Mre11 complex

and exonuclease 1 for activation of the Mec1 signaling pathway. Mol. Cell.

Biol. 24, 10016–10025. doi:10.1128/MCB.24.22.10016-10025.2004

Nakajima, N., Abe, K., 1995. Genomic structure of the mouse A-type lamin gene

locus encoding somatic and germ cell-specific lamins. FEBS Lett. 365, 108–

114. doi:10.1016/0014-5793(95)00453-G

Nandakumar, J., Cech, T.R., 2013. Finding the end: recruitment of telomerase to

telomeres. Nat. Rev. Mol. Cell Biol. 14, 69–82. doi:10.1038/nrm3505

Navarro, C.L., Cadiñanos, J., De Sandre-Giovannoli, A., Bernard, R., Courrier, S.,

Boccaccio, I., Boyer, A., Kleijer, W.J., Wagner, A., Giuliano, F., Beemer, F.A.,

Freije, J.M., Cau, P., Hennekam, R.C.M., López-Otín, C., Badens, C., Lévy, N.,

2005. Loss of ZMPSTE24 (FACE-1) causes autosomal recessive restrictive

dermopathy and accumulation of Lamin A precursors. Hum. Mol. Genet. 14,

1503–1513. doi:10.1093/hmg/ddi159

Page 254: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

239

Navarro, C.L., De Sandre-Giovannoli, A., Bernard, R., Boccaccio, I., Boyer, A.,

Geneviève, D., Hadj-Rabia, S., Gaudy-Marqueste, C., Smitt, H.S., Vabres, P.,

Faivre, L., Verloes, A., Van Essen, T., Flori, E., Hennekam, R., Beemer, F.A.,

Laurent, N., Le Merrer, M., Cau, P., Lévy, N., 2004. Lamin A and ZMPSTE24

(FACE-1) defects cause nuclear disorganization and identify restrictive

dermopathy as a lethal neonatal laminopathy. Hum. Mol. Genet. 13, 2493–

503. doi:10.1093/hmg/ddh265

Navarro, C.L., Esteves-Vieira, V., Courrier, S., Boyer, A., Duong Nguyen, T., Huong,

L.T., Meinke, P., Schroder, W., Cormier-Daire, V., Sznajer, Y., Amor, D.J.,

Lagerstedt, K., Biervliet, M., van den Akker, P.C., Cau, P., Roll, P., Levy, N.,

Badens, C., Wehnert, M., De Sandre-Giovannoli, A., 2013. New ZMPSTE24

(FACE1) mutations in patients affected with restrictive dermopathy or

related progeroid syndromes and mutation update. Eur J Hum Genet.

doi:10.1038/ejhg.2013.258

Nawrot, T.S., Staessen, J.A., Gardner, J.P., Aviv, A., 2004. Telomere length and

possible link to X chromosome. Lancet 363, 507–510. doi:10.1016/S0140-

6736(04)15535-9

Nielsen, P.E., Egholm, M., Berg, R.H., Buchardt, O., 1991. Sequence-selective

recognition of DNA by strand displacement with a thymine-substituted

polyamide. Science (80-. ). 254, 1497–1500. doi:10.1126/science.1962210

Nordfjäll, K., Larefalk, A., Lindgren, P., Holmberg, D., Roos, G., 2005. Telomere

length and heredity: Indications of paternal inheritance. Proc. Natl. Acad.

Sci. U. S. A. 102, 16374–16378. doi:10.1073/pnas.0501724102

Notaro, R., Cimmino, A., Tabarini, D., Rotoli, B., Luzzatto, L., 1997. In vivo

telomere dynamics of human hematopoietic stem cells. Proc. Natl. Acad.

Sci. U. S. A. 94, 13782–13785.

Novelli, G., Muchir, A., Sangiuolo, F., Helbling-Leclerc, A., D’Apice, M.R., Massart,

C., Capon, F., Sbraccia, P., Federici, M., Lauro, R., Tudisco, C., Pallotta, R.,

Scarano, G., Dallapiccola, B., Merlini, L., Bonne, G., 2002. Mandibuloacral

dysplasia is caused by a mutation in LMNA-encoding lamin A/C., American

journal of human genetics. doi:10.1086/341908

Nussenzweig, A., Nussenzweig, M.C., 2007. A Backup DNA Repair Pathway

Moves to the Forefront. Cell 131, 223–225. doi:10.1016/j.cell.2007.10.005

Olive, M., Harten, I., Mitchell, R., Beers, J.K., Djabali, K., Cao, K., Erdos, M.R., Blair,

C., Funke, B., Smoot, L., Gerhard-Herman, M., MacHan, J.T., Kutys, R.,

Virmani, R., Collins, F.S., Wight, T.N., Nabel, E.G., Gordon, L.B., 2010.

Page 255: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

240

Cardiovascular pathology in Hutchinson-Gilford progeria: Correlation with

the vascular pathology of aging. Arterioscler. Thromb. Vasc. Biol. 30, 2301–

2309. doi:10.1161/ATVBAHA.110.209460

Olovnikov, A.M., 1973. A theory of marginotomy. The incomplete copying of

template margin in enzymic synthesis of polynucleotides and biological

significance of the phenomenon. J. Theor. Biol. 41, 181–190.

doi:10.1016/0022-5193(73)90198-7

Opresko, P.L., Otterlei, M., Graakjær, J., Bruheim, P., Dawut, L., Kølvraa, S., May,

A., Seidman, M.M., Bohr, V.A., 2004. The werner syndrome helicase and

exonuclease cooperate to resolve telomeric D loops in a manner regulated

by TRF1 and TRF2. Mol. Cell 14, 763–774. doi:10.1016/j.molcel.2004.05.023

Ottaviani, A., Schluth-bolard, C., Gilson, E., Magdinier, F., 2010. D4Z4 as a

prototype of CTCF and lamins-dependent insulator in human cells. Nucleus

1, 30–36.

Ouellette, M.M., McDaniel, L.D., Wright, W.E., Shay, J.W., Schultz, R. a, 2000. The

establishment of telomerase-immortalized cell lines representing human

chromosome instability syndromes. Hum. Mol. Genet. 9, 403–11.

Padiath, Q.S., Fu, Y.H., 2010. Autosomal Dominant Leukodystrophy Caused by

Lamin B1 Duplications. A Clinical and Molecular Case Study of Altered

Nuclear Function and Disease. Methods Cell Biol. 98, 337–357.

doi:10.1016/S0091-679X(10)98014-X

Palm, W., de Lange, T., 2008. How shelterin protects mammalian telomeres.

Annu. Rev. Genet. 42, 301–34. doi:10.1146/annurev.genet.41.110306.130350

Pare, G.C., Easlick, J.L., Mislow, J.M., McNally, E.M., Kapiloff, M.S., 2005. Nesprin-

1alpha contributes to the targeting of mAKAP to the cardiac myocyte

nuclear envelope. Exp. Cell Res. 303, 388–399.

doi:10.1016/j.yexcr.2004.10.009

Patel, A.A., Steitz, J.A., 2003. Splicing double: insights from the second

spliceosome. Nat. Rev. Mol. Cell Biol. 4, 960–970. doi:10.1038/nrm1259

Patel, J.T., Bottrill, A., Prosser, S.L., Jayaraman, S., Straatman, K., Fry, A.M.,

Shackleton, S., 2014. Mitotic phosphorylation of SUN1 loosens its

connection with the nuclear lamina while the LINC complex remains intact.

Nucleus 5, 462–473. doi:10.4161/nucl.36232

Page 256: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

241

Paull, T.T., Rogakou, E.P., Yamazaki, V., Kirchgessner, C.U., Gellert, M., Bonner,

W.M., 2000. A critical role for histone H2AX in recruitment of repair factors

to nuclear foci after DNA damage. Curr. Biol. 10, 886–895.

doi:10.1016/S0960-9822(00)00610-2

Perner, S., Brüderlein, S., Hasel, C., Waibel, I., Holdenried, A., Ciloglu, N.,

Chopurian, H., Nielsen, K.V., Plesch, A., Högel, J., Möller, P., 2003.

Quantifying telomere lengths of human individual chromosome arms by

centromere-calibrated fluorescence in situ hybridization and digital

imaging. Am. J. Pathol. 163, 1751–1756. doi:10.1016/S0344-0338(04)80508-

9

Perrini, B., Piacentini, L., Fanti, L., Altieri, F., Chichiarelli, S., Berloco, M., Turano, C.,

Ferraro, A., Pimpinelli, S., 2004. HP1 controls telomere capping, telomere

elongation, and telomere silencing by two different mechanisms in

Drosophila. Mol. Cell 15, 467–476. doi:10.1016/j.molcel.2004.06.036

Pollex, R.L., Hegele, R. a, 2004. Hutchinson-Gilford progeria syndrome. Clin.

Genet. 66, 375–81. doi:10.1111/j.1399-0004.2004.00315.x

Pyrpasopoulou, A., Meier, J., Maison, C., Simos, G., Georgatos, S.D., 1996. The

lamin B receptor (LBR) provides essential chromatin docking sites at the

nuclear envelope. EMBO J. 15, 7108–7119.

Quijano-Roy, S., Mbieleu, B., Bönnemann, C.G., Jeannet, P.Y., Colomer, J., Clarke,

N.F., Cuisset, J.M., Roper, H., De Meirleir, L., D’Amico, A., Yaou, R. Ben,

Nascimento, A., Barois, A., Demay, L., Bertini, E., Ferreiro, A., Sewry, C.A.,

Romero, N.B., Ryan, M., Muntoni, F., Guicheney, P., Richard, P., Bonne, G.,

Estournet, B., 2008. De novo LMNA mutations cause a new form of

congenital muscular dystrophy. Ann. Neurol. 64, 177–186.

doi:10.1002/ana.21417

Raz, V., Carlotti, F., Vermolen, B.J., van der Poel, E., Sloos, W.C.R., Knaän-Shanzer,

S., de Vries, A. a F., Hoeben, R.C., Young, I.T., Tanke, H.J., Garini, Y., Dirks,

R.W., 2006. Changes in lamina structure are followed by spatial

reorganization of heterochromatic regions in caspase-8-activated human

mesenchymal stem cells. J. Cell Sci. 119, 4247–56. doi:10.1242/jcs.03180

Raz, V., Vermolen, B.J., Garini, Y., Onderwater, J.J.M., Mommaas-Kienhuis, M. a,

Koster, A.J., Young, I.T., Tanke, H., Dirks, R.W., 2008. The nuclear lamina

promotes telomere aggregation and centromere peripheral localization

during senescence of human mesenchymal stem cells. J. Cell Sci. 121, 4018–

28. doi:10.1242/jcs.034876

Page 257: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

242

Reddel, C.J., Weiss, a S., 2004. Lamin A expression levels are unperturbed at the

normal and mutant alleles but display partial splice site selection in

Hutchinson-Gilford progeria syndrome. J. Med. Genet. 41, 715–7.

doi:10.1136/jmg.2004.019323

Redwood, A.B., Perkins, S.M., Vanderwaal, R.P., Feng, Z., Biehl, K.J., Gonzalez-

Suarez, I., Morgado-Palacin, L., Shi, W., Sage, J., Roti-Roti, J.L., Stewart, C.L.,

Zhang, J., Gonzalo, S., 2011. A dual role for A-type lamins in DNA double-

strand break repair. Cell Cycle 10.

Richmond, T.J., Davey, C.A., 2003. The structure of DNA in the nucleosome core.

Nature 423, 145–150. doi:10.1038/nature01595

Riemer, D., Dodemont, H., Weber, K., 1993. A nuclear lamin of the nematode

Caenorhabditis elegans with unusual features; cDNA cloning and gene

organization. Eur. J. Cell Biol. 62, 214–223.

Riemer, D., Stuurman, N., Berrios, M., Hunter, C., Fisher, P.A., Weber, K., 1995.

Expression of Drosophila lamin C is developmentally regulated: analogies

with vertebrate A-type lamins. J. Cell Sci. 108 ( Pt 1, 3189–3198.

Riethman, H., Ambrosini, A., Paul, S., 2005. Human subtelomere structure and

variation. Chromosom. Res. 13, 505–15. doi:10.1007/s10577-005-0998-1

Riethman, H.C., Spais, C., Buckingham, J., Grady, D., Moyzis, R.K., 1993. Physical

analysis of the terminal 240 kb of DNA from human chromosome 7q.

Genomics 17, 25–32. doi:10.1006/geno.1993.1278

Rober, R., Sauter, H., Weber, K., Osborn, M., 1990. Cells of the cellular immune

and hemopoietic system of the mouse lack lamins A/C: distinction versus

other somatic cells. J. Cell Sci. 95, 587–598.

Rober, R., Weber, K., Osborn, M., 1989. Differential timing of nuclear lamin A/C

expression in the various organs of the mouse embryo and the young

animal: a developmental study. Development 105, 365–378.

Rodriguez, S., Coppedè, F., Sagelius, H., Eriksson, M., 2009. Increased expression

of the Hutchinson-Gilford progeria syndrome truncated lamin A transcript

during cell aging. Eur. J. Hum. Genet. 17, 928–37. doi:10.1038/ejhg.2008.270

Rogakou, E.P., Boon, C., Redon, C., Bonner, W.M., 1999. Megabase chromatin

domains involved in DNA double-strand breaks in vivo. J. Cell Biol. 146,

905–915. doi:10.1083/jcb.146.5.905

Page 258: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

243

Roth, D.B., Wilson, J.H., 1986. Nonhomologous recombination in mammalian

cells: role for short sequence homologies in the joining reaction. Mol. Cell.

Biol. 6, 4295–4304. doi:10.1128/MCB.6.12.4295.Updated

Rybanska-Spaeder, I., Reynolds, T.L., Chou, J., Prakash, M., Jefferson, T., Huso,

D.L., Desiderio, S., Franco, S., 2013. 53BP1 is limiting for NHEJ repair in

ATM-deficient model systems that are subjected to oncogenic stress or

radiation. Mol. Cancer Res. 11, 1223–34. doi:10.1158/1541-7786.MCR-13-

0252-T

Saleh-Gohari, N., Helleday, T., 2004. Conservative homologous recombination

preferentially repairs DNA double-strand breaks in the S phase of the cell

cycle in human cells. Nucleic Acids Res. 32, 3683–3688.

doi:10.1093/nar/gkh703

Samassekou, O., Gadji, M., Drouin, R., Yan, J., 2010. Sizing the ends: Normal

length of human telomeres. Ann. Anat. 192, 284–91.

doi:10.1016/j.aanat.2010.07.005

Samassekou, O., Ntwari, A., Hébert, J., Yan, J., 2009. Individual telomere lengths

in chronic myeloid leukemia. Neoplasia 11, 1146–1154.

doi:10.1593/neo.09836

Sasseville, A.M., Raymond, Y., 1995. Lamin A precursor is localized to

intranuclear foci. J. Cell Sci. 108, 273–285.

Scaffidi, P., Misteli, T., 2006. Lamin A-dependent nuclear defects in human

aging. Science (80-. ). 312, 1059–1063.

Scaffidi, P., Misteli, T., 2005. Reversal of the cellular phenotype in the premature

aging disease Hutchinson-Gilford progeria syndrome. Nat. Med. 11, 440–5.

doi:10.1038/nm1204

Scharner, J., Gnocchi, V.F., Ellis, J. a, Zammit, P.S., 2010. Genotype-phenotype

correlations in laminopathies: how does fate translate? Biochem. Soc. Trans.

38, 257–62. doi:10.1042/BST0380257

Scharner, J., Lu, H.C., Fraternali, F., Ellis, J.A., Zammit, P.S., 2014. Mapping

disease-related missense mutations in the immunoglobulin-like fold

domain of lamin A/C reveals novel genotype-phenotype associations for

laminopathies. Proteins Struct. Funct. Bioinforma. 82, 904–915.

doi:10.1002/prot.24465

Page 259: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

244

Schirmer, E.C., Florens, L., Guan, T., Yates, J.R., Gerace, L., 2003. Nuclear

membrane proteins with potential disease links found by subtractive

proteomics. Science (80-. ). 301, 1380–1382. doi:10.1126/science.1088176

Schirmer, E.C., Gerace, L., 2005. The nuclear membrane proteome: Extending the

envelope. Trends Biochem. Sci. doi:10.1016/j.tibs.2005.08.003

Schoeftner, S., Blasco, M.A., 2010. Chromatin regulation and non-coding RNAs

at mammalian telomeres. Semin. Cell Dev. Biol.

doi:10.1016/j.semcdb.2009.09.015

Schoppee Bortz, P.D., Wamhoff, B.R., 2011. Chromatin immunoprecipitation

(ChIP): Revisiting the efficacy of sample preparation, sonication,

quantification of sheared dna, and analysis via pcr. PLoS One 6, 26015.

doi:10.1371/journal.pone.0026015

Sedelnikova, O. a, Horikawa, I., Zimonjic, D.B., Popescu, N.C., Bonner, W.M.,

Barrett, J.C., 2004. Senescing human cells and ageing mice accumulate DNA

lesions with unrepairable double-strand breaks. Nat. Cell Biol. 6, 168–70.

doi:10.1038/ncb1095

Segura-Totten, M., Kowalski, A.K., Craigie, R., Wilson, K.L., 2002. Barrier-to-

autointegration factor: Major roles in chromatin decondensation and

nuclear assembly. J. Cell Biol. 158, 475–485. doi:10.1083/jcb.200202019

Sfeir, A., de Lange, T., 2012. Removal of Shelterin Reveals the Telomere End-

Protection Problem. Science (80-. ). doi:10.1126/science.1218498

Sfeir, A.J., Chai, W., Shay, J.W., Wright, W.E., 2005. Telomere-end processing the

terminal nucleotides of human chromosomes. Mol. Cell 18, 131–138.

doi:10.1016/j.molcel.2005.02.035

Shackleton, S., Lloyd, D.J., Jackson, S.N., Evans, R., Niermeijer, M.F., Singh, B.M.,

Schmidt, H., Brabant, G., Kumar, S., Durrington, P.N., Gregory, S., O’Rahilly,

S., Trembath, R.C., 2000. LMNA, encoding lamin A/C, is mutated in partial

lipodystrophy. Nat. Genet. 24, 153–6. doi:10.1038/72807

Shackleton, S., Smallwood, D.T., Clayton, P., Wilson, L.C., Agarwal, a K., Garg, A.,

Trembath, R.C., 2005. Compound heterozygous ZMPSTE24 mutations

reduce prelamin A processing and result in a severe progeroid phenotype.

J. Med. Genet. 42, 36. doi:10.1136/jmg.2004.029751

Shagin, D.A., Rebrikov, D. V, Kozhemyako, V.B., Altshuler, I.M., Shcheglov, A.S.,

Zhulidov, P.A., Bogdanova, E.A., Staroverov, D.B., Rasskazov, V.A., Lukyanov,

Page 260: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

245

S., 2002. A novel method for SNP detection using a new duplex-specific

nuclease from crab hepatopancreas. Genome Res. 12, 1935–1942.

doi:10.1101/gr.547002

Shalev, S.A., De Sandre-Giovannoli, A., Shani, A.A., Levy, N., 2007. An association

of Hutchinson-Gilford progeria and malignancy. Am. J. Med. Genet. Part A

143, 1821–1826. doi:10.1002/ajmg.a.31803

Sharma, A., Singh, K., Almasan, A., 2012. Histone H2AX phosphorylation: A

marker for DNA damage. Methods Mol. Biol. 920, 613–626.

doi:10.1007/978-1-61779-998-3-40

Shay, J.W., Bacchetti, S., 1997. A survey of telomerase activity in human cancer.

Eur. J. Cancer 33, 787–91. doi:10.1016/S0959-8049(97)00062-2

Shimi, T., Butin-Israeli, V., Adam, S.A., Hamanaka, R.B., Goldman, A.E., Lucas, C.A.,

Shumaker, D.K., Kosak, S.T., Chandel, N.S., Goldman, R.D., 2011. The role of

nuclear lamin B1 in cell proliferation and senescence. Genes Dev. 25, 2579–

2593. doi:10.1101/gad.179515.111

Shimi, T., Koujin, T., Segura-Totten, M., Wilson, K.L., Haraguchi, T., Hiraoka, Y.,

2004. Dynamic interaction between BAF and emerin revealed by FRAP, FLIP,

and FRET analyses in living HeLa cells. J. Struct. Biol. 147, 31–41.

doi:10.1016/j.jsb.2003.11.013

Shimi, T., Pfleghaar, K., Kojima, S., Pack, C.-G., Solovei, I., Goldman, A.E., Adam,

S.A., Shumaker, D.K., Kinjo, M., Cremer, T., Goldman, R.D., 2008a. The A- and

B-type nuclear lamin networks: microdomains involved in chromatin

organization and transcription. Genes Dev. 22, 3409–21.

doi:10.1101/gad.1735208

Shimi, T., Pfleghaar, K., Kojima, S.I., Pack, C.G., Solovei, I., Goldman, A.E., Adam,

S.A., Shumaker, D.K., Kinjo, M., Cremer, T., Goldman, R.D., 2008b. The A- and

B-type nuclear lamin networks: Microdomains involved in chromatin

organization and transcription. Genes Dev. 22, 3409–3421.

doi:10.1101/gad.1735208

Shoeman, R.L., Traub, P., 1990. The in vitro DNA-binding properties of purified

nuclear lamin proteins and vimentin. J. Biol. Chem. 265, 9055–9061.

Shumaker, D., 2003. The nucleoskeleton: lamins and actin are major players in

essential nuclear functions. Curr. Opin. Cell Biol. 15, 358–366.

doi:10.1016/S0955-0674(03)00050-4

Page 261: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

246

Shumaker, D.K., Dechat, T., Kohlmaier, A., Adam, S. a, Bozovsky, M.R., Erdos,

M.R., Eriksson, M., Goldman, A.E., Khuon, S., Collins, F.S., Jenuwein, T.,

Goldman, R.D., 2006. Mutant nuclear lamin A leads to progressive

alterations of epigenetic control in premature aging. Proc. Natl. Acad. Sci.

U. S. A. 103, 8703–8. doi:10.1073/pnas.0602569103

Shumaker, D.K., Lee, K.K., Tanhehco, Y.C., Craigie, R., Wilson, K.L., 2001. LAP2

binds to BAF·DNA complexes: Requirement for the LEM domain and

modulation by variable regions. EMBO J. 20, 1754–1764.

doi:10.1093/emboj/20.7.1754

Simha, V., Agarwal, A.K., Oral, E.A., Fryns, J.P., Garg, A., 2003. Genetic and

phenotypic heterogeneity in patients with mandibuloacral dysplasia-

associated lipodystrophy. J. Clin. Endocrinol. Metab. 88, 2821–2824.

doi:10.1210/jc.2002-021575

Simon, D.N., Wilson, K.L., 2013. Partners and post-translational modifications of

nuclear lamins. Chromosoma. doi:10.1007/s00412-013-0399-8

Simon, D.N., Zastrow, M.S., Wilson, K.L., 2010. Direct actin binding to A- and B-

type lamin tails and actin filament bundling by the lamin A tail. Nucleus 1,

264–272. doi:10.4161/nucl.11799

Sinensky, M., Fantle, K., Trujillo, M., McLain, T., Kupfer, A., Dalton, M., 1994. The

processing pathway of prelamin A. J. Cell Sci. 107, 61–67.

Singh, M., Hunt, C.R., Pandita, R.K., Kumar, R., Yang, C.-R., Horikoshi, N., Bachoo,

R., Serag, S., Story, M.D., Shay, J.W., Powell, S.N., Gupta, A., Jeffery, J.,

Pandita, S., Chen, B.P.C., Deckbar, D., Löbrich, M., Yang, Q., Khanna, K.K.,

Worman, H.J., Pandita, T.K., 2013. Lamin A/C depletion enhances DNA

damage-induced stalled replication fork arrest. Mol. Cell. Biol. 33, 1210–22.

doi:10.1128/MCB.01676-12

Slagboom, P.E., Droog, S., Boomsma, D.I., 1994. Genetic determination of

telomere size in humans: a twin study of three age groups. Am. J. Hum.

Genet. 55, 876–882.

Smallwood, D.T., Shackleton, S., 2010. Lamin A-linked progerias: is farnesylation

the be all and end all? Biochem. Soc. Trans. 38, 281–286.

Smilenov, L.B., Dhar, S., Pandita, T.E.J.K., 1999. Altered Telomere Nuclear Matrix

Interactions and Nucleosomal Periodicity in Ataxia Telangiectasia Cells

before and after Ionizing Radiation Treatment. Mol. Cell. Biol. 19, 6963–

6971.

Page 262: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

247

Solovei, I., Wang, A.S., Thanisch, K., Schmidt, C.S., Krebs, S., Zwerger, M., Cohen,

T. V., Devys, D., Foisner, R., Peichl, L., Herrmann, H., Blum, H., Engelkamp, D.,

Stewart, C.L., Leonhardt, H., Joffe, B., 2013. LBR and lamin A/C sequentially

tether peripheral heterochromatin and inversely regulate differentiation.

Cell 152, 584–598. doi:10.1016/j.cell.2013.01.009

Sosa, B.A., Rothballer, A., Kutay, U., Schwartz, T.U., 2012. LINC complexes form

by binding of three KASH peptides to domain interfaces of trimeric SUN

proteins. Cell 149, 1035–1047. doi:10.1016/j.cell.2012.03.046

Soutoglou, E., Dorn, J.F., Sengupta, K., Jasin, M., Nussenzweig, A., Ried, T.,

Danuser, G., Misteli, T., 2007. Positional stability of single double-strand

breaks in mammalian cells. Nat. Cell Biol. 9, 675–682. doi:10.1038/ncb1591

Spann, T.P., Goldman, A.E., Wang, C., Huang, S., Goldman, R.D., 2002. Alteration

of nuclear lamin organization inhibits RNA polymerase II-dependent

transcription. J. Cell Biol. 156, 603–608. doi:10.1083/jcb.200112047

Spann, T.P., Moir, R.D., Goldman, a E., Stick, R., Goldman, R.D., 1997. Disruption

of nuclear lamin organization alters the distribution of replication factors

and inhibits DNA synthesis. J. Cell Biol. 136, 1201–12.

Stewart, C., Burke, B., 1987. Teratocarcinoma stem cells and early mouse

embryos contain only a single major lamin polypeptide closely resembling

lamin B. Cell 51, 383–392. doi:10.1016/0092-8674(87)90634-9

Stewart, S. a, Ben-Porath, I., Carey, V.J., O’Connor, B.F., Hahn, W.C., Weinberg, R.

a, 2003. Erosion of the telomeric single-strand overhang at replicative

senescence. Nat. Genet. 33, 492–6. doi:10.1038/ng1127

Stierlé, V., Couprie, J., Östlund, C., Krimm, I., Zinn-Justin, S., Hossenlopp, P.,

Worman, H.J., Courvalin, J.C., Duband-Goulet, I., 2003. The carboxyl-

terminal region common to lamins A and C contains a DNA binding

domain. Biochemistry 42, 4819–4828. doi:10.1021/bi020704g

Stixová, L., Matula, P., Kozubek, S., Gombitová, A., Cmarko, D., Raška, I., Bártová,

E., 2012. Trajectories and nuclear arrangement of PML bodies are

influenced by A-type lamin deficiency. Biol. Cell 104, 418–432.

doi:10.1111/boc.201100053

Stuurman, N., Heins, S., Aebi, U., 1998. Nuclear lamins: their structure, assembly,

and interactions. J. Struct. Biol. 122, 42–66. doi:10.1006/jsbi.1998.3987

Page 263: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

248

Stuurman, N., Meijne, A.M., van der Pol, A.J., de Jong, L., van Driel, R., van

Renswoude, J., 1990. The nuclear matrix from cells of different origin.

Evidence for a common set of matrix proteins. J. Biol. Chem. 265, 5460–

5465.

Stuurman, N., Sasse, B., Fisher, P.A., 1996. Intermediate Filament Protein

Polymerization: Molecular Analysis of Drosophila Nuclear Lamin Head-to-

Tail Binding 117, 1–15.

Sui, X., Kong, N., Wang, Z., Pan, H., 2013. Epigenetic regulation of the human

telomerase reverse transciptase gene: A potential therapeutic target for the

treatment of leukemia (Review). Oncol. Lett. doi:10.3892/ol.2013.1367

Surrallés, J., Hande, M.P., Marcos, R., Lansdorp, P.M., 1999. Accelerated telomere

shortening in the human inactive X chromosome. Am. J. Hum. Genet. 65,

1617–1622. doi:10.1086/302665

Swift, J., Ivanovska, I.L., Buxboim, A., Harada, T., Dingal, P.C.D.P., Pinter, J.,

Pajerowski, J.D., Spinler, K.R., Shin, J.-W., Tewari, M., Rehfeldt, F., Speicher,

D.W., Discher, D.E., 2013. Nuclear lamin-A scales with tissue stiffness and

enhances matrix-directed differentiation. Science (80-. ). 341, 1240104.

doi:10.1126/science.1240104

Symington, L.S., Gautier, J., 2011. Double-Strand Break End Resection and Repair

Pathway Choice. Annu. Rev. Genet. doi:10.1146/annurev-genet-110410-

132435

Tahara, H., Kusunoki, M., Yamanaka, Y., Matsumura, S., Ide, T., 2005. G-tail

telomere HPA: simple measurement of human single-stranded telomeric

overhangs. Nat. Methods 2, 829–831. doi:10.1038/nmeth797

Taimen, P., Pfleghaar, K., Shimi, T., Möller, D., Ben-Harush, K., Erdos, M.R., Adam,

S. a, Herrmann, H., Medalia, O., Collins, F.S., Goldman, A.E., Goldman, R.D.,

2009. A progeria mutation reveals functions for lamin A in nuclear

assembly, architecture, and chromosome organization. Proc. Natl. Acad. Sci.

U. S. A. 106, 20788–93. doi:10.1073/pnas.0911895106

Takai, H., Smogorzewska, A., de Lange, T., 2003. DNA damage foci at

dysfunctional telomeres. Curr. Biol. 13, 1549–56.

Takata, M., Sasaki, M.S., Sonoda, E., Morrison, C., Hashimoto, M., Utsumi, H.,

Yamaguchi-Iwai, Y., Shinohara, a, Takeda, S., 1998. Homologous

recombination and non-homologous end-joining pathways of DNA

double-strand break repair have overlapping roles in the maintenance of

Page 264: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

249

chromosomal integrity in vertebrate cells. EMBO J. 17, 5497–508.

doi:10.1093/emboj/17.18.5497

Taniura, H., Glass, C., Gerace, L., 1995. A chromatin binding site in the tail

domain of nuclear lamins that interacts with core histones. J. Cell Biol. 131,

33–44.

Taylor, M.R., Fain, P.R., Sinagra, G., Robinson, M.L., Robertson, A.D., Carniel, E., Di

Lenarda, A., Bohlmeyer, T.J., Ferguson, D.A., Brodsky, G.L., Boucek, M.M.,

Lascor, J., Moss, A.C., Li, W.L., Stetler, G.L., Muntoni, F., Bristow, M.R.,

Mestroni, L., 2003. Natural history of dilated cardiomyopathy due to lamin

A/C gene mutations. J. Am. Coll. Cardiol. 41, 771–780.

Tomaska, L., Nosek, J., Kramara, J., Griffith, J.D., 2009. Telomeric circles: universal

players in telomere maintenance? Nat. Struct. Mol. Biol. 16, 1010–1015.

doi:10.1038/nsmb.1660

Toth, J.I., Yang, S.H., Qiao, X., Beigneux, A.P., Gelb, M.H., Moulson, C.L., Miner,

J.H., Young, S.G., Fong, L.G., 2005. Blocking protein farnesyltransferase

improves nuclear shape in fibroblasts from humans with progeroid

syndromes. Proc. Natl. Acad. Sci. U. S. A. 102, 12873–12878.

doi:10.1073/pnas.0505767102

Turgay, Y., Champion, L., Balazs, C., Held, M., Toso, A., Gerlich, D.W., Meraldi, P.,

Kutay, U., 2014. SUN proteins facilitate the removal of membranes from

chromatin during nuclear envelope breakdown. J. Cell Biol. 204, 1099–1109.

doi:10.1083/jcb.201310116

Uhlírová, R., Horáková, A.H., Galiová, G., Legartová, S., Matula, P., Fojtová, M.,

Varecha, M., Amrichová, J., Vondrácek, J., Kozubek, S., Bártová, E., 2010.

SUV39h- and A-type lamin-dependent telomere nuclear rearrangement. J.

Cell. Biochem. 109, 915–26. doi:10.1002/jcb.22466

Uziel, T., Lerenthal, Y., Moyal, L., Andegeko, Y., Mittelman, L., Shiloh, Y., 2003.

Requirement of the MRN complex for ATM activation by DNA damage.

EMBO J. 22, 5612–5621. doi:10.1093/emboj/cdg541

Vakoc, C.R., Mandat, S.A., Olenchock, B.A., Blobel, G.A., 2005. Histone H3 lysine 9

methylation and HP1gamma are associated with transcription elongation

through mammalian chromatin. Mol. Cell 19, 381–391.

doi:10.1016/j.molcel.2005.06.011

Varela, I., Cadiñanos, J., Pendás, A.M., Gutiérrez-Fernández, A., Folgueras, A.R.,

Sánchez, L.M., Zhou, Z., Rodríguez, F.J., Stewart, C.L., Vega, J. a, Tryggvason,

Page 265: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

250

K., Freije, J.M.P., López-Otín, C., 2005. Accelerated ageing in mice deficient

in Zmpste24 protease is linked to p53 signalling activation. Nature 437,

564–8. doi:10.1038/nature04019

Varley, H., Pickett, H.A., Foxon, J.L., Reddel, R.R., Royle, N.J., 2002. Molecular

characterization of inter-telomere and intra-telomere mutations in human

ALT cells. Nat. Genet. 30, 301–305. doi:10.1038/ng834

Vasa-Nicotera, M., Brouilette, S., Mangino, M., Thompson, J.R., Braund, P.,

Clemitson, J.-R., Mason, A., Bodycote, C.L., Raleigh, S.M., Louis, E., Samani,

N.J., 2005. Mapping of a major locus that determines telomere length in

humans. Am. J. Hum. Genet. 76, 147–151. doi:10.1086/426734

Vaughan, A., Alvarez-Reyes, M., Bridger, J.M., Broers, J.L., Ramaekers, F.C.,

Wehnert, M., Morris, G.E., Whitfield WGF, Hutchison, C.J., 2001. Both emerin

and lamin C depend on lamin A for localization at the nuclear envelope. J.

Cell Sci. 114, 2577–2590.

Vaziri, H., Benchimol, S., 1998. Reconstitution of telomerase activity in normal

human cells leads to elongation of telomeres and extended replicative life

span. Curr. Biol. 8, 279–282. doi:10.1016/S0960-9822(98)70109-5

Vaziri, H., Schächter, F., Uchida, I., Wei, L., Zhu, X., Effros, R., Cohen, D., Harley,

C.B., 1993. Loss of telomeric DNA during aging of normal and trisomy 21

human lymphocytes. Am. J. Hum. Genet. 52, 661–667.

Verdun, R.E., Crabbe, L., Haggblom, C., Karlseder, J., 2005. Functional human

telomeres are recognized as DNA damage in G2 of the cell cycle. Mol. Cell

20, 551–561. doi:10.1016/j.molcel.2005.09.024

Verdun, R.E., Karlseder, J., 2006. The DNA Damage Machinery and Homologous

Recombination Pathway Act Consecutively to Protect Human Telomeres.

Cell 127, 709–720. doi:10.1016/j.cell.2006.09.034

Vergnes, L., Péterfy, M., Bergo, M.O., Young, S.G., Reue, K., 2004. Lamin B1 is

required for mouse development and nuclear integrity. Proc. Natl. Acad.

Sci. U. S. A. 101, 10428–10433. doi:10.1073/pnas.0401424101

Verstraeten, V.L.R.M., Ji, J.Y., Cummings, K.S., Lee, R.T., Lammerding, J., 2008.

Increased mechanosensitivity and nuclear stiffness in Hutchinson-Gilford

progeria cells: effects of farnesyltransferase inhibitors. Aging Cell 7, 383–93.

doi:10.1111/j.1474-9726.2008.00382.x

Page 266: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

251

Villa, R., Daidone, M.G., Motta, R., Venturini, L., De Marco, C., Vannelli, A.,

Kusamura, S., Baratti, D., Deraco, M., Costa, A., Reddel, R.R., Zaffaroni, N.,

2008. Multiple mechanisms of telomere maintenance exist and differentially

affect clinical outcome in diffuse malignant peritoneal mesothelioma. Clin.

Cancer Res. 14, 4134–4140. doi:10.1158/1078-0432.CCR-08-0099

Vodenicharov, M.D., Wellinger, R.J., 2006. DNA Degradation at Unprotected

Telomeres in Yeast Is Regulated by the CDK1 (Cdc28/Clb) Cell-Cycle Kinase.

Mol. Cell 24, 127–137. doi:10.1016/j.molcel.2006.07.035

Vogel, V., Sheetz, M., 2006. Local force and geometry sensing regulate cell

functions. Nat. Rev. Mol. Cell Biol. 7, 265–275. doi:10.1038/nrm1890

Walne, A.J., Vulliamy, T., Beswick, R., Kirwan, M., Dokal, I., 2008. TINF2 mutations

result in very short telomeres: Analysis of a large cohort of patients with

dyskeratosis congenita and related bone marrow failure syndromes. Blood

112, 3594–3600. doi:10.1182/blood-2008-05-153445

Wang, C., Jurk, D., Maddick, M., Nelson, G., Martin-Ruiz, C., von Zglinicki, T.,

2009. DNA damage response and cellular senescence in tissues of aging

mice. Aging Cell 8, 311–323. doi:ACE481 [pii]\r10.1111/j.1474-

9726.2009.00481.x

Wang, F., Stewart, J.A., Kasbek, C., Zhao, Y., Wright, W.E., Price, C.M., 2012.

Human CST Has Independent Functions during Telomere Duplex

Replication and C-Strand Fill-In. Cell Rep. 2, 1096–1103.

doi:10.1016/j.celrep.2012.10.007

Wang, R.C., Smogorzewska, A., De Lange, T., 2004. Homologous recombination

generates t-loop-sized deletions at human telomeres. Cell 119, 355–368.

doi:10.1016/j.cell.2004.10.011

Weber, K., Plessmann, U., Ulrich, W., 1989. Cytoplasmic intermediate filament

proteins of invertebrates are closer to nuclear lamins than are vertebrate

intermediate filament proteins; sequence characterization of two muscle

proteins of a nematode. EMBO J. 8, 3221–3227.

Weng, N.P., Granger, L., Hodes, R.J., 1997. Telomere lengthening and telomerase

activation during human B cell differentiation. Proc. Natl. Acad. Sci. 94,

10827–32.

Wheeler, M.A., Davies, J.D., Zhang, Q., Emerson, L.J., Hunt, J., Shanahan, C.M.,

Ellis, J.A., 2007. Distinct functional domains in nesprin-1alpha and nesprin-

2beta bind directly to emerin and both interactions are disrupted in X-

Page 267: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

252

linked Emery-Dreifuss muscular dystrophy. Exp. Cell Res. 313, 2845–2857.

doi:10.1016/j.yexcr.2007.03.025

Witt, D.R., Hayden, M.R., Holbrook, K.A., Dale, B.A., Baldwin, V.J., Taylor, G.P.,

1986. Restrictive dermopathy: a newly recognized autosomal recessive skin

dysplasia. Am. J. Med. Genet. 24, 631–648. doi:10.1002/ajmg.1320240408

Wu, D., Flannery, A.R., Cai, H., Ko, E., Cao, K., 2014. Nuclear localization signal

deletion mutants of lamin A and progerin reveal insights into lamin A

processing and emerin targeting. Nucleus 5, 66–74. doi:10.4161/nucl.28068

Wu, P., Takai, H., de Lange, T., 2012. Telomeric 3′ overhangs derive from

resection by Exo1 and Apollo and fill-in by POT1b-associated CST. Cell 150,

39–52. doi:10.1016/j.cell.2012.05.026.Telomeric

Wyllie, F.S., Jones, C.J., Skinner, J.W., Haughton, M.F., Wallis, C., Wynford-

Thomas, D., Faragher, R.G., Kipling, D., 2000. Telomerase prevents the

accelerated cell ageing of Werner syndrome fibroblasts. Nat. Genet. 24, 16–

17. doi:10.1038/71630

Xin, H., Liu, D., Wan, M., Safari, A., Kim, H., Sun, W., O’Connor, M.S., Songyang, Z.,

2007. TPP1 is a homologue of ciliate TEBP-beta and interacts with POT1 to

recruit telomerase. Nature 445, 559–562. doi:10.1038/nature05469

Yang, D., He, Q., Kim, H., Ma, W., Songyang, Z., 2011. TIN2 protein dyskeratosis

congenita missense mutants are defective in association with telomerase. J.

Biol. Chem. 286, 23022–23030. doi:10.1074/jbc.M111.225870

Yang, S.H., Bergo, M.O., Toth, J.I., Qiao, X., Hu, Y., Sandoval, S., Meta, M.,

Bendale, P., Gelb, M.H., Young, S.G., Fong, L.G., 2005. Blocking protein

farnesyltransferase improves nuclear blebbing in mouse fibroblasts with a

targeted Hutchinson-Gilford progeria syndrome mutation. Proc. Natl. Acad.

Sci. U. S. A. 102, 10291–6. doi:10.1073/pnas.0504641102

Yang, S.H., Meta, M., Qiao, X., Frost, D., Bauch, J., Coffinier, C., Majumdar, S.,

Bergo, M.O., Young, S.G., Fong, L.G., 2006. A farnesyltransferase inhibitor

improves disease phenotypes in mice with a Hutchinson-Gilford progeria

syndrome mutation. J. Clin. Invest. 116, 2115–2121.

doi:10.1172/JCI28968.efficacy

Yang, S.H., Qiao, X., Fong, L.G., Young, S.G., 2008. Treatment with a

farnesyltransferase inhibitor improves survival in mice with a Hutchinson-

Gilford progeria syndrome mutation. Biochim. Biophys. Acta 1781, 36–9.

doi:10.1016/j.bbalip.2007.11.003

Page 268: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

253

Ye, J.Z.S., Donigian, J.R., Van Overbeek, M., Loayza, D., Luo, Y., Krutchinsky, A.N.,

Chait, B.T., De Lange, T., 2004. TIN2 binds TRF1 and TRF2 simultaneously

and stabilizes the TRF2 complex on telomeres. J. Biol. Chem. 279, 47264–

47271. doi:10.1074/jbc.M409047200

Ye, Q., Callebaut, I., Pezhman, a, Courvalin, J.C., Worman, H.J., 1997. Domain-

specific interactions of human HP1-type chromodomain proteins and inner

nuclear membrane protein LBR. J. Biol. Chem. 272, 14983–9.

Ye, Q., Worman, H.J., 1996. Interaction between an integral protein of the

nuclear envelope inner membrane and human chromodomain proteins

homologous to Drosophila HP1. J. Biol. Chem. 271, 14653–14656.

doi:10.1074/jbc.271.25.14653

Ye, Q., Worman, H.J., 1994. Primary structure analysis and lamin B and DNA

binding of human LBR, an integral protein of the nuclear envelope inner

membrane. J. Biol. Chem. 269, 11306–11311.

Yeager, T.R., Neumann, A.A., Englezou, A., Huschtscha, L.I., Noble, J.R., Reddel,

R.R., 1999. Telomerase-negative immortalized human cells contain a novel

type of promyelocytic leukemia (PML) body. Cancer Res. 59, 4175–4179.

Young, S.G., Fong, L.G., Michaelis, S., 2005. Prelamin A, Zmpste24, misshapen

cell nuclei, and progeria--new evidence suggesting that protein

farnesylation could be important for disease pathogenesis. J. Lipid Res. 46,

2531–2558. doi:10.1194/jlr.R500011-JLR200

Zhang, Q., Bethmann, C., Worth, N.F., Davies, J.D., Wasner, C., Feuer, A.,

Ragnauth, C.D., Yi, Q., Mellad, J.A., Warren, D.T., Wheeler, M.A., Ellis, J.A.,

Skepper, J.N., Vorgerd, M., Schlotter-Weigel, B., Weissberg, P.L., Roberts,

R.G., Wehnert, M., Shanahan, C.M., 2007. Nesprin-1 and -2 are involved in

the pathogenesis of Emery - Dreifuss muscular dystrophy and are critical

for nuclear envelope integrity. Hum. Mol. Genet. 16, 2816–2833.

doi:10.1093/hmg/ddm238

Zhao, Y., Hoshiyama, H., Shay, J.W., Wright, W.E., 2008. Quantitative telomeric

overhang determination using a double-strand specific nuclease. Nucleic

Acids Res. 36, 14. doi:10.1093/nar/gkm1063

Zheng, R., Ghirlando, R., Lee, M.S., Mizuuchi, K., Krause, M., Craigie, R., 2000.

Barrier-to-autointegration factor (BAF) bridges DNA in a discrete, higher-

order nucleoprotein complex. Proc. Natl. Acad. Sci. U. S. A. 97, 8997–9002.

doi:10.1073/pnas.150240197

Page 269: Relationships between nuclear lamins and telomere … between nuclear lamins and telomere biology in progeroid laminopathies and ... of telomere shortening in progeria …

254

Zhou, Z., Du, X., Cai, Z., Song, X., Zhang, H., Mizuno, T., Suzuki, E., Yee, M.R.,

Berezov, A., Murali, R., Wu, S.L., Karger, B.L., Greene, M.I., Wang, Q., 2012.

Structure of Sad1-UNC84 homology (SUN) domain defines features of

molecular bridge in nuclear envelope. J. Biol. Chem. 287, 5317–5326.

doi:10.1074/jbc.M111.304543

Zhu, J., Wang, H., Bishop, J.M., Blackburn, E.H., 1999. Telomerase extends the

lifespan of virus-transformed human cells without net telomere

lengthening. Proc. Natl. Acad. Sci. U. S. A. 96, 3723–3728.

doi:10.1073/pnas.96.7.3723

Zuleger, N., Boyle, S., Kelly, D., 2013. Specific nuclear envelope transmembrane

proteins can promote the location of chromosomes to and from the

nuclear periphery. Genome Biol. 14, R14. doi:10.1186/gb-2013-14-2-r14

Zwerger, M., Jaalouk, D.E., Lombardi, M.L., Isermann, P., Mauermann, M.,

Dialynas, G., Herrmann, H., Wallrath, L., Lammerding, J., 2013. Myopathic

lamin mutations impair nuclear stability in cells and tissue and disrupt

nucleo-cytoskeletal coupling. Hum. Mol. Genet. 22, 2335–2349.

doi:10.1093/hmg/ddt079