200979151316888

30
1 Running title: In Vitro Derived Human Sperm Title: Derivation of Human Sperm from Embryonic Stem Cells JAE HO LEE 1, 2, 7 , MAJLINDA LAKO 1, 2, 7 , LYLE ARMSTRONG 1, 2 , MARY HERBERT 1, 2, 3 , MANYU LI 4 , WOLFGANG ENGEL 4, 8 , Xin Zhang 1, 2 , DAVID ELLIOTT 2 , MIODRAG STOJKOVIC 5 , JOHN PARRINGTON 6 , ALISON MURDOCH 1, 3 , TOM STRACHAN 2 , KARIM NAYERNIA 1, 2, 9 1 North East England Stem Cell Institute (NESCI), Newcastle University, Newcastle upon Tyne, UK 2 Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, UK 3 Department of Reproductive Medicine, Newcastle University, Newcastle upon Tyne, UK 4 Institute of Human Genetics, University of Göttingen, Germany 5 Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain 6 Department of Pharmacology, University of Oxford, Oxford, UK 7 These authors contributed equally to this work. 8 Wolfgang Engel did not work with human embryonic stem cells 9 Corresponding Author: Prof. Dr. Karim Nayernia Chair of Stem Cell Biology NESCI/IHG International Centre for Life Newcastle University, Newcastle upon Tyne Central Parkway Newcastle upon Tyne NE1 3BZ UK Telephone: 0044-191-2418643 Fax: 0044-191-2418810 Email: [email protected] Acknowledgement: This study was supported by Newcastle University, Newcastle upon Tyne and ONE NorthEast. Key words: Human embryonic stem cells, Differentiation, Germ cells, Sperm Page 1 of 30 Stem Cells and Development In Vitro Derivation of Human Sperm from Embryonic Stem Cells (doi: 10.1089/scd.2009.0063) This article has been peer-reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.

Upload: adenmy

Post on 11-Nov-2015

214 views

Category:

Documents


1 download

TRANSCRIPT

  • 1

    Running title: In Vitro Derived Human Sperm

    Title: Derivation of Human Sperm from Embryonic Stem Cells

    JAE HO LEE1, 2, 7, MAJLINDA LAKO1, 2, 7, LYLE ARMSTRONG1, 2, MARY HERBERT1, 2, 3, MANYU LI4, WOLFGANG ENGEL4, 8, Xin Zhang1, 2, DAVID

    ELLIOTT2, MIODRAG STOJKOVIC5, JOHN PARRINGTON6, ALISON MURDOCH1, 3, TOM STRACHAN2, KARIM NAYERNIA1, 2, 9

    1 North East England Stem Cell Institute (NESCI), Newcastle University, Newcastle upon Tyne, UK

    2 Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, UK

    3 Department of Reproductive Medicine, Newcastle University, Newcastle upon Tyne, UK

    4 Institute of Human Genetics, University of Gttingen, Germany

    5 Centro de Investigacin Prncipe Felipe (CIPF), Valencia, Spain

    6 Department of Pharmacology, University of Oxford, Oxford, UK

    7 These authors contributed equally to this work.

    8 Wolfgang Engel did not work with human embryonic stem cells

    9 Corresponding Author:

    Prof. Dr. Karim Nayernia Chair of Stem Cell Biology NESCI/IHG International Centre for Life Newcastle University, Newcastle upon Tyne Central Parkway Newcastle upon Tyne NE1 3BZ UK Telephone: 0044-191-2418643 Fax: 0044-191-2418810 Email: [email protected]

    Acknowledgement: This study was supported by Newcastle University, Newcastle upon Tyne and ONE NorthEast.

    Key words: Human embryonic stem cells, Differentiation, Germ cells, Sperm

    Page 1 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 2

    ABSTRACT

    Differentiation of human embryonic stem cells (hESCs) to specific cell types can be

    achieved using methods that mimic in vivo embryonic developmental programs.

    Human egg and sperm cells (gametes) are derived from a founder population of

    germline stem cells (GSCs), primordial germ cells (PGCs), that are set aside early in

    embryogenesis which give raise to gonocytes in males. After birth, gonocytes

    differentiate to adult male germline stem cells, spermatogonial stem cells (SSCs),

    which can self-renew and generate sperm cells, are unique stem cells in that they are

    solely dedicated to transmit genetic information from generation to generation.

    Understanding the mechanisms of germ cell specification, development and its

    differentiation to sperm is important for elucidating the causes of male infertility.

    Here, we developed an in vitro strategy for establishing of male GSCs from human

    embryonic stem cells. These in vitro derived GSCs express markers which are

    specific for PGCs, SSCs, meiotic and postmeiotic germ cells indicating maturation of

    the primordial germ cells to haploid male gametes. In vitro derived germ cells are able

    to enter meiosis and generate haploid motile sperm-like cells in vitro. While full

    potential of the human ES derived germ cells and sperm remains to be demonstrated,

    this in vitro modeling of human gametogenesis provides a new approach for studying

    biology of human germ cells and establishment of therapeutic approaches in

    reproductive medicine.

    Page 2 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 3

    INTRODUCTION

    Germ cells are a pre-requisite for fertility [1, 2]. Derivation of both male and female

    gametes in vitro raises the possibility of using these gametes to gain a better

    understanding of basic reproductive biology and, in particular, to extend the potential

    for therapeutic cloning, transgenic technologies and the treatment of infertility. Our

    understanding of the molecular regulation of normal germ cell development in

    mammals has progressed significantly due to the utility of the mouse as a model

    system [3-5]. However, the molecular and cellular regulation of human germ cell

    development is almost completely unknown due to the lack of an in vitro model. The

    creation of such a model will provide powerful tools for undertaking new types of

    reproductive studies, and might permit the development of new technologies and

    novel approaches in assisted reproductive medicine. Human embryonic stem cells

    (hESCs) could potentially fill this need, as embryonic stem cells (ESCs) in general

    have a remarkable capacity for pluripotency. Recent studies have reported that

    successful differentiation of mouse ESCs into primordial germ cells (PGCs) as well as

    into mature male [6-8] and female [9] gametes can be achieved in vitro and in vivo.

    Hubner et al. reported the first study of in vitro gamete production [9]. These authors

    cultured mouse ESCs carrying a Pou5f1-reporter gene without feeder cells or added

    growth factors. The formation of ovarian follicle-like structures was observed, which

    included oocyte-like cells expressing Pou5f1 and Ddx4. These cells also expressed

    meiotic marker genes (e.g., Dmc1, Sycp3), and were released from follicles after 34

    weeks of culture. Although the germ cell potential of oocyte-like cells was not tested

    by fertilization, structures that closely resemble blastocysts emerged in 6 weeks,

    Page 3 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 4

    suggesting spontaneous parthenogeneic activation. Thus, this study suggested that

    ESC-derived cells have a potential to undergo gametogenesis in vitro.

    Toyooka et al. next reported the derivation of male germ cells from mouse ESCs [6].

    These authors took a different approach from Hubners, and used embryoid bodies

    (EBs) as starting material. Using ESCs carrying a Ddx4-reporter construct, the authors

    induced EBs and found that Ddx4+ cells spontaneously appeared, suggesting the

    presence of cells with characteristics of postmigratory PGCs in EBs. Ddx4+ cells

    were purified and aggregated with male genital ridge cells of wild-type embryos.

    Following implantation into adult mouse testes, the cell aggregates formed

    seminiferous tubules that supported complete spermatogenesis derived from purified

    Ddx4+ cells. Although the ability of spermatozoa to activate eggs was not examined,

    this study suggested that the germ line specification and the emergence of

    postmigratory PGCs can occur spontaneously or be induced in EBs.

    Using EBs, Geijsen et al. also suggested a spontaneous emergence of male PGCs

    from mouse ESCs in vitro [7]. Furthermore, the authors detected and isolated haploid

    cells from EBs, and showed that the injection of the cells into eggs resulted in the

    formation of blastocyst-like structures. Although the isolated cells did not resemble

    spermatozoa and analyses of further embryonic development were not completed, this

    study suggested that male PGCs arise from ESCs and spontaneously become

    postmeiotic cells that are capable of activating eggs.

    Recently, Nayernia et al. reported not only the induction of male gametes from ESCs

    but also successful production of offspring [8]. These authors cultured mouse ESCs,

    which were transfected with a Stra8 (stimulated by retinoic acid gene 8)-reporter

    construct, and later, also with a Prm1 (protamine 1)-reporter construct, in monolayer

    culture. Following repeated RA treatments and selection of Stra8+ cells, they isolated

    Page 4 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 5

    Prm1+ cells and injected them into eggs. When the resulting 65 embryos were

    transferred to surrogate mothers, seven live pups carrying the Prm1-reporter gene

    were derived, which apparently had growth abnormalities and died within 5 months

    after birth.

    The ability of human ESCs to enter the germ line was examined by Clark et al. [10].

    These authors generated EBs from female and male human ESCs and found using

    PCR or immunohistochemistry that some cells in EBs expressed marker genes

    specific to different stages of germ line development (e.g., DDX4, DAZL).

    Furthermore, cells expressing a meiotic marker gene, SYCP3, were identified in

    human EBs. Thus, this study suggested the possibility that human ESCs of both sexes

    may spontaneously enter the germ line and undergo meiosis.

    In present study, we have developed a strategy for the establishment of germline stem

    cells from human embryonic stem cells which are able to enter and complete meiosis

    and produce sperm-like cells.

    MATERIALS AND METHODS

    Culture of Human Embryonic Stem Cells

    Human ESC were grown on mitotically inactivated mouse embryonic fibroblasts and

    passaged essentially as described [11, 12]. The human ESC medium contains

    Knockout-DMEM (Invitrogen, Paisley, UK), 100 M -mercaptoethanol

    (Sigma,Dorset UK), 1 mM L-glutamine (Invitrogen), 100 mM non-essential amino

    acids (Invitrogen), 20% serum replacement (SR, Invitrogen), 1% penicillin-

    streptomycin (Sigma) and 4 ng/ml FGF2 (Invitrogen).Medium was changed daily.

    Human ESC were passaged by incubation in 1 mg/ml collagenase IV (Invitrogen) for

    Page 5 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 6

    5-8 minutes at 37 C or mechanically dissociated and then removed to freshly

    prepared feeders.

    In Vitro Derivation of Male Germ Cells

    Construction of the Stra8-EGFP construct was previously described [13]. Human

    ESCs were maintained under antibiotic free conditions for at least 3 passages before

    transfection. On the day of transfection, human ESC were incubated with collagenase

    IV (1mg/ml; Sigma) for five minutes to generate small clumps which were

    subsequently transfected using the Nucleofector kit from Amaxa (mouse embryonic

    stem cell transfection kit) using manufacturers instructions. Two days after the

    transfection, stable clones were selected using G418 selection (150 g/ml) for 10 days.

    The efficiency of transfection was 7 colonies for Ncl-3 and 10 colonies for Ncl-4 per

    1,000,000 cells. The resulting colonies were pooled together to generate the stable

    cells lines Ncl3-Stra8 and Ncl4-Stra8.

    Thereafter, cells were cultured on a feeder layer of mitomycin C-inactivated mouse

    embryonic fibroblasts with human ESCs medium. To induce differentiation, hESCs

    were spontaneously differentiated for 3 weeks with differentiation ESCs medium,

    which was replaced 20% serum replacement (SR, Invitrogen) with 20% FBS

    (Hyclone) and excluded 4 ng/ml FGF2 (Invitrogen). After this, these ESCs were

    cultured with conditioned medium containing retinoic acid (RA) (Sigma) at a final

    concentration of 106 M for another 3 weeks. Differentiated GFP-positive cells were

    sorted by FACS. Sorted cells were cultured in medium containing Knockout-DMEM

    (Invitrogen, Paisley, UK), 1 mM L-glutamine (Invitrogen), 100 mM non-essential

    amino acids (Invitrogen), 20% serum replacement (SR, Invitrogen), 1% penicillin

    streptomycin (Sigma), 1,000 U/ml human recombinant leukemia inhibitory factor

    Page 6 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 7

    (LIF; Sigma) and 4 ng/ml human recombinant basic fibroblast growth factor (hrbFGF;

    GIBCO). Furthermore, 10 mM forskolin (Sigma) is a mediator that enhances

    intracellular cAMP levels by stimulating adenylate cyclase (protein kinase A) and it

    has also been reported to significantly promote PGC proliferation [14, 15]. In

    particular, to help aggregation, sorted cells were cultured in 96 well plate and

    centrifuged at 1400 rpm for 4 min and for ROCK inhibitor treatment, Y-27632

    (Calbiochem; water soluble) was added to culture medium at 10 mM 1 h before

    detaching the cells from the feeder layer and also upon seeding the cells onto a

    newMEF layer. A single halfday treatment of Y-27632 was sufficient for enhanced

    survival of dissociated hES cells in low-density adhesion culture on MEF cells [16].

    Multi-cellular colonies emerged in approximately 20-25 days.

    SemiQuantitative RTPCR Analysis

    Total RNA was isolated by using the RNeasy Mini Kit (Qiagen) according to

    manufacturers instructions. Single strand cDNAs were prepared from 5 g RNA

    using a reverse transcriptase reaction (Invitrogen, Karlsruhe, Germany) and analysis of

    gene expression was done RT-PCR with Platinum Taq (Invitrogen). The primer pairs

    are shown in Table 1.

    Real-Time RT-PCR Analysis

    Total RNA was isolated using the RNeasy Mini Kit (Qiagen) according to

    manufacturers instructions and For mRNA, reverse transcription was performed

    using random primers and the Superscript III kit (Invitrogen). Real-time quantitative

    PCR for mRNA was conducted with 1 l of cDNA, 10 l of QuantiTect SYBR Green

    PCR Master Mix, 1 l each of specific 10 M forward and reverse primers (MWG

    Page 7 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 8

    Biotech, Ebersberg, Germany), and 7 l of sterile water (all reagents from Qiagen,

    Hilden, Germany) using primer sets shown in Table 1.

    The data were analyzed by using the RelQuant software (Roche Applied Science).

    Genomic PCR

    PCR analysis was performed on genomic DNA extracted from human ESCs with

    NEO, EGFP and Stra8-EGFP-specific primers: GFP (5-GAC CCT GAA GTT CAT

    CTG CA-3, 5-ACT CCA GCA GGA CCA TGT GAT-3), NEO (5-AGG CTA

    TTC GGC TAT GAC TG-3, 5-TCA TCC TGA TCG ACA AGA CC-3), Stra8-

    EGFP (5-CGT AGT GTG CCA AAC TGA TGT GG-3, 5-CAG GGT CAG CTT

    GCC GTA GGT GG-3), respectively. The cycling conditions were as follows: 5 min

    at 94C, five cycles of 30 s at 94C, 30 s at 5761C, 1 min at 72C, 30 cycles of 30 s

    at 94C, 45 s at 56C, 1 min at 72C and 72C for 10 min.

    Flow Cytometry

    Single cell suspensions were generated using acutase (Chemicon). Single cells were

    suspended in PBS at concentration of 106 cells/ml. Cells were washed again three

    times and resuspended in PBS before being analysed with FACS Calibur instrument

    (BD) equipped with CellQuest software. For haploidization, cells were prepared as

    described above and stained with using a Kit supplied by Partec (CyStain DNA). Cell

    count was ensured to be below 5x106/ml. After that, extraction buffer (200ul to 100ul

    of cells) was added for 30 mins at room temperature. For counter staining, 800ul of

    DAPI added and incubated in the dark for 30 mins. The sample was analysed at 10PSI

    using a 60mw UV laser (355) at cell count of 200 cells / second.

    Immunohistochemistry

    Page 8 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 9

    Cytospin preparations of single cell suspension were made by using approximately

    10,000 cells per slide. Cells were fixed in 4% paraformaldehyde solution for 10 min at

    room temperature, washed three times in PBS and then incubated with an anti-Piwil2

    [24], TSPY [25], RBM [26], hnRNPG-T [26], PLCzeta [27], antibody in a

    humidifying chamber at 4C overnight. After washing, culture slides were incubated

    with a 1:300 FITC-conjugated in goat anti-sheep IgG, 1:300 FITC-conjugated in goat

    anti-rabbit IgG and/or 1:300 Cy3-conjugated in goat anti-rabbit IgG (Sigma-Aldrich)

    secondary antibody. Slides were washed three times in PBS and mounted in DAPI

    mounting solution (Vector Laboratories, Burlingame, CA, USA).

    Fluorescence In Situ Hybridization Analysis

    Moving cells were washed with PBS and then resuspended in 30% glycerol-PBS.

    Briefly, cells were fixed with 0.5% formaldehyde-PBS for 2 hrs and decondensed in

    10 mM DTT, 0 to 0.5 mg/ml heparin in PBS for 30 minutes at room temperature.

    Treatment with DTT-heparin induces uniform nuclear swelling, while preserving

    nuclear shape; the higher the heparin concentration, the higher the resulting nuclear

    decondensation. Such swelling is a prerequisite to perform FISH in moving cells.

    Decondensed cells were loaded onto microscope slides, air-dried, washed in 2x SSC at

    37 C for 2 mins, water, dehydrated in a 70% to 100% ethanol series for each 1 min

    and air-dried again. For denaturation of the FISH probe, a wet towel was placed in a

    sealed box/container and leave in a 37 C incubator. A hot plate was set at 75 C and

    allowed to reach temperature before moving onto the next step. After that, probe (7 l

    buffer + 2 l dH20 + 1 l probe) 10 l was aliquoted into the tube and spun for a few

    seconds in a micro-centrifuge. 10ul of probe was distributed onto slide and place a

    22x22mm coverslip (0-thickness) and the edges of the cover-slip were sealed with

    Page 9 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 10

    vulcanizing rubber glue solution and then left to dry at 37 C for approximately 5

    minutes. Finally, the cover-slip placed face down onto the slide and left for 1 min

    for the probe to denature. The slide was put in humid box and left in a 37 C incubator

    overnight. For post hybridization wash, approximately 50mls of post hybridization

    buffer was washed into a coplin jar and placed it into a cool water bath before setting

    the temperature to 72 C. 50mls of 2x SSC was aliquoted into a coplin jar, carefully

    removed vulcanizing rubber glue solution and cover-slip from slide, washed slide in

    2xSSC for 2mins using tweezers and mounted slide with preferably Vectashield anti-

    fade mountant with DAPI.

    Poseidon fluorescent labeled specific DNA probes SE X (DXZ1)/SE Y (DYZ3) and

    SE 18 (D18Z1)-specific probes from KREATECH Biotechnology. SE X/SE Y

    KB20030 is a ready made mix of the two probes with the X centromere being labelled

    in green and the Y centromere being labelled in red. Additionally, SE 18 (D18Z1)

    KB-20018 is labelled in red. DNA from appropriate, chromosome-specific probes are

    first labeled with reporter molecules. The labeled DNA probe is then hybridized to the

    metaphase chromosomes or interphase nuclei on a slide. After washing, the specimen

    is screened for the reporter molecules by fluorescence microscopy.

    RESULTS AND DISCUSSION

    Embryonic stem (ES) cells are pluripotent cell lines established from undifferentiated

    embryonic cells characterized by nearly unlimited self-renewal and differentiation

    capacity. During differentiation in vitro, ES cells were found to be able to develop

    into specialized somatic cells types and to recapitulate processes of early embryonic

    Page 10 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 11

    development. Despite the well-recognized pluripotency of hESC, it has proven rather

    challenging to direct the differentiation of hESC into specific lineages. However, an

    ESC-based strategy could permit the generation of an unlimited supply of desirable,

    fully functional cell types from an abundant, renewable, and readily-accessible source.

    The differentiation of hESC into specific lineages could provide a tool for studying

    the molecular mechanisms underlying lineage specification, early embryonic

    developmental pathways, and the pathological basis of genetic disorders, as well as

    provide a source of transplantable cells and tissue. In all these studies, strategies for

    manipulating stem cell differentiation (selection of differentiated cell types) are based

    on knowledge of the mechanisms by which lineage decisions are made during early

    embryogenesis. A combination of approaches, including analysis of molecular

    markers specifically expressed in relevant cell types and morphological and/or

    histochemical approaches should be used to define differentiated cell types. Therefore,

    in order to identify germ cells in different stages of formation and differentiation

    during human ES differentiation, we compiled a list of markers that are germ cell-

    specific (not expressed in somatic lineages), and germ cell-enriched (highly expressed

    in germ cells with limited expression in somatic cells) and are expressed in defined

    stages of germ cell development in vivo (from primordial germ cells to sperm). From

    this list, a profile of the expected sequential expression of markers were generated that

    would define development of the male germ cell lineage during ES cell differentiation

    in vitro (Fig. 1)

    Mimicking the cellular interactions of the embryo by providing appropriate signalling

    molecules in culture has enabled the differentiation of ES cells to be directed

    predominately toward particular lineages. Multistep strategies incorporating the

    provision of soluble factors known to influence lineage choices in the embryo,

    Page 11 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 12

    coculture with other cells or tissues, genetic modification, and selection for desirable

    cell types by reporter gene constructs have allowed the production of ES cell derived

    specific cell types. We used a promoter based selection strategy that can visualize the

    initial step of male germ cell formation. We used promoter region of Stra8 gene for

    generation of reporter construct (Fig. 2A). The Stra8 gene encodes a cytoplasmic

    protein and is expressed specific to the developing male gonad during mouse

    embryogenesis. In adult mouse, its expression is restricted to the premeiotic germ

    cells.

    Two XY (hES-NCL3, hES-NCL4) [12] and one XX (H9) [28] human ES cell lines

    were transfected with the Stra8-EGFP fusion gene and stable cell lines were

    established (Fig. 2B, C). After culture of cells in conditioned medium containing

    retinoic acid (RA), positive cells were sorted by fluorescence-activated cell sorting

    (FACS), and after culturing, expression of green fluorescence protein (GFP) under the

    control of the germline stem cell specific Stra8 promoter region [13] was observed

    (Fig. 2D-O). The expression of Stra8 gene which is required for spermatogenesis is

    directly related to the availability of retinoic acid.

    RT-PCR analyses were employed to evaluate the expression in the GFP+ sorted cells

    of different markers for premeiotic, meiotic, and postmeiotic male germ cells. Positive

    expression was found for markers for the premeiotic (OCT4 [29], GDF3 (Growth and

    Differentiation Factor 3) [30], C-KIT [29], STELLA [30], VASA [31, 32], , PLZF [20],

    PIWIL2 [14, 24], DAZL (Deleted in Azoospermia Like) [31]), meiotic ( synaptonemal

    complex protein (SCP3) [33], DMC1 [34], acrosin (ACR) [22], BOULE [35], PGK2

    [36]) and haploid (TP2 [23], protamine 1 (PRM1) [23]) male germ cells (Fig. 3A).

    These results could be confirmed by quantitative real-time RT-PCR analysis (Fig. 3B).

    Page 12 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 13

    Therefore, Stra8-EGFP positive cells appear to be heterogeneous and contain

    premeiotic, meiotic and postmeiotic cells.

    Expression of germ cell-specific genes in the sorted cells revealed that these cells are

    able to differentiate to different stages of postnatal male germ cell development. On

    protein level, male germ cell differentiation could be confirmed by

    immunohistochemical analysis with antibodies against VASA [31, 32], RNA Binding

    Motif (RBM) [37], PIWIL2 [14, 24], Testis Specific Protein Y-encoded (TSPY) [25,

    38], HNRNPG-T [26] , Synaptonemal Complex Protein 1 (SCP1) [39], PLCzeta [40]

    and PRM1 [23] proteins which are expressed in premeiotic, meiotic and postmeiotic

    stages (Fig. 4).

    To investigate whether ES-derived germ cells undergo meiosis in vitro, we

    determined the DNA content of the cells by flow cytometric analysis. As shown in

    Figure 5A and 5B, about 3.1 % of cells were haploid indicating completion of meiotic

    process (Fig. 5). In contrast, in germline stem cells derived from XX embryonic stem

    cells (H9), only male specific premeiotic and early meiotic markers could be observed,

    indicating that XX derived germ cells are not able to enter meiosis in vitro (Fig. 5).

    Motility is a characteristics function of the male gamete, which allows sperm to

    actively reach and penetrate the female gamete. Motile cells were released to the hES

    cell culture medium with human recombinant leukemia inhibitory factor (LIF), human

    recombinant basic fibroblast growth factor (hrbFGF), forskolin and ROCK inhibitor

    treatment, Y-27632 after culturing the cells for two months in the hES cell culture

    medium with retinoic acid (RA) indicating formation of the specialized structure of

    the sperm flagellum (Supplemental Video online). The formation of tail-like

    structures could be observed in some motile cells (Fig. 6A). Meiosis of these cells was

    confirmed by fluorescence in situ hybridization (FISH) analysis on chromosomes 18,

    Page 13 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 14

    X and Y (Fig. 6B). Using the X-Y-18 assay, chromosome X is green, chromosome Y

    is red and 18 is red. A normal human sperm carrying 23 single chromosome is labeled

    in chromosome X, Y and 18 respectively and ES-IVD is also shown in each labeled

    chromosome X, Y and 18 like normal human sperm.

    These results clearly demonstrate that human embryonic stem cells are able to

    differentiate to adult male germline stem cells, spermatogonial stem cells, which can

    undergo meiosis and produce motile haploid sperm-like cells. These models are very

    useful tools for understanding of molecular and cellular mechanisms of male

    infertility, establishment of therapeutic approaches for male infertility and provide

    testing systems to examine toxicological effects of drugs on human germ cells.

    Approximately 1 in 7 couples have fertility problems. In 20% of couples, low sperm

    count/quality is the only cause of infertility, and it is a contributory factor in a further

    25% of couples. In 30-50% of men, no cause is identified for the poor sperm

    characteristics. Although assisted reproductive technologies such as IVF and ICSI

    have dramatically improved the prospects for infertile couples, there are some types

    of male factor infertility that remain untreatable. These include arrested

    spermatogenesis which may be due to defective germ cells, an abnormal testicular

    environment or aberrations in endocrine pathways regulating testis function. One of

    the essential bases for treatment of male infertility is the understanding of the human

    spermatogenesis by modelling human germ cell development. There has previously

    been no robust cell-based model for examining the genetic and epigenetic

    mechanisms of human germ cell formation.

    Human spermatogenesis consists of three biologically significant processes: stem cell

    self-renewal and differentiation, meiosis, and haploid cell morphogenesis.

    Understanding the molecular mechanisms behind these processes might provide clues

    Page 14 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 15

    to the puzzle of species preservation and evolution. From the biological point of view,

    in vitro derived germ cells provide a very useful experimental tool for undertanding of

    basic mechanisms of male germ cell specification, development and differntiation

    including meiosis, germ cell morphogenesis and epigenetics. We demonstarted clearly

    that germ cells dereived from XY hES cells are able to enter meiosis and complete

    this process. In contrast, germ cells derived from XX hES cells express spermatogonia

    specific markers, but are not able to complete meiotic process in vitro. There are

    several lines of evidence demonstrating that genes on Y chromosomes are essential

    for meiosis. DAZ (Deleted in Azoopsermia) and BOULE proteins paly imortant role

    during meiosis. The defects in spermatogenesis that are associated with an absence of

    BOULE or DAZ function have substantial similarities. Spermatocytes are formed in

    boule homozygotes, but fail to udergo meiotic devisions. In individuals deletd for

    DAZ containing region of Yq, the postmeiotic stages of spermatogenesis are similary

    rarer of absent [41, 42].

    Our results clearly demonstrated that germ cells derived from XY human embryonic

    stem cells undergo meiotic division and produce haploid sperm-like cells. However,

    these proof-of-principle experiments should be followed by detailed molecular,

    cellular, morphological and functional analysis in further experiments.

    DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

    The authors indicate no potential conflicts of interests

    ACKNOWLEDGEMENTS

    This research was supported by The University of Newcastle upon Tyne, One

    NorthEast and BBSRC. We thank I. Dimmick, Ch. Mueller and J. Barel for excellent

    Page 15 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 16

    technical assistance. We thank J. Schmidtke, D. Elliott and H. Cooke for providing

    antibodies against Tspy and hnRNPG-T, Dazl and RBM antibodies.

    REFERENCES

    1. McLaren A. (2000). Establishment of germ cell lineage in mammals. J Cell

    Physiol 182:141-143.

    2. Surani MA. (2007). Germ cells: the eternal link between generations. C R

    Biol 330:474-478.

    3. Grootegoed JA, Baarends WM, Roest HP and Hoeijimakers JH. (1998).

    Knockout mouse model and gametogenesis failure. Mol Cell Endocrinol

    145:161-166.

    4. Cooke HJ and Saunders PT. (2002). Mouse models of male infertility. Nat

    Rev Genet 3:790-801.

    5. OBryan MK and de Kretser D. (2006). Mouse models for genes involved in

    impaired spermatogeneis. Int J Androl 29:76-89.

    6. Toyooka Y, Tsunekawa N, Akasu R and Noce T. (2003). Embryonic stem

    cells can form germ cells in vitro. Proc Natl Acad Sci (USA) 100:11457-

    11462.

    7. Geijsen N, Horoschak M, Kim K, Gribnau J, Eggan K and Daley GQ. (2004).

    Derivation of embryonic germ cells and male gametes from embryonic stem

    cells. Nature 427:148-154.

    8. Nayernia K, Nolte J, Michelmann HW, Lee JH, Rathsack K, Drusenheimer N,

    Dev A, Wulf G, Ehrmann IE, Elliott DJ, Okpanyi V, Zechner U, Haaf T,

    Meinhardt A and Engel W. (2006). In vitro-differentiated embryonic stem

    cells give rise to male gametes that can generate offspring mice. Dev Cell 11:

    125-132.

    Page 16 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 17

    9. Hbner K, Fuhrmann G, Christenson LK, Kehler J, Reinbold R, De La Fuente

    R, Wood J, Strauss JF 3rd, Boiani M and Schler HR. (2003). Derivation of

    oocytes from mouse embryonic stem cells. Science 300:1251-1256.

    10. Clark AT, Bodnar MS, Fox M, Rodriquez RT, Abeyta MJ, Firpo MT and Pera

    RA. (2004). Spontaneous differentiation of germ cells from human embryonic

    stem cells in vitro. Hum Mol Genet 13:727-739.

    11. Stojkovic M, Lako M, Stojkovic P, Stewart R, Przyborski S, Armstrong L,

    Evans J, Herbert M, Hyslop L, Ahmad S, Murdoch A and Strachan T. (2004).

    Derivation of human embryonic stem cells from day-8 balstocysts recovered

    after three-step in vitro culture. Stem Cells 22:790-797.

    12. Zhang X, Stojkovic P, Przyborski S, Cooke M, Armstrong L, Lako M and

    Stojkovic M. (2006). Derivation of human embryonic stem cells from

    developing and arrested embryos. Stem Cells 24:2669-2676.

    13. Nayernia K, Li M, Jaroszynski L, Khusainov R, Wulf G, Schwandt I,

    Korabiowska M, Michelmann HW, Meinhardt A and Engel W. (2004). Stem

    cell based therpeutical approach of male infertility by teratocarcinoma derived

    germ cells. Hum Mol Genet 13:1451-1460.

    14. De Felici M, Dolci S and Pesce M. (1993). Proliferation of mouse primordial

    germ cells in vitro: a key role for cAMP. Dev Biol 157:277-280.

    15. Dolci S, Pesce M and De Felici M. (1993). Combined action of stem cell

    factor, leukemia inhibitory factor, and cAMP on in vitro proliferation of

    mouse primordial germ cells. Mol Reprod Dev 35:134-139.

    16. Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T,

    Takahashi JB, Nishikawa S, Nishikawa S, Muguruma K and Sasai Y. (2007).

    Page 17 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 18

    A ROCK inhibitor permits survival of dissociated human embryonic stem

    cells. Nature Biotechnology 25:681-686.

    17. Sasaki T, Shiohama A, Minoshima S and Shimizu N. (2003). Identification of

    eight members of the Argonaute family in the human genome small star, filled.

    Genomics 82:323-330.

    18. Tung JY, Luetjens CM, Wistuba J, Xu EY, Reijo Pera RA and Gromoll J.

    (2006). Evolutionary comparison of the reproductive genes, DAZL, BOULE,

    in primates with and without DAZ. Dev Genes Evol 216:158-168.

    19. Eberhart CG, Maines JZ and Wasserman SA. (1996). Meiotic cell cycle

    requirment for a fly homologue of human Deleted in Azoospermia Nature

    381:783-785.

    20. Costoya JA, Hobbs RM, Barna M, Cattoretti G, Manova K, Sukhwani M,

    Orwig KE, Wolgemuth DJ and Pandolfi PP. (2004). Essential role of Plzf in

    maintenance of spermatogonial stem cells. Nat Genet 36:653-659.

    21. Ravel C, Chantot-Bastaraud S, El Houate B, Berthaut I, Verstraete L, De

    Larouziere V, Loureno D, Dumaine A, Antoine JM, Mandelbaum J, Siffroi

    JP and McElreavey K. (2007). Mutations in the protamine 1 gene associated

    with male infertility. Mol Hum Reprod 13:461-464.

    22. Keime S, Adham IA and Engel W. (1990). Nucleotide sequence and exon-

    intron organization of the human proacrosin gene. Eur J Biochem 190:195-200.

    23. Choudhary SK, Wykes SM, Kramer JA, Mohamed AN, Koppitch F, Nelson

    JE and Krawetz SA. (1995). A haploid expressed gene cluster exists as a

    single chromatin domain in human sperm. J Biol Chem 270: 8755-8762.

    24. Lee JH, Schtte D, Wulf G, Fzesi L, Radzun HJ, Schweyer S, Engel W and

    Nayernia K. (2006). Stem-cell protein Piwil2 is widely expressed in tumors

    Page 18 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 19

    and inhibits apoptosis through activation of Stat3/Bcl-XL pathway. Hum Mol

    Genet 15:201-211.

    25. Skawran B, Schubert S, Dechend F, Vervoorts J, Nayernia K, Lscher B and

    Schmidtke J. (2005). Characterization of a human TSPY promoter. Mol Cell

    Biochem 276:159-167.

    26. Elliott DJ. (2004). The role of potential splicing factors including RMBY,

    RBMX, hnRNPG-T and STAR proteins in spermatogenesis. Int J Androl

    27:328-334.

    27. Swann K, Saunders CM, Rogers NT and Lai FA. (2006). PLCzeta(zeta): a

    sperm protein that triggers Ca2+ oscillations and egg activation in mammals.

    Semin Cell Dev Biol 17:264-273.

    28. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ,

    Marshall VS and Jones JM. (1998). Embryonic stem cell lines derived from

    human blastocysts. Science 282:1145-1147.

    29. Kerr CL, Hill CM, Blumenthal PD and Gearhart JD. (2008). Expression of

    pluripotent stem cell markers in the human fetal testis. Stem Cells 26:412-421.

    30. Clark AT, Rodriguez RT, Bodnar MS, Abeyta MJ, Cedars MI, Turek PJ, Firpo

    MT and Reijo Pera RA. (2004). Human STELLAR, NANOG, and GDF3

    genes are expressed in pluripotent cells and map to chromosome 12p13, a

    hotspot for teratocarcinoma. Stem Cells 22:169-179.

    31. Anderson RA, Fulton N, Cowan G, Coutts S and Saunders PT. (2007).

    Conserved and divergent patterns of expression of DAZL, VASA and OCT4

    in germ cells of the human fetal ovary and testis. BMC Dev Biol 7:136.

    Page 19 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 20

    32. Zeeman AM, Stoop H, Boter M, Gillis AJ, Castrillon DH, Oosterhuis JW and

    Looijenga LH. (2002). VASA is a specific marker for both normal and

    malignant human germ cells. Lab Invest 82:159-166.

    33. Yuan L, Liu JG, Zhao J, Brundell E, Daneholt B and Hg C. (2000). The

    murine SCP3 gene is required for synaptonemal complex assembly,

    chromosome synapsis, and male fertility. Mol Cell 5:73-83.

    34. Masson JY, Davies AA, Hajibagheri N, Van Dyck E, Benson FE, Stasiak AZ,

    Stasiak A and West SC. (1999). The meiosis-specific recombinase hDmc1

    forms ring structures and interacts with hRad51. EMBO J 18:6552-6560.

    35. Luetjens CM, Xu EY, Rejo Pera RA, Kamischke A, Nieschlag E and Gromoll

    J. (2004). Association of meiotic arrest with lack of BOULE protein

    expression in infertile men. J Clin Endocrinol Metab 89: 1926-1933.

    36. McCarrey JR, Kumari M, Aivaliotis MJ, Wang Z, Zhang P, Marshall F and

    Vandeberg JL. (1996). Analysis of the cDNA and encoded protein of the

    human testis-specific PGK2 gene. Dev Genet 19:321-332.

    37. Elliott DJ, Oghene K, Makarov G, Makarova O, Hargreave TB, Chandley AC,

    Eperon IC and Cooke HJ. (1998). Dynamic changes in the subnuclear

    organisation of pre-mRNA splicing proteins and RBM during human germ

    cell development. J Cell Sci 111:1255-1265.

    38. Schnieders F, Drk T, Arnemann J, Vogel T, Werner M and Schmidtke J.

    (1996). Testis-specific protein,Y-encoded (TSPY) expression in testicular

    tissues. Hum Mol Genet 5:1801-1807.

    39. Kondoh N, Nishina Y, Tsuchida J, Koga M, Tanaka H, Uchida K, Inazawa J,

    Taketo M, Nozaki M, Nojima H, Matsumiya K, Namiki M, Okuyama A and

    Nishimune Y. (1997). Assignment of synaptonemal complex protein 1 (SCP1)

    Page 20 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 21

    to human chromosome 1p13 by fluorescence in situ hybridization and its

    expression in the testis. Cytogenet Cell Genet 78:103-104.

    40. Saunders CM, Swann K and Lai FA. (2007). PLCzeta, a sperm-specific PLC

    and its potential role in fertilization. Biochem Soc Symp 74:23-36.

    41. Eberhart CG, Maines JZ and Wasserman SA. (1996). Meiotic cell cycle

    requirment for a fly homologue of human Deleted in Azoospermia. Nature

    381:783-785.

    42. Reynolds N and Cooke HJ. (2005). Role of the DAZ genes in male fertility.

    Reprod Biomed Online 10:72-80.

    Online Supplemental Video: Release of motile sperm-like cells in medium

    Page 21 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 22

    LEGEND TO FIGURES

    FIG. 1. Schematic representation of the different stages of germ cell differentiation in

    prenatal and postnatal development. Expected expression patterns of genes used to

    predict each stage of germ cell development are shown by the name of the gene

    together with the black bars extending to the right of each gene name.

    FIG. 2. Isolation of germ cells derived human embryonic stem cells. (A) Schematic

    depiction of the Stra8-EGFP reporter gene harbouring a 1.4 kb promoter region of

    Stra8. (B, C) Genomic PCR using specific primers for GFP, neomycin (NEO) and

    Stra8-EGFP regions on DNA isolated from transfected human embryonic stem cell

    lines H9, hES-NCL3 (NCL3), hES-NCL4 (NCL4) and untransfected human

    embryonic stem cell (-). Phase contrast (left), fluorescence (middle) and merged

    images (right) of Stra8-EGFP+ sorted cells growing on a embryonic fibroblast layer

    show expression of green fluorescence protein under germline stem cell specific

    promoter region, (D-F) Stimulation of differentiation of germ cells treatment with

    retinoic acid (RA) on spontaneous differentiation of hES-NCL4 (NCL4) and (J-L)

    H9 cell lines. (G-I) After FACS, Stra8-EGFP (Green) expression in colonized hES-

    NCL4 (NCL4) and (M-O) H9 cell lines. Bar, 50 m.

    FIG. 3. (A) Transcription of germ cell specific genes in hES cell derived germ cells.

    RT-PCR analysis of parental XY (1) and XX (2) embryonic stem cells under

    nondifferentiating conditions compared to the sorted cells in differentiating conditions

    (4:XY, 7:XX). Expression of premeiotic (OCT4, GDF3 (growth/differentiation factor

    3), C-KIT, VASA, STELLA, PLZF (promyelocytic leukemia zinc finger), PIWIL2

    Page 22 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 23

    (piwi-like 2), DAZL, (deleted-in-azoospermia-like)); meiotic (SCP3 (synaptonemal

    complex protein 3), DMC1 (disrupted meiotic cDNA), ACR (acrosin), BOULE, and

    PGK2 (phosphoglycerate kinase 2)) and haploid (TP2 (transition nuclear protein 2)

    and PRM1 (protamine 1)) genes could be detected in XY hES derived germ cells (4).

    In XX hES cell derived germ cells, only expression of premeiotic germ cells and

    SCP3 could be observed (7). RT-PCR analysis of mouse fibroblast (3), human testis

    (5) and human heart (6) served as controls. (B) Quantitative expression analysis of

    germ cell specific genes in undifferentiated XY and XX ES cells, germ cells derived

    from XY and XX ES cells, mouse fibroblast, human testis and human heart served as

    controls. A clear increase of expression of germ cell specific genes ( Stra8, c-Kit,

    Vasa, plzf, Piwil2, SCP3, PGK2 and Prm1) has been detected.

    FIG. 4. Expression of germ cell specific markers in differentiated hES cells. (A-I)

    Immunohistochemical analysis showed expression of premeiotic (VASA, RBM,

    PIWIL2, TSPY), meiotic (hnRNPG-T (GT), SYCP1 (synaptonemal complex protein

    1) and sperm (PLCzeta, PRM1 (protamine 1)) specific proteins. (A-F and H, I) Bar,

    20 m. (G) Bar, 50 m.

    FIG. 5. Haploidization of hES cell derived germ cells. (A) Identification of haploid

    (1N), diploid (2N) and tetraploid (4N) cell populations in ES-derived germ cells. XY

    hES derived germ cells (XY-IVD) show about 3.1% haploid cells, whereas XX hES

    (XX-IVD) derived germ cells show no haploid cells, DNA content of human sperm

    and human Leucocytes serve as controls. (B) Balk diagram of FACS analysis.

    Page 23 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 24

    FIG. 6. Formation of haploid sperm-like cells. (A) Magnification of cells from

    supernatant showing formation of tail and head in sperm-like cells. (B) Fluorescence

    in situ hybridization analysis of ES-derived sperm-like cells (lower part) and control

    sperm cells (upper part) have been shown by fluorescence in situ hybridization using

    probes specific for chromosome 18 (red), X (green) and Y (red). Here, probes are

    used for the mixture of chromosome X (DXZ1) / chromosome Y (DYZ3) together

    and chromosome 18 (D18Z1). Bar scales: in (A) 5 m, in (B) 1 m.

    Page 24 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 25

    FIG. 1.

    Page 25 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 26

    FIG. 2.

    Page 26 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 27

    FIG. 3.

    A

    B

    Page 27 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 28

    FIG. 4.

    FIG. 5.

    Page 28 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 29

    FIG. 6.

    Page 29 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.

  • 30

    Page 30 of 30St

    em C

    ells

    and

    Dev

    elop

    men

    tIn

    Vitr

    o D

    eriv

    atio

    n of

    Hum

    an S

    perm

    from

    Em

    bryo

    nic

    Stem

    Cel

    ls (do

    i: 10.1

    089/s

    cd.20

    09.00

    63)

    This

    artic

    le h

    as b

    een

    peer

    -revi

    ewed

    and

    acc

    epte

    d fo

    r pub

    licat

    ion,

    but

    has

    yet

    to u

    nder

    go c

    opye

    ditin

    g an

    d pr

    oof c

    orre

    ctio

    n. T

    he fi

    nal p

    ublis

    hed

    vers

    ion

    may

    diff

    er fr

    om th

    is pr

    oof.