drosophila nedd4-long reduces amphiphysin levels in ...€¦ · chapter 3: results ... c2 –...

107
Drosophila Nedd4-long reduces Amphiphysin levels in muscles and leads to impaired T-tubule formation by Frozan Safi A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Biochemistry University of Toronto © Copyright by Frozan Safi 2015

Upload: others

Post on 19-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

  • Drosophila Nedd4-long reduces Amphiphysin levels in

    muscles and leads to impaired T-tubule formation

    by

    Frozan Safi

    A thesis submitted in conformity with the requirements

    for the degree of Master of Science

    Department of Biochemistry

    University of Toronto

    © Copyright by Frozan Safi 2015

  • ii

    Drosophila Nedd4-long reduces Amphiphysin levels in

    muscles and leads to impaired T-tubule formation

    Frozan Safi

    Master of Science

    Department of Biochemistry

    University of Toronto

    2015

    Abstract

    Drosophila Nedd4 (dNedd4) is a HECT ubiquitin ligase with two main splice isoforms: dNedd4

    short (dNedd4S) and long (dNedd4Lo). We previously showed that while dNedd4S promotes

    neuromuscular synaptogenesis, dNedd4Lo inhibits it. To delineate the cause of this inhibition,

    we searched for binding partners to the N-terminal unique region of dNedd4Lo in embryo

    lysates using mass-spectrometry and identified Amphiphysin (dAmph). dAmph is a postsynaptic

    protein containing SH3-BAR domains, which regulates muscle T-tubule formation in flies. We

    validated the interaction by co-immunoprecipitation and showed direct binding between

    dAmph-SH3 domain and dNedd4Lo-N-terminus. Accordingly, dNedd4Lo was co-expressed

    with dAmph postsynaptically and at muscle T-tubules. Moreover, expression of dNedd4Lo in

    the muscle during embryonic development led to disappearance of dAmph and to impaired T-

    tubules formation, phenocopying amph null mutants. This effect was not seen in muscles

    expressing dNedd4S or a catalytically-inactive dNedd4Lo(CA). We propose that dNedd4Lo

    destabilizes dAmph in muscles, leading to impaired T-tubule formation.

  • iii

    Acknowledgments

    I would like to thank my supervisor, Dr. Daniela Rotin, for her guidance and encouragement

    throughout my Master’s. I am grateful for your mentorship and support. I want to thank my

    supervisory committee members, Dr. Gabrielle Boulianne and Dr. Bill Trimble for their

    insightful suggestions and comments during the committee meetings.

    Thank you to Dr. Alina Kotler for your unwavering guidance, support and mentorship

    throughout this project. Thanks to Dr. Avinash Persaud for teaching me a lot of the lab

    techniques and for the helpful suggestions throughout the project. A special thanks to Yunan

    Zhong, who performed the mass spectrometry screen.

    I’m especially grateful for the support of my fellow lab members. The past few years have been

    memorable because I got to share them with: Avi, Chong, Chen, Anthony and Ruth, who made

    my Master’s study an enjoyable and fun experience. I will miss our buffet lunches at Joe’s and

    Mandarin!

    My project involved a lot of fly genetics, which could not be done without the Boulianne lab,

    who provided all the necessary reagents for my fly work. I want to thank Mike, David and

    Oxana for sharing with me their knowledge in fly genetics. A special thanks to Brenda for

    providing me with the Amph transgenic lines and antibody.

    Thanks to my family and friends for their patience and support throughout my Master’s!

  • iv

    Table of Contents

    Abstract ...........................................................................................................................................ii

    Acknowledgments ......................................................................................................................... iii

    List of Tables .................................................................................................................................. vi

    List of Figures ................................................................................................................................vii

    Abbreviations .................................................................................................................................. ix

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

    1. Introduction ................................................................................................................................. 1

    1.1 Drosophila Muscle Development ........................................................................................ 1

    1.2 Ubiquitination ...................................................................................................................... 4

    1.2.1 Role of Ubiquitination System in NM synaptogenesis............................................ 5

    1.3 The Nedd4 family of E3 ubiquitin ligases ........................................................................... 6

    1.3.1 Physiological Functions of Mammalian Nedd4 ...................................................... 9

    1.3.2 Drosophila Nedd4 family members ...................................................................... 10

    1.4 Muscle Contraction ............................................................................................................ 23

    1.5 Amphiphysin ..................................................................................................................... 26

    1.5.1 Mammalian Amphiphysin ..................................................................................... 26

    1.5.2 Drosophila Amphiphysin ...................................................................................... 30

    1.6 Rationale ............................................................................................................................ 33

    Chapter 2: Materials and Methods ............................................................................................ 34

    2. Methodology ............................................................................................................................. 34

    2.1 Fly Stocks .......................................................................................................................... 34

    2.2 Plasmid Constructs ............................................................................................................ 34

    2.3 Mass Spectrometry ............................................................................................................ 38

  • v

    2.4 In vitro binding assay between dAmph variants and dNedd4Lo N-terminus ................... 39

    2.5 Co-immunoprecipitation of dNedd4Lo and dAmph in Drosophila Schneider 2 tissue culture cells ........................................................................................................................ 39

    2.6 Third instar larval fillet preps and immunofluorescent staining of body wall muscles ..... 41

    2.7 Western Blot analysis of muscle protein levels of dAmph in dNedd4 transgenic larvae.................................................................................................................................. 43

    2.8 Statistical Analysis............................................................................................................. 43

    Chapter 3: Results ....................................................................................................................... 45

    3. Results ....................................................................................................................................... 45

    3.1 dAmph interacts with dNedd4Lo....................................................................................... 45

    3.2 dNedd4Lo N-terminus directly binds to the SH3 domain of dAmph ................................ 53

    3.3 dAmph is enriched postsynaptically and co-localizes with dNedd4Lo ............................. 56

    3.4 dAmph lacking its SH3 domain is mislocalized in muscles .............................................. 59

    3.5 dNedd4Lo regulates the levels of dAmph in the postsynaptic region ............................... 62

    3.6 dNedd4Lo regulates the organization of muscle T-tubules ............................................... 68

    3.7 dAmph protein levels in the muscle are reduced in flies overexpressing dNedd4Lo ....... 72

    Chapter 4: Discussion and Future Directions ........................................................................... 77

    4. Discussion and Future Directions ............................................................................................. 77

    4.1 Validating interaction of dNedd4Lo with other target proteins from the mass spectrometry screen ........................................................................................................... 81

    5. Conclusion ................................................................................................................................ 83

    References...................................................................................................................................... 84

  • vi

    List of Tables

    Table 1: Antibody Staining ........................................................................................................... 40

    Table 2: Fly line crosses ............................................................................................................... 42

    Table 3: Mass spectrometry identification of binding partners to Nedd4Lo unique N-terminus

    and Middle regions. ....................................................................................................................... 46

  • vii

    List of Figures

    Figure 1: Neuromuscular system of the Drosophila melanogaster larva ....................................... 2

    Figure 2: Phylogenetic relationship between Nedd4 and Nedd4-2 ubiquitin ligases among

    different species. .............................................................................................................................. 7

    Figure 3: Schematic representation of dNedd4S and dNedd4Lo unique regions and Akt

    phosphorylation sites. .................................................................................................................... 12

    Figure 4: dNedd4 regulates Comm endocytosis from the muscle membrane. ............................. 15

    Figure 5: Neuromuscular innervation and locomotion defects in larvae overexpressing

    dNedd4Lo in the muscle. ............................................................................................................... 18

    Figure 6: The inhibitory role of dNedd4Lo in NM synaptogenesis is regulated by its N-terminus

    and middle region, not by dAkt phosphorylation. ......................................................................... 20

    Figure 7: T-tubules form junctional triads that are in close association with the sarcoplasmic

    reticulum and mediate excitation-contraction (EC) coupling. ....................................................... 24

    Figure 8: Domain structure of Mammalian Amphiphysin I and Drosophila Amphiphysin

    (dAmph). ........................................................................................................................................ 27

    Figure 9: Drosophila Amphiphysin is required for muscle T-tubule organization. ..................... 31

    Figure 10: The GAL4/UAS system for targeted expression of dNedd4 in the muscles. ............. 36

    Figure 11: dNedd4Lo co-immunoprecipitates with dAmph ......................................................... 51

    Figure 12: Drosophila Amphiphysin directly binds the N-terminus of dNedd4Lo via its SH3

    domain. .......................................................................................................................................... 54

  • viii

    Figure 13: Drosophila Amph and dNedd4 are enriched postsynaptically at the neuromuscular

    junctions and in muscle T-tubules. ................................................................................................ 57

    Figure 14: dAmph(ΔSH3) mislocalizes in Drosophila muscles. ................................................ 60

    Figure 15: Postsynaptic levels of dAmph are significantly reduced in Drosophila muscles

    overexpressing dNedd4Lo. ............................................................................................................ 63

    Figure 16: Muscle-specific overexpression of dNedd4Lo leads to decreased V5-dAmph and

    Dlg levels in the postsynaptic region. ............................................................................................ 66

    Figure 17: Overexpression of dNedd4Lo in muscles impairs the transverse tubule network. ..... 69

    Figure 18: Muscle protein levels of dAmph are reduced in flies overexpressing dNedd4Lo. ..... 73

    Figure 19: Muscle protein levels of V5-dAmph are reduced in flies overexpressing dNedd4Lo.75

  • ix

    Abbreviations

    BAR- bin–amphiphysin–rvs

    BIN1- bridging integrator 1

    BMP- bone morphogenetic protein

    BSA- bovine serum albumin

    C2 – conserved region 2; a Ca2+-binding domain first identified in Ca2+ responsive forms of

    protein kinase C

    Ca+2

    - calcium

    Cav1.2- L-type calcium channel

    CMN2- centronuclear myopathy

    Co-IP- co-immunoprecipitation

    Comm- commissureless

    dAkt- Drosophila Akt

    dAmph- Drosophila Amphiphysin

    dAmph SH3(W->A)- tryptophan -> alanine SH3 domain mutant of dAmph

    dAmph ΔSH3- dAmph with SH3 domain deleted

    DHPR- dihydropyridine receptors

    DLG- discs large

    dNedd4 – Drosophila Nedd4

    dNedd4Lo – dNedd4 long isoform

  • x

    dNedd4Lo S->A – dNedd4L (S39->A, S543->A)

    dNedd4Lo (CA)-catalytically inactive dNedd4Lo

    dNedd4Lo ∆Mid - dNedd4L with its unique middle 159 amino acid-long sequence deleted

    dNedd4Lo Middle (Mid)- residues 304-473 of dNedd4Lo

    dNedd4Lo N-terminus (Nterm)- residues 1-63

    dNedd4Lo ΔNterm – dNedd4L with its unique N-terminal 63 amino acid-long sequence

    dNedd4S – dNedd4 short isoform

    dNedd4S S->A – dNedd4S (S653->A)

    DPP- decapentaplegic

    DS- donkey serum

    E1 – ubiquitin activating enzyme

    E2 – ubiquitin conjugating enzyme

    E3 – ubiquitin ligase

    EC- excitation-contraction

    EDTA- ethylenediaminetetraacetic acid

    ENaC- epithelial Na+ channel

    ERAD- endoplasmic reticulum associated protein degradation

    Faf- fat facets

    FGFR1- Fibroblast growth factor receptor 1

    FLAG-epitope DYKDDDDK

  • xi

    GST- glutathione S-transferase

    HA- hemagglutinin tag YPYDVPDYA

    HECT- homologous to E6-associated protein C-terminus

    HEPES- 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

    Hiw- highwire

    His- poly-histidine tag

    HRP- horseradish peroxidase

    IFM- indirect flight muscles

    ISN- intersegmental nerve branch

    LUBAC- linear ubiquitin chain assembly complex

    MAD- mothers against decapentaplegic

    mEPP- miniature endplate potential

    MS- mass spectrometry

    Myc- epitope EQKLISEEDL

    NGS- normal goat serum

    NM- neuromuscular

    NMJ- neuromuscular junction

    PBS- phospho-buffered saline

    PBT- 0.1% Tween-20 in 1x PBS

    PCR- polymerase chain reaction

  • xii

    PFA- paraformaldehyde

    PMSF- phenylmethylsulfonyl fluoride

    PY motif – the sequence L/PPxY that binds to WW domains

    RBR- RING-between-RING

    RING- really interesting new gene

    RIPA- radioimmunoprecipitation assay buffer

    RNAi- RNAi interference

    Robo- roundabout

    RT – room temperature (22-23oC)

    RTK- receptor tyrosine kinase

    RyR- ryanodine receptors

    S2- Drosophila Schneider 2 cells

    SDS-PAGE- sodium dodecyl sulfate polyacrylamide gel electrophoresis

    SH3- src homology 3 domain

    SH3 dAmph- SH3 domain of dAmph

    SH3PX1- drosophila sorting nexin 9

    SNX9- sorting nexin 9

    SN- segmental nerve branch

    SR- sarcoplasmic reticulum

    SSR- subsynaptic reticulum

  • xiii

    Su(dx)- suppressor of Deltex

    SV- synaptic vesicle

    SVE- synaptic vesicle endocytosis

    TN- transverse nerve

    TNF- tumor necrosis factor

    Tris-HCl – tris hydrochloride

    T-tubule- transverse-tubule

    UAS- upstream activating sequence

    UPS- ubiquitin proteasome system

    V5- GKPIPNPLLGLDST epitope

    WASP- wiskott–aldrich syndrome protein

    WT- wildtype

  • 1

    Chapter 1: Introduction

    1. Introduction

    1.1 Drosophila Muscle Development

    Muscle development is a complex process that in body wall muscles of Drosophila begins by

    mesoderm specification of myoblasts, which is under the control of several transcription factors

    and signalling molecules such as Wingless and decapentaplegic (DPP; a bone morphogenetic

    protein 2 (BMP2) and BMP4 orthologue). Myoblast founder cells fuse to fusion competent

    myoblasts to generate multinucleated muscle cells (Collins & DiAntonio, 2007). The muscle size

    is determined by the number of such fusion events, while muscle identity is controlled by the

    expression of a group of transcriptional regulators termed identity genes. These muscles then

    attach to specialized tendon cells within the epidermis (Collins & DiAntonio, 2007) and are

    innervated to allow them to respond to stimuli arriving from the CNS. Drosophila larvae contain

    30 unique body wall muscles innervated by 32 well-characterized motor neurons (Fig. 1) by a

    process called neuromuscular (NM) synaptogenesis, each adapting a defined innervation

    pathway (Collins & DiAntonio, 2007). The growth cones of these motor neurons reach their

    muscle targets during late embryogenesis, and then differentiate into neuromuscular junction

    synapses. The site of interaction (synapse) between the axon and muscle is called a bouton.

    While many proteins are involved in the regulation of NM synaptogenesis in flies (DiAntonio et

    al., 2001), the role of the ubiquitin system in this process is not well characterized. It is known

    that the deubiquitinating enzyme Fat facet (DiAntonio et al., 2001) and the RING-family

    ubiquitin ligase Highwire (Wan et al., 2000) negatively regulate synapse formation and function.

  • 2

    Figure 1: Neuromuscular system of the Drosophila melanogaster larva (A) Schematic

    representation of an abdominal muscle hemisegment showing all 30 muscles that are innervated

    by one or more of the 5 nerve branches, ISN: intersegmental nerve branch, SNa-d: segmental

    nerve branch a-d; TN: transverse nerve. (B) Confocal microscopy image of third instar larva

    body wall muscles stained with rhodamine-phalloidin showing the symmetrical and segmental

    organization of the muscles stained with an actin probe. (C) Zoomed-in image of (B) showing

    morphological details of the typical muscle pattern present in each hemisegment (numbers

    identify single muscles) (Adapted from Peron et al., 2009)

  • 3

    12 13

  • 4

    1.2 Ubiquitination

    Ubiquitination is the covalent attachment of the 76 amino acid protein ubiquitin to one or more

    lysine residues in the substrate protein, which can signal proteins for degradation, alter their

    localization, trafficking, or function. A covalent bond is formed between the carboxyl group of

    the carboxy-terminal Gly residue of ubiquitin and the ε-amino group of an internal Lys in the

    substrate (Hershko & Ciechanover, 1998). Substrate proteins can be mono-ubiquitinated, multi-

    monoubiquitinated, or poly-ubiquitinated, and the type of ubiquitylation determines the fate of

    the protein. Ubiquitin itself contains seven lysine residues that can form different ubiquitin

    linkage types; for example poly-ubiquitination on Lys48 of ubiquitin targets substrates for

    degradation by the 26S proteosome, while mono-ubiqutination or polyubiquitylation through

    Lys63 are associated with endocytosis and vesicular sorting (Hicke & Dunn, 2003). There is also

    another form of ubiquitin linkage called linear ubiquitination, which does not involve any of the

    lysine residues in the ubiquitin molecule, but occurs between the amino-terminal methionine of

    one ubiquitin and the carboxy-terminal glycine of the next in the chain (Kirisako et al., 2006).

    Linear ubiquitin, chains also known as M1-linked chains, are generated by a novel type of E3

    ubiquitin ligase called the linear ubiquitin chain assembly complex (LUBAC). Linear ubiquitin

    chain linkages have been shown to be important in several innate and adaptive immune signaling

    pathways, including the tumor necrosis factor (TNF) pathway (Gerlach et al., 2011).

    Ubiquitination is an ATP dependent process and it is initiated by the E1-activating enzyme,

    which binds ubiquitin and activates it by adding an ATP before transferring it to an E2-

    conjugating enzyme. Next, the E2 binds directly to the substrate protein or to an E3-ligase that is

    bound to the substrate protein. Finally, the E3 ligase protein removes the ubiquitin from E2 and

    attaches it to the substrate protein (Hershko and Ciechanover, 1998; Deshaies and Joazeiro,

    2009). Organisms contain only a few E1s, many E2s and a large number of E3s, which dictate

    the specificity of the ubiquitination system (Li et al., 2008). E3s are responsible for substrate

    recognition and transfer of ubiquitin onto them, either indirectly (e.g. RING and U-box E3

    ligases) or directly (e.g. HECT E3 ligases (*Rotin & Kumar, 2009). RING (really interesting

    new gene) and U-box (a modified RING motif without the full complement of Zn2+

    -binding

  • 5

    ligands) function as scaffolds to bring the E2 and the substrate in close proximity and facilitate

    the direct transfer of ubiquitin from the E2 to the substrate. In contrast, HECT (homologous to

    E6-associated protein C-terminus) contain a conserved catalytic Cys residue that accepts

    ubiquitin from E2s before directly ubiquitinating the substrate (Ardley & Robinson, 2005).

    Recently, a third class of E3 ubiquitin ligases known as the RING-between-RINGs (RBRs) was

    discovered. RBRs are a subclass of RING-containing E3 ligases and they use the hybrid

    RING/HECT mechanism to transfer ubiquitin to substrates. The proposed mechanism for RBR

    ubiquitination involves binding of the ubiquitin-conjugated E2 to the RING1 domain of the RBR

    E3 ligase. The ubiquitin is then transferred from the E2 to the E3 RING2 domain from which it

    is transferred to the substrate (Wenzel & Klevit, 2012).

    1.2.1 Role of Ubiquitination System in NM synaptogenesis

    The development of functional synapses requires a balance between protein synthesis and

    degradation, which is usually mediated by the ubiquitin proteasome system (UPS) (Speese, 2003;

    Kawabe and Brose, 2011). Ubiquitination of specific pre- and post-synaptic proteins and their

    degradation by the UPS has been shown to be important for neuronal development (Haas and

    Broadie, 2008). Highwire (hiw) is a putative ubiquitin ligase in flies that localizes at synaptic

    terminals and regulates development of the Drosophila NMJ (DiAntonio et al., 2001). Hiw

    functions as a negative regulator of synaptic growth, as loss of function Hiw mutants exhibit

    overgrown synaptic terminals but weakened synaptic transmission (Wan et al., 2000). There is a

    strong genetic interaction between the gene encoding a deubiquitinating enzyme fat facets (faf)

    and hiw in Drosophila (DiAntonio et al., 2001). Neuronal overexpression of faf causes a large

    increase in the number of synaptic boutons, an elaboration of the synaptic branching pattern and

    defects in neurotransmitter release, which is similar to the hiw loss-of-function (DiAntonio et al.,

    2001).

    Other E3 ligase complexes that regulate synaptic development include the anaphase-promoting

    complex/cyclosome which regulates synaptic size and transmission by controlling the

    presynaptic levels of Liprin-alpha (van Roessel et al. 2004) and postsynaptic levels of glutamate

    receptors (Juo & Kaplan, 2004), respectively, at the fly NMJs. In addition, work from our lab

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3831508/#R26http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3831508/#R26

  • 6

    showed that the short isoform of the fly HECT E3 ubiquitin ligase Nedd4 (dNedd4), regulates

    NMJ formation by promoting ubiquitination and internalization of Commissureless (Comm)

    (*Ing et al. 2007), while the long isoform of Nedd4 inhibits NM synaptogenesis (*Zhong et al.

    2011) (See below for detail).

    1.3 The Nedd4 family of E3 ubiquitin ligases

    Nedd4 family members comprise a common C2-WW (n)-HECT domain architecture and belong

    to the HECT family of ubiquitin ligases (Fig. 2). The C2 domain of Nedd4 targets proteins to

    phospholipid membranes in a calcium dependent manner (*Plant et al., 1997; *Plant et al., 2000;

    Dunn et al., 2004), and can also be involved in protein-protein interactions (Morrione et al.,

    1999). Furthermore, the C2 domain is responsible for intramolecular regulatory interactions with

    the HECT domain (Wiesner et al., 2007; Wang et al., 2010; *Persaud et al., 2014). The HECT

    domain is the catalytic domain and is composed of ~350 amino acids located on the C-terminal

    region of HECT E3 ligases (Huibregtse et al., 1995). This domain mediates the interaction with

    E2s (Kar et al., 2012; Sheng et al., 2012) and forms a thioester complex with ubiquitin through a

    cysteine residue (Scheffner & Staub, 2007; *Rotin & Kumar, 2009; Huibregtse et al., 1995;

    Schwarz et al., 1998) to catalyze the attachment of ubiquitin onto substrate proteins. The WW

    domains are protein-protein interaction modules, containing two conserved tryptophan (W)

    residues, which interact primarily with proline rich PPxY (PY) motifs or phospho-

    serine/threonine residues in substrates (Sudol et al., 1995). The number of WW domains in

    Nedd4 proteins varies between family members and species; for instance, human Nedd4 proteins

    contain four WW domains, whereas mouse Nedd4-1 and Drosophila contains three WW

    domains (Yang and Kumar, 2010).

  • 7

    Figure 2: Phylogenetic relationship between Nedd4 and Nedd4-2 ubiquitin ligases among

    different species. Phylogenetic tree representing the conserved modular protein domains of

    Nedd4 and Nedd4-2 among species. The length of each box is proportional to the domain size. c:

    chicken (Gallus gallus); d: fruitfly (Drosophila melanogaster); h: human (Homo sapiens); m:

    mouse (Mus musculus); x: frog (Xenopus tropicalis); and z: zebrafish (Danio rerio) (Adapted

    from Yang & Kumar, 2010).

  • 8

  • 9

    1.3.1 Physiological Functions of Mammalian Nedd4

    There are nine Nedd4 family members in mammals including Nedd4 (also known as Nedd4-1)

    and Nedd4L (also known as Nedd4-2). Nedd4 is known to regulate key signaling pathways, such

    as cellular growth and proliferation that are important in animal development and cancer

    (Reviewed by *Rotin & Kumar, 2009). A few examples are provided below to illustrate the

    identified roles of Nedd4 proteins. Nedd4L regulates the cell surface stability of the epithelial

    Na+ channel (ENaC) by interacting with the PY motifs of ENaC, promoting its endocytosis and

    degradation by ubiquitination (*Staub et al., 1996; Kamynina et al., 2001; *Lu et al., 2007).

    Regulation of ENaC by Nedd4L prevents Na+ overload in epithelial cells and is necessary for

    salt and fluid balance in the body, which is perturbed in PY motif deletion or mutants of ENaC

    that cause the hereditary hypertension disease known as Liddle syndrome (Lifton et al., 2001).

    Nedd4L also regulates ENaC in the lung epithelia since knockout of Nedd4L in this region

    causes mice to develop cystic fibrosis-like symptoms due to elevated ENaC abundance, which

    can be rescued when treated with an ENaC inhibitor, amiloride (*Kimura et al., 2011).

    Nedd4-1 regulates the signalling of the receptor tyrosine kinase (RTK) fibroblast growth factor

    receptor 1 (FGFR1), which controls cellular proliferation and differentiation. FGFR1 contains a

    novel site (VL***PSR) that binds the WW3 domain of Nedd4-1, leading to the ubiquitination

    and down-regulation of FGFR1 at the cell surface, and terminating downstream signalling

    (*Persaud et al., 2011). Accordingly, mutant FGFR1 lacking the recognition motif, FGFR1(Δ6),

    cannot bind Nedd4-1 and thus is retained on the plasma membrane where it can constitutively

    activate downstream signalling in response to FGF stimulation. Expression of FGFR1(Δ6) in

    zebrafish embryos induces defects in brain development, similar to a constitutively active

    FGFR1 (*Persaud et al., 2011). Activation of FGFR1 or EGFR leads to c-Src-mediated

    phosophorylation of Nedd4-1 on several tyrosines, including one in the C2 domain and one in

    the HECT domain, which dissociates the intramolecular inhibitory interaction between the

    HECT and C2 domains, leading to enhanced ubiquitin ligase activity of Nedd4-1 and as a result

    enhanced endocytosis and degradation of FGFR1 (*Persaud et al., 2014).

  • 10

    Nedd4-1 is also involved in the development of the mammalian nervous system, as mutant mice

    deficient in Nedd4 show significant reduction in the skeletal muscle fiber sizes and motorneuron

    numbers. Furthermore, Nedd4 mutant mice contain synapses that are composed of numerous

    smaller nerve terminal profiles and motorneurons that defasciculate upon reaching their target

    muscle. Accordingly, electrophysiological analyses reveal functional impairments of

    neuromuscular (NM) synaptic activities (Liu et al., 2009). Nedd4 mutant NMJ impairments

    include increased spontaneous miniature endplate potential (mEPP) frequency, decreased

    response to membrane depolarization, increase in the number, but decrease in the diameter, of

    pre-synaptic nerve terminal branches. These ultrastructural changes are consistent with

    functional alternation of the NMJs in Nedd4 mutants (Liu et al., 2009). Interestingly, Nedd4-1

    also plays an important role in the regulation of denervation-induced skeletal muscle atrophy in

    animals. Nedd4-1 skeletal muscle-specific knockout mice demonstrate partial protection from

    muscle atrophy, show heavier denervated muscle weights and large type II fibre cross sectional

    areas (Nagpal et al., 2012).

    Nedd4-1 also regulates dendrite development in mammalian neurons as Nedd4-1 deficient mice

    show a decrease in dendrite length and complexity, causing a decrease in the number of

    functional synapses and synaptic transmission (Kawabe et al., 2010). Nedd4-1 forms a ternary

    complex with the serine/threonine kinase TNIK, and Rap2A, which leads to ubiquitination of

    Rap2A by Nedd4-1. Nedd4-1-mediated ubiquitination of Rap2A inhibits its function, which

    reduces the activity of Rap2 effector kinases of the TNIK family and promotes dendrite growth

    (Kawabe et al., 2010).

    1.3.2 Drosophila Nedd4 family members

    Drosophila melanogaster contain three Nedd4 family members, which include Nedd4 (dNedd4),

    suppressor of Deltex (Su(dx), also known as ITCH or AIP4), and Smurf (also known as Lack).

    dNedd4 regulates neuromuscular synaptogenesis by targeting Commisureless for degradation

    (see below). dNedd4 also antagonizes Notch signaling by promoting ligand-independent

    endocytosis and degradation of Notch, as well as its positive regulator Deltex (Sakata et al.,

  • 11

    2004). Notch signaling is also negatively regulated by Su(dx), which is involved in sorting of

    Notch to the late endosome for lysosomal degradation (Wilkin et al., 2004). Smurf negatively

    regulates DPP signalling during embryonic dorsal–ventral patterning by mediating degradation

    of DPP-activated MAD, a Smad protein that transduces the signal of DPP (Liang et al., 2003).

    Moreover, mutations of Smurf are lethal and lead to defects in hindgut organogenesis (Podos et

    al., 2001).

    1.3.2.1 Drosophila Nedd4 (dNedd4)

    In mammals, Nedd4 and Nedd4-2 are encoded by different genes (Kamynina et al., 2001), while

    flies have a single dNedd4 gene, which undergoes alternative splicing to produce several splice

    isoforms, including two prominent isoforms: dNedd4-short (dNedd4S) and dNedd4-long

    (dNedd4Lo) (Fig.3). Differences between the two isoforms of dNedd4 include an alternate start

    codon site resulting in a longer N-terminal region in dNedd4Lo, and an extra exon inserted

    between the WW1 and WW3 domains (*Zhong et al., 2011).

  • 12

    Figure 3: Schematic representation of dNedd4S and dNedd4Lo unique regions and Akt

    phosphorylation sites. Both dNedd4S and dNedd4Lo contain a C2 domain, three WW domains

    and a HECT domain. dNedd4Lo contains two unique regions absent in dNedd4S, an N-terminal

    region (Nterm) and an extra sequence between the WW1 and WW3 domains (Mid). Both

    dNedd4S and dNedd4Lo contain putative Akt phosphorylation sites, however, only the N-

    terminal unique sequence of dNedd4Lo is phosphorylated by dAkt (*Zhong et al., 2011).

  • 13

  • 14

    1.3.2.2 dNedd4S regulation of Commisureless

    dNedd4S was shown to be important for axon guidance at the midline by controlling the levels of

    Commisureless (Comm), a regulator of Roundabout (Robo) (Keleman et al., 2002). Upon axon

    crossing, the surface levels of Robo increase, which prohibits midline recrossing (Kidd et al.,

    1999). The expression of Robo is regulated in part by Comm, which binds to and is regulated by

    dNedd4. In accord, an intact dNedd4 binding site and ubiquitin acceptor sites within Comm are

    required for Comm to regulate Robo (Myat et al., 2002). However, another study has found that

    ubiquitination of Comm by dNedd4 is not involved in axon guidance at the midline (Keleman et

    al., 2005). Therefore, the role of dNedd4 and ubiquitination in midline crossing remains

    controversial.

    In addition to being expressed in axons, all muscles of Drosophila embryos express Comm

    during the period of motoneuron-muscle interactions (Wolf et al., 1998). However, Comm must

    be internalized from the muscle surface prior to synaptogenesis (Wolf et al., 1998). Earlier work

    from our lab showed that dNedd4S promotes endocytosis of Comm from the muscle surface, as

    wildtype Comm is expressed in intracellular vesicles in the muscle, but Comm containing

    mutations in the two PY motifs (L/PPXY) responsible for binding dNedd4 [Comm (2PYAY)],

    or bearing LysArg mutations in all Lys residues that serve as ubiquitin acceptor sites [Comm

    (10KR)], fail to endocytose and display aberrant muscle innervation (Fig. 4) (*Ing et al.,

    2007). RNAi-mediated knockdown of dNedd4 expression or expression of catalytically inactive

    dNedd4(CA), bearing a Cys-to-Ala mutation in the conserved Cys of the HECT domain,

    results in the accumulation of Comm at the muscle surface and synaptic innervation defects,

    similar to those observed in Comm (2PYAY) or the Comm (10KR) mutant. The role of

    Nedd4 in regulating neuromuscular (NM) synaptic activities is conserved in mice, although

    mammals do not have Comm.

  • 15

    Figure 4: dNedd4 regulates Comm endocytosis from the muscle membrane. (A) Motor

    neurons innervating muscles 12 and 13 expressing Comm mutants: Comm (2PY->AY) or Comm

    (10K->R) exhibit aberrant branching (indicated by yellow arrows). RNAi-mediated knockdown

    of muscular dNedd4 expression yields synaptic mutant phenotypes similar to those observed in

    the Comm(2PYAY) or the Comm(10KR) mutant. (B) Comm (WT) accumulates in

    intracellular vesicles, while Comm mutants: Comm (2PY->AY) and Comm (10K->R)

    accumulate at the plasma membrane of the muscle, where they co-localize with the plasma

    membrane marker concanavalin A (ConA), shown in green. (C) Schematic representing dNedd4

    mediated endocytosis of WT Comm in intracellular vesicles, Comm (2PY->AY) and Comm

    (10K->R) fail to endocytose and thus accumulate on the plasma membrane (From *Ing et al.,

    2007).

  • 16

  • 17

    1.3.2.3 dNedd4 regulation of neuromuscular synaptogenesis

    Recently, our lab showed that while dNedd4S is essential for NM synaptogenesis (*Ing et al.,

    2007), the dNedd4Lo isoform inhibits it, resulting in impaired locomotion and larval lethality.

    Specifically, muscle-specific overexpression of dNedd4Lo causes abnormal motor neuron

    innervation along the SNb branch on body wall muscles 13->12, such as inappropriate backward

    innervation from muscles 12->13 and increased number of nerve branches on muscle 12 (Fig. 5)

    (*Zhong et al., 2011).

    In order to determine the mechanism used by dNedd4Lo to inhibit NM synaptogenesis and

    muscle function, our lab examined the unique N-terminal region of dNedd4Lo, which contains a

    putative Akt phosphorylation site (S39) that is phosphorylated by Drosophila Akt (dAkt) (Fig.

    6A). Mutating the dAkt phosphorylation site of dNedd4Lo did not alter the muscle innervation

    defects (Fig. 6A) (*Zhong et al., 2011). Therefore, regulation of the negative role of dNedd4Lo

    in NM synaptogenesis is not mediated by dAkt phosphorylation. In addition, the unique N-

    terminal or middle regions of dNedd4Lo did not bind to the HECT domain, nor inhibit the

    catalytic activity of dNedd4S in vitro, hence dNedd4Lo does not inhibit NM synaptogenesis by

    inhibiting the catalytic activity of dNedd4S. Furthermore, overexpression of dNedd4Lo did not

    affect internalization of Comm in S2 cells. Therefore, dNedd4Lo does not act by interfering with

    the interaction of dNedd4S with its substrate, Comm.

    The unique N-terminal and middle region, as well as the HECT domain of dNedd4Lo, regulate

    the adverse function of dNedd4Lo in NM synaptogenesis. Removing either the N-terminal or the

    middle region or creating an inactivating mutation in the HECT domain (Cys->Ala) rescued the

    lethality and alleviated the muscle innervation defects (Fig6B). Therefore, the unique N terminal

  • 18

    Figure 5: Neuromuscular innervation and locomotion defects in larvae overexpressing

    dNedd4Lo in the muscle. (A) dNedd4 null mutant lethality is partially rescued with dNedd4S,

    but not dNedd4Lo, expression driven by Actin-GAL4 throughout the embryo. (B) Muscle-

    specific overexpression of dNedd4Lo (dNedd4Lo/5-GAL4) leads to aberrant synaptic innervation

    along the SNb branch from body wall muscles 13->12 of third instar larvae (motoreneurons

    shown in red). (C) Overexpression of dNedd4Lo but not dNedd4S inhibits larval locomotion as

    measured by total path length (From *Zhong et al., 2011).

  • 19

  • 20

    Figure 6: The inhibitory role of dNedd4Lo in NM synaptogenesis is regulated by its N-

    terminus and middle region, not by dAkt phosphorylation. (A) Schematic representation of

    dNedd4Lo showing its consensus Akt phosphorylation sites that were mutated to alanine in the

    phosphorylation mutant: dNedd4Lo (S39A,S645A). Mutation of the dAkt phosphorylation sites

    in dNedd4Lo (S39A,S645A) fails to reduce the innervation defects. (B) Schematic representation

    of dNedd4Lo mutants: dNedd4LoΔMiddle (Mid) and dNedd4LoΔN-terminus (Nterm) that has

    its unique middle and N-terminal sequence deleted, respectively. Muscle innervation defects are

    decreased in embryos/larvae expressing dNedd4Lo lacking its Nterm, Mid region or its catalytic

    activity in the muscle (From *Zhong et al., 2011).

  • 21

  • 22

    and middle regions of dNedd4Lo mediate its negative function on NM synaptogenesis and larval

    locomotion, a process that also requires active dNedd4Lo. Thus, we suspect that the unique

    regions of dNedd4Lo regulate NM synaptogenesis and muscle function by an unknown

    mechanism, possibly by interacting with other proteins or substrates (*Zhong et al., 2011).

    To identify such putative interacting partners, Yunan Zhong from our lab incubated embryo

    lysates with purified (immobilized) unique regions of dNedd4Lo (N terminus and Middle

    regions) and identified several interacting proteins by mass spectrometry (See Results section).

    Three of these proteins comprised BAR-SH3 domain architecture: Syndapin, Sorting Nexin 9

    (SH3PX1) and, importantly, Amphiphysin (dAmph), which was a binding partner of the unique

    N-terminal region of dNedd4Lo. In Drosophila, dAmph regulates muscle architecture and

    function, as detailed in the following sections.

  • 23

    1.4 Muscle Contraction

    Muscle contraction occurs when an action potential propagates along the sarcolemma (muscle

    fiber membrane) and triggers the release of calcium from intracellular calcium stores present in

    the sarcoplasmic reticulum (SR) (Al-Qusairi & Laporte, 2011). This Excitation-Contraction

    coupling is mediated by the transverse-tubule (T-tubule) network, which forms regularly-spaced

    perpendicular invaginations toward the interior of the muscle fibre, where they make physical

    contact with the SR in a region called the terminal cisternae, or triad (Fig. 7A). At the triad

    junctions, voltage-dependent Ca2+

    channels in the T-tubular membranes known as

    dihydropyridine receptors (DHPRs) detect the depolarization and transduce it into a signal for

    opening Ca2+

    release channels (also called ryanodine receptors (RyRs)) on the closely opposed

    SR membrane. RyR activation causes Ca2+

    efflux from the SR into the cytosol, which diffuses

    and binds to the regulatory subunit of troponin, thereby removing troponin’s inhibitory effect on

    the contractile proteins, actin and myosin, and allowing muscle contraction (Fig. 7B) (Al-Qusairi

    & Laporte, 2011). Several genes/proteins have been identified that are involved in T-tubule

    biogenesis, including Amphiphysin 2 (BIN1), and its Drosophila orthologue dAmph (Al-Qusairi

    & Laporte, 2011).

  • 24

    Figure 7: T-tubules form junctional triads that are in close association with the

    sarcoplasmic reticulum and mediate excitation-contraction (EC) coupling. (A) Left:

    Electron micrograph of a triad junction showing a T-tubule flanked on both sides by a

    sarcoplasmic reticulum terminal cisternae element. Arrows indicate electron-dense regions

    corresponding to the ryanodine receptor-dihydropyridine receptor complex. Right: Schematic

    representing muscle sarcomere and surrounding membranes. T-tubules are shown in gray as

    specialized invaginations of the muscle plasma membrane surrounded by the sarcoplasmic

    reticulum network shown in blue (Al-Qusairi & Laporte, 2011). (B) A schematic of the skeletal

    muscle fiber illustrating the sequence of EC coupling. An action potential originating from a

    motor neuron propagates into the muscle fiber interior via a specialized system of tubular

    membranes (called T-tubules), which are extensions of the sarcolemma that form specialized

    intracellular junctions called triads. At the triad junctions, voltage-dependent Ca2+

    channels in

    the T-tubular membranes known as dihydropyridine receptors (DHPRs) detect the depolarization

    and activate close by ryanodine receptors (RyRs) on the SR membrane, which causes an efflux

    of Ca2+

    into the cytosol where it can induce muscle contraction (Adapted from Judith A. Heiny).

  • 25

  • 26

    1.5 Amphiphysin

    Amphiphysins are members of the BAR-SH3 domain-containing family of proteins, encoded by

    a single gene in flies (dAmph), two genes in mammals (Amph I & IIa/IIb (BIN1)) and two in

    yeast (Rvs167/Rvs161) (Fig. 8) (Leventis et al., 2001). The N-terminal BAR (Bin-Amphiphysin-

    Rvs) domain forms homodimers, binds negatively charged curved membranes and promotes

    their further curvature and tubulation (Peter et al., 2004). The structure of the Drosophila

    amphiphysin BAR domain has been solved and it is a crescent-shaped dimer that binds

    preferentially to highly curved and negatively charged membranes. Amphiphysin belongs to the

    N-BAR family of proteins because it contains an N-terminal amphipathic helix and BAR

    domain, which allows it to drive membrane curvature in vitro and in vivo (Peter et al., 2004). The

    C-terminal SH3 domain is a protein: protein interaction module that usually binds proline-rich

    motifs (Stamenova et al., 2007). The SH3 domain of Amph can bind ubiquitin (Stamenova et al.,

    2007), which is interesting since proteins with ubiquitin binding domains/motifs are usually

    themselves targeted for ubiquitination, often by Nedd4 family members (Woelk et al., 2006).

    1.5.1 Mammalian Amphiphysin

    Amphiphysins were originally suggested to function in endocytosis, particularly in synaptic

    vesicle (SV) recycling during neurotransmission, by interacting with clathrin/dynamin, a

    function critical for recycling of SVs for successive rounds of neurotransmission (Lichte et al.,

    1992). Amphs were shown to bind nerve termini (Ramjuan et al., 1997) and to bind to several

    components of the endocytic machinery in mammalian cells (e.g. clathrin, endophilin, AP2,

  • 27

    Figure 8: Domain structure of Mammalian Amphiphysin I and Drosophila Amphiphysin

    (dAmph). The central region on Amph I contains binding sites for endophilin, the AP-2, and

    clathrin, while the SH3 domain of Amph I contains the dynamin/synaptojanin binding site. These

    binding regions are absent in the Drosophila orthologue, dAmph (adapted from Craft et al.,

    2008).

  • 28

  • 29

    dynamin (Ramjuan et al., 1997). In accord, mice lacking Amph I exhibit decreased SV recycling

    (Di Paolo et al., 2002), although Bin1 (splice isoform of Amph IIb) knockout mice are lethal but

    did not show endocytic defects and lack the clathrin/AP-2 binding motifs (Fig. 8) (Muller et al.,

    2003). Bin1 is highly expressed in the heart, where it localizes to a specialized membrane system

    of the heart known as the transverse tubule (T-tubule) network that facilitates muscle contraction

    (Muller et al., 2003).

    1.5.1.1 Bridging integrator 1 (Bin1)

    The BAR domain of the isoform of mammalian Amphiphysin 2, Bridging integrator 1 (Bin1), is

    known to be involved in T-tubule formation in cardiac and skeletal muscles (Caldwell et al.,

    2014; Lipsett et al., 2015). Bin1 induces tubular plasma membrane invaginations when expressed

    in non-muscle cells and segregates in tubular portions of the T-tubule system in developing

    myotubes (Lee et al., 2002). Bin1 is mutated in a subset of centronuclear myopathies, a disease

    in which patients exhibit muscle weakness and mislocalized nuclei within muscle fibre (Bohm et

    al., 2010). Patients with Bin1 mutation in these myopathies have severely disorganized T-tubule

    networks (Toussaint et al., 2011).

    Cardiac Bin1 has been implicated in calcium channel trafficking and formation of the inner

    membrane folds of the cardiac T-tubules (Hong et al., 2014; Lipsett et al., 2015). Bin1 localizes

    to cardiac T-tubules with the L-type calcium channel, Cav1.2, by tethering dynamic

    microtubules to membrane scaffolds, allowing targeted delivery of Cav1.2 to cardiac T-tubules

    (Hong et al., 2010). Knockdown of Bin1 reduces surface Cav1.2 and delays development of the

    calcium transient (Hong et al., 2010). In cardiomyopathy, decrease in Bin1 alters T-tubule

    morphology and can cause arrhythmia (Hong et al., 2014). Mice with cardiac Bin1 deletion show

    decreased T-tubule folding, which leads to free diffusion of local extracellular ions, prolonging

    action-potential duration and increasing susceptibility to arrhythmias (Hong et al., 2014).

    Bin1 is also important for the maintenance of intact T-tubule structure and Ca²⁺ homeostasis in

    adult skeletal muscle (Tjondrokoesoemo et al., 2011). Adult mouse skeletal muscles with Bin1

    knockdown display swollen T-tubule structures, alterations to intracellular Ca²⁺ release and

  • 30

    compromised coupling between the voltage-gated calcium channel, DHPR, and the intracellular

    calcium channel, RyR1 (Tjondrokoesoemo et al., 2011).

    1.5.2 Drosophila Amphiphysin

    DAmph was recently shown to participate in plasma membrane remodeling during cleavage

    furrow ingression that is required for de novo formation of cells in Drosophila embryo (Su et al.,

    2013). The BAR domain of dAmph is required for the formation of endocytic tubules that form

    at the cleavage furrow tips. In accord, amphiphysin null embryos fail to form tubules, which

    correlates with faster cleavage furrow ingression rates (Su et al., 2013).

    Interestingly, studies in Drosophila have shown enrichment of dAmph postsynaptically at

    neuromuscular junctions and in a reticular pattern in larval and adult indirect flight muscles,

    where it colocalizes with the T-tubule marker, Discs Large (Leventis et al., 2001; Zelhof et al.,

    2001; Razzaq et al., 2001). Moreover, dAmph is not involved in SVs recycling, since Amph

    mutants lack obvious endocytic defects in SV recycling (Leventis et al., 2001; Zelhof et al.,

    2001; Razzaq et al., 2001), most likely because (like its orthologue Bin1) it lacks the

    clathrin/AP2 binding motifs present in Amph I. DAmph is, however, required for normal

    localization of the postsynaptic proteins Discs large, lethal giant larvae and Scribble (Zelhof et

    al., 2001), as well as the organization of the excitation-contraction coupling machinery of

    muscles (Razzaq et al., 2001). Accordingly, dAmph mutant larvae and flies show severe

    locomotor defects and flight impairments, indicative of defects in muscle function (Leventis et

    al., 2001; Razzaq et al., 2001). Amph null flies show a disordered T-tubule network that is

    largely longitudinal, with expanded tubules and few transverse elements (Fig. 9) (Razzaq et al.,

    2001). RyR localization in the muscle is also disrupted in Amph null flies (Razzaq et al., 2001).

    In accord, expression of wildtype Amph partially rescues the T-tubule network and locomotion

    defects of Amph null flies (Razzaq et al., 2001).

  • 31

    Figure 9: Drosophila Amphiphysin is required for muscle T-tubule organization. (A)

    Amphiphysin is localized on muscle T-tubules in larval body wall muscles and adult indirect

    flight muscles (IFM), Amph (green) and actin (red). (B) Confocal microscope sections showing

    localization of amphiphysin (Amph, green) with the T-tubule marker, Discs-large (DLG, red) in

    larval body wall muscles of wildtype (WT) and Amph null flies. The T-tubule network in Amph

    null flies is largely longitudinal, with expanded tubules and few transverse elements (From

    Razzaq et al., 2001). (C) Schematic diagram illustrating reduced transverse elements in Amph

    null flies.

  • 32

  • 33

    1.6 Rationale

    I hypothesize that dNedd4Lo is a suppressor of dAmph, which regulates stability of dAmph and

    hence muscle architecture and function. In order to elucidate the mechanism of dNedd4Lo

    induced inhibition of muscle function, I investigated:

    1. If dNedd4Lo interacts with dAmph in Drosophila S2 cells and in flies, and if it is a

    substrate for dNedd4Lo

    2. The levels of dAmph in flies overexpressing dNedd4Lo postsynaptically and in muscles

    3. The T-tubule architecture of muscles in flies overexpressing dNedd4Lo

  • 34

    Chapter 2: Materials and Methods

    2. Methodology

    2.1 Fly Stocks

    Fly strains and crosses are depicted in Table 2. Overexpression of dNedd4 or dAmph was

    achieved using the Gal4/UAS system and the early muscle driver, 24B-Gal4 (Figure 10). Flies

    were maintained at room temperature on standard food. All experiments were performed at 25oC.

    All flies used in this experiment have a wild type genetic background because dNedd4 null

    (dNedd4T121FS

    homozygote) flies are homozygous lethal at the embryonic stage and cannot be

    rescued with dNedd4Lo overexpression (Zhong et al., 2011).

    2.2 Plasmid Constructs

    I. Generation of N-terminal and Middle unique sequences of dNedd4Lo: GST tagged

    dNedd4Lo N-terminus (Nterm, residues 1-63) and dNedd4Lo Middle (Mid, residues

    304-473) region were subcloned into the pQE30 vector for expression in bacteria.

    II. Generation of WT, SH3 WA mutant, ΔSH3 and SH3 dAmph: The PCR-amplified

    DNA fragments corresponding to WT dAmph, ∆SH3 dAmph (residues 1-522), and

    SH3 dAmph (residues 523-602) were subcloned into pDEST17 with an N-terminal

    His tag using the Gateway Cloning System (Life technology). PCR-amplified full-

    length WA dAmph SH3 mutant (W580A) was synthesized using the Quick

    Change Site Directed Mutagenesis Kit (Stratagene).

    III. Generation of HA-dAmph WT in pRmHA-3 vector for expression in S2 cells: The

    expressed sequence tag (EST) LD19810, representing the cDNA of Amphiphysin

    was obtained from Berkeley Drosophila Genome Project. PCR amplified full-length

    dAmph containing EcoRI and KpnI sites was subcloned into pRmHA-3 vector for

  • 35

    expression in S2 cells. An N-terminal HA tag was inserted using the Quick Change

    Site Directed Mutagenesis Kit (Stratagene).

  • 36

    Figure 10: The GAL4/UAS system for targeted expression of dNedd4 in the muscles.

    The GAL4/UAS system is based on the properties of the yeast GAL4 transcription factor, which

    activates transcription of its target genes by binding to the upstream activating system, UAS,

    cis-regulator sites. In Drosophila, the two components are carried in separate lines. The driver

    lines provide tissue-specific GAL4 expression and the responder lines carry the coding sequence

    for the gene of interest under the control of the UAS sites. In all the experiments described, the

    muscle-specific driver 24B-GAL4 is used to drive expression of UAS-dNedd4S, UAS-

    dNedd4Lo or dNedd4Lo C->A mutant in the muscles (Adapted from Johnston, 2002).

  • 37

  • 38

    2.3 Mass Spectrometry

    Bacterially expressed GST tagged dNedd4Lo Nterm and Mid region were lysed in sonication

    buffer (1xPBS, 10 g/ml aprotinin, 5 g/ml pepstatin, 1 g/ml leupeptin, 50 g/ml DNase I,

    1mg/mL lysozyme, and 1mM PMSF) at 4oC. Cell lysates were incubated with Glutathione

    sepharose beads for 1 hr at 4oC. After incubation, beads were washed with low salt HNTG (20

    mM HEPES pH 7.5, 150 mM NaCl, 10% glycerol and 0.1% Triton-X100) and 1xPBS. Proteins

    were eluted in 1xPBS (pH 8.0) containing reduced glutathione for 1 hr at 4oC.

    I. Drosophila embryo collection and preparation: Wildtype (WT) adult flies were placed

    on grape fruit agar plates with yeast for embryo collection (0-24 hrs) at 22oC in fly cages.

    ~300 embryos were collected and dechorionated in 50% bleach for 2 min. Embryos were

    lysed in RIPA Buffer (% NP-40, 0.5% Na deoxycholate, 0.1% SDS, 50 mM Tris-HCl,

    pH 7.4, 150mM NaCl, 10% Glycerol and 2 mM EDTA) plus protease inhibitor cocktail

    (Roche Complete tablet) on ice.

    II. Mass Spectrometry Analysis of Drosophila Embryo Lysate using dNedd4Lo N-terminal

    and Middle regions: Bacterially expressed and purified GST dNedd4Lo Nterm, Mid or

    GST control on Glutathione sepharose beads were incubated with embryo lysates (~100

    embryos) at 4oC for 2.5 hours and then washed with 1xPBS. Samples were run on SDS-

    PAGE and Coommassie blue -stained prior to analysis by a Thermo LTQ-Orbitrap

    Hybrid Mass Spectrometer (Hospital for Sickkids, SPARC BioCentre). X! Tandem

    (version: Cyclone, 2010.12.01.2) was set up to search the Drosophila melanogaster

    database in order to identify the peptides fragments generated from trypsin digestion. The

    mass spectrometry data was analyzed using the Scaffold 4.4 software (Proteome Software

    Inc).

  • 39

    2.4 In vitro binding assay between dAmph variants and

    dNedd4Lo N-terminus

    Bacterially expressed and purified His tagged WT dAmph, ΔSH3 dAmph, SH3 dAmph, or W-

    >A dAmph SH3 mutant was incubated with bacterially expressed GST-dNedd4Lo Nterm at 4°C

    for 2 hrs. Supernatant was removed and the beads were washed with low salt HNTG followed by

    1xPBS. Following SDS-PAGE, membranes were blocked and incubated with mouse anti-His to

    detect dAmph. Mouse anti-glutathione S transferase (GST) antibody and HRP-conjugated goat

    anti-mouse antibody were used to detect bacterially expressed GST-dNedd4Lo Nterm (Table 1).

    2.5 Co-immunoprecipitation of dNedd4Lo and dAmph in

    Drosophila Schneider 2 tissue culture cells

    Two mL of sub-confluent Drosophila S2 tissue culture cells (~1.0x106

    cells) were transferred

    into 6-well plates and allowed to grow for ~16 hrs to obtain ~90% confluency prior to

    transfection. Effectene transfection kit (Qiagen) was used to transfect FLAG-dNedd4S or FLAG-

    dNedd4Lo and HA-dAmph in pRmHa3 vectors into S2 cells using standard protocol. 0.4µg of

    DNA each for dNedd4 and dAmph were transfected per well. 24 hours post-transfection, 500µM

    CuSO4 was added in each well to induce expression of the transfected dNedd4 and dAmph

    under the metallothionein promoter. 48 hours post-transfection, cells were lysed in lysis buffer

    (150 mM NaCl, 50 mM HEPES, 1% Triton-X, 10% glycerol, 1.5 mM MgCl2 and 1.0 mM

    EGTA) plus

  • 40

    Table 1: Antibody Staining

    Primary Antibody Blocking 1

    o antibody

    (dilution)

    Source of 1o

    antibodies

    2o antibody (dilution)

    3rd

    instar Larval Muscle Immunohistochemistry

    Mouse anti-Dlg 2%BSA+2% NGS 4F3 anti-discs

    large (1:200) Iowa University

    Goat anti mouse- Alexa

    Fluor488

    (1:600)

    (Invitrogen)

    Mouse anti-Flag 2%BSA+2%NGS Anti-Flag (1:250) Sigma

    Goat anti mouse-Cy3

    (1:600) (Jackson

    ImmunoResearch

    Laboratories, Inc)

    HRP 2%BSA+2%NGS Anti-HRP Alexa

    Fluor488

    (1:400)

    Jackson

    ImmunoResearch

    Laboratories, Inc

    Rabbit anti-V5 2%BSA+2%DS Anti-V5 (1:200) Millipore

    Donkey anti rabbit-cy3

    (1:600) (Jackson

    ImmunoResearch

    Laboratories, Inc)

    Rabbit anti-Myc 2%BSA+2%DS Anti-Myc (1:250) Millipore

    Donkey anti rabbit-cy3

    (1:600) (Jackson

    ImmunoResearch

    Laboratories, Inc)

    Rabbit anti-Amph 2%BSA+2%DS Anti-Amph

    (1:100) G. Boulianne

    Donkey anti rabbit-cy3

    (1:600) (Jackson

    ImmunoResearch

    Laboratories, Inc)

    Western Blotting

    Mouse anti-actin 3% skim milk Anti-actin, JLA20

    (1:100) Iowa University

    Anti-mouse HRP

    (1:10,000)

    Mouse anti-HA 3% skim milk Anti-HA Covance Anti-mouse HRP

    (1:2000) (1:10,000)

    Mouse anti-Flag 3% skim milk Anti-Flag

    (1:5000) Sigma

    Anti-mouse HRP

    (1:10,000)

    Mouse anti-GST 5% dry milk Anti-GST

    (1:5000) Covance

    Anti-mouse HRP

    (1:10,000)

    Mouse anti-His 3% skim milk Anti-His (1:1000) Qiagen Anti-mouse HRP

    (1:10,000)

    Rabbit anti-Amph 3% skim milk Anti-Amph

    (1:500) G. Boulianne

    Anti-rabbit HRP

    (1:10,000)

  • 41

    protease inhibitors (1 mM PMSF, 1 µg/ml each of aprotinin, leupeptin and pepstatin A).

    dNedd4S or dNedd4Lo was immunoprecipitated (IP) with M2 anti-Flag agarose beads and the

    membrane was immunoblotted with anti HA antibody. Flag-dNedd4S and dNedd4Lo were

    detected using M2 anti-Flag antibody and HRP-conjugated goat anti-mouse antibody (Table 1).

    2.6 Third instar larval fillet preps and immunofluorescent staining

    of body wall muscles

    Wandering third instar larvae were dissected using a standard fillet preparation technique (Brent

    et al., 2009), fixed in 4% paraformaldehyde (PFA) (20 min), washed in PBT (0.1% Tween-20 in

    PBS), blocked (2% BSA, 2% Normal Goat Serum (NGS) and/or 2% Donkey Serum (DS) in

    PBT) for 1 hr at RT prior to overnight incubation with primary antibodies at 4oC. See Table 1 for

    antibodies and dilutions used, and Table S1 for fly line crosses used in this study. Muscle

    segments and postsynaptic regions from muscle 6/7 were visualized using a Zeiss AxioVert

    200M confocal microscope with a Zeiss objective lens set to 100x (oil imaging medium),

    numerical aperture 1.4 or 63x (water imaging medium), numerical aperture 1.3. Confocal data

    was acquired at RT with a Hamamatsu C9100-13 EM-CCD camera and images were analyzed

    and deconvolved to reduce background signal using Volocity 6.3.0 (Perkin Elmer).

    I. Analysis of postsynaptic dAmph and T-tubule architecture in Drosophila larva:

    Larvae were incubated overnight with rabbit anti-Amph antibody and mouse anti-

    Dlg antibody at 4oC. The samples were incubated for 2 hrs with Cy3-conjugated

    donkey anti-rabbit and Alexa488

    conjugated goat anti-mouse antibody to visualize

    dAmph and Dlg, respectively (Table 1).

  • 42

    Table 2: Fly line crosses

    Overexpression of dNedd4 in the muscle for immunohistochemistry and western blot analysis

    dAmph5E3

    (Amph null,

    Leventis et al., 2001)

    Control for anti amph antibody

    W1118

    (Wild type ) [source:

    G. Boulianne]

    X 24B-GAL4 (driver control) [source: Bloomington]

    24B-Gal4 X 24B-Gal4 (driver control)

    UAS-dNedd4S-Flag

    (Zhong et al., 2011)

    X 24B-GAL4

    UAS-dNedd4Lo WT-Flag

    (Zhong et al., 2011)

    X 24B-GAL4

    UAS-dNedd4Lo CA-Flag

    (Zhong et al., 2011)

    X 24B-GAL4

    Overexpression of dAmph and dNedd4 in the muscle for immunohistochemistry

    UAS-dAmph WT-V5

    [source: G. Boulianne]

    X 24B-GAL4

    UAS-dAmph WT-V5 X UAS-dNedd4S-Flag; 24B-GAL4

    UAS-dNedd4Lo WT-Flag;

    UAS-dAmph WT-V5

    X 24B-GAL4

    UAS-dAmph WT-V5 X UAS-dNedd4Lo CA-Flag; 24B-GAL4

    UAS-dAmph ΔSH3-Myc

    [source: G. Boulianne]

    X 24B-GAL4

  • 43

    II. Co-localization of V5-dAmph and Flag-dNedd4Lo: Larvae were immunostained with

    M2 anti-Flag antibody and rabbit anti-V5 primary antibodies overnight at 4oC. The

    samples were incubated for 2 hrs with Cy3-conjugated goat anti-mouse antibody,

    Alexa Fluor488

    conjugated goat anti-rabbit antibody, and Alexa Fluor647

    HRP to label

    the presynaptic region, and visualized by confocal microscopy (See Table 1).

    III. Postsynaptic localization of V5-dAmph and MYC-ΔSH3 dAmph: Larvae were

    immunostained with mouse anti-Dlg and rabbit anti-V5 or rabbit anti-MYC primary

    antibodies overnight at 4oC. The samples were incubated for 2 hrs with Cy3-

    conjugated donkey anti-rabbit antibody, Alexa Fluor488

    conjugated goat anti-mouse

    antibody, and Alexa Fluor647

    HRP (Table 1). Co-localization of Dlg and dAmph (V5-

    WT or MYC-ΔSH3) was assessed by Volocity 6.3.0 (Perkin Elmer) and expressed in

    terms of the Pearson's correlation coefficients.

    2.7 Western Blot analysis of muscle protein levels of dAmph in

    dNedd4 transgenic larvae

    Third instar larvae were collected (35 larvae per line, or 10 larvae for the overexpressed V5-

    dAmph), washed in 1x PBS and manually homogenized in lysis buffer plus protease inhibitors (1

    mM PMSF, 1 µg/ml each of aprotinin, leupeptin, and pepstatin A) and 40µM MG132, and 0.2

    mM chloroquine on ice. Samples were examined by Western blotting, using rabbit anti-Amph or

    mouse anti-V5 antibody and M2 anti-Flag antibody to detect dAmph and dNedd4, respectively

    (Table 1). The amount of dAmph was determined relative to Actin using Image Studio software

    after subtracting the background signal from Amph null flies for the endogenous dAmph.

    2.8 Statistical Analysis

    I. Quantification of postsynaptic levels of dAmph in dNedd4 transgenic fly: The

    postsynaptic region was identified as the area demarcated by anti-Dlg immunostaining,

  • 44

    and the intensity of endogenous or overexpressed (using V5 antibodies) dAmph in this

    region was measured using Volocity 6.3.0 software (Perkin Elmer). The intensity of

    dAmph and Dlg in this region was standardized to the levels of dAmph in WT or 24B-

    Gal4 flies for overexpressed V5-dAmph. For endogenous dAmph levels, the intensity of

    dAmph measured in Amph null was subtracted from the intensity of dAmph measured

    for each sample to eliminate background signal from the dAmph antibody. Student’s t-

    test was performed to analyze levels of dAmph in the presence of dNedd4 isoforms and

    the dNedd4Lo(CA) mutant. A p-value of ≤ 0.05 was considered significant.

    II. Quantification of defective muscle T-tubules in dNedd4Lo transgenic fly: The indicated

    numbers of muscle 6/7 hemi-segments for Amph null (28), wildtype (80), dNedd4S (30),

    dNedd4Lo (39) and dNedd4Lo(CA) (94) were analyzed and compared to wildtype to

    determine whether segments appeared normal or abnormal. Muscle segments that showed

    reduced transverse tubules numbers were defined as abnormal. GraphPad Prism 5.0

    software (GraphPad Software, San Diego, CA) and Fisher's exact test in a 2×2

    contingency table were used to analyze the data for T-tubule defects of body wall muscle

    segments. The number of transverse (T) tubules that appeared to branch from a

    longitudinal tubule segment were also counted blindly. For each muscle segment

    analyzed, the mean T-tubules along three different longitudinal segments of a hemi-

    segment were counted. GraphPad Prism 5.0 software and student’s t-test was performed

    to quantify the number of T-tubules branching along segments of longitudinal tubules.

  • 45

    Chapter 3: Results

    3. Results

    3.1 DAmph interacts with dNedd4Lo

    Previously, we found that dNedd4S promotes neuromuscular (NM) synaptogenesis (Ing

    et al 2007), but that dNedd4Lo negatively regulates this process and inhibits muscle function

    (larval locomotion) (Zhong et al., 2011). The unique N-terminal and Middle region, as well as

    the HECT domain of dNedd4Lo, are involved in regulating its adverse function in

    synaptogenesis (Zhong et al., 2011). Since this adverse effect was not caused by inhibition (by

    dNedd4Lo) of the HECT domain of dNedd4S, nor by AKT phosphorylation of dNedd4Lo

    (Zhong et al 2011), we suspected that the unique regions of dNedd4Lo might negatively regulate

    NM synaptogenesis and muscle function by interacting with other proteins. To identify such

    putative interacting partners, embryo lysates were incubated with purified (immobilized) unique

    regions of dNedd4Lo (N-terminus and Middle regions), and several high confidence (>95%)

    interacting proteins were identified by mass spectrometry (Table 3). Drosophila Amphiphysin

    (dAmph), which contains BAR and SH3 domains, was identified as a binding partner of the

    unique N-terminal region of dNedd4Lo (Table 3 and Fig. 1A). To validate the MS results, we

    tested whether dAmph can bind dNedd4Lo by co-immunoprecipitation (co-IP). We transiently

    expressed HA-tagged dAmph and Flag-tagged dNedd4Lo or dNedd4S in Drosophila S2 cells,

    and found binding (co-IP) of dAmph with dNedd4Lo, but not dNedd4S (Fig. 1B).

  • 46

    Table 3: Mass spectrometry identification of binding partners to Nedd4Lo unique N-

    terminus and Middle regions. (A) Bacterially expressed GST tagged dNedd4Lo N-terminus

    (Nterm), residues 1-63, or dNedd4Lo Middle (Mid), residues 304-473, were incubated with

    embryo lysates (~100 embryos) and analyzed by a Thermo LTQ-Orbitrap Hybrid Mass

    Spectrometer. The BAR-SH3 domain containing proteins: dAmph, syndapin as well as sorting

    nexin 9 were identified as interacting proteins of the N-terminus and middle region of

    dNedd4Lo, respectively, using X! Tandem to search the Drosophila melanogaster database. (B)

    dAmph peptides identified as binding partners to dNedd4Lo N-terminus in the MS analysis.

  • 47

    Total Spectral Counts

    Identified Proteins (80)

    Accession

    Number

    MW,

    kDa GST GST

    GST

    dNedd4-

    Lo,

    Middle

    GST

    dNedd4-

    Lo,

    Middle

    GST

    dNedd4-

    Lo, N-

    Term

    GST

    dNedd4-

    Lo, N-

    Term

    Nedd4, isoform H [Drosophila melanogaster] gi|221512757 113 0 0 378 410 123 173

    heat shock protein cognate 3, isoform A

    [Drosophila melanogaster]

    gi|24641402

    (+3) 72 2 5 38 41 46 49

    SH3PX1 [Drosophila melanogaster] gi|21355219 63 0 0 53 67 0 0

    Rm62, isoform A [Drosophila melanogaster]

    gi|24644479

    (+6) 79 0 0 39 39 0 0

    glutathione S transferase D1, isoform A

    [Drosophila melanogaster]

    gi|17737923

    (+1) 24 4 3 11 12 11 11

    CG6459 [Drosophila melanogaster] gi|20130085 29 0 3 20 18 0 2

    Prp19 [Drosophila melanogaster] gi|17647459 55 0 0 31 29 0 0

    heat shock protein cognate 4, isoform A

    [Drosophila melanogaster]

    gi|17737967

    (+5) 71 0 4 29 26 18 20

    glutathione S transferase E6 [Drosophila

    melanogaster] gi|19922532 25 0 0 21 24 0 0

    CG31012, isoform C [Drosophila

    melanogaster] gi|28571934 92 0 0 0 0 16 21

    ribosomal protein S4, isoform A [Drosophila

    melanogaster]

    gi|24663668

    (+1) 29 0 0 17 20 0 0

    beta-Tubulin at 56D, isoform B [Drosophila

    melanogaster] gi|24655737 50 4 9 0 3 0 0

    heat shock protein cognate 5 [Drosophila

    melanogaster] gi|24653595 74 0 0 5 7 6 6

    ribosomal protein S17 [Drosophila

    melanogaster] gi|17647889 15 0 2 9 9 2 3

    Roe1 [Drosophila melanogaster] gi|24653432 24 0 0 0 0 8 6

    CG6905, isoform A [Drosophila melanogaster]

    gi|19922992

    (+1) 93 0 0 13 12 0 0

    ribosomal protein S3A, isoform A [Drosophila

    melanogaster] gi|17864162 30 0 0 10 9 0 2

    belle [Drosophila melanogaster] gi|17985987 85 0 0 12 16 0 0

    ribosomal protein S9, isoform A [Drosophila

    melanogaster]

    gi|24661707

    (+1) 23 0 0 11 5 0 0

    ribosomal protein S15Aa, isoform D

    [Drosophila melanogaster]

    gi|17975567

    (+4) 15 0 0 7 6 0 0

    receptor of activated protein kinase C 1

    [Drosophila melanogaster] gi|17137396 36 0 0 7 11 0 0

    ribosomal protein L5, isoform A [Drosophila

    melanogaster]

    gi|116007382

    (+1) 34 0 0 7 10 0 0

    ribosomal protein S8 [Drosophila

    melanogaster] gi|24651181 24 0 0 7 7 0 0

    cabeza [Drosophila melanogaster] gi|24642436 39 0 0 7 7 0 0

    string of pearls [Drosophila melanogaster] gi|17136734 29 0 0 5 8 0 0

    actin 5C, isoform B [Drosophila melanogaster]

    gi|17530805

    (+4) 42 0 7 0 0 2 3

    growl, isoform A [Drosophila melanogaster] gi|21355167 60 0 0 6 6 0 0

    Table 3: Mass spectrometry identification of binding partners to Nedd4Lo

    unique N-terminus and Middle regions

    A. Performed by Yunan Zhong

  • 48

    ribosomal protein L23A [Drosophila

    melanogaster] gi|24655502 29 0 0 8 4 0 2

    ribosomal protein LP0 [Drosophila

    melanogaster] gi|17737731 34 0 0 3 7 0 0

    ribosomal protein L26 [Drosophila

    melanogaster] gi|21357853 17 0 0 0 4 0 0

    alpha-Tubulin at 84B [Drosophila

    melanogaster]

    gi|17136564

    (+2) 50 3 4 0 0 0 0

    argonaute 2, isoform B [Drosophila

    melanogaster]

    gi|24664664

    (+1) 137 0 0 4 3 0 0

    ribosomal protein S15, isoform A [Drosophila

    melanogaster] gi|19922390 17 0 0 5 4 0 0

    syndapin [Drosophila melanogaster] gi|28571785 56 0 0 0 0 4 5

    ribosomal protein L19, isoform A [Drosophila

    melanogaster]

    gi|17136322

    (+1) 24 0 0 5 5 0 0

    heterogeneous nuclear ribonucleoprotein at

    87F, isoform A [Drosophila melanogaster]

    gi|17136624

    (+1) 39 0 0 3 9 0 0

    downstream of receptor kinase, isoform A

    [Drosophila melanogaster]

    gi|17136708

    (+5) 24 0 0 0 0 4 6

    amphiphysin [Drosophila melanogaster] gi|17647155 66 0 0 0 0 4 4

    ribosomal protein L22 [Drosophila

    melanogaster] gi|17137152 31 0 0 5 4 0 0

    ribosomal protein S25, isoform A [Drosophila

    melanogaster]

    gi|24645865

    (+1) 13 0 0 3 2 0 0

    Fmr1, isoform A [Drosophila melanogaster]

    gi|19922726

    (+4) 76 0 0 4 5 0 0

    vasa intronic gene, isoform A [Drosophila

    melanogaster]

    gi|17737419

    (+3) 53 0 0 5 4 0 0

    glutathione S transferase D5 [Drosophila

    melanogaster] gi|45549270 25 0 0 2 4 0 2

    capping protein alpha [Drosophila

    melanogaster] gi|19922662 33 0 0 0 0 4 6

    capping protein beta [Drosophila

    melanogaster] gi|17136938 31 0 0 0 0 4 4

    elongation factor 1alpha48D, isoform A

    [Drosophila melanogaster]

    gi|17137572

    (+1) 50 0 0 0 3 0 0

    ribosomal protein L23 [Drosophila

    melanogaster] gi|17647883 15 0 0 5 5 0 0

    ribosomal protein L11 [Drosophila

    melanogaster] gi|17137026 21 0 0 3 4 0 0

    glyceraldehyde 3 phosphate dehydrogenase 2,

    isoform B [Drosophila melanogaster]

    gi|17933600

    (+1) 35 2 3 0 2 0 0

    ribosomal protein S7, isoform A [Drosophila

    melanogaster]

    gi|21358459

    (+3) 22 0 0 0 4 0 0

    ribosomal protein S23 [Drosophila

    melanogaster] gi|22024141 16 0 0 0 2 0 0

    histone H1 [Drosophila melanogaster]

    gi|24585669

    (+21) 26 0 0 2 4 0 0

    beta'-coatomer protein [Drosophila

    melanogaster] gi|24584107 103 0 2 0 0 0 0

    ypsilon schachtel [Drosophila melanogaster] gi|24663131 38 0 0 3 2 0 0

    SF2 [Drosophila melanogaster] gi|21358097 28 0 0 2 5 0 0

    Qm, isoform C [Drosophila melanogaster]

    gi|221513692

    (+1) 26 0 0 0 3 0 0

  • 49

    ribosomal protein S11, isoform C [Drosophila

    melanogaster]

    gi|24652922

    (+1) 18 0 0 0 2 0 0

    ribosomal protein L24 [Drosophila

    melanogaster] gi|19921254 18 0 0 0 4 0 0

    mitochondrial ribosomal protein L11

    [Drosophila melanogaster] gi|17737961 22 0 0 3 2 0 0

    Aly [Drosophila melanogaster] gi|21356157 28 0 0 0 2 0 0

    ribosomal protein L12, isoform A [Drosophila

    melanogaster]

    gi|17864452

    (+2) 18 0 0 3 2 0 0

    ribosomal protein S13, isoform A [Drosophila

    melanogaster]

    gi|17136832

    (+1) 17 0 0 3 2 0 0

    trailer hitch [Drosophila melanogaster] gi|45550607 69 0 0 4 2 0 0

    ribosomal protein S6, isoform B [Drosophila

    melanogaster] gi|17737290 28 0 0 2 3 0 0

    ribosomal protein L29, isoform A [Drosophila

    melanogaster]

    gi|17137274

    (+2) 9 0 0 4 2 0 0

    ribosomal protein L13, isoform A [Drosophila

    melanogaster]

    gi|17647879

    (+1) 25 0 0 2 2 0 0

    CG11784 [Drosophila melanogaster] gi|20129811 26 0 0 0 3 0 0

    CG12030 [Drosophila melanogaster] gi|19923002 39 0 0 2 0 0 0

    CG6995, isoform A [Drosophila melanogaster]

    gi|45550823

    (+1) 73 0 0 2 3 0 0

    ribosomal protein L30, isoform A [Drosophila

    melanogaster]

    gi|17864264

    (+1) 12 0 0 3 3 0 0

    ribosomal protein L31, isoform B [Drosophila

    melanogaster]

    gi|19921922

    (+2) 15 0 0 0 3 0 0

    ribosomal protein S27A [Drosophila

    melanogaster] gi|17136574 18 0 0 2 0 0 0

    transport and golgi organization 4 [Drosophila

    melanogaster] gi|18859793 53 0 0 0 2 0 0

    twinstar [Drosophila melanogaster] gi|17136986 17 3 0 0 0 0 0

    glutathione S transferase E3 [Drosophila

    melanogaster] gi|24654975 25 0 0 2 0 0 0

    ribosomal protein LP1 [Drosophila

    melanogaster] gi|17136320 12 0 0 2 0 0 0

    alpha-coatomer protein, isoform A [Drosophila

    melanogaster]

    gi|17137608

    (+1) 139 0 2 0 0 0 0

    vig2, isoform D [Drosophila melanogaster]

    gi|161078610

    (+3) 45 0 0 0 2 0 0

    fibrillarin [Drosophila melanogaster] gi|17647425 35 0 0 0 2 0 0

    protein on ecdysone puffs, isoform B

    [Drosophila melanogaster]

    gi|17864514

    (+1) 78 0 0 0 2 0 0

  • 50

    Unique peptides of Amphiphysin identified as interactors of dNedd4Lo N-terminus

    Sequence Prob X! Tandem

    (R)HSFQNLQANANK(R) 100% 4.958607

    (K)GREQLEEAR(R) 98% 3.3372421

    (K)ATTTTQTSPTEDK(A) 95% 2.7447276

    (R)VIEYDDPEDQEEGWLMGQK(E) 100% 10.207608

    (K)GLFPANFTRPI(-) 100% 4.2676063

    B.

  • 51

    Figure 11: dNedd4Lo co-immunoprecipitates with dAmph

    (A) Schematic representation of dNedd4S and dNedd4Lo showing the C2, WW(x3) and HECT

    domains. dNedd4Lo contains unique N-terminal (Nterm) and Middle (Mid) regions that are

    absent in dNedd4S. The Drosophila Amphiphysin (dAmph) protein was identified as an

    interacting partner of the unique N-terminal region of dNedd4Lo by mass spectrometry. (B)

    DAmph binds to dNedd4Lo in Drosophila S2 cells: Drosophila S2 cells were transfected (Tfxn)

    with Flag-tagged dNedd4S or dNedd4Lo and HA-tagged dAmph. Transfected cells were lysed

    and the lysates were immunoprecipitated (IP) with anti-Flag (dNedd4) antibodies and

    immunoblotted with anti-HA to detect co-immunoprecipitated dAmph (upper panel).

    Precipitated Flag-dNedd4 proteins are shown in the second panel. DAmph expression in the

    lysates was verified using anti-HA antibodies; actin was used as a loading control (lower panels).

  • 52

  • 53

    3.2 dNedd4Lo N-terminus directly binds to the SH3 domain of

    dAmph

    DAmph contains a C-terminal SH3 domain, a domain that usually binds proline-rich

    motifs (Grabs et al., 1997). dNedd4Lo N-terminus contains a Pro-rich sequence with a perfect

    consensus motif for SH3 binding (PRPPPR). We therefore tested in vitro binding of a purified

    SH3 domain of dAmph to the purified N-terminus of dNedd4Lo. Purified HIS tagged full-length

    (WT) dAmph, SH3 domain deletion mutant (dAmph-𝛥𝑆𝐻3), SH3 mutant (dAmph SH3(W->A),

    containing a mutation in the conserved Trp required for binding the Pro-rich motif), or the SH3

    domain of dAmph alone (SH3 dAmph) (Fig. 12A), were incubated with purified GST-tagged

    dNedd4Lo N-terminus (residues 1-63). Our results show that WT dAmph and the dAmph-SH3

    domain bound to GST-dNedd4Lo N-terminus, while the dAmph(𝛥𝑆𝐻3) or dAmph-SH3(W->A)

    mutants did not (Fig 12B). This demonstrates that the SH3 domain of dAmph directly binds the

    N-terminus of dNedd4Lo.

  • 54

    Figure 12: Drosophila Amphiphysin directly binds the N-terminus of dNedd4Lo via its SH3

    domain. (A) Schematic representation of dNedd4Lo, the unique N-terminal region of dNedd4Lo

    (Nterm, residues 1-63), wildtype dAmph (WT dAmph), the SH3 mutant containing a tryptophan

    to alanine substitution in the SH3 domain (dAmph SH3(W->A)), SH3 deletion mutant (ΔSH3

    dAmph) and the SH3 domain alone (SH3 dAmph). (B) The SH3 domain of dAmph mediates

    direct binding to the N-terminal region of dNedd4Lo: HIS-tagged purified dAmph (WT, ΔSH3,

    SH3(W->A) or SH3 domain alone) were incubated with GST beads or GST fused to dNedd4Lo

    N-terminal region (GST dNedd4Lo(Nterm)). DAmph was detected using HIS antibodies and

    dNedd4Lo(Nterm) using GST antibodies.

  • 55

  • 56

    3.3 dAmph is enriched postsynaptically and co-localizes with

    dNedd4Lo

    Drosophila dAmph is enriched postsynaptically at neuromuscular junctions (NMJs) and

    in muscle T-tubules (Leventis et al., 2001), and our earlier work detected dNedd4 expression in

    larval muscles (Ing et al., 2007). To investigate the biological importance of the

    dAmph:dNedd4Lo interaction in vivo, we first analyzed co-localization of dAmph and

    dNedd4Lo in the postsynaptic region and the muscle T-tubule network. It has been previously

    shown that endogenous dAmph co-localizes with the postsynaptic marker Dlg at NMJs (Leventis

    et al., 2001). Here, we overexpressed Flag-tagged dNedd4Lo and V5-tagged dAmph in flies

    using the muscle-specific driver 24B-Gal4, and examined larval NMJs located on muscles 6/7 by

    immunohistochemistry with antibodies against Flag (dNedd4), V5 (dAmph) and HRP

    (presynaptic marker). dNedd4Lo co-localized with dAmph in muscles but not with HRP

    labeling, which is specific for (presynaptic) neuronal membranes (Fig. 13A). To verify that V5-

    dAmph is localized postsynaptically and in muscle T-tubules, we examined NMJs using

    antibodies against V5, HRP, and a postsynaptic/T-tubule marker Disc large (Dlg) (Fig. 13B).

    Similar to what was previously observed with endogenous dAmph (Leventis et al., 2001), V5-

    dAmph co-localized with Dlg labeling (Fig.13B), indicating that dNedd4Lo co-localizes with

    dAmph in the postsynaptic region and muscle T-tubules.

  • 57

    Figure 13: Drosophila Amph and dNedd4 are enriched postsynaptically at the

    neuromuscular junctions and in muscle T-tubules.

    (A) dNedd4Lo is expressed in the muscles and the postsynaptic region of the neuromuscular

    junction (NMJ) where it co-localizes with Drosophila Amphiphysin (dAmph). (B) DAmph is

    enriched postsynaptically in NMJ and in muscle T-tubules where it co-localizes with

    postsynaptic and T-tubule marker Dlg. Expression was driven by 24B-Gal4 in larval muscles,

    dNedd4Lo (UAS-dNedd4Lo) was detected with an anti-Flag antibody (green) and dAmph (UAS-

    V5-dAmph) was detected with V5 antibody (red). Synaptic boutons from muscles 6/7 were

    analyzed. Presynaptic marker: HRP (blue). Scale bar: 3 m.

  • 58

  • 59

    3.4 dAmph lacking its SH3 domain is mislocalized in muscles

    Since we found that the interaction between dNedd4Lo and dAmph is mediated by the

    SH3 domain of dAmph, we expect this interaction to be lost in vivo in dAmph(ΔSH3) mutant

    flies. To determine the localization of the dAmph(ΔSH3) mutant in Drosophila, we analyzed

    UAS transgenic fly lines that overexpress MYC-tagged dAmph(ΔSH3) in muscles. In contrast to

    wildtype dAmph, dAmph(ΔSH3) did not co-localize in muscle T-tubules with Dlg. Deletion of

    the SH3 domain of dAmph shifts its localization from the muscle T-tubules and postsynaptic

    region to a region near the plasma membrane of the muscles (Fig. 14A and B). Quantification of

    the Pearson’s correlation coefficient further shows that dAmph WT co-localizes with the

    postsynaptic marker Dlg (r= 0.83) (Fig. 14C), but that the SH3 domain deletion mutant

    dAmph(ΔSH3) shows a significantly reduced co-localization (r=0.45). These results indicate that

    the SH3 domain of dAmph is important for its localization in the postsynaptic region and muscle

    T-tubules.

  • 60

    Figure 14: dAmph(ΔSH3) mislocalizes in Drosophila muscles.

    (A) ΔSH3 dAmph (red) localization with Dlg (green) is reduced in the postsynaptic region and

    muscle T-tubules. (B) Wild-type (WT) dAmph (red) is enriched postsynaptically and in muscle

    T-tubules, where it co-localizes with Dlg (green). Muscle overexpression of dAmph(ΔSH3) and

    WT dAmph was achieved using 24B-Gal4 muscle driver. dAmph(ΔSH3) and WT dAmph were

    visualized in third instar larva postsynaptic regions of muscle 6/7 using anti-MYC and anti-V5

    antibodies, respectively. Presynaptic marker: HRP (blue). Scale bar: 3 m. (C) dAmph ΔSH3 or

    WT colocalization with the postsynaptic marker Dlg is expressed in terms of the Pearson’s

    correlation coefficient. Graph illustrates changes in the mean Pearson's coefficients of dAmph

    V5-WT or MYC-ΔSH3 with Dlg (WT dAmph, n=24;ΔSH3 dAmph, n=30). Error bars indicate

    SEM. *** denotes p

  • 61

  • 62

    3.5 dNedd4Lo regulates the levels of dAmph in the postsynaptic

    region

    As dNedd4Lo is an E3 ubiquitin ligase, we investigated the possibility that it regulates

    levels of dAmph in vivo in the postsynaptic region. Thus, we analyzed the levels of endogenous

    dAmph in our UAS transgenic fly lines that overexpress dNedd4S, dNedd4Lo and

    dNedd4Lo(CA) (a catalytically inactive mutant generated by mutating the catalytic cysteine in

    the HECT domain). The flies used in this experiment have a wild type genetic background

    because dNedd4 null (dNedd4T121FS

    ) flies are homozygous lethal during embryogenesis and

    cannot be rescued with dNedd4Lo overexpression (Zhong et al., 2011).

    If dNedd4Lo regulates dAmph, then we expect dAmph abundance in the postsynaptic

    region to be reduced in flies overexpressing dNedd4Lo but not the dNedd4Lo(CA) mutant or

    dNedd4S. We examined larval NMJs located on muscles 6/7 by immunohistochemistry with

    antibodies against dAmph and Dlg. w1118

    (wildtype) flies and damph5E3

    (Amph null) transgenic

    flies that have their entire amph gene deleted were used as controls (Leventis et al., 2001). The

    mean intensity of dAmph that overlapped with Dlg labeling in the postsynaptic region was used

    to quantify the levels of dAmp