US20120009156A1 - Germline stem cell banking system - Google Patents

Germline stem cell banking system Download PDF

Info

Publication number
US20120009156A1
US20120009156A1 US12/940,888 US94088810A US2012009156A1 US 20120009156 A1 US20120009156 A1 US 20120009156A1 US 94088810 A US94088810 A US 94088810A US 2012009156 A1 US2012009156 A1 US 2012009156A1
Authority
US
United States
Prior art keywords
cells
tissue
cell
testes
germline
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/940,888
Other languages
English (en)
Inventor
Fariborz Izadyar
Joel Marh
Chad Maki
Jason Pacchaiarotti
Thomas Ramos
Kyle Howerton
Jadelind Wong
Marnie Olmstead
Johnny Yung-Chiong Chow
Constance Yuen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
HARVARDIANAMD CONSULTING Inc
PrimeGen Biotech LLC
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US12/940,888 priority Critical patent/US20120009156A1/en
Assigned to PRIMEGEN BIOTECH LLC DBA REPROCYTE reassignment PRIMEGEN BIOTECH LLC DBA REPROCYTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MARH, JOEL, YUEN, CONSTANCE, MAKI, CHAD, HOWERTON, KYLE, IZADYAR, FARIBORZ, OLMSTEAD, MARNIE, PACCHIAROTTI, JASON, RAMOS, THOMAS, WONG, JADELIND
Publication of US20120009156A1 publication Critical patent/US20120009156A1/en
Assigned to HARVARDIANAMD CONSULTING, INC. reassignment HARVARDIANAMD CONSULTING, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHOW, JOHN YUNG-CHIONG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/0609Oocytes, oogonia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/061Sperm cells, spermatogonia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • C12N2500/84Undefined extracts from animals from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/06Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/31Pituitary sex hormones, e.g. follicle-stimulating hormone [FSH], luteinising hormone [LH]; Chorionic gonadotropins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the present invention relates to a system of banking germline stem cells from patients at risk of infertility due to damage to their germline stem cells and restoration of fertility in the patients at a later date. Specifically, the present invention relates to collection and isolation of germline stem cells, processing and cryopreservation and determination of reproductive capacity of the cells. Also disclosed are methods for restoring fertility in the patients from whom the germline stem cells were isolated.
  • Germline stem cells reside in the reproductive organs, i.e., the ovaries and testes, represent potentially one of the most important and protected classes of stem cells in the mammalian body. Genetic conservation and high telomerase activity has been reported in stem cells derived from these tissues, as well as, extensive DNA modification with chromatin chromosomal modifications. Scientists have differed about what types of stem cells are resident in adult reproductive tissues, as well as, their potentiality in differentiation.
  • Chemotherapy and radiation treatments not only target cancerous cells, but also rapidly dividing cells.
  • the rapidly dividing germ cells are highly sensitive to these exposures.
  • germline stem cells are similarly sensitive and are acutely and dose-dependently depleted following radiation exposure. Low doses of cytotoxic drugs or irradiation deplete the differentiating spermatogonia while less sensitive spermatogonial stem cells as well as spermatocytes and spermatids may survive.
  • the differentiating germ cells can continue their maturation into sperm cells and can re-colonize the seminiferous tubules with stem cells which are generated from the surviving stem cell population.
  • spermatogeneis may only be restored in a very few seminiferous tubules, thereby limiting fertility.
  • Patients will be permanently infertile after complete depletion of testicular stem cells.
  • the impact on spermatogenesis manifests itself most acutely at, or before, the time of puberty because sperm cannot be typically obtained and cryopreserved as in postpubertal males. Even a few stem cells can re-colonize the seminiferous tubules if given sufficient time to re-initiate spermatogenesis.
  • the limited therapeutic options of preserving fertility and normal ovarian function under various adverse conditions are invasive, such as, for example, cryopreservation of ovarian tissue fragments or single oocytes, and often require prior hormonal therapy, which can be medically inappropriate for many women with hormonally responsive tumors.
  • the first is the grafting of immature tissue (either ovarian or testis) tissue fragments onto the surviving tissue and the second is based on isolation and transplantation of stem cells.
  • Germ cell transplantation has been developed in rodent animal models. Microinjection of germ cells from mice or closely-related species into the seminiferous tubules of a mouse re-stimulated spermatogenesis from donor spermatogonial stem cells. The spermatogonial cells can be cryopreserved or cultured prior to transfer. Similarly cyropreserved ovarian cortical tissue has been transplanted in sheep and human with the resulting resumption of estrus and the birth of live offspring following normal matings.
  • a germline cell banking system is needed in humans to restore reproductive potential in patients without the ability to bank mature sperm or ova.
  • the present disclosure provides a germline cell banking system comprising methods for collection, isolation, expansion, processing, cryopreservation, assay, release and implantation of germline stem cells from both males and females.
  • a method for banking germline stem cells comprising collecting gonadal tissue from a mammal at risk of loss of fertility; transporting said gonadal tissue to a central banking facility; processing said gonadal tissue; cryopreserving said gonadal tissue; and determining fertility potential of said cryopreserved gonadal tissue.
  • the gonadal tissue is testicular tissue.
  • processing of the testicular tissue comprises preparing approximately 0.5-2.0 mm 2 pieces of testicular tissue.
  • processing of the testicular tissue comprises digesting said testicular tissue with at least one enzyme selected from the group consisting of collagenase, DNase, hyaluronidase and trypsin; and inactivating said at least one enzyme, thereby obtaining a suspension of dissociated testicular cells.
  • determining fertility potential is the ability of the testicular tissue to differentiate into mature sperm in vitro. In another embodiment, determining fertility potential is determining the characteristics of testicular cells by at least one of expression of meiotic and postmeiotic markers and size and morphology.
  • the gonadal tissue is ovarian tissue.
  • processing of the ovarian tissue comprises preparing approximately 0.5-3.0 mm 2 pieces of ovarian tissue.
  • the processing further comprises digesting the ovarian tissue pieces with at least one enzyme selected from the group consisting of collagenase, DNase, hyaluronidase and trypsin; and inactivating the at least one enzyme, thereby obtaining a suspension of dissociated ovarian cells.
  • determining fertility potential is performed by the ability of said ovarian tissue to differentiate into mature ova in vitro. In another embodiment, determining fertility potential is determining the characteristics of ovarian cells by at least one of expression of meiotic and postmeiotic markers and size and morphology.
  • the method further comprises the step of implanting the cryopreserved gonadal tissue into residual gonadal tissue in the same individual from which the gonadal tissue was isolated.
  • a germline stem cell banking system comprising means for collecting gonadal tissue; means for transporting gonadal tissue; means for processing gonadal tissue to isolate germline stem cells; means for cryopreserving gonadal tissue or said germline stem cells; and means for determining fertility potential of cryopreserved gonadal tissue or said cryopreserved germline stem cells.
  • FIG. 1 depicts enrichment of GFP (green fluorescent protein) positive subpopulations of testicular stem cells isolated from the transgenic OG2 mouse using flow cytometry.
  • GFP green fluorescent protein
  • FIG. 2 depicts morphological changes during the development of multipotent germ cell (mGC) lines in culture.
  • Mouse Oct-4+/GFP+ cells were observed in cell preparations before culture ( FIG. 2A ; arrows; Day 1-3).
  • FIG. 2B Shortly after culture, down regulation of Oct-4 was observed FIG. 2B ; Day 3-7).
  • FIG. 2C Day 7-15; FIG. 2D Day 15-20.
  • FIG. 2E Approximately three weeks after culture, colonies containing small round cells were formed ( FIG. 2E ; Day 20-30). Up-regulation of Oct-4 was observed about one month after culture ( FIG. 2F ; Day 30-40).
  • FIGS. 2G-I Images of three established mGC lines derived from neonatal OG2, adult OG2 or neonatal OG2-LacZ are presented in FIGS. 2G-I ( FIG. 2G , Neonatal OG2; FIG. 2H , Adult OG2; FIG. 2I , OG2 LacZ), respectively. Scale bars: 50 ⁇ m.
  • FIG. 4 depicts phenotypic and molecular characterization of mGCs. Immunolocalization of pluripotent and germ cell markers are depicted in FIGS. 4A-4D for Oct-4, FIGS. 4E-4H for Nanog, FIGS. 4M-4O for SSEA-1 and FIGS. 4I-4L for the germ cell marker, VASA. Scale bars: FIGS. 4A-4H : 25 ⁇ m; FIG. 4I-4O : 20 ⁇ m. Expression of pluripotent and germ-specific markers determined by RT-PCR is shown in FIG. 4Q . The Western blot analysis of protein contents of Oct-4, Nanog and Sox2 in mGC cells before and after immunoprecipitation (IP) is presented in FIG. 4P .
  • IP immunoprecipitation
  • FIG. 7 depicts the spontaneous differentiation of mGCs. Gastulation of embryoid body (EB; FIG. 7A ) and the expression of markers indicative of polarized epithelium (E-cadherin and laminin1; FIGS. 7B-7C ) and early development of the three germ layers, i.e., ectoderm (ZIC1, PAX6, SOX1), endoderm (GATA4, FOXA2) and mesoderm (BRACHYURY, BMP4 and COL2A1) are shown in FIG. 7D-7F .
  • ectoderm ZIC1, PAX6, SOX1
  • endoderm GATA4, FOXA2
  • BRACHYURY mesoderm
  • FIG. 7A and 7I 50 ⁇ m
  • FIG. 7C and 7E 30 ⁇ m
  • FIG. 7B and 7D 25 ⁇ m
  • FIG. 7G , 7 H and 7 L 45 ⁇ m
  • FIG. 7K 12 ⁇ m.
  • FIG. 9 depicts the formation of teratomas after transplantation of mouse ES cells ( FIG. 9A-9F ) but not multipotent LacZ-GFP mGCs ( FIG. 9G-9L ) into the skin, muscle and testis.
  • the morphology of ESC-derived teratomas was identified by H&E staining on thin paraffin sections, whereas the fate of transplanted multipotent germ cells (mGCs) was identified by LacZ staining shown in blue.
  • mGCs multipotent germ cells
  • FIG. 9M-9R Cross section of the normal testis of an immune deficient mouse is shown in FIG. 9M .
  • FIG. 9M One month after busulfan treatment the majority of the seminiferous tubules are depleted from endogenous spermatogenesis ( FIG. 9N ).
  • Testes of a mouse transplanted with freshly isolated Oct-4+ cells showed spermatogenesis in more than 50% of seminiferous tubule cross sections indicating the presence of cells with SSC property in this population ( FIG. 90 ).
  • FIG. 9P While more than 80% of seminiferous tubules of the mice transplanted with Oct-4+/c-Kit ⁇ cells showed some degree of spermatogenesis ( FIG. 9P ), the majority of tubule cross sections of the mice received Oct-4+/c-Kit+ cells were empty ( FIG. 9Q ). Transplanted mGC also failed to repopulate recipient testes indicating that they do not have SSC properties ( FIG. 9R ). Scale bars: FIG. 9A and 9K : 275 ⁇ m; FIG. 9B , 9 D and 9 I: 60 ⁇ m; FIG. 9C : 140 ⁇ m; FIG. 9E : 100 ⁇ m; FIG. 9F : 50 ⁇ m; FIG. 9G and 9L : 125 ⁇ m; FIG. 9H and 9J : 40 ⁇ m; FIG. 9M-9R : 60 ⁇ m.
  • FIG. 10 depicts chimera formation after incorporation of mGCs into blastocysts and host embryos.
  • the incorporation of LacZ-GFP + mGC cells during early embryonic development and blastocyst formation is presented in FIG. 10A-10D .
  • the majority of the GFP-lacZ cells injected at 8-cell stage have been incorporated at day two of the embryonic development (arrow head) and some cells have not been incorporated yet (arrows).
  • GFP+ cells were further found at day 3.5 incorporated in inner cell mass (arrow) of the blastocyst.
  • An example of four chimeric embryos showing different degree of chimerism is shown in FIG. 10E as whole embryo staining.
  • FIG. 10H-10K show the chimeric pattern in dissected organs
  • FIG. 10L-10O show the chimeric cell population in histological sections in the brain, heart, liver and gonadal ridge (chimeric LacZ-GFP cells appear in blue).
  • Amplification of the GFP and LacZ DNA in tissues of the chimeric pups is shown in FIG. 10P and 10Q, respectively.
  • FIG. 11 depicts graphically in a flow cytometric plot the results of freshly isolated neonatal and adult ovarian cells from transgenic OG2 mice wherein the Oct-4 promoter drives expression of GFP.
  • FIG. 11A shows adult ovarian germline stem cells as depicted graphically with the fluorescence intensity of GFP.
  • FIG. 11B shows neonatal ovarian germline stem cells as depicted graphically with the fluorescence intensity of GFP (levels of c-Kit on GFP+ cells).
  • FIG. 11C depicts graphically the fluorescence intensity of neonatal GFP+ cells expressing c-Kit, also known as CD117.
  • FIG. 12 depicts germline stem cells identifiable by expression of green fluorescent protein in the ovary of a transgenic OG2 mouse at day 2 after birth.
  • FIGS. 12A and 12B show total fluorescence and
  • FIG. 12C shows a computer enhanced cross sectional image with removal of auto-fluorescence.
  • FIG. 13 depicts the results of RT-PCR analysis of mRNA isolated from mouse embryonic stem cells (lane 3), mouse embryonic fibroblasts (MEF, lane 4), GFP+ germline stem cells (lane 5) and GFP ⁇ cells (lane 6) isolated from an OG2 transgenic mouse.
  • FIG. 14 depicts microscopic images of isolated and substantially purified ovarian germline stem cells established and growing as colonies on a feeder layer of MEF cells.
  • FIG. 14A shows colonies after 4 days in culture;
  • FIG. 14B shows a first type of representative colony morphology;
  • FIG. 14C shows a second type of representative colony morphology;
  • FIG. 14D shows colony morphology after passage with collagenase;
  • FIG. 14E shows a third type of representative colony morphology after collagenase passage having a clearly defined border;
  • FIG. 14F shows a fourth type of representative colony morphology after collagenase passage having a poorly defined border;
  • FIG. 14G shows colony morphology after passage #1;
  • FIG. 14H shows colony morphology after passage #2;
  • FIG. 14I shows colony morphology after passage #3;
  • FIGS. 14J and 14K show two different magnifications of ovarian germline stem cell colonies after passage #4.
  • FIG. 15 depicts immunocytochemical staining of isolated and substantially purified ovarian germline stem cells, stained to reveal expression of pluripotent stem cell marker Oct-4 ( FIG. 15A ); pluripotent stem cell marker Nanog ( FIG. 15B ); germ cell marker VASA ( FIG. 15C ); and pluripotent stem cell marker alkaline phosphatase ( FIG. 15D ).
  • FIG. 16 depicts images of differentiating ovarian germline stem cells.
  • FIG. 16A shows GFP+ cells resembling primary oocytes growing at the center of the female germ cell colony;
  • FIG. 16B shows images of follicle-like structures;
  • FIG. 16C shows GFP+ cells resembling primary oocytes growing in the vicinity of the female germ cell colony; and
  • FIG. 16 depicts a pigmented colony.
  • FIG. 17 graphically depicts in a scatter plot the results of a flow cytometric size analysis of the cultured ovarian cells of FIG. 16 confirming and characterizing the large (>15 ⁇ m) oocyte-like cells: FIG. 17A depicting MEF control cells ( ⁇ 15 ⁇ m); and FIG. 17B depicting the oocyte-like cells of FIG. 16 .
  • FIG. 18 depicts immunohistochemical localization of spermatogonial stem cell and germline cell markers in adult primate testes.
  • FIG. 19 depicts the distribution of cells positively stained with stem cell markers at the basement membrane of seminiferous tubules of primate testes.
  • FIG. 20 depicts phenotypic characterization of primate germline stem cells using flow cytometry.
  • FIG. 21 depicts flow cytometric analysis of primate germline stem cells.
  • FIG. 22 depicts GFR ⁇ +/VASA+ cells in different enriched primate germline cell populations.
  • FIG. 23 depicts the carboxyfluorescein diacetate succinimidyl ester (CSFE) activity of subpopulations of primate germline stem cells.
  • CSFE carboxyfluorescein diacetate succinimidyl ester
  • FIG. 24 depicts repopulation of busulfan-treated primate testes with primate germline stem cells: seminiferous tubules of recipient mice transplanted by non sorted cells ( FIG. 24A ); cells sorted by triple markers ( FIG. 24B ); SSEA-4+ sorted cells ( FIG. 24C ); and sham transplanted control testes ( FIG. 24D ).
  • FIG. 26 depicts the quantitative PCR analysis of PLZF expression ( FIG. 26A ) and telomerase activity ( FIG. 26B ) in primate germline stem cells.
  • FIG. 27 depicts the percentages of proliferating primate germline stem cells determined by proliferating cell nuclear antigen (PCNA).
  • PCNA proliferating cell nuclear antigen
  • FIG. 28 depicts the gene expression profile of subpopulations of primate germline stem cells.
  • FIG. 29 depicts the morphology of an expanded primate germline stem cell colony 10 days after culture on MEF feeder layer ( FIG. 29A ); SSEA-4 staining of expanded primate germline stem cell colonies ( FIGS. 29B and C); and GFR- ⁇ staining of an expanded primate germline stem cell colony after passage 4 ( FIG. 29D )
  • FIG. 30 depicts human testicular tissue THT stained with SSEA-4 ( FIG. 30A ) and VASA ( FIG. 30B ).
  • FIG. 31 depicts THT stained with GFR- ⁇ ( FIG. 31A ) and VASA ( FIG. 31B ).
  • FIG. 32 depicts THT stained for VASA ( FIG. 32A ) and Nanog ( FIG. 32B ).
  • FIG. 33 depicts THT stained for SSEA-4 ( FIG. 33A ) and ⁇ 6-integrin ( FIG. 33B ).
  • FIG. 34 depicts negative controls for FIGS. 30-33 consisting of human testis sections stained only with secondary antibody.
  • FIG. 35 depicts THT SSEA-4+ magnetic bead sorted cells transplanted into busulfan-treated recipient mouse testes and after one month stained for SSEA-4 ( FIG. 35A ) and human nuclear protein ( FIG. 35B ).
  • FIG. 36 depicts THT SSEA-4+ magnetic bead sorted cells transplanted into busulfan treated recipient mouse testes and after one month stained for ⁇ 6-integrin ( FIG. 36A ) and human nuclear protein ( FIG. 36B ).
  • FIG. 37 depicts THT SSEA-4+magnetic bead sorted cells transplanted into busulfan treated recipient mouse testes and after one month stained for SSEA-4 ( FIG. 37A ) and ⁇ 6-integrin ( FIG. 37B ).
  • FIG. 38 depicts the negative control for FIGS. 35-37 consisting of human testis sections stained only with second antibody.
  • FIG. 39 depicts the weight of testes from control and transplanted animals.
  • FIG. 40 depicts fertile ( FIG. 40A ) and infertile ( FIG. 40B ) testes.
  • FIG. 41 depicts the stage of spermatogenesis (percent empty and partially full tubules) in fertile and infertile animals.
  • FIG. 42 depicts the percentage of GFP positive tubules in infertile and fertile mice.
  • FIG. 43 depicts the morphology and cell surface marker analysis of adult human testicular cells. Note the morphology of the testes obtained from the obstructive azoospermic men ( FIG. 43A ) are similar to normal human testes ( FIG. 43B ). Also after isolation cells with similar morphologies were obtained from both normal testes ( FIG. 43C ) and testes collected from azoospermic patients ( FIG. 43D ). Note SSCs were present in both testes isolates and could be identified as round cells with large nucleus: cytoplasm ratio, 1-3 nucleoli and cytoplasmic inclusions. Flow cytometry analysis of surface markers SSEA-4, CD49f and CD90 in isolated cells from adult human testes ( FIG.
  • FIG. 43E-H Distinct populations of SSEA-4+, CD49f+and CD90+ cells were found in the adult human testicular biopsies and no population of double stained cells for CD49f and CD90 was found in the adult human testes ( FIG. 43E-F ). Histogram representation of four independent flow analyses is presented in FIG. 43I .
  • FIG. 44 depicts the immunohistochemical localization of spermatogonial stem cell markers in adult human testes.
  • Co-localization of SSEA-4 and CD49f at the basement membrane of seminiferous tubules FIG. 44A-C ).
  • SSEA-4 is specifically localized in subpopulation of spermatogonia at the basement membrane of the seminiferous tubules persumabely the SSCs. All SSEA-4+ cells were also positive for CD49f, while there are some CD49f+ cells that are SSEA-4 ⁇ .
  • C-Kit was found in both cells located at the basement membrane of seminiferous tubules and in more advanced germ cells ( FIG. 44E-F ).
  • Co-localization of SSEA-4 and c-Kit revealed that some of the SSEA-4+ cells possess c-Kit and some are c-Kit ⁇ .
  • Co-localization of CD49f with c-Kit ( FIG. 44G-I ) showed that the majority of CD49f+ cells at the basement membrane of tubular cross sections were also positively stained for c-Kit (arrows).
  • Expression pattern of Nanog in adult human testes was similar to c-Kit and it was present in both undifferentiated and differentiated germ cells ( FIG. 44K and L).
  • Co-localization of SSEA-4 with Nanog showed that some of the SSEA-4+ cells in adult human testes are Nanog+.
  • FIG. 45 depicts the quantitative RT-PCR analysis and telomerase activity of enriched population of SSCs isolated from adult human testes.
  • SSEA-4+ cells showed significantly (P ⁇ 0.05) higher expression levels of SSC specific genes including GFR- ⁇ 1, C-Ret, GPR-125 and hTERT ( FIG. 45A ).
  • c-Kit was remarkably increased in the SSEA-4+ cells as compared to the negative cells.
  • Telomerase activity of SSEA-4+ cells was also significantly (P ⁇ 0.01) higher than freshly isolated non sorted cells ( FIG. 45B ).
  • FIG. 46 depicts the expression of specific markers in human spermatogonial stem cells repopulating in the mouse testes.
  • the identity of human cells in the mouse testes was detected by human nuclear protein (HNP) antibody ( FIG. 46A ). Note all the human cells colonized mouse testes are positively stained for germ cell specific marker VASA ( FIG. 46B and C). Some of the human cells at the basement membrane of the mouse testes co-localized CD49f and some were negative for this marker ( FIG. 46D-F ). Co-localization of SSEA-4 with c-Kit showed that all the SSEA-4+ cells in the mouse testes express c-Kit ( FIG. 46G-I ).
  • FIG. 47 depicts the expression pattern of cell surface markers used for characterization of human spermatogonial stem cells by flow cytometry. There was a minute amount of cells (1-2%) found in adult human testes expressing GFR- ⁇ 1, CD24, CD117 and CD166. BCRP, CD29, MHCI and MHCII were moderately (2-5%) expressed in adult human testicular cells. CD90, CD49f, CD34, and SSEA-4 were abundantly found at the surface of cells isolated from adult human testes. The value represents the actual amount of positive cells minus any auto fluorescent events.
  • FIG. 48 depicts the viability of cells isolated from mouse testes before and after tissue freezing.
  • FIG. 49 depicts the average viability of thawed porcine testicular cells frozen at different cell concentrations.
  • FIG. 50 depicts the viability of thawed human testicular cells shipped in three different media for three different times ( FIG. 50A ) and at different cell concentrations ( FIG. 50B ).
  • FIG. 51 depicts the percent recovery of total cells ( FIG. 51A ) and viability ( FIG. 51 B) from frozen samples in different concentrations and shipping conditions.
  • FIG. 52 depicts the change in average testicular cell viability of shipped rat testes over time.
  • FIG. 53 depicts flow cytometric analysis of GFP+ sperm and tubules in the right ( FIG. 53A ) and left ( FIG. 53B ) testes of an infertile mouse.
  • Committed refers to cells which are considered to be permanently committed to a specific function. Committed cells are also referred to as “terminally differentiated cells.”
  • culture refers to the propagation of cells under controlled conditions such that cell division and increase in cell numbers occurs.
  • Differentiation refers to the adaptation of cells for a particular form or function. In cells, differentiation leads to a more committed cell.
  • Embryonic Stem Cell refers to any cell that is totipotent and derived from a developing embryo that has reached the developmental stage to have attached to the uterine wall. In this context embryonic stem cell and pre-embryonic stem cell are equivalent terms.
  • Embryonic stem cell-like (ESC-like) cells are totipotent cells not directly isolated from an embryo. ESC-like cells can be derived from primordial sex cells that have been isolated and expanded.
  • Expanded refers to a growing culture of cells that has increased in cell number from its original concentration.
  • Fetal Stem Cell refers to a cell that is multipotent and derived from a developing multi-cellular fetus that is no longer in early or mid-stage organogenesis.
  • Germline Cell As used herein, “germ cell”, “germline cell,” or “germline tissue” refers to a reproductive cell such as a spermatocyte or an oocyte, or a cell that will develop into a reproductive cell or a tissue containing such cells.
  • Germline precursor stem cell refers to a reproductive cell such as a precursor of a spermatogonial stem cells or an oocyte precursor stem cell.
  • Germline stem cell refers to a reproductive cell such as a spermatogonial stem cell (SSC) or an oocyte precursor stem cell.
  • Gonad refers to any of the paired organs in animals that produce reproductive cells (gametes). These include female ovaries, which produce eggs, and male testes, which produce sperm.
  • Long-term culture refers to the propagation of cells under controlled conditions for longer than at least two months or more than 10 passages.
  • the long-term cultures are cultured for more than 4 months, more than 6 months or more than 1 year.
  • the long-term cultures are passaged for more than 15 passages, more than 18 passages or more than 20 passages.
  • the duration of the long-term cultures is highly dependent on the individual cells and there can be variability from cell line to cell line.
  • Maturation refers to a process of coordinated biochemical steps leading toward a terminally differentiated cell type.
  • Multipotent refers to cells that can give rise to several other cell types, but those cell types are limited in number.
  • An example of multipotent cells is hematopoietic cells—blood stem cells that can develop into several types of blood cells but cannot develop into brain cells.
  • Multipotent adult progenitor cells refers to multipotent cells isolated from the bone marrow which have the potential to differentiate into cells of the ectoderm, mesoderm and endodermal lineages.
  • Pluripotent refers to cells that can give rise to any cell type except the cells of the placenta or other supporting cells of the uterus.
  • Post-natal Stem Cell refers to any cell that is derived from a multi-cellular organism after birth.
  • Pre-Pubescent As used herein, “pre-pubescent” or “pre-pubertal” refers to individuals who have not yet entered puberty. Onset of puberty is associated with high gonadotrophin releasing hormone (GnRH) pulsing, which precedes the rise in sex hormones, lutenizing hormone (LH) and follicle stimulating hormone (FSH). Puberty begins consistently at around 47 kg for girls and 55 kg for boys. Although there is a wide range of normal ages, on average, girls begin the process of puberty about 1-2 years earlier than boys (with average ages of 9 to 14 for girls and 10 to 17 for boys), and reach completion in a shorter time with girls usually having completed puberty by age 17.
  • GnRH gonadotrophin releasing hormone
  • LH lutenizing hormone
  • FSH follicle stimulating hormone
  • primordial germ cell refers to cells present in early embryogenesis that are destined to become germ cells.
  • primordial germline sex stem cell also referred to in short form as a “germline sex cell” abbreviated PGLSC, refers to a cell that is derived from adult male or female reproductive tissue, and which is able to generate germline stem cells and their progeny as evidenced by its ability to repopulate reproductively sterile testicular or ovarian tissues after e.g. radiation or chemotherapy. Germline sex cells can be quiescent or actively dividing in adult reproductive tissues.
  • Reprogramming refers to the resetting of the genetic program of a cell such that the cell exhibits pluripotency and has the potential to produce a fully developed organism. In addition this reprogramming gives the cell undergoing reprogramming characteristics that would normally not be expressed or found in the cell in its pre-programming state.
  • selection refers to fluorescence-activated cell sorting, magnetic bead sorting or other means of collecting cells bearing a particular marker profile
  • Sex Cell refers to diploid or haploid cells derived from the mammalian male or female reproductive tissues. Representative examples of these cells include male gonocytes, female gonocytes, oogonia, type-A spermatogonia and Type-B spermatogonia.
  • Somatic Cell refers to any tissue cell in the body except sex cells and their precursors.
  • Somatic Stem Cells As used herein, “somatic stem cells” refers to diploid multipotent or pluripotent stem cells. Somatic stem cells are not totipotent stem cells.
  • stem cells refers to cells capable of self-renewal (the ability to go through numerous cycles of cell division while maintaining the undifferentiated state and being at least multipotent (the capacity to differentiate into more than one specialized cell type.
  • substantially pure refers to a population of cells wherein greater than 75%, greater than 85%, greater than 90%, greater than 95%, greater than 98% or greater than 99% of the cells have the desired characteristic(s).
  • Totipotent refers to cells that contain all the genetic information needed to create all the cells of the body plus the placenta. Human cells have the capacity to be totipotent only during the first few divisions of a fertilized egg.
  • the present disclosure provides methods for collection, isolation, expansion, processing, cryopreservation, assay, release and transplantation of germline stem cells from both males and females for the purposes of fertility preservation in patients at risk for loss of fertility.
  • Chemotherapy and radiation therapy used in the treatment of cancers, as well as other non-malignant diseases (such as, but not limited to, sickle cell anemia and thalassemia) prior to bone marrow stem cell transplantation has a well-documented, detrimental effect on the fertility of patients undergoing these therapy. These treatments may permanently impair the reproductive capability of patients, presenting a 30% chance that the patients will become infertile.
  • the present disclosure provides methods for banking testicular gonadal tissue and/or germline stem cells from pre-pubescent patients.
  • tissue and germline stem cell banking would be desired in males include, but is not limited to, bilateral cryptorchidism, testicular torsion, undescended testis, varicocele, cancer, including reproductive and non-reproductive cancers, cytotoxic therapy, bone marrow transplant.
  • the donor is a pre-pubertal male. In another embodiment, the donor is a post-pubertal male.
  • the present disclosure provides methods for banking ovarian tissue and cells for pre-pubescent patients and ovarian tissue, follicles and cells from adult patients.
  • disorders or situations in which tissue and germline stem cell banking would be desired in females include disorders and situations that directly or indirectly affect fertility such as, but is not limited to, ovarian cysts, ectopic pregnancies, hysterectomy, cancer, including reproductive and non-reproductive cancers, cytotoxic therapy, bone marrow transplant, ovariectomies, endometriosis, or for family planning purposes if woman wants to preserve reproductive potential but is approaching or passing her reproductive prime.
  • the donor is a pre-pubertal female.
  • the donor is a post-pubertal female.
  • oocytes or eggs can be removed and cryopreserved for use with assisted reproductive technologies, should the patient become infertile after treatment.
  • viability of frozen and thawed human oocytes is quite low due to inherent difficulties in freezing a very large cell with low surface to volume ratio which does not allow cryoprotectant to penetrate the cell membrane easily. Therefore, tissue, primordial follicle, or ovarian cell (which may contain germline stem cells) banking is a viable option instead of, or in addition to, oocyte freezing.
  • cryopreserved ovarian tissue or follicles can be transplanted back into patients if they become infertile, at a time after treatment when they are ready to restore their fertility.
  • the frozen tissue or follicles can be used in an ex host maturation process described herein and in co-pending application ______ entitled “Ex host maturation of germline stem cells” (Attorney Docket No.
  • the germline cells disclosed herein are isolated from gonadal tissues from mammals including but not limited to, rodents, domesticated animals, dogs, cats and primates.
  • the term “primates” includes, but is not limited to, humans.
  • Germline cell markers include, but are not limited to, VASA, promyelocytic leukemia zinc factor (PLZF), glial derived neurotrophic factor receptor ⁇ 1 (GFR- ⁇ 1), ⁇ 6-integrin, Thy-1, CD9, CD90, CD49f, Dolichos biflourus agglutinin (DBA), neural cell adhesion molecule (NCAM), germ cell nuclear antigen 1 (GCNA1) and DAZL.
  • VASA promyelocytic leukemia zinc factor
  • GFR- ⁇ 1 glial derived neurotrophic factor receptor ⁇ 1
  • ⁇ 6-integrin ⁇ 6-integrin
  • Thy-1 ⁇ 6-integrin
  • CD9 ⁇ 6-integrin
  • CD90 CD49f
  • CD49f Dolichos biflourus agglutinin
  • DBA Dolichos biflourus agglutinin
  • NCAM neural cell adhesion molecule
  • Pluripotent cell markers include, but are not limited to, Oct-4 (POU5F1), Nanog, alkaline phosphatase, SSEA-4, TRA1-60 and TRA1-81.
  • germline cells can also be isolated based on the expression of germline and/or pluripotent stem cell genes.
  • Germline stem cell genes include, but are not limited to, telomerase, VASA, c-RET, c-Kit, PLZF, DAZL and GFR- a1.
  • Pluripotent cell genes include, but are not limited to, Oct-4, Nanog, Dppa-5, Sox2, alkaline phosphatase and Crypto.
  • Additional embodiments presented herein include the long-term culture of certain populations of germline cells such that long-term multipotent or pluripotent cells lines are generated. These cells lines can be used as a source of cells for differentiation into tissue-specific lineages.
  • Long term culture of the instant germline cells comprises the steps of isolating a substantially pure population of the desired germline cell based on expression, or lack of expression, of germline and/or pluripotent cell markers and germline and/or pluripotent genes; culturing the cells in growth medium as disclosed in the Examples section which allow continued cell division while maintaining an undifferentiated multipotent or pluripotent state.
  • the long-term cultures described herein can be cryopreserved for future uses.
  • isolated, substantially pure populations of germline cells or germline cell lines can be used for therapeutic applications in regenerative medicine.
  • Germline cells disclosed herein are capable of forming more differentiated germline cells such as spermatocytes, spermatids and sperm in male, and follicles and oocytes in female and are capable of re-populating a sterile reproductive organ in vivo.
  • the germline tissue banking system of the present disclosure comprises the following components and/or steps: 1) Tissue collection; 2) Transport of tissue to central banking location; 3) Processing of tissue at central bank; 4) Cryopreservation of tissue and isolated cells at central bank; and 5) Quality assurance and release of tissue and/or isolated cells.
  • An optional sixth step comprises re-implantation of tissue and/or isolated cells into the donor.
  • Germline cells are retrieved from testes prior to the initiation of a cytotoxic procedure or soon after an injury which can lead to the destruction of the germ cell tissue. At least one gram of seminiferous tubule tissue is removed under surgical aseptic conditions and anesthesia. Physical and enzymatic digestion of the tissue to form a single cell suspension can be performed either prior to or after shipment of the tissue to the central banking facility.
  • testicular seminiferous tubule tissue is cut into approximate 10 ⁇ 10 mm pieces and up to 5 pieces are placed in a sterile tube containing sterile shipping media. In another embodiment, the tissue is maintained in a single piece and placed in a sterile container containing sterile shipping media.
  • Shipping media includes any media which can support the viability of testicular seminiferous tubule tissue and/or dissociated cells for up to 24 hr.
  • tissue maintenance media include PBS, FRS, DMEM supplemented with HEPES and antibiotics (penicillin and streptomycin) as well as proprietary media disclosed in U.S. Patent Application Publication No. 2007-0020759, which is incorporated by reference herein for all it contains regarding tissue culture media.
  • Germline cells are retrieved from ovaries prior to the initiation of a cytotoxic procedure, soon after an injury which can lead to the destruction of the germ cell tissue or at the time of ovariectomy or hysterectomy. At least one gram of ovarian tissue is removed under surgical aseptic conditions and general anesthesia. Physical and enzymatic digestion of the tissue to form a single cell suspension can be performed either prior to or after shipment of the tissue to the central banking facility.
  • ovarian tissue is cut into approximate 10 ⁇ 10 mm pieces and up to five pieces are placed in a sterile tube containing sterile shipping media. In another embodiment, the tissue is maintained in a single piece and placed in a sterile tube containing sterile shipping media.
  • Shipping media includes any media which can support the viability of ovarian tissue and/or dissociated cells for up to 24 hr.
  • tissue maintenance media include PBS, FRS, DMEM supplemented with HEPES and antibiotics (penicillin and streptomycin) as well as proprietary media disclosed in U.S. Patent Application Publication No. 2007/0020759.
  • the harvested testicular or ovarian tissue is kept at approximately 4° C. and transported to the central banking facility such that it arrives at the central banking facility within 24 hr of tissue harvesting.
  • Our data show that the viability of cells does not change after 48 hr of shipment and declines after 72 h of shipment. We prefer to have the tissue within 24 hr of harvesting.
  • Harvested ovarian and testicular tissue is enzymatically dissociated, assessed for viability and cell markers prior to cryopreservation.
  • germline stem cells are enriched from the tissue prior to cryopreservation.
  • total testicular or ovarian cells, without enrichment for germline stem cells are cryopreserved.
  • enrichment for germline stem cells is conducted by flow cytometric sorting using antibodies specific for male and/or female germline stem cells.
  • a variety of media and procedures can be used for cryopreserving gonadal tissue and/or germline stem cells.
  • germline stem cells are cryopreserved in a solution comprising at least one cryoprotectant including, but not limited to, dimethyl sulfoxide (DMSO), ethylene glycol, glycerol, and propanediol; at least one culture medium including but not limited to, DMEM, MEM and proprietary media disclosed above; at least one additional agent including, but not limited to, sucrose, dextran, a serum substitute and HEPES buffer.
  • the solution comprises CryoStorTM CS-10 media (BioLife Solutions Inc., Bothell, Wash.).
  • the serum substitute is Knockout Serum Replacement (Invitrogen 10828-028).
  • the cells are then frozen at a controlled rate or by a “manual” process.
  • the controlled-rate freezing procedure begins by turning the controlled-rate freezer on and setting up the freezing program for tissue or cell freezing.
  • the controlled-rate freezer will use liquid nitrogen to decrease the temperature in an internal chamber (and thus decrease the temperature of any contents of that chamber).
  • the freezing program for cells begins by cooling the internal chamber to 4° C. and holding it there until prompted to continue the procedure. While the controlled-rate freezer is cooling, cells are suspended in a cryopreservation media cooled to 4° C. That cell suspension is aliquoted into cryovials, 1 mL per cryovial. The cryovials are then labeled and placed in the controlled-rate freezer chamber and the program is prompted to continue.
  • the chamber is temperature is held at 4° C. for an additional 10 minutes.
  • the chamber is cooled at a rate of ⁇ 1° C./minute until the temperature reaches ⁇ 80° C.
  • the chamber then cooled at a rate of ⁇ 50° C./minute until it reaches a temperature of ⁇ 120° C.
  • the temperature of the frozen cells will equilibrate to ⁇ 120° C.
  • the cryovials of frozen cells are then transferred to a liquid nitrogen Dewar for long-term storage.
  • the manual freezing procedure begins by preparing a “Mr. Frosty” freezing container (Nalgene) which is used to slowly freeze the cells.
  • the “Mr. Frosty” container is a polycarbonate unit that provides the critical, repeatable ⁇ 1° C./minute cooling rate required for successful cell cryopreservation and recovery.
  • the base of the “Mr. Frosty” is filled with 250 mL of 100% isopropanol.
  • the tube rack is placed on top, the lid is screwed down over the tube rack and the “Mr. Frosty” is placed at 4° C. for at least 1 hour before being used.
  • the cells are suspended in a cryopreservation media cooled to 4° C. That cell suspension is aliquoted into cryovials, 1 mL per cryovial.
  • cryovials are then labeled and placed in the pre-cooled “Mr. Frosty”.
  • the “Mr. Frosty” is placed back at 4° C. for 10 minutes.
  • the “Mr. Frosty” is then placed in a ⁇ 80° C. freezer overnight. After the overnight stay in the ⁇ 80° C. freezer, the cryovials are transferred to a liquid nitrogen Dewar for long-term storage.
  • Potency, purity, identity, viability and stability assays are performed on the samples prior to cryopreservation, or at specific time points (for stability), and upon release to ensure that the tissue or cells are in proper condition for transplantation.
  • These assays quantify specific relevant markers in the tissue/cells sample that provide information on the amount of germline stem cells present (to extrapolate the “potency” potential to repopulate testes if transplanted), viability of cells, and optionally DNA analysis to confirm identity of the cells.
  • Stability assays consist of small samples of tissue or cells being thawed at regular intervals to check quality of cryopreservation.
  • Transplanting the isolated substantially pure population of germline stem cells into the recipient is accomplished by direct injection using standard injection means known to persons of ordinary skill in the art.
  • support cells such as Leydig or Sertoli cells that provide hormonal stimulus to spermatogonial differentiation
  • These transferred support cells are unmodified, or, alternatively, are genetically modified.
  • These transferred support cells can be autologous or heterologous to either the donor or recipient testis.
  • An example concentration of cells in the transfer fluid can easily be established by simple experimentation, but will likely be within the range of about 10 3 -10 10 cells per ml.
  • the cells are be introduced into the vas deferens, the rete testis or the seminiferous is performed be done manually.
  • a suitable dyestuff or bubbles can optionally be incorporated into the carrier fluid for easy identification of satisfactory delivery of the transplanted germline stem cells to testes.
  • An ultrasound equipped with appropriate transducer may be helpful for placing the needle in the injection site.
  • Suitable cell transplant vehicles are known to persons of ordinary skill in the art and include molecules such as serum albumin, cholesterol and/or lecithin, selenium and inorganic salts as well as serum components and/or growth factors and/or cytokines.
  • the cell transplant vehicle has a pH which is roughly physiologic, i.e. 7.0 to 7.6.
  • the instant cellular compositions may to be administered alone or in combination with one or more pharmaceutically acceptable carriers, in either single or multiple doses.
  • suitable pharmaceutical carriers may include inert biodelivery gels or biodegradable semi-solid matrices, as well as diluents or fillers, sterile aqueous solutions and various nontoxic solvents.
  • the pharmaceutically acceptable carriers generally perform three functions: (1) to maintain and preserve the cells in the instant cellular composition; (2) to retain the cells at a tissue site in need of regeneration, restoration or rejuvenation; and (3) to improve the ease of handling of the instant composition by a practitioner, such as, but not limited to, improving the properties of an injectable composition or the handling of a surgical implant.
  • compositions formed by combining an instant cellular composition with a pharmaceutically acceptable carrier may be administered in a variety of dosage forms such as injectable solutions, and the like.
  • the pharmaceutical carriers can, if desired, contain additional ingredients such as binders, excipients, and the like.
  • the aqueous solution is preferably suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • Such aqueous solutions are suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal injection.
  • the subject sterile aqueous media employed are obtainable by standard techniques well known to those skilled in the art.
  • Ovarian germline stem cells could be isolated from patients prior to chemo or radiation therapy, expanded in number in vitro and kept frozen until the patient requires them to regain their fertility.
  • Cells isolated from human ovary are transplanted directly into the ovarian surface epithelium using a fine needle or alternatively cells could be aggregated in a fibrin clot.
  • Fibrin clots are prepared from patients own blood and support the viability and survival of cells during the implantation process. Fibrin clots could be transplanted back to the patient and grafted under the ovarian bursa.
  • OSE ovarian surface epithelium
  • tissue collection OSE is dissected from the rest of the ovarian tissue using scalpels to a 0.5 cm ⁇ 0.5 cm pieces with the diameter of 1 mm. These pieces are frozen using a vitrification procedure and kept frozen until patients need them for fertility restoration. When needed, the OSE pieces are thawed and sutured together to make a larger piece and grafted to the surface of the infertile ovary.
  • ovarian follicles at different developmental stages can be frozen and either re-implanted into the patient or be used for in vitro culture into mature follicles. Oocytes from these follicles could then be fertilized by ICSI (intracytoplasmic sperm injection) or other assisted reproductive techniques. Transplantation of ovarian germline stem cells is a permanent fertility restoration.
  • ICSI intracytoplasmic sperm injection
  • Female germline stem cells isolated from human ovaries can be differentiated into primordial follicles using an aggregation without growth factors and ⁇ -mercaptoethanol in ultra-low adhesive culture dishes following by 3D culture in sodium alginate or fibrin clot in the presence of growth hormone. To further support development of germ cells to mature egg, these cells are aggregated with immature granulosa cells from prepubertal age donors. This method allows development of primary oocytes to fertilizable metaphase-II oocytes. For isolation of granulosa cells, ovaries from prepubertal human ovaries are dissected and granulosa cells are separated from germ cells by a two step differential adhesion procedure.
  • testicular germline stem cells are differentiated in a surrogate animal or in vitro to further developed stages and be used for ICSI or other assisted reproductive technologies.
  • SSCs spermatogonial stem cells
  • ECM tubule extracellular matrix
  • spermatogenesis has already been started in prepubescent patients but not completed, partially mature germ cells that already entered meiosis (primary and secondary spermatocytes) can be stored and used for further differentiation to round or elongated spermatids.spermatids then could be used for ICSI allowing fertilization and subsequent embryo development.
  • the disclosed methods are suitable for use in any mammals including, but not limited to, human, mice, domesticated animals such as cattle, swine, sheep and goats, dogs, cats, etc.
  • a germline cell bank or depository or storage facility is a place where germline cells or germline stem cells are kept for safe keeping.
  • the storage facility may be designed in such a way that the stem cells are kept safe in the event of natural disasters, over prolonged periods of time and in a controlled environment.
  • a depository e.g., a germline cell bank
  • a depository for storing germline cell units in anticipation of a need to restore fertility in the donor.
  • the scope of the disclosed methods includes not only the provision of such a depository, but also to methods for administering such a depository.
  • a germline cell unit includes any indexing and retrieval system so that the germline cells or tissues can be stored.
  • the germline cells are stored in such way that a storage facility will be able to store hundreds, thousands and even millions of different germline cell units.
  • Identifying information may include type identifier information, patient identifier information and/or genotype of phenotypic information. It may also include maintenance information. It may include information such as the volume of the germline cell unit or the total number of cells per unit.
  • This patient identifier indication may be in the form of a name or a secret identifier code.
  • Each germline cell unit e.g., particular unit from an individual, will be indexed in a manner for reliable and accurate identification and retrieval.
  • Any conventional indexing system may work as long as it is reliable and accurate.
  • each container for each germline cell unit may be marked with alphanumeric codes, bar codes, or any other cognizable method or combinations thereof.
  • At a location in the depository and outside the depository there may be an accessible and readable listing of information enabling identification of each unit and its location in the depository and enabling identification of the source and/or type of unit.
  • This indexing system can be managed in any way known in the art, e.g., manually or non-manually, e.g. a computer and conventional software can be used.
  • the germline cell bank comprises a system for storing a plurality of records associated with a plurality of individuals and a plurality of germline cell units. Each record may contain type information, genotypic information or phenotypic information associated with the germline cell units or specific individuals.
  • Storage of the germline cell units may be short term or long term.
  • the stored germline cells are cryogenically preserved. Any storage method may be used providing that the stored product retain viability for the therapeutic purposes discussed herein.
  • germline cell units are known in the art and can be used according to the present disclosure. There is no upper limit on the number of germline cell units than can be stored in one particular bank. In one embodiment, hundreds of germline cell units from different individuals will be stored at one bank or storage facility.
  • a facility for storing germline cell products may be quite small, yet still store many samples for a large number of people.
  • germline cell units are stored in such a way to minimize the amount of space needed.
  • the storage facility may include means for any method of organizing, and indexing the stored products, e.g., automated robotic retrieval and/or the manipulation of germline cell units.
  • the facility may include micromanipulation devices for processing such germline cell units. More than one storage facility may be used to store germline cell units. These facilities may each be at a different location miles apart. In some embodiments, the storage facilities may be in different states, in different countries.
  • the storage facilities may be underground or aboveground.
  • Fault tolerance computers and redundant systems may be used throughout the storage facilities to eliminate potential problems and to provide a fail-safe system.
  • Known conventional technologies can be used for efficient storage and retrieval of the germline cell units, e.g., Machine Vision, Robotics, Automated Guided Vehicle System, Automated Storage and Retrieval Systems, Computer Integrated Manufacturing, Computer Aided Process Planning, Statistical Process Control.
  • Less sophisticated storage facilities may be used as well, e.g., large areas maintained at appropriate temperatures contain numerous racks on which are indexed and stored the germline cell units
  • a depository, storage facility or germline cell bank may include one or more storage units for storing the germline cell samples and one or more processing stations for processing the germline cell units for storage or for transplantation. Processing may be done by an inside unit or outsource unit, for example a laboratory which specializes in germline cell processing and storage. In some embodiments, the entire process, acquiring, processing, typing, recording and storing the stem cells is managed by a data processing and control unit.
  • the operating of this system may be managed with the assistance of dedicated software.
  • the software may manage enrollment of the potential recipient, acquisition of the germline cell units from donors, database management, storage monitoring, quality checking.
  • this system may operate on a typical computer system.
  • the computer system may include input devices, e.g., a keyboard or mouse, a processor, e.g., a general purpose processor or a more developed processor with increased database processing capabilities, an internal memory, e.g., RAM, ROM, and external storage, e.g., disks, CD, ROMs, ASICs, External RAM, external ROM.
  • the computer system is capable of running on any operating system.
  • the database system stores information for each germline cell unit in the bank.
  • the information is stored in association with each unit.
  • the information may be associated with a particular donor, for example, an identification of the donor and the donor's medical history.
  • the information may be sample type information.
  • the information might include the volume of the germline cell unit or the total cells count in the product.
  • the information stored with each sample is searchable and identifies the sample in such a way that it can be located and supplied to the client immediately.
  • C-Kit a tyrosine kinase receptor
  • SCF ligand stem cell factor
  • C-Kit is expressed in PGCs from their initial segregation to their arrival at the genital ridge. In postnatal mouse testes, it has been reported that c-Kit can be used as a marker for differentiation of undifferentiated and differentiating type A spermatogonia.
  • Combinations of Oct-4 and c-Kit allow the isolation of two distinct populations in germline stem cells: one containing more primitive germ cells or germline progenitors (Oct-4+/Kit+) and other contains germline stem cells destined to be SSCs (Oct-4+/c-Kit ⁇ ) and with the ability to regenerate a sterile testis.
  • the molecular and phenotypic characteristics of these cells were analyzed both before and after culture and compared their ability to generate multipotent cell lines under a defined culture condition with a mixture of growth factors.
  • the functionality of these subpopulations and their descendent mGC lines to repopulate recipient testes was evaluated using spermatogonial stem cell transplantation technique.
  • GSC Male germline stem cells
  • mGC multipotent germ cell
  • a mGC line was generated from neonatal wild type CD-1 mice indicating that the method is not limited to transgenic OG2 mice.
  • Selected cell lines were frozen/thawed and propagated for more than 40 passages with an estimated cell doubling time of 72 hr (using both manual cell count and GFP sorting ( FIG. 3 ).
  • the number of GFP+ cells were analyzed by FACS ( FIG. 3E ).
  • C-Kit+/GFP+ cells were separated from the c-Kit ⁇ /GFP+ cells by flow cytometry ( FIG. 1D-E ) and cultured on MEF feeders. Only c-Kit+ populations generated mGC colonies and no cell line could be generated from the c-Kit ⁇ pool.
  • removal of GDNF resulted to smaller colonies indicating the role of GDNF in self renewal of the mGCs.
  • removal of FGF2 resulted in differentiation of the colonies, indicating possible role of FGF2 in maintenance of the mGCs in their undifferentiated stage. Removal of LIF or EGF did not affect either the expansion or differentiation of the mGCs.
  • FIGS. 4A-4D The majority of cells in the mGC colonies expressed Oct-4 ( FIGS. 4A-4D ), Nanog ( FIGS. 4E-4H ), VASA ( FIG. 41-4L ), and SSEA-1 ( FIGS. 4M-4O ). They also expressed pluripotent genes Sox2, DPPa5, Rex-1, eRas, and Cripto along with germline specific markers, including Stella, Daz1, Vasa and cRet ( FIG. 4Q ). In addition, the expression of Oct-4, Nanog and Sox2 was confirmed by Western blot analysis ( FIG. 4P ).
  • the mouse cell line at passage 20 showed high telomerase activity ( FIG. 5A , similar to ESC) and normal karyotype (40, XY) ( FIG. 5B ).
  • the global gene expression and imprinting patterns of the mGCs were also analyzed before and after culture and compared with that of ESC. Interestingly, culture conditions did not change the imprinting pattern of the mGCs in all the DMR (differentially methylated region) sites tested. In contrast to mouse ESC that showed only a partial androgenetic imprinting, the mGCs clearly exhibited a 100% androgenetic imprinting pattern ( FIG. 6 ). Somewhat surprisingly, microarray analysis showed that the global gene expression pattern of the mGCs had 87% similarity before and after culture.
  • FIG. 7A When mGCs were aggregated to form embryoid bodies (EBs), gastrulation was observed within 9-15 days ( FIG. 7A ).
  • Cells in the EBs expressed early developmental markers including E-cadherin and laminin1 (markers of polarized epithelium ( FIG. 7B-7C ), Zic1, PAX6 and Sox1 (early ectoderm markers, FIG. 7D and 7F ), GATA4 and FoxA2 (early endoderm markers, FIG. 7E-7F ), and Brachyury, BMP4 and COL2A1 (early mesoderm markers, FIG. 7F ).
  • E-cadherin and laminin1 markers of polarized epithelium ( FIG. 7B-7C ), Zic1, PAX6 and Sox1 (early ectoderm markers, FIG. 7D and 7F ), GATA4 and FoxA2 (early endoderm markers, FIG. 7E-7F ), and Brachyur
  • mGC lines could be induced to differentiate into neural cells representing neural progenitors (nestin, neuroD1), neurons (MAP2, NF-68, GAD67) and glial cells (GFAP, MBP, A2B5, O4, NG2) as shown in FIGS. 8A-8G and 8 J. They could also be induced to form cardiomyocytes (troponin1, cardiac myosin, desmin, Nkx2.5, GATA4, FIGS. 8I and 8L ) or chondrocytes (collagen Xa1, and staining by alcian blue, FIG. 8H and 8K ).
  • FIG. 9N One month after busulfan treatment, the majority of the seminiferous tubules were depleted from endogenous spermatogenesis ( FIG. 9N ). While 73% of seminiferous tubules of mice transplanted with Oct-4+/c-Kit ⁇ cells showed some degree of spermatogenesis ( FIG. 9O ), the majority of tubule cross-sections of the mice receiving Oct-4+/c-Kit ⁇ cells were empty ( FIG. 9P ). A CSFE-tagged positive colony shortly after transplantation of Oct-4+/c-Kit ⁇ cells is depicted in FIG. 9R . No spermatogenesis was found in the majority of seminiferous tubules of the recipient mice testes transplanted with the mGCs, indicating these cells do not have SSC properties ( FIG. 9Q ).
  • mice For teratoma formation, equal numbers of mouse ESC (as positive control) or Oct-4-GFP/LacZ mGCs were injected into the skin, muscle and testes of different groups of nude mice (1 ⁇ 10 6 cells/site). All recipient mice (6/6) receiving ESC developed teratomas in all three tissue types. In contrast, none of the mice (0/20) receiving mGCs gave rise to teratomas (FIGS. 9 A- 9 F). Six weeks after transplantation, Oct-4-GFP/LacZ cells, were found in skin, muscle and testicular tissues ( FIGS. 9G-9I ). These data show that mGCs, unlike ESC, are non-tumorigenic.
  • the intensity of LacZ staining was much higher in chimeric embryos received mouse ES cells than those injected with multipotent germ cell lines.
  • the distribution of chimeric cells is also demonstrated in histological sections of the brain, heart, gonadal ridge and liver ( FIG. 10L-10O ).
  • the intensity and number of LacZ+ cells was much higher in chimeric embryos injected with LacZ-ES cells than those injected with LacZ-GS cells.
  • Confirmation of Oct-4-GFP/LacZ chimeric tissues was supported by the presence of GFP DNA sequence in the ectodermal (brain), mesodermal (heart), endodermal (liver) and testis of the chimeric pups ( FIG. 10P ), as well as the presence of LacZ DNA ( FIG. 10Q ) in all 4 tissues.
  • Multipotent germ cell lines can be generated from adult mouse testes without reprogramming growth factors; indicating the possible presence of a subpopulation of cells with pluripotent characteristics in the adult testes.
  • germline sex cell lines and germline precursor cells from post-natal mouse germline stem cells were derived with some, but not all, of the pluripotent characteristics of ESC. Both of these cell line types are distinctively different from the multipotent germ cell lines obtained by the other laboratories, most notably, with regard to the extent of pluripotentiality and teratoma formation.
  • mouse Oct-4+/c-Kit+ germline cell lines expressed pluripotent genes Nanog and crypto but at 1000-fold and 5000-fold lower levels than in ESC.
  • the germ cell lines expressed oncogenes including, but not limited to, p53, Eras, Bak, Int-2 and c-myc, but the expression levels were several fold lower than with ES cells.
  • the germ cell lines did not form teratomas upon transplantation in vivo, but they did form limited chimeric cell populations in mouse embryos.
  • the Oct-4+/c-Kit+ germline precursor cells retain their germ cell properties and thereby differ from ESC and other previously reported testicular cells: namely, 1) the derived germline precursor cell lines have a cell cycle time that doubles their cell numbers in about 72 hr (determined by both GFP sorting and manual counting), and this cell cycle time is more similar to that of germline stem cells and is about three times longer than that of the ESC; 2) based on global gene expression analysis in arrays, the instant germline precursor cells seem to have molecular characteristics different from those in ESC or other multipotent germ cell lines.
  • the instant germline precursor cell lines showed significantly higher expression level of germline specific genes (Vasa, Plzf, GFR- ⁇ 1, DazI) and lower expression level of pluripotent genes (Oct-4, Nanog, Dppa-5, Sox2, Crypto); 3) these cell lines are more dependent on GDNF for their self renewal than LIF or FGF2.
  • GDNF has been proposed to be the key regulator of the self renewal of male germline stem cells, while LIF and FGF2 play crucial role in self renewal of ESC; 4), the expression level of SSEA-1 in these cell lines was lower than the level found either in mouse ES cells or other multipotent germ cell line as reported.
  • SSEA-1 may be involved in tumor invasion and metastasis in certain animal model systems suggesting that higher expression may reflect higher potential for tumorigenesis; and 5) multipotent GCs exhibited an androgenic imprinting pattern that is different from mouse ESC or other mGC lines reported by other laboratories.
  • the transgenic mouse model allowed the isolation germline stem cells from both neonatal and adult testes based on their Oct-4 expression.
  • the germline stem cells were further fractionated into two subpopulations according to their expression of c-Kit with the following observations: 1) only the Oct-4-/GFP+ cells that possess the c-Kit receptor molecule responded to culture and generated multipotent germ cell lines; and, 2) only the c-Kit ⁇ subpopulations repopulated the testis after spermatogonial stem cell transplantation.
  • the results clearly indicate the presence of at least two distinct subset of germline stem cells within reproductive tissues: (1) a c-Kit+ pool with the ability to become multipotent germline stem cells, i.e.
  • germline precursor cells as well as, (2) a subset of germline stem cells that have lost their c-Kit expression and acquired the ability to colonize the testis, i.e., germline sex cells.
  • germline stem cells in the reproductive organs are either present in different developmental stages, or alternatively, they possess differing abilities to respond to growth factor signaling and/or transcription factors.
  • testicular cells Isolation of testicular cells.
  • the testes of either neonatal mouse pups (2-5 days after birth) or adult mice were sterilely removed from the body.
  • the capsule of the testes was removed and the seminiferous tubules were suspended in an enzyme solution consisting of 1 mg/mL collagenase 1A and 10 units/mL DNase in PBS.
  • the testes were digested at 37° C. in a water bath until all tubules were digested.
  • the reaction was stopped with fetal bovine serum (FBS).
  • FBS fetal bovine serum
  • MEFs mouse embryonic fibroblast feeders
  • MEFs were made by standard procedures using 12.5 dpc CD-1 mouse embryos. The embryos were eviscerated before trypsinization, and the dissociated cells were plated onto 150-mm plates with plating density at approximately 1.5 embryos per plate. After the initial plating, MEFs were split 1:5 and then frozen (passage 1). Thawed MEFs (P1) were passed only once for expansion purposes prior to mitomycin C treatment. MEF feeders were plated in a density of 50-60 ⁇ 10 3 per cm 2 . New MEF feeders were used for pluripotent germ cell culture every 7-10 days. All the animal experiments followed the guide lines for the care and use of laboratory animals (National Research Council).
  • telomerase activity was detected by PCR-based assay using TRAPEZE Detection Kit (Chemicon).
  • telomere extension Two microliters of cell extract at 750 ⁇ g/ ⁇ l was added to a total volume of 50 ⁇ l PCR reaction mix containing TRAP Reaction Buffer, dNTPs, substrate oligonucleotide, telomerase primer, internal standard primer, and Taq polymerase.
  • positive control 2 ⁇ l of mESC cell extract was added to the reaction mix, and CHAPS lysis buffer alone and heat inactivated telomerase were used as negative control for each experimental sample. Each sample was incubated at 30° C. for 30 min for telomerase extension, followed by PCR amplification.
  • proliferating cells were incubated in culture with 0.1 ⁇ g/ml KaryoMAX Colcemid (Invitrogen) for 3-4 hr before they were re-suspended in hypotonic solution (0.075M KCL) and incubated at room temperature for 10 min. Cells were then resuspended in cold fixative (3:1 methanol: acetic acid) and stored at 4° C. for at least 30 min. Following washing with fixative, cells were applied to clean glass slides and air dried. Metaphase chromosomes were prepared and karyotypes created using an Applied Spectral Imaging Band View digital imaging system.
  • the EBs were cultured in PM medium for four days before they were cultured in the serum-free N1 medium for lineage selection: i.e., DMEM/F12 (Invitrogen) supplemented with ITS (insulin, 10 mg/l; transferrin, 5.5 mg/l; selenium, 0.67 mg/l) and fibronectin (50 ⁇ g/ml).
  • DMEM/F12 Invitrogen
  • ITS insulin, 10 mg/l
  • transferrin 5.5 mg/l
  • selenium 0.67 mg/l
  • fibronectin 50 ⁇ g/ml
  • N1-treated cell aggregates were transferred to gelatin-coated culture plates in N2 medium for expansion of neural progenitor cells (N1 medium with ITS, without fibronectin and supplemented with 10 ng/ml bFGF).
  • EBs were cultured for two weeks in the presence of different cardiogenic compounds including 0.06 M DMSO, 5 mM 5′-aza-2′-deoxy-cytidine (AZA) and 25-50 ⁇ M cardiogenol-C.
  • AZA 5 mM 5′-aza-2′-deoxy-cytidine
  • cardiogenol-C 25-50 ⁇ M cardiogenol-C.
  • the morphology of cells was analyzed and samples were taken both for gene expression analysis by RT-PCR and immunohistochemical staining.
  • Chondrocyte differentiation of mGSCs was induced by adding a chondrogenic induction medium (Chondrogenic SingleQuots, Cambrex) supplemented with 10 ng/ml TGF-3 ⁇ and 20% FBS.
  • Immunocytochemical (ICC) and immunohistochemical (IHC) staining Cultured cells were fixed in 4% paraformaldehyde for 10-30 min at room temperature and stored in PBS at 4° C. For fluorescent immunocytochemistry, cells were permeablized with 1 ⁇ Cytoperm (BD Biosciences) or 0.2% Triton X-100 for 15 min and subsequently incubated in 2% (w/v) bovine serum albumin (BSA), 2% (v/v) normal goat serum (GS)/ 1x Cytoperm-PBS for 30-60 min both at room temperature.
  • BSA bovine serum albumin
  • GS normal goat serum
  • RT-PCR cDNA was synthesized with the Sensiscript RT Kit, and PCR was performed with HotStarTaq DNA Polymerase. All PCR reactions began with an initial incubation at 95° C. for 15 min to activate the enzyme. This was followed by 35 cycles of 95° C. for 15 sec, the appropriate annealing temperature for 1 min and 72° C. for 1 min, which was then followed by 1 cycle of 72° C. for 10 min for final extension.
  • Reactions were carried out using an iCyclerTM Thermal Cycler (Bio-Rad). The procedure for RT-PCR was carried out using specific primers including, Oct-4, Nanog, Rex-1, DPPa5, Dazl, ⁇ actin, Nkx2.5, Nestin, Mab2, and GFAP.
  • specific primers including, Oct-4, Nanog, Rex-1, DPPa5, Dazl, ⁇ actin, Nkx2.5, Nestin, Mab2, and GFAP.
  • GADPH was used as a house keeping gene for cellular samples and ⁇ -actin or interleukin-2 (IL-2) was used in mouse embryos.
  • Imprinting patterns in mGSCs and mESCs were determined by a PCR-based analysis. PCR amplification of each dimethylated region (DMR) from bisulfite-treated DNAs was carried out by specific primers. For analysis of the imprinted genes the UVP image software was used to quantify the band intensity. For GFP and LacZ amplification, individual tissue from chimeric embryos were carefully collected by dissection, minced into small pieces, and placed in DNA extraction buffer (DNeasy kit) for DNA isolation and purification according to the manufacturer's protocol.
  • DMR dimethylated region
  • UVP image software was used to quantify the band intensity.
  • GFP and LacZ amplification individual tissue from chimeric embryos were carefully collected by dissection, minced into small pieces, and placed in DNA extraction buffer (DNeasy kit) for DNA isolation and purification according to the manufacturer's protocol.
  • mice To test the functionality of mGCs for regeneration of spermatogenesis, spermatogonial stem cell transplantation was used. Twenty 6-8 weeks immune deficient nude male mice (Harlan) were treated with busulfan (40 mg/kg) and used as recipients. One month after busulfan treatment, 2 ⁇ 10 5 cells were transplanted into the seminiferous tubules via rete testis injection. Four mice received mGCs (GFP sorted cells). Four other mice were injected with freshly isolated GFP+ sorted cells. Four mice were transplanted with freshly isolated GFP+/c-Kit+ sorted cells, and four mice were injected with freshly isolated GFP+/c-Kit ⁇ sorted cells.
  • mice served as sham control and were not injected.
  • One month after transplantation the animals were sacrificed and testes were harvested and used for histological evaluations. To evaluate the efficiency of transplantation, total number of tubular cross sections with spermatogenesis was counted.
  • ES cells were injected in some mice.
  • approximately 1 ⁇ 10 6 cells were injected.
  • Mice were sacrificed six weeks later, and tissues were harvested for morphological and histological analysis.
  • mice Oct-4+/GFP+/LacZ GSCs The ability of mouse Oct-4+/GFP+/LacZ GSCs to form chimeric cell populations was determined after injection into host blastocysts, or by their aggregation with morula-stage embryos or eight-cell stage embryos. Blastocyst injections of 15-20 cells were carried out using day-3.5 blastocysts collected from CD-1 mice. After injection, blastocysts were transferred (7-8 blastocysts in each horn of the uterus) into 2.5-day pseudopregnant CD-1 females, previously mated with vasectomized males.
  • lacZ cells were examined in different areas of the chimeric 12.5 dpc embryos by the ⁇ -galactocidase staining kit (Sigma).
  • lacZ and GFP PCR were performed in DNAs isolated from the brain, heart, liver and gonadal ridges of the chimeric embryos formed from Oct-4-GFP/LacZ cells.
  • Spermatogenesis is a highly regulated process in which undifferentiated germ cells classified as spermatogonial stem cells (SSC) divide and mature to produce spermatozoa.
  • SSC spermatogonial stem cells
  • a s (A single ) spermatogonia are considered to be the resident stem cells responsible for spermatogenesis as they are capable of both self-renewal and differentiation.
  • histological studies of primate and human cells demonstrate two different distinct types of nuclear staining resident on the basement membrane of the testicular seminiferous tubular epithelium, i.e., designated as A dark and A pale spermatogonia.
  • Rhesus monkey testes were used for characterization of primate germline stem cells. Immunohistochemical examination, surface markers and fluorescence activated cell sorting were used to identify, characterize and substantially purify germline stem cells from adult Rhesus monkey testes. The presence of germline stem cells in each cell population was confirmed using telomerase, RT-PCR and immunohistochemical staining with the germ specific GFP/LacZ marker VASA and SSC-specific marker GFR- ⁇ 1. Spermatogonial transplantation was used to define the functional capacities of cell populations before and after enrichment.
  • Antibodies specific for ⁇ 6-integrin stained cells located adjacent to the basement membrane in the seminiferous tubules, as well as the seminiferous tubular basement membrane ( FIG. 18 ). An average of thirteen ⁇ 6-integrin+ cells was found in each seminiferous tubule histologic cross section. Within seminiferous tubular sections the majority of ⁇ 6-integrin+ cells were also VASA+, confirming their germline stem cell status. Quantitatively, there were more ⁇ 6-integrin+ cells per tubular cross section than GFR- ⁇ 1 and co-localization studies showed that about 60% of ⁇ 6-integrin+ cells were also GFR- ⁇ 1+.
  • the GFR- ⁇ -1 cell surface marker was specifically expressed in cells located at the basement membrane of the seminiferous tubules. All of the GFR- ⁇ -1+ cells were also positive for germ cell specific marker VASA.
  • SSEA-4+ cells were located at the basement membrane of adult primate seminiferous tubules and these cells also were positive for VASA staining. The majority of SSEA-4+ cells were also ⁇ 6-integrin+. There was also significant co-localization between SSEA-4 and GFR- ⁇ 1 showing that SSEA-4 is an important cell surface marker for germline stem cells. About 40% of spermatogonial cells at the basement membrane of the seminiferous tubule in primate testes histologic cross-sections expressed SSEA-4.
  • Nanog was expressed in abundant in primate testes. Nanog appeared as a nuclear staining and was colocalized with VASA in almost all germ cells in seminiferous tubules. Nanog expression was stronger in advanced germ cells located at the lumen of seminiferous tubules compared to undifferentiated germ cells located at the basement membrane. Co-localization study of Nanog and GFR- ⁇ 1 showed that all the germline stem cells showed a low level of Nanog expression.
  • CD90 antibodies stained only the basement membrane and did not stain any cellular structure in the testes.
  • the immunohistochemical characterization of primate testicular samples showed that germline stem cells in the adult primate testes are positive for ⁇ 6-integrin, SSEA-4 and GFR- ⁇ 1 and are negative for c-Kit.
  • collagenase A (1 mg/mL)
  • DNase 10 U/mL
  • the undigested tissue was further digested in an enzyme cocktail consisting of 1.5 mg/mL collagenase A, 1.5 mg/mL hyaluronidase Type V (Sigma), 0.5 mg/mL trypsin (Worthington Biochemical Corporation), and 10 units/mL DNAse in DMEM in a reciprocating 37° C. water bath for 20 min.
  • Digested and undigested tissue was passed through a 70 ⁇ m strainer into FBS (fetal bovine serum; Hyclone) to inactivate enzymes. After centrifugation at 400 ⁇ g for 10 min, the cell pellets were resuspended in DMEM+10% FBS and placed in tissue culture coated 15 cm dishes in a 5% CO 2 /95% air humidified incubator.
  • FBS fetal bovine serum
  • Flow cytometry was used to identify cell surface markers specific for adult primate testicular germline stem cells ( FIG. 20 ). Contrary to reports from other investigators with non-primate SSCs, freshly isolated adult primate testicular cells did not express epithelial cell adhesion/activating molecule (EpCAM). However, germline stem cells were identified as a very small portion of the total adult primate testicular cell population, less than 1% of the total testicular cell isolate, by virtue of expression on their cell surface of the GDNF receptor GFR- ⁇ 1. Similarly, contrary to experience with murine testicular germline stem cells (Example 1) freshly isolated adult primate germline stem cells did not express c-Kit.
  • EpCAM epithelial cell adhesion/activating molecule
  • adult primate germline stem cells (about 2% of the isolated testicular cell population), expressed cell surface carbohydrate determinants bound by Dolichos biflourus agglutinin (DBA), a lectin.
  • DBA Dolichos biflourus agglutinin
  • the adult primate germline stem cells expressed the CD9, CD90 and CD49f cell surface markers ( FIG. 21 ).
  • c-Kit was gated as the negative/parent sorting window against which were plotted both CD90+ and CD49f+ to identify the double positive CD90+/CD49f+ cells. Sorting for double positive cells resulted in isolation of germline stem cells, present as about 5.77% of total cells in the adult primate testicular isolates. The latter CD90+/CD49f+ double positive cells were collected for further use. Additional purification was achieved by selecting for c-Kit ⁇ cells that were positive for SSEA4, resulting in isolation of a second substantially purified cell population that constituted about 2% of the total adult primate testicular cells.
  • a yet additional purification was achieved by selecting for c-Kit ⁇ cells that were positive for all of CD90, CD49f and SSEA4, resulting in isolation of a third substantially purified cell population that constituted about 1.47% of the total adult primate testicular cells.
  • testicular cells were sorted as follows: (i) ⁇ 6-integrin+; (ii) CD-90+; (iii) CD-90+/ ⁇ 6-integrin+/c-Kit ⁇ (triple sort); and (iv) SSEA-4+ cells.
  • the different isolated and purified testicular cell populations were tested for the presence of germ cell marker VASA and SSC marker GFR- ⁇ 1 ( FIG. 22 ).
  • Non-sorted cells contained about 70% VASA+ cells, but only 10% of these cells stained positive for GFR- ⁇ 1.
  • Sorting for just ⁇ 6-integrin resulted in a significant increase in cells with both germline and SSC markers, i.e., populations with 42.6% VASA+ and GFR- ⁇ 1+ cells.
  • Sorting for CD-90 alone or in combination with c-Kit ⁇ also significantly increased the proportion of VASA+/GFR- ⁇ 1+ cells to 30% and 46.4% respectively.
  • Sorting for SSEA4+ alone also resulted in an enrichment for cells expressing germline and SSC markers, i.e., sorted cell populations in which 37.5% of the cells were VASA+ and GFR ⁇ -1+.
  • the functional properties of different primate testicular cell populations were determined before and after substantial purification by testing for their ability to repopulate the basement membrane of seminiferous tubules in the testes of immunodefficient nude mice treated with the chemotherapeutic drug busulfan.
  • busulfan 40 mg/kg.
  • 2 ⁇ 10 5 adult primate testicular cells were transplanted into the seminiferous tubules via rete testis injection.
  • mice received a transplant consisting of freshly isolated non-sorted cells; three mice received a transplant consisting of freshly isolated c-Kit ⁇ /SSEA-4+ sorted cells; and, three mice received a transplant consisting of freshly isolated c-Kit ⁇ /SSEA-4 ⁇ sorted cells.
  • CSFE carboxyfluorescein diacetate succinimidyl ester
  • mice Two weeks after transplantation, mice were sacrificed and the number of CSFE+ cell colonies was determined microscopically in histologic sections of the mouse testes ( FIG. 23 ). Theoretically, if germline stem cells have a cell cycle time of about 72 hr, at two weeks post-transplantation the cells should have undergone 2-3 cell doublings, resulting in colonies of about 4-8 cells. For statistical analysis the ANOVA test was applied and p ⁇ 0.05 was considered significant.
  • the DNA content of the two populations was investigated using flow cytometry ( FIG. 25 ).
  • This analysis showed that the SSEA-4+ cell population had a cellular DNA content resembling that of cells in G0-G1 stage of the cell cycle.
  • cells having the Thy-1 and ⁇ 6-integrin cell surface markers had two discrete and different DNA contents, resembling either the G0-G1 stage of the cell cycle or the S phase.
  • the data show that SSEA-4+ cell population with SSC cell surface markers represents a quiescent population of progenitor germline stem cells, while the Thy-1+ and ⁇ 6-integrin+ population of cells represents an actively dividing population of SSCs ( FIG. 27 ).
  • ⁇ 6-integrin+cells enriched by selecting ⁇ 6-integrin+cells showed a very high level of co-localization with GFR- ⁇ 1, confirming the findings using immunohistochemical methods to identify germline stem cells in testes sections.
  • Expression of ⁇ 6-integrin on primate SSC indicates that this marker is conserved among the species as mouse, marmoset and human SSC also possess this marker on their cell surface. Localization of some ⁇ 6-integrin+ cells within interstitial cells outside the tubules indicates that this marker alone can not be used for isolation of highly pure populations of SSC from adult primate testes.
  • SSCs share some but not all phenotypic and molecular characteristics with other stem cells, in particular hematopoetic stem cells.
  • the flow cytometry analysis using a variety of cell surface markers, revealed that in the adult Rhesus monkey testes, there are distinct cell populations expressing ⁇ 6-integrin and Thy-1 and the majority of cells in primate testes were c-Kit ⁇ . Immunohistochemical staining of primate testes also showed that all the cells along the basement membrane of seminiferous tubule were c-Kit ⁇ indicating that ⁇ 6-integrin+cells are c-Kit ⁇ .
  • Sorting for ⁇ 6-integrin or Thy-1 alone resulted in enrichment of SSC markers as shown by immunohistochemical staining, RT-PCR and telomerase assay.
  • sorting the ⁇ 6-integrin+, Thy-1+ and c-Kit ⁇ cells resulted to the highest expression level of SSC marker PLZF as shown by quantitative RT-PCR ( FIG. 26A ) and the most elevated telomerase activity ( FIG. 26B ), indicating that combination of these markers enrich SSC in several folds.
  • SSEA-4+ cells there were also a clear population of SSEA-4+ cells in the primate testes, which also showed a high level of telomerase activity and expressed high level of both germ and SSC markers ( FIG. 28 ).
  • Immunohistochemical staining showed that SSEA-4+ cells also located at the basement membrane of seminiferous tubules and are highly co-localized with ⁇ 6-integrin and GFR- ⁇ 1.
  • Flow cytometric analysis showed that there are about 5-7% of ⁇ 6-integrin+, Thy-1+, c-Kit ⁇ sorted cells in adult primate testes while only 2-3% SSEA-4+ cells are present. This is also consistent with immunohistochemical data on testes sections showing that there are significantly less SSEA-4+ cells found per tubule cross section than the ⁇ 6-integrin+ cells.
  • SSEA-4 is stage specific embryonic antigen and is predominantly found in pluripotent cells like embryonic stem cells. Interestingly all the SSEA-4 ⁇ cells co-expressed germ cell marker VASA, however only a fraction of these cells co-localize with GFR- ⁇ 1 indicating that this marker expresses only on subpopulations of spermatogonial stem cells in monkey testis.
  • SSEA-4+ population of germline stem cells have different DNA contents from the Thy-1+, ⁇ 6-integrin+ cells. While SSEA-4+ cells had DNA profile similar to the actively dividing cells, Thy-1+, ⁇ 6-integrin+ cells showed an accumulated number of cells arrested in the S phase of the cell cycle. Moreover, SSEA-4+ cells showed significantly higher proliferation activity as shown by PCNA staining than the Thy-1+, ⁇ 6-integrin+ cells.
  • Pluripotent marker Nanog which has an essential role in maintaining ES cells in their undifferentiated stage was abundantly expressed in primate testes. Nanog expression in all germ cells and not only in SSC indicates a different role for this transcription factor in germline stem cells compared to ES cells. It has been shown that deletion of Nanog in germ cells induces apoptosis rather than differentiation indicating that Nanog is a survival factor for germ cells.
  • PLZF While GDNF regulates SSC self renewal through up regulation of BCL6b transcription factor, PLZF maintains SSC self renewal with a yet unknown mechanism.
  • Promyelocytic leukemia zinc factor (PLZF) is shown to inhibit cell growth at the G1/S transition and transit through S-phase by suppression of cyclin A which is available in a variety of cell types.
  • PLZF is also shown to inhibit P21 another regulator of G1/S transition.
  • RAR- ⁇ Retinoic acid receptor alfa
  • RAR- ⁇ Retinoic acid receptor alfa
  • Flow cytometry sorting was accomplished using an InFlux Cell Sorter.
  • InFlux Cell Sorter For surface characterization and sorting, cells were stained with antibody reagents specific for stem cell surface markers and spermatogonial stem cell markers in non-primate species including anti-CD90-FITC, anti-CD49f-PE, and anti-CD117-APC.
  • markers analyses cells were stained for 30 min in complete medium on ice, washed once in cold staining buffer, resuspended in complete culture medium and kept on ice until cytofluorimetric analysis.
  • Primate germ cell magnetic sorting The population of primate germ cells was enriched by tagging with magnetic microbeads and passing the cells through a magnetic column. Freshly isolated primate testicular cells were labeled with biotinylated antibodies for SSEA-4 or for ⁇ 6-integrin and Thy-1 (Ebioscience, Abcam, BD Pharrmigen, respectively). Once biotinylated, the cells were labelled with streptavidin magnetic microbeads (Miltenyi Biotec). Magnetically labeled cells were selected for by passing the cells through a column in the presence of a magnet. Magnetically labeled cells were removed from the column by removing the column from the magnet, freeing the cells to be washed off of the column.
  • This process was successful in enriching the population of cells positive for each of the markers up to 22 ⁇ the original percentage in freshly isolated cells.
  • magnetic sorting could provide a population as high as 90% purely labeled cells.
  • This enrichment process was used in conjugation with fluorescent flow cytometry. By magnetically sorting the cell isolation before performing fluorescent flow cytometry the amount of time needed to sort out fluorescently labeled cells was greatly reduced and the number of fluorescently labeled cells that could be sorted out was greatly increased.
  • Tissues were fixed overnight in 4% paraformaldehyde (PF; Electron Microscopy Science); transferred into 20% sucrose (Sigma) and frozen in OCT (VWR). Cryosections were prepared at 8 ⁇ m thickness and stored at ⁇ 80° C. Sorted cells were fixed in 4% PF, re-suspended in 100 mM sucrose at approximately 25,000 cells/10 ⁇ l; 10 ⁇ l aliquots were transferred onto ornithine/lysine-coated glass slides; and, the slides were placed on a 37° C. hot plate until dry. Slides were stored at ⁇ 80° C. until analysis.
  • PF paraformaldehyde
  • VWR OCT
  • the cells in testicular sections and in FACS sorted samples were permeabilized using 0.1% Triton-X100 and blocked in either a solution containing 2% BSA and 5% sheep serum, or alternatively, in a solution containing 2% BSA, 5% goat serum and 0.1% Triton-X100.
  • DAPI Invitrogen
  • cells were preserved using Permafluor (Beckman Coulter). Distribution of surface markers in tissue sections and sorted cells was evaluated using an Olympus BX-61 microscope fitted with SlideBookTM imaging software.
  • cDNA template 5 ng was used in a 25 ⁇ L reaction volume with Quantitect Sybr Green PCR master mix (Qiagen) and the samples were amplified using a BioRad iCycler. Each sample was assayed in triplicate and normalized to a GAPDH control.
  • telomerase assay The SYBR Green real time quantitative telomeric repeat amplification protocol (RQ-TRAP) was employed. Tissue or cells pellets were washed once in PBS, resuspended and homogenized in 1 ⁇ Chaps lysis buffer containing RNaseOut Inhibitor (Invitrogen), at a final concentration of 1000 cells/ ⁇ L and 400 units/mL of the RNaseOut Inhibitor. After 25 min of incubation on ice, the cell lysates were centrifuged at 4° C. in a microfuge at 16,000 rpm for 10 min.
  • RQ-TRAP SYBR Green real time quantitative telomeric repeat amplification protocol
  • the supernatant was transferred to a fresh microcentrifuge tube and the protein concentrations determined by measuring absorbance at 280 nm using an ND-1000 spectrophotometer (Nanodrop).
  • Telomerase reaction volumes were 25 ⁇ L in a solution containing 500 ng protein lysate, Quantitect SYBR Green PCR mix, 1 ⁇ g TS primer, 0.5 ⁇ g ACX primer and nuclease-free distilled water. Each sample was tested in triplicate along with a no template control (lysis buffer), a positive control (ESC cells), and a standard curve prepared from aliquots of human ESC lysate that contained 1000 ng, 200 ng, 40 ng, 8 ng or 1.6 ng of protein.
  • the threshold cycle values were determined from semi-log amplification plots (log increase in fluorescence versus cycle number) and compared with standard curve.
  • the software default setting for the threshold was 10 times the mean of the standard deviation of the fluorescence reading of each well over the first 10 cycles, excluding cycle 1. Telomerase activities for different primate testicular cell samples were read from the standard curve and/or expressed as a percentage of the values recorded with human ESC lysate standards.
  • Germline stem cells (Example 2) after isolation were transferred to MEF plates and cultured in different serum free media including Mouse Serum Free Medium (MSFM), Rat Serum Free Medium (RSFM) or MEM-X® media. The morphological changes of the cells and the number of germ cell colonies per well was counted during culture. Half of the medium was changed every other day.
  • MSFM Mouse Serum Free Medium
  • RSFM Rat Serum Free Medium
  • MEM-X® media MEM-X® media.
  • FIG. 29A Ten days after culture, flat colonies appeared in all media types ( FIG. 29A ). Colonies in MEM-X maintained their morphology better than other two culture media. The number of colonies found in non-sorted population was lower than sorted cells. Among the cell surface markers tested, SSEA-4 and triple stained cells resulted in colony formation. Depletion of SSEA-4+ cells from triple sorted cells resulted to very few colonies; however depletion of the triple sort phenotype from SSEA-4+ cells did not change colony formation ability. Cells positive for both SSEA-4 and triple sort formed highest number of colonies in culture and cells depleted from SSEA-4 and triple sort did not form any colony. The colonies were then stained for SSEA-4 ( FIG. 29B-C ), GFR- ⁇ ( FIG. 29D ) and ⁇ 6-integrin
  • Ovarian germ-line stem cells were substantially purified by collecting GFP-positive cells by flow cytometry identifying green fluorescent intensity ( FIG. 11A ), gating three channels for c-Kit (R2, R3 and R4) ( FIG. 11B ); and then sorting R3 for c-Kit intensity ( FIG. 11C ).
  • FIG. 11A Using flow cytometry, GFP/Oct-4+ cells were detected in neonatal ( FIG. 11A ) and adult ( FIG. 11B ) mice indicating the presence of germline stem cells in postnatal ovary. The percentage of germline stem cells in the mouse ovary significantly diminished with age. While 1-2% GFP+ cells were found in the ovaries of the neonatal mice, only 0.05% were present in the adult ovary. Among the Oct-4+ cells, 60% were negative or expressed low level of c-Kit and 40% showed high level of c-Kit expression ( FIG. 11C ), indicating the presence of two populations among germline stem cells. Immunohistochemical analysis revealed that GFP-Oct-4+ cells are present throughout the ovarian epithelium ( FIGS.
  • RT-PCR analysis showed that GFP+ cells isolated from neonatal mouse ovary expressed both pluripotent marker Oct-4 and germ cell markers VASA and c-Kit confirming the presence of germline stem cells in this population, while the GFP- cells showed only the expression of germ cell markers ( FIG. 13 ).
  • GFP+cells at the onset of culture like ESC, express Oct-4 ( FIG. 13 ).
  • the marker c-Kit has been associated with male germline stem cells in certain prior scientific reports.
  • Oct-4+ cells 60% were negative or expressed low level of c-Kit and 40% showed high level of c-Kit expression.
  • GFP+ cells isolated from neonatal mouse ovary expressed both pluripotent marker Oct-4 and germ cell markers VASA and c-Kit . The combined results confirm the presence of germline stem cells in the GFP+ cell population isolated from the ovaries of OG2 mice.
  • FIG. 14A-14C and 14 E were clearly defined boundaries
  • FIG. 14F did not. Representative of the clear-border and non-clear border colonies were picked ( FIG. 14B ) and passaged on MEFs using collagenase ( FIG. 14D ). After passage, cells assembled into distinctive colonies recognizable by a tight oval central grouping of small round cells surrounded by flat tightly packed cells having a more epitheliod shape ( FIG. 14G-14I ). This colony appearance was continued beyond passage 4 ( FIG. 14J-14K ).
  • GFP+ colonies tolerated enzymatic digestion using collagenase and generated new colonies. However, they did not tolerate trypsinization and the majority of the colonies differentiated after trypsin treatment. GFP ⁇ cells showed only the expression of germ cell markers and not stem cell markers. After several passages (for example, passage 15) these differentiated colonies retained their morphology, but most cells no longer expressed GFP, suggesting down-regulation of the Oct-4 promoter and possible differentiation. Only a few cells in each colony, mainly large cells in the center of the colony, showed GFP expression. With time, these GFP+ cells appeared to form very large, up to 40 ⁇ m, oval cells that were resident in structures having morphologic similarity to ovarian follicles ( FIG. 16A , 16 B).
  • GFP+ cells were cultured on MEF feeders in PM-1TM medium in a concentration of 5000-10000 per well of a 4-well plate. Culture was maintained at 37° C. and half of the medium was changed every other day. Every two weeks cells were transferred either mechanically or enzymatically (collagenase) to a new MEF plate.
  • ovarian germ-line stem cells Freshly isolated GFP-positive cells were used for telomerase assay and gene expression profiling. For ovarian histology, ovaries were fixed in 4% paraformaldehyde (PFA) in 1M sucrose overnight at 4° C. and mounted in cryostat freezing medium. Five micron sections were prepared and localization of the GFP+ cells was determined using fluorescent microscopy. Localization of germline stem cells in the ovary was confirmed by Oct-4 and VASA double labeling. For immunocytochemistry (ICC), cultured ovarian germ-line stem cells were fixed in 2% PFA for 30 min at room temperature, washed in PBS and kept at 4° C. To characterize cultured ovarian germ-line stem cells, VASA, Oct-4, Nanog and alkaline phosphatase staining was performed using bright field ICC, as described further below.
  • PFA paraformaldehyde
  • ICC immunocytochemistry
  • cDNA template 5 ng was used in a 25 ⁇ L reaction volume with Quantitect Sybr Green PCR master mix and the reaction mixtures were amplified using a BioRad iCycler. Each sample was assayed in triplicate and normalized to a GAPDH control.
  • testes collected as testicular biopsies from patients with non-obstructive azospermia or remnant of testes tissue collected after orchiectomy were used for this study. All the tissues were donated with the informed consent of the patients. Tissues were transferred in PBS-antibiotics at 4° C. within 24 hr of collection. The procedure of processing human testicular tissue is similar to that for primate as disclosed in Example 2.
  • a tissue sample was taken for ICC, and two small pieces of testes were taken for RNA and DNA extractions. Following cell isolation and determination of viability and cell number, samples were taken for RNA and DNA analysis. Methods for ICC, RNA and DNA extractions are similar to the primate as disclosed in Example 2. In addition, cells were labeled for expression of cell surface markers previously developed for separation of primate germline stem cells were used by magnetic cell sorting and flow cytometry. Antibodies and methods used for flow cytometry are similar to that used for separation of primate germline stem cells as disclosed in Example 4.
  • the population of germ cells was also enriched by tagging with magnetic microbeads and passing the cells through a magnetic column.
  • Freshly isolated testicular cells were labeled with biotinylated antibodies for SSEA-4 or for ⁇ 6-integrin and Thy-1. Once biotinylated, the cells were labeled with streptavidin magnetic microbeads. Magnetically-labeled cells were selected for by passing the cells through a column in the presence of a magnet.
  • Magnetically-labeled cells were removed from the column by removing the column from the magnet and freeing the cells to be washed off of the column. This process was successful in enriching the population of cells positive for each of the markers up to 22 ⁇ he original percentage in freshly isolated cells. In addition, magnetic sorting could provide a population as high as 90% purely labeled cells. This enrichment process was used in conjugation with fluorescent flow cytometry. By magnetically sorting the cells before performing fluorescent flow cytometry, the amount of time needed to sort fluorescently-labeled cells was greatly reduced and the number of fluorescently-labeled cells that could be sorted was greatly increased.
  • Sorted cells were then used for RT-PCR and DNA analysis. Also, some samples cells were subjected to a spermatogonial transplantation assay using immunodefficient mice as recipients. The techniques for spermatogonial stem cell transplantation are similar to the mouse and primate as disclosed in Examples 1 and 2.
  • THT whole human testes tissue
  • SSEA-4 and VASA THT stained for GFR- ⁇ and VASA
  • THT stained for VASA and Nanog FIG. 32
  • human bHT testes biopsy tissue
  • SSEA-4 and ⁇ 6-integrin FIG. 33
  • Negative controls consisted of human testis sections stained only with secondary antibody ( FIG. 34 ).
  • THT SSEA-4+ magnetic bead sorted cells were transplanted into busulfan-treated recipient mouse testes and after one month were sectioned and stained for the following markers: SSEA-4 and human nuclear protein (HNP, FIG. 35 ); ⁇ 6-integrin and HNP ( FIG. 36 ); SSEA-4 and ⁇ 6-integrin ( FIG. 37 ).
  • Negative controls consisted of THT transplanted cells in mouse testis sections stained only with secondary antibody ( FIG. 38 ). All stains contain a general nuclear dye.
  • Flow cytometry analysis confirmed immunohistochemical observation and positive populations for ⁇ 6-integrin were found in samples collected from human testes. In addition, a distinct population of Thy-1+ cells was found. Co-localization of Thy-1 and ⁇ 6-integrin showed that there are three subpopulations of Thy-1+ cells within human testes: 1) ⁇ Thy-1 medium and ⁇ 6-integrin low, 2) a Thy-1 high and ⁇ 6-integrin medium, and 3) a Thy-1 high and ⁇ 6-integrin negative. Most of the ⁇ 6-integrin+ cells were Thy-1 ⁇ . There were also clear population of SSEA-4+ (10-12%) and GFR- ⁇ + (1-5%) cells found in human testes. Magnetic sorting significantly enhanced the percentage of SSEA-4+ cells to 44% indicating a 4 fold increase for this marker.
  • Quantitative RT-PCR analysis revealed that among the samples tested SSEA-4+ cells and GFR- ⁇ + cells express highest levels of spermatogonial stem cell markers including C-RET, PLZF, and TERT and germ cell markers including VASA and DAZL. Telomerase activity is indicative of spermatogonial stem cells.
  • Spermatogonial stem cell transplantation revealed that SSEA-4+ cells colonize testes of recipient mice and repopulate, indicating that these cells are functional spermatogonial stem cells.
  • FIG. 54 depicts the average growth of male ( FIG. 54A ) and female ( FIG. 54B ) F1 mice created through breeding of mice derived from male GSC transplantation. These growth rates are similar to the growth rates of the parental mice as provided by the vendor (data not shown).
  • Human testicular cells were enriched for spermatogonial stem cells (SSCs) by magnetic sorting for SSEA-4 and the enriched population was microinjected into the testes of a recipient mouse. One month after transplantation, testes were harvested and cryosections were made. The identity of human cells in the mouse testes was determined using human nuclear protein (HNP) antibody conjugated to alexa-488. Co-localization of HNP with germ cell, somatic cells, stem cells and pluripotent markers (Table 3) were used to assess the phenotypic characteristics of human SSC in the mouse testes.
  • HNP human nuclear protein
  • Extensive colonization of human cells as detected by HNP staining in the mouse testes indicates the presence of highly enriched population of SSC in the SSEA-4 magnetic sorted cells. All the human cells colonized the mouse testes were positively stained for germ cell marker VASA and negatively stained for LHR, a marker for testicular sertoli and leydig cells. This indicates that all the colonized cells are germ cells. Among the markers used in this study only 15% of the human cells expressed SSEA-4, 31% expressed ⁇ 6-integrin and 45% of them expressed GPR125 on their surface. Almost all the human cells colonized mouse testes expressed c-Kit indicating that this marker is needed for SSC self renewal.
  • Luteinizing Hormone Receptor does not co-localize with monkey, human, or mouse germ cells as indicated by VASA, SSEA4, GFR ⁇ , and ⁇ 6-integrin co-localization studies. LHR is expressed on Sertoli and Leydig cells as indicated by the location and morphology of those positive cells. Connexin-43 appears to stain both germ cells located at the basement membrane, Sertoli cells, and possibly Leydig cells.
  • testicular tissue containing spermatogonial stem cells are obtained as in Example 6. Either isolated cells or testicular tissue are cryopreserved. Additionally, small segments of seminiferous tubules are preserved for use in ex host maturation.
  • the cells or tissues are cryopreserved in a solution comprising at least one cryoprotectant including, but not limited to, dimethyl sulfoxide (DMSO), ethylene glycol, glycerol, and propanediol; at least one culture medium including but not limited to, DMEM, MEM and proprietary media disclosed above; at least one additional agent including, but not limited to, sucrose, dextran, a serum substitute and HEPES buffer.
  • the solution comprises CryoStorTM CS-10 media (BioLife Solutions Inc.).
  • the cells or tissues are frozen by either conventional methods or at a controlled rate.
  • cryopreserved germline stem cells are suspended in 1.5 mL of DMEM with a DMSO concentration of 10%. The cell suspension is then diluted to 50 mL with media to bring the DMSO concentration to 0.3%. The cells are then centrifuged, the media aspirated, and the cells resuspended in media to bring the residual DMSO concentration to no more than 0.012%.
  • testes In order to determine whether testes should be cryopreserved in cell suspension or tissue form, 2-5 day old prepubertal mouse testes were obtained and some testes were cryopreserved as tissue and some was dissociated to a single cell suspension prior to cryopreservation. A controlled rate freezing protocol was used in both instances. After thaw, the total cell number recovered, the viability and cell surface markers were analyzed to determine the proportion of germ cells, somatic cells and germline stem cells in each suspension. The results indicate that cryopreservation of cells are better than tissue. While cryopreserved mouse testes cells are about 95% viable, cells isolated from frozen tissue are about 80% viable ( FIG. 48 ).
  • testes from adult human were also studied. Testes sample from young adult men (two samples) undergoing transsexual surgery were used for banking. Also testes biopsies (three samples) donated from a fertility clinic were used for tissue and cell banking. Testes cells demonstrated high viability after thaw using controlled rate freezing and viability was not affected by cell density or different transport media but viability was reduced after 72 hr of transportation ( FIGS. 50A and B).
  • testes cells are better maintained after cryopreservation using manual freezing or by slow freezing in a controlled environment. 2-5 days old prepubertal mouse and 3 months old prepubertal porcine testes were used for this experiment and some cells were frozen manually and some with a controlled rate freezing protocol. After thaw, the total cell number recovered, the viability and cell surface markers were analyzed to determine the proportion of germ cells, somatic cells and germline stem cells in each suspension. Cryopreservation of murine cells under controlled freezing protocols produce cells with similar characteristics to manual freezing (Table 4).
  • ⁇ 1 mm ⁇ 1 mm
  • DMSO ethylene glycol
  • SS serum substitute
  • Vitrification will be conducted according to the following protocol: (1) equilibrate in 7.5% EG and 7.5% DMSO in 20% SS for 25 min, (2) equilibrate in 20% EG and 20% DMSO in 0.5M sucrose for 15 min or until tissue sinks, and (3) place tissue on metal strip and submerged directly into liquid nitrogen.
  • the metal strips are immersed directly in 1M sucrose at 37° C. for at least 1 min and then are transferred into 0.5M sucrose for 5 min at room temperature. The thawed tissue is then washed twice in the base medium without cryoprotectant.
  • Ovarian tissue from prepubescent and adult monkeys is used in this study. Upon arrival ovaries are dissected and cortex is separated from the medulla. Ovarian cortex is then cut into small pieces (2 ⁇ 2 mm). Half of the pieces are used for tissue freezing and the other half are subjected to tissue chopper and enzymatic digestion for isolation of follicles and cells. Different cryoprotectants and freezing protocols are used. The viability and survival of cells and follicles frozen under different conditions is analyzed after thaw by flow cytometry and for apoptosis.
  • Germ cell transplantation represents a functional reconstitution assay for male germline stem cells and as such has vastly increased our ability to study the biology of stem cells in the testis and define phenotypes of infertility.
  • rodents the technique has now been used in a number of animal species, including domestic mammals, chicken and fish.
  • the optimal transplantation number of GFP germ cells were determine for transplantation into busulfan treated mice.
  • the optimum transplantation cell number of GFP germ cells is determined by mating of the transplanted GFP germ cell nude male mouse with a nude female mouse.
  • the GFP+offspring from those matings will determine if the transplanted GFP germ cells restored fertility and produced viable GFP offspring.
  • the GFP offspring are then used to validate the GFP germ cell transplantion assay by comparing the parameters described below with the total cells transplanted and/or other parameters (marker expression, testes weight, etc.). After GFP pups are born, those GFP offspring from the transplanted GFP germ cell sire are used to verify fertility and germline transmission of GFP germ cells into the next progeny by mating again with nude mice.
  • Each individual GFP+ transplanted male mouse is mated with 4 nude female mice. Two females are mated with each male every night. The four females are cycled every night depending on their estrus cycle. The males will mate for three weeks, have a one week break, and begin another three weeks of mating if the females have not formed a vaginal plug. These procedures will give the females the optimum opportunity to become pregnant, while the break periods will allow the male to recover from continuous matings. Each morning, the mice are checked for vaginal plugs as a positive indicator of successful mating. This mating procedure is continued for 5-7 months post transplantation. If no females become pregnant, matings will continue for an additional 2 months.
  • the matings stop and testes from the GFP transplanted mice are analyzed by flow analysis, sperm analysis for GFP, and histology to determine the number of GFP+ and SSC+ marker expressed in the testis.
  • the F1 progeny are also assayed for GFP using fluorescence and PCR. As nude mice do not have fur, any pup with fur should have been generated from the GFP positive sperm.
  • F1 progeny In order to assess the fertility and germline transmission of GFP or coat color from the F1 progeny, they are allowed to mature to the age of 2 months and then mated with 2 nude mice (male or female) for two months to ensure pregnancies. After the F1 mice produce F2 progeny, the F2 GFP+ and/or coat color+ mice will be screened for GFP with fluorescence and PCR. This will verify trans-generational germline transmission of the GFP+ cells. F2 progeny which prove to be GFP+ are then sacrificed and the reproductive organs collected for histology to verify complete spermatogenesis with GFP+ sperm.
  • mice used in the potency assay were kept mating for 5 months and some of them have proven to be fertile and some did not induce any pregnancies and were thus scored as infertile (by means of natural mating; these mice may have had low sperm count). These animals were sacrificed and testes were surgically removed and sperm was collected and frozen. Frozen sperm were later used for separation of GFP+ sperm by flow cytometry to be used for intra cytoplasmic sperm injection (ICSI). Testes were first weighted and then analyzed under a dissection microscope equipped with fluorescent light suitable for GFP and CFP detection. After gross examination testes were fixed in 4% PFA and frozen sections were made for histological examination.
  • ICSI intra cytoplasmic sperm injection
  • Testes of the transplanted animals were significantly heavier than the non transplanted control group ( FIG. 39 ).
  • testes weight increased significantly after to transplantation Histological and gross examination both showed the extensive repopulation of GFP+ cells in all the transplanted animals. While GFP score based on gross examination showed a positive correlation with fertility, the percentage of GFP+ tubules showed a reverse correlation. There is a positive correlation between the number of tubules with full spermatogenesis and fertility and a negative correlation between the number of empty tubules and fertility.
  • testes weight, GFP score, and testes histology are reliable indicators of fertility and can be used in future for determination of the predicted success of transplantation.
  • mice In male mice that have been sterilized by busulfan treatment, 77.78% of the mice can have their fertility restored by testicular cell transplantation surgery. A range of approximately 101,563 to 1,000,000 cells transplanted into each testis results in restoration of fertility. Also, the range of GFR- ⁇ + injected cells that can result in fertility restoration is 41 to 6,675, and the range of c-Kit ⁇ / ⁇ 6-integrin+ injected cells that can restore fertility is 22,648 to 166,500. SSEA-1+ cells do not appear to be present in pre-pubertal mice and therefore did not correlate to the fertility after transplantation, but the same marker for the human (SSEA-4) may be crucial for fertility restoration. Also, Thy-1 and Triple Stained (Thy-1+, c-Kit ⁇ , ⁇ 6-integrin+) cells did not correlate with fertility restoration in the mouse, but may be important in the human.
  • the objective of this project is to find out whether ovarian tissue is more successfully transplanted in tissue or cell suspension form.
  • Mice were ovariectomized and some received grafts of fresh ovaries, some received grafts of frozen/thawed ovaries, some received a graft of a clot containing fresh ovarian cells and follicles, some received a graft of a clot containing frozen/thawed ovarian cells and follicles, and some remain intact as control for normal breeding.
  • Vascular Endothelial Growth Factor (VEGF) was added to the clots to promote vascularization and angiogenesis of the grafted clots.
  • testis In larger mammals, however the anatomy and size of the testis is different. First, the rete testis is located in the testis in the close vicinity of testicular blood supply. Secondly, there are multiple efferent ducti connecting the rete testis to epidydimis. Finally the size of testes requires higher volume of cell suspension to fill in the testes (milliliter rather than microliter volumes). In bovine and monkey, ultrasound guided methods have been developed for transplantation of SSCs into the testes by injection of a large needle into the testes lumen. However these protocols are inefficient and invasive as in both methods the injection in some cases results damage to the rete testis and hemorrhage.
  • a microinjection device for human testes allowing access from outside to human testis segments, including the epidydimis, and having the ability to maneuver into individual efferent ductus allowing access to rete testes without damaging the arteries and veins.
  • This device consists of two parts: An endoscopic capillary catheter, which has a light source and a camera allowing the operator to guide the catheter, and an internal narrower catheter that passes through the ductus efferent and transfers the cell suspension into the rete testes.
  • the catheter is inserted into the epidydimis via a small incision and is guided to the efferent dustus.
  • an ultrasound contrast solution Prior to cell injection, an ultrasound contrast solution (Levovist) is injected and the flow of the solution is monitored by an ultrasound device to ensure that the solution is passing through the rete testis into the seminiferous tubules.
  • This device is non-invasiveness and reliable access to human testicular lumen.
  • This device can also be used for diagnostic purposes of male infertility, for example finding the exact location of obstructive azospermia.
  • the device can also be used to collect cells and tissue from epidydimis and rete testes in a less invasive manner.
  • Prepubescent patients are at a greater risk of losing their fertility since they have not completed spermatogenesis.
  • Their seminiferous epithelium contains only Sertoli cells and different types of spermatogonia, among which are the stem cells. Because of the absence of mature gametes, cryo-preservation of immature tissue is currently the only means by which fertility may be preserved in young boys.
  • Testis cell transplantations have been performed using donor testis from a wide variety of animals, mostly using immunocompromised mice as the recipient.
  • the animals that have been used as donors in experiments where the recipient was mice includes; mice, rats, hamsters, rabbits and dogs, cattle, monkeys, and humans. Progeny derived from the donor testis cells has only been shown in mice and rats.
  • germline stem cells can be auto-transplanted into the patient after chemotherapy and/or radiation therapy.
  • autotransplantation of germline cells from cancer patients poses the risk of transmission of malignant cells. Therefore, germline cells should be completely isolated from malignant cells.
  • a method, based on flow cytometry sorting, that differentially selects for germline stem cells and to purify them from cancer cells that differentially selects for germline stem cells and to purify them from cancer cells.
  • the results of the assay will allow decisions about the amount of stem cells needed to restore fertility. It is further used to evaluate stability, viability and potency of germline stem cells at the time of tissue collection and before release.
  • a mouse model is used since surface markers for spermatogonial stem cells in this species are well characterized and a transplantation technique is available. This transplantation technique will test functionality of germline stem cells in the busulfan-treated immunodeficient mouse testes allowing complete progression of spermatogenesis of the donor cells in the recipient animal.
  • LHR Leutinizing hormone receptor
  • the objective of this study is to enrich germline stem cells while removing any tumor cells from a patient sample, a method is developed that differentially selects and isolates germline stem cells from a heterogeneous cell population; to quantify the selection process to a point where contaminating cancer cells have been depleted enough for clinical application. This includes the evaluation of how many cancer cells are required to initiate tumor growth.
  • a method for disease specific immunophenotyping will be developed. To do this the specific cell surface marker expression for a specific cancer type will be assessed and used in the depletion of cancer cells from a patient sample.
  • the main concern of this procedure is the isolation of germline stem cells.
  • a procedure for the negative selection of germ cells from leukemic mice by flow cytometry sorting has been established with antibodies against two surface markers expressed in blood cancer cells including MHC class I and common leukocyte antigen (CD45).
  • MHC class I MHC class I
  • CD45 common leukocyte antigen
  • This procedure leads to successful transplantation of mouse germline cells into recipient testes without transmission of leukemia in mice.
  • human germ cells are positively selected with specific markers for germ cells such as CD90, CD49f, SSEA-4 and GFR- ⁇ .
  • other indicators for cancer can be employed such as DNA ploidy detection.
  • the goal of this model is to restore fertility without reintroducing cancer into a patient.
  • the experiments will determine the feasibility of a positive or negative selection for germline stem cells, a threshold when a tumor can be re-transplanted in a mouse model and a disease specific surface marker expression topology.
  • germline stem cell populations To reach conclusions about the ability of different germline stem cell populations to restore fertility in busulfan-treated testis, a potency assay is needed. Each of the populations was compared to a negative control to determine the increase in efficency. This also allows conclusions about the necessity of co-transplanted cell types such as Leydig cells, Sertoli cells or myoid cells. Increasing amounts of germline stem cells should yield a higher efficiency of transplanted cells and restoration of fertility. To be able to unambiguously recognize donor cells from the recipient, germline stem cells are isolated from transgenic GFP mice (NAGY, Jackson Labs) testes. To mimic prepubescent human patients, juvenile male mice between 7-10 days were used for collection of germline stem cells.
  • mice were used for mating 8 weeks post transplantation. Each male mated with two nude females. The generated offspring were assayed for their GFP expression or fur color to demonstrate what the originating strain was. The offspring from each transplanted mouse were observed for eventual abnormalities. Results are depicted in Table 9.
  • OTT Ovarian tissue transplantation
  • POF premature ovarian failure
  • OF ovarian function
  • Another proposed indication is to prolong the reproductive life in otherwise healthy women.
  • the potential uses proposed for harvested ovarian tissue are: in vivo/in vitro maturation of primordial follicles, xenografting of ovarian tissue, or using a novel method to subsequently differentiate germ cells while using blood plasma clots and grafting them with ovarian tissue for maturation and development of oocytes.
  • mice To develop a method of differentiating female mouse OG2 germ cells into follicles and/or oocytes by engrafting mouse OG2 germ cells with ovarian tissue cells together in blood plasma clots onto functional ovaries or by developing them engrafting into the subcutaneous space on the back of nude mice.
  • the goal of the experiment is to determine if plasma clots function as a graft medium to differentiate germ cells into immature or mature oocytes while co-culturing them with functional ovaries.
  • mice are naturally mated to see if there are any functional oocytes that have differentiated, are fertilizable, and able to yield live pups from the transplanted cells.
  • clot conditions co-grafted to host ovaries in the ovarian bursa and subcutaneous space are used: 1) cultured OG2 female GS cells on MEF (Day 0); 2) early EB's (2 day old) from cultured OG2 female GS Cells; 3) late oocyte-like cells from EB's (6 day old) from cultured OG2 female GS cells; 4) freshly isolated ovarian cells and follicles from 4-6 day old FVB GFP mice (used as a positive control).
  • Germ Cell Embryoid Formation A 6-well plate containing a culture of OG2 female colonies is obtained at ⁇ 80% confluence (colonies had minimal contact with each other). The media is aspirated and the wells are washed once with PBS and 700 ⁇ L of warm trypsin was added to each well. The plate is then placed in a 37° C. incubator for 4 min. Each well of the 6-well plate is triturated to wash OG2 female cells off of the MEF layer and then breakup the MEF layer as much as possible. After each well is triturated and all wells were micropipetted into a 50 mL conical tube containing an equal volume (4.2 mL) of PM1TM+15% FBS+GFs.
  • One mL of PM-1+15% FBS+GFs is used to wash out the first three wells of the 6-well plate and combined with cells from the previous step and this step is repeated for the last three wells of the 6-well plate.
  • the cells are then spun down at 400 ⁇ g for 5 min and the supernatant aspirated with vacuum aspiration and then a 200 ⁇ L pipette is used to aspirate the remaining supernatant.
  • the cells are resuspended in 8 mL of PM-1+15% FBS, no ⁇ -mercaptoethanol, no GFs.
  • Two mL of this cell suspension is pipetted into each of 4 wells of a 6-well non-adhesive plate and the plate is placed in a 37° C. incubator for 2-9 days with 50% media change every two days. The media is changed by pipetting 1000 ⁇ L of media in wells.
  • Ovarian Tissue Ovaries of 4-8 day old LacZ/Oct4 GPF OG2 mice are isolated and bisected with fine dissection scissors. The bisected ovaries are transferred to 2 ml of HEPES-buffered DMEM containing 5% FCS and collagenase (1.5 mg/ml). Ovaries in digestion solution are incubated into a 37° C. water bath for 30 min, with gentle pipetting every 10 min. 12 ml of HEPES-buffered DMEM containing 5% FCS is added to stop the digestion of ovaries and digestion mixture after 30 min. The cells are spun at 80 ⁇ G for 10 min at 4° C. Supernatant is removed and cells washed twice by centrifugation.
  • Preparing Ovarian Support Cells Mixture with Germ Cells for Differentiation The digested cells from one ovary are used as support cells for germ cells for each condition. 100K GS cells from each condition are added to one completely digested OG2/LacZ ovarian cell pellet. For only late EB from GS Cells (Day 6), instead of using 100K GS cells, 100 oocyte-like cells (40-70 ⁇ m) are manually picked and added to one completely digested OG2/LacZ ovarian cell pellet. The cell mixture is mixed completely with gentle tapping. Cells are spun at 80 ⁇ G for 10 min at 4° C. then all the supernatant is removed.
  • the prepared cell mixtures are spun down at 80 ⁇ G for 5 min. The supernatant is carefully removed and 20 ⁇ l of venous plasma is added. Re-suspend cells in venous plasma by gentle tapping to vortex cells and plasma. Remove 20 ⁇ l of plasma and cells and make a drop on 6 cm dish and add 0.5 ⁇ l of 1M CaCl 2 to the plasma drop. Cover the dish and place plasma drop with cells into 37° C. incubator. Incubate for 30 min to allow clot to form. After 30 min of incubation clots hardened to a gelatin like consistency and are ready for transplantation into the ovarian bursa next to the ovary.
  • a 6-8 week nude mouse is anesthetized with 0.5 ml of avertin. Wipe the back of the recipient mouse with 70% ethanol and then make two single small longitudinal incisions (less than 1 cm) in the skin with fine dissection scissors near the midline at the level of the last rib (one incision to the right of the midline and the other to the left of the midline). Slide the skin to the left or right until the incision is over the ovary (orange-pink) or fat pad (white), both of which are visible through the body wall.
  • the removed clots are fixed in 5 ml of 4% paraformaldhyde, embedded in OTC, cryo-sectioned at 8 ⁇ m and stained with heamatoxylin and eosin to shoe if differentiation and propagation of follicles are forming by morphology. Slides are stained with oocyte specific antibodies if necessary.
  • Transplanted nude mice are checked 21 days after surgery and then daily to determine if mouse was in estrus. Closure of the introitus is taken as an indication of estrogen deficiency and re-opening as a sign that oestrogenic follicles has emerged in the graft. After vaginal opening or 3 weeks post-operation (whichever was earlier), host females are paired with fertile males. The females are inspected daily for signs of mating (vaginal plugs). Those that mated re allowed to litter.
  • SSCs Spermatogonial stem cells maintain spermatogenesis by self renewal and continuous production of spermatozoa during the entire life. Histological and ultra structural studies revealed that in non-primate mammals, the A s (A single) spermatogonia are considered to be the stem cells of spermatogenesis. Upon division of the A spermatogonia, the daughter cells either migrate away from each other and become two new stem cells, or stay together through an intercellular bridge and become A-paired (Apr) spermatogonia. The Apr spermatogonia develop further into chains of four, eight or 16 A-aligned (Aal) spermatogonia. The Aal spermatogonia differentiate into Al spermatogonia and after six mitotic divisions result in A2, A3, A4 and, finally, B spermatogonia, which give rise to spermatocytes at the last mitotic division.
  • enriched populations of SSCs from the adult primate testes were characterized and isolated.
  • selected markers found at the surface of the primate SSCs the identity of different populations of SSCs in the adult human testes was investigated.
  • enriched populations of human SSCs were transplanted into the recipient mouse testes and the identity of the repopulating human spermatogonial stem cells in the recipient mouse testes was investigated.
  • Testicular tissues devoid of tumor contamination were obtained from patients who underwent an orchiectomy and were generously donated by two patients.
  • Testicular biopsies were obtained from patients undergoing the TESE (testicular biopsy and testicular sperm extraction) procedure. All patients have signed the informed consent form prior tissue collection. A small portion of tissue was extracted and used in this study. Testicular tissues and biopsies were surgically removed, placed in PBS supplemented with penicillin/streptomycin and transported from as little as two hr to overnight on ice. Testicular tissue samples were taken for histology and molecular biological analysis.
  • Seminiferous tubules of the remaining tissue were finely minced and digested with collagenase A (1 mg/mL) and DNase (10 U/mL) in a reciprocating 37° C. water bath for 15 min. After collagenase digestion, the undigested tissue was allowed to settle and cells in the supernatant were removed. The undigested tissue was further digested in an enzyme cocktail consisting of 1.5 mg/mL collagenase A, 1.5 mg/mL hyaluronidase type V, 0.5 mg/mL trypsin, and 10 U/mL DNase in DMEM in a reciprocating 37° C. water bath for 20 min. After straining out remaining undigested tissue, isolated cells were centrifuged at 400 g for 10 min.
  • Flow cytometry and magnetic sorting were stained with selected stem cell markers used for characterization of primate SSCs including CD90-FITC, CD49f-PE, and CD117-APC and SSEA-4 (Table 10). Cells were stained for 30 min in MEM+HEPES+5% FBS (complete medium) on ice, washed once, and re-suspended in complete medium and kept on ice until flow analysis. Flow analysis was accomplished on an InFlux Cell Sorter. Fluorescein (FITC) and phycoerythrin (PE) were excited with a 488 nm 200 mW laser and emission was collected with 530/40 and 580/30 band pass filters, respectively.
  • FITC Fluorescein
  • PE phycoerythrin
  • Allophycocyanin was excited with a 638 nm 25 mW laser and emission was collected with a 670/40 band pass filter.
  • cells up to 200 ⁇ 10 6 ) were resuspended in DMEM+10% FBS and SSEA-4-biotin was added (1:200) and was incubated on ice for 1 hr.
  • Labeling buffer containing PBS, BSA (0.5%) and 2 mM EDTA is prepared and degassed for 10 min. Labeling buffer was added to the SSEA-4 stained cells and centrifuged at 400g for 10 min.
  • SSEA-4 cells resuspended in 1.8 mL of buffer and 200 ⁇ L of streptavidin microbeads was added.
  • Tissues were fixed overnight in 4% paraformaldehyde (PFA) and transferred into 20% sucrose for overnight equilibration. Tissues were frozen in OCT compound and cryosections were prepared at 8 ⁇ m thickness and stored at ⁇ 80° C.
  • PFA paraformaldehyde
  • OCT compound 4% paraformaldehyde
  • sections were washed in PBS and stained with Mayer hematoxilin for 5 min, washed with distilled water for 5 min and mounted using an aqueous mounting medium. The sections were then analyzed using brightfield microscopy.
  • testicular sections were blocked and permeabilized using 0.1% Triton-X/2% BSA/5% sheep serum.
  • RNA Extraction and Real Time PCR Analysis Total cellular RNA was isolated using RNeasy Mini Kit (Qiagen Inc.) according to the manufacturer's recommendations. The isolated RNA was then transcribed to cDNA using the Quantitect RT kit (Qiagen) and later purified with the QIAquick PCR purification kit (Qiagen). For each RT-PCR reaction, 20 ng of cDNA template was used in a 25 ⁇ L reaction volume with HotStar Taq Plus (Qiagen) and respective primers (Table 11). All targets were amplified for 30 cycles. Amplification products were identified by size on a 2% agarose gel.
  • cDNA template 5 ng was used in a 25 ⁇ L reaction volume with Quantitect Sybr Green PCR master mix (Qiagen) and run on a BioRad iCycler. Each sample was assayed in triplicate and normalized to a GAPDH control.
  • the SYBR Green real time quantitative telomeric repeat amplification protocol has been adapted from Wege et al (Wege H, Chui M S, Le H T, Tran J M, Zern M A (2003) SYBR Green real-time telomeric repeat amplification protocol for the rapid quantification of telomerase activity. Nucl. Acids Res. 31, e3.). Tissue or cell pellets were washed once in PBS and re-suspended and homogenized in a prepared lysis buffer containing 1 ⁇ Chaps lysis buffer and 400 U/ml RNaseOut Inhibitor at a volume of 1,000 cells/ ⁇ l.
  • the cell lysates were centrifuged at max speed for 10 min at 4° C. The supernatant was then transferred to a new micro centrifuge tube and the protein concentrations were determined at A280 nm with the ND-1000 spectrophotometer. Reactions were done in 25 ⁇ l volumes containing 500 ng protein lysate, Quantitect SYBR Green PCR mix (Qiagen), 1 ⁇ g TS primer, 0.5 ⁇ g ACX primer, and nuclease-free water.
  • each sample was tested in triplicate along with a no template control (lysis buffer), a positive control (ESC cells), and a standard curve prepared from Human ESC protein lysates (1000 ng, 200 ng, 40 ng, 8 ng, 1.6 ng).
  • a no template control lysis buffer
  • ESC cells positive control
  • a standard curve prepared from Human ESC protein lysates (1000 ng, 200 ng, 40 ng, 8 ng, 1.6 ng).
  • the threshold cycle values were determined from semi-log amplification plots (log increase in fluorescence versus cycle number) and compared with the standard curve.
  • the software default setting for the threshold is 10 times the mean of the standard deviation of the fluorescence reading of each well over the first 10 cycles, excluding cycle 1. Telomerase activity was expressed as a percentage relative to human ESCs.
  • mice testes Eight week old immune deficient Athymic Nude-Foxn1 nu male mice (Harlan) were treated with a single intraperitoneal busulfan injection (40 mg/kg) and were used as recipients. One month after busulfan treatment, 0.3-0.8 ⁇ 10 6 SSEA-4+ magnetically sorted adult human testicular cells were transplanted into the seminiferous tubules via rete testis injection. Four weeks after transplantation, the mice were sacrificed and the testes were fixed in 4% PFA and cryosections were made. The identity of human spermatogonial stem cells in the mouse testes was recognized using human nuclear protein antibody in combination with other stem cell or germ cell markers. All animal experiments were conducted in accordance with the National Research Council's Guidelines for the Care and Use of Laboratory Animals.
  • spermatogonial stem cells Isolation and enrichment of spermatogonial stem cells.
  • Cells isolated from testicular biopsies collected from obstructive azoospermic men showed similar morphology and distribution of spermatogenic cells to the normal human testes indicating that spermatogenesis is in progress in these patients.
  • On average 0.5 ⁇ 10 6 cells was isolated from each sample with the viability of 87%.
  • Spermatogonial stem cells were morphologically detectable among the other cells as round cells with large nucleus to cytoplasm ratio, 1-3 nucleoli and cytoplasmic Inclusions ( FIG. 39 ).
  • For enrichment of spermatogonial stem cells dissociated cells from adult human testicular tissues were analyzed for various cell surface markers using flow cytometry.
  • SSEA-4 has been shown to be expressed on SSCs in the adult Rhesus monkey testes, was abundantly expressed in the human testes. Human testicular cells isolated from both testicular biopsies and donated tissues were tested and it was found that 13.3 ⁇ 1.4% of cells express SSEA-4 on their surface.
  • Another subset of SSC markers that have been described in rodent and primate testes are CD49f ( ⁇ 6-integrin), CD90 (Thy-1), CD117 (c-Kit), and in combination CD49f+/CD90+/CD117 ⁇ (Triple Stain).
  • testicular biopsies have a similar morphology when compared to the donated tissues after hematoxylin-eosin staining ( FIG. 39A-B ).
  • Human testicular tissues were taken for immunohistochemical examination to better understand the distribution and marker expression of human SSCs.
  • SSEA-4 and CD49f also known as ⁇ 6-integrin, have interesting staining patterns in adult human testes, which is very similar to previous observations in the monkey testes ( FIG. 40 ).
  • telomere activity indicates their high level of telomerase activity and their repopulation capability.
  • SSEA-4 positively sorted cells were compared with none sorted cells from the testis against human embryonic stem cells (hESCs) for telomerase activity ( FIG. 41B ).
  • the none sorted cells showed an average of 10.4% ⁇ 10.32% telomerase activity as compared to hESCs (100%), while SSEA-4+ cells had about 5-fold more expression as compared to the none sorted cells 54.6% (+/ ⁇ 7.8%), which is also approximately 2-fold less than hESC telomerase expression. This supports the finding of upregulated h-TERT expression and suggests at least prolonged replication capabilities in SSEA-4 positive cells.
  • ⁇ 6-integrin+ cells two populations of ⁇ 6-integrin+ cells, ⁇ 6-integrin+/SSEA4+ and ⁇ 6-integrin+/SSEA-4 ⁇ , are SSCs that can repopulate the recipient testis.
  • ⁇ 6-integrin+ cells only account for about a quarter of the integrated cells, meaning that about 75% of the SSCs that have integrated have yet to be characterized with a surface marker by flow cytometry. Although almost all of the integrated cells stain positively for c-Kit, it is localized in the nucleus in most cells and c-Kit+ cells were not found by flow cytometry ( FIG.
  • SSEA-4 sorted cells have higher expression level of all the SSC specific genes and a high level of telomerase activity indicating the presence of spermatogonial stem cells in this population.
  • SSCs isolated from mouse and adult primate testes express CD49f and CD90 and are negative for CD117.
  • Expression of CD49f and CD90 in isolated human testes cells has already been reported.
  • This immunohistochemical study revealed that CD49f in human testes was localized along the basement membrane of seminiferous tubules suggesting that this marker is expressing in both the SSCs as well as the differentiating type A spermatogonia.
  • CD49f positive cells outside seminiferous tubules indicates that CD49f, although expressed in human SSCs, is not a specific marker and cannot be used alone for enrichment of SSCs from human testes.
  • Flow cytometry analysis confirmed immunohistochemical staining and showed that there are distinct populations of cells within the adult human testes positively stained for CD49f or CD90, however in contrast to primate there was no population of double positive cells present in the human testes. Similar to SSEA-4, the percentage of CD49f+ and CD90+ cells also was much higher in the adult human testes as compared to the monkey testes.
  • C-Kit is a tyrosine kinase membrane protein which is expressed in hematopoietic stem cells and progenitor cells and in several non hematopoietic tissues including gonads.
  • SCF ligand stem cell factor
  • Double localization of SSEA-4 with c-Kit showed that there are two populations of SSEA-4+ cells in the adult human testes, one with and the other without c-Kit expression. Based on the studies in the mouse SSCs are shown to be c-Kit negative.
  • SSEA-4+ cells are the actively dividing population of spermatogonial stem cells capable of repopulating recipient mouse testes.
  • Human SSCs were purified by SSEA-4 magnetic sorting and SSEA-4+ cells were transplanted into the testes of busulfan treated recipient mouse testes.
  • SSEA-4+ cells were found at the basement membrane of the majority of mouse seminiferous tubules following transplantation indicating the presence of functional SSCs in this population.
  • all the human cells colonized recipient testes were c-Kit+, indicating that only the c-Kit+fraction of SSEA-4 sorted cells were able to colonize recipient testes and therefore are the active SSCs in the human testes.
  • RT-PCR analysis also revealed that SSEA-4 sorted cells have a very high expression level of c-Kit and FGFR3.
  • Expression of c-Kit in human SSCs might indicate the involvement of this receptor and its ligand SCF in colonization and/or repopulation of human SSCs.
  • c-Kit and SCF are key regulators of germ cell migration adhesion and proliferation.
  • high expression of FGFR3 in human SSCs might indicates involvement of its ligand in proliferation and self-renewal of human SSCs.
  • Fibroblast growth factors (FGFs) and their receptors (FGFRs) are key signaling molecules for early embryonic and germ cell development.
  • FGFs has been shown to promote survival and maintenance of mouse and human SSCs.
  • FGF9 is a very potent ligand for FGFR3. Therefore addition of FGF9 and SCF to the culture medium of human SSCs might be beneficial for their survival and proliferation in vitro.
  • Nanog in human testes was not only limited to the undifferentiated SSCs but was also localized in all the germ cells even in the lumen of the seminiferous tubules. This observation is very similar to the adult primate testes suggesting a different role for transcription factor Nanog in the advanced germ cells. The nature of such a role for Nanog is yet to be determined, however it has been reported that pluripotent marker Oct-4 is a survival factor for germ cells and its down regulation will result in apoptosis and cell death rather than differentiation.
  • mouse testes cannot provide appropriate environment to support complete spermatogensis from higher species, its basement membrane of seminiferous tubules has the ability to selectively attract and house human spermatogonial stem cells in the manner very similar to the human testes.
  • repopulating spermatogonial stem cells in the adult human testes have phenotypic characteristics of SSEA-4+, CD49f+, CD90+, GPR-125+ and c-Kit+.
  • About one third of SSCs express Nanog indicating the existence of populations of spermatogonial stem cells in the adult human testes with pluripotent characteristics. The results have direct implications for isolation and purification of spermatogonial stem cells from adult human testes for clinical applications, culture expansion or differentiation purposes.
  • expression of pluripotent markers in subpopulations of human SSCs indicates potential application of these cells for cell replacement therapy and tissue regeneration.
  • testicular tissue is collected under different conditions. First, during surgery to correct undescended testes or torsion, a small piece of testicular tissue weighing approximately 1 gram is removed under sterile conditions. For testicular cancer patients, a larger piece of testicular tissue is removed during surgery to remove the cancerous growth. For cancer patients, thalassemia or sickle-cell anemia patients or patients with other non-malignant hematological indications that require stem cell transplantation, surgery is performed to remove the portion of testicular tissue.
  • a small incision is made on the anterior surface of the tunica albuginea of the testis with a 15-blade scalpel, approximately 1-2 mm in length.
  • the testis is manually compressed (squeezed) to produce egress of seminiferous tubular tissue.
  • the expressed tissue is captured and placed in the tube and returned via courier in the temperature controlled boxes provided to the processing facility.
  • the small incision in the tunica albuginea is then closed with absorbable suture material to reapproximate the tunica albuginea and to provide hemostasis (control of any bleeding from the incision).
  • Post-Surgical Follow-Up The patients are followed up after surgery for signs of complications in the recovery room, once 10-14 days following their procedures, at 6 months, 12 months, and then yearly thereafter during ongoing enrollment in the study. Should the patient/family elect to discontinue enrollment, follow-up will be based on normal clinical indications for follow-up. At each anniversary, the patients will be assessed for ongoing testicular growth/lack of atrophy either while they continue enrollment or until they are comfortable performing their own physical examinations for this endpoint. The parents and patients will be instructed at each visit how to perform testicular self-examination. Patients will be instructed to return for follow-up and counseling at such time as they decide they wish to pursue fertility. Early post-operative complications of hematoma (bleeding) will be assessed in the recovery room and at the first visit post-operatively. A scrotal ultrasound will be performed at the six month visit to assess testicular circulation and size.
  • the tissue is picked up by a representative and transported to the processing facility.
  • the tissue is placed inside a pre-qualified shipper capable of maintaining a temperature of from 2° C. to 8° C. for 48 hours.
  • the tissue is transported via personal vehicle and processed immediately once it is at the processing center.
  • Tissue Processing is performed inside of a Class II Type 2A biosafety cabinet inside of an ISO Class 7 Clean Room Environment. All materials necessary to perform the tissue processing are placed inside the biosafety cabinet prior to processing and any additional items are sterilized if they are needed. An assistant aids the operator and ensures proper passage of materials into and out of the biosafety cabinet. The operator, being sterilely gowned, will not touch anything outside of the biosafety cabinet or anything that has not been previously sterilized. First a small piece of tissue is removed for morphological analysis and the remaining tissue is weighed on a scale inside of the biosafety cabinet.
  • the isolated cells are tested to determine the percentage of cells that are viable. This will be done during the counting process with two types of viability assays: Trypan blue dye exclusion viability assay and 7AAD flow cytometric assay:
  • the cells are prepared for single-color or multi-color staining with monoclonal/polyclonal antibodies. If necessary, complete the secondary antibody staining. After the last washing step re-suspend the cells 1 ml (for 10 million cells) of buffer for analysis. Add 7-AAD stock solution to a final concentration (1 ⁇ g) to each tube and incubate for 10 minutes. Keep the samples in this solution at 4° C. protected from light until analysis on the flow cytometer. Samples with ⁇ 20% viability will be cryogenically frozen only after consulting with the medical director.
  • Cells are assessed under a phase contrast microscope for morphological analysis to determine health, quality, size and general appearance of the cells. The efficiency of digestion of the tissue to single cells and the presence of cell clumps are also analyzed. Under phase contrast, healthy cells maintain a bright, round structure and do not appear dark or fragmented. More detailed information about the intracellular structures, including the nucleus and cytoplasm, can be obtained with Hoffmann or Normarski microscopes. Under these microscopes, germline cells appear round with a large nucleus-to-cytoplasm ratio, 1-3 nucleoli and cytoplasmic inclusions. Sertoli cells are big and irregularly shaped, while Leydig cells have a lot of lipid droplets in their cytoplasm which look like small vesicles.
  • Flow Cytometric Markers Sperm: The X-chromosome is larger than the Y-chromosome. A fluorescent DNA dye (Hoechst 33342) is applied to sperm and incubated for 20 minutes. The flow cytometer separates them based on their relative brightness; XX is brighter than XY.
  • Germ Line Cells Human and Primate SSEA4, CD49f, CD114, CD90, GFR- ⁇ , Rat and Mouse CD49f, SSEA1, GFR- ⁇ .
  • Leydig Cells These cells lack specific markers, but one that we have used is luteinizing hormone receptor (LHR)
  • Staining Protocol Suspend cells in a concentration of 5-10 ⁇ 10 6 cells per ml of staining media. Blocking is done with FBS and/or specific antibody to Fc receptors. Add primary antibody to a final concentration of 1 ⁇ g per million cells. Incubate 30 min on ice in the dark. Wash. Add secondary antibody to 0.5 ⁇ g per million cells (if needed). Incubate 30 min on ice in the dark. Wash. Suspend to a concentration of 5-10 ⁇ 10 6 cells per ml of staining media. Keep on ice in the dark until analysis
  • the germline cell immunohistochemical markers include, but are not limited to, VASA.
  • Other IHC markers for paraffin or frozen sectioning staining for spermatogonial germline cell markers are SSEA-4, CD49f and for germ cells markers are VASA, c-Kit, GFR- ⁇ , and PLZF. These markers are used in an assay to assess the presence of different types of germline cells and their potential to restore fertility.
  • Fluorescent Staining Protocol for frozen tissue sections Use the Super PAP Pen to draw a clean border around the tissue section. Wash tissue section with PBS twice to remove OCT. Block using 5% goat or sheep serum+1% BSA+0.1% Triton-X in PBS for 1 hr at room temperature (block solution should be made within 3 days and stored at 4° C.). Same block solution without Triton-X can be used for surface markers. Incubate in primary antibody in 1% BSA overnight at 4° C. Wash section 3X with PBS for 5 min. Incubate in secondary antibody in 1% BSA in PBS for 1 hr at room temperature. Remove secondary antibody. Stain with nuclear stain [Hoechst (1:5000), DAPI (1:5000), ToPro-3 (1:200)]. Wash section 3 ⁇ with PBS for 5 min.
  • the germline cell molecular markers include, but are not limited to, VASA.
  • HLA A/B/DR Typing & ABO/Rh Typing are performed to confirm identity of the patient.
  • HLA Typing is performed using DNA-based methods to confirm identity of the patient.
  • One tube of collected blood is sent to an ASHI-approved lab for HLA typing.
  • one tube of collected blood is sent to a CLIA-approved lab for ABO/Rh typing.
  • the tubes are sent from the processing facility according to protocol.
  • Infectious disease testing will be performed to ensure safety as follows: Serological: HIV-1/2, HTLV-I/II, HCV, HBsAg, HBc, Syphilis, CMV, Chagas Disease; NAT: HIV, HCV and WNV.
  • Serological HIV-1/2, HTLV-I/II, HCV, HBsAg, HBc, Syphilis, CMV, Chagas Disease; NAT: HIV, HCV and WNV.
  • One tube of collected blood will be sent to a CLIA-approved lab for infectious disease testing; fungi, aerobic and anaerobic bacteria culture testing, speciation & antibiotic susceptibility testing (if culture is positive); microbiological culture will be performed to ensure safety. If culture is positive, speciation and susceptibility testing will be performed so the patient can be put on appropriate antibiotic therapy at the time of transplant.
  • the cells isolated from the tissue processing are suspended in a special media containing clinical grade cryoprotectants designed for cryopreservation of germline cells.
  • a controlled-rate freezer is then used to slowly freeze the cells at ⁇ 1° C./minute from +4° C. to ⁇ 60° C., and at ⁇ 10° C./minute from ⁇ 60° C. to ⁇ 90° C.
  • the cells are then placed in temporary quarantine storage liquid nitrogen vapor phase between ⁇ 150° C. and ⁇ 196° C.
  • Cryopreserved products are stored in the vapor phase of a temporary quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C.
  • a temporary quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C.
  • the contaminated product is removed to the vapor phase of a permanent quarantine liquid nitrogen tank.
  • the temporary quarantine tank changes status to permanent storage tank.
  • Products that are found to be contaminated with tumor cells, aerobic bacteria, anaerobic bacteria, or fungi, or reactive to HIV-1/2, HTLV-I/II, HCV, HBsAG, HBc, and WNV, are stored permanently in the vapor phase of a permanent quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C. Assent will be required to study enrollees for hepatitis and HIV testing. Any positive results will be disclosed to the consented parties and referral for appropriate counseling and treatment will be provided.
  • Study analysis will include determination of loss of cells and viability for processing and cryopreservation (pre-freeze comparison with post-thaw); determination of product stability in cryogenic storage (post-thaw analysis at different time points after freezing); determination of rate of infertility for various indications; determination of rate of adverse reaction or complication for the surgical collection procedure; determination of the effect of the age of the subjects to viability of the cells after thawing; determination of the effect of efficient cryopreservation protocol on viability after thaw.
  • Protocol Stoppage Rules The accrual of patients for the trial will be suspended and subjected to review by the IRB if there are any deaths or severe adverse reactions directly resulting from the surgical collection procedure. Only if the IRB deems it reasonable to re-initiate the trial will accrual of patients resume.
  • Patient Enrollment Research subjects will be enrolled in the study on a physician's recommendation. Once it is determined that subjects fit into the criteria for the study the subject will complete the consent (and assent in the case of a minor) process.
  • ovarian tissue is collected under different conditions. During surgery, a small piece of ovarian cortex tissue weighing approximately 1-2 grams is removed under sterile conditions. For conditions that require ovarioctomies or ovariohysterectomies such as ovarian cyst patients, a larger piece of ovarian tissue is removed during surgery to remove the cancerous growth or the entire ovary, or both ovaries. For cancer patients, thalassemia or patients with other non-malignant hematological indications that require stem cell transplantation, an elective surgery is performed to remove the portion of ovarian tissue or an ovary.
  • the tissue is picked up by a representative and transported to the processing facility.
  • the tissue will be placed inside a pre-qualified shipper capable of maintaining a temperature of 2° C. to 8° C. for 48 hours.
  • Tissue Processing Once the tissue is at processing facility, the tissue is mechanically and enzymatically digested inside of a Class II Type A2 biosafety cabinet inside of an ISO class 7 clean room environment under sterile conditions.
  • the shipping media is transferred to a Petri dish and floating follicles and eggs are collected under a binocular microscope.
  • the cortex of the ovary is sliced to small thin pieces and is prepared for cryopreservation. To dissect and isolate small follicles and cells, the ovary is cut in small pieces and enzymatically digested.
  • Isolated cells and follicles are then further processed for cryopreservation All materials necessary to perform the tissue processing are placed inside the biosafety cabinet prior to processing and any additional items are sterilized if they are needed.
  • the operator being sterilely gowned, does touch anything outside of the biosafety cabinet or anything that has not been previously sterilized.
  • An assistant aids the operator and ensure proper passage of materials into and out of the biosafety cabinet.
  • Stability is the characterization of the ovarian cells after processing. It includes viability and morphological analysis. Approximately less than half a million cells are used for testing and once a year for 10 years the sample is re-assayed.
  • the isolated cells are tested to determine the percentage of cells that are viable. This is done during the counting process with two types of viability assays: trypan blue dye exclusion viability assay (samples with ⁇ 50% viability are cryogenically frozen only after consulting with the medical director) and 7AAD flow cytometric assay (samples with ⁇ 20% viability are cryogenically frozen only after consulting with the medical director).
  • Oocytes are assessed under a phase contrast microscope for morphological analysis to determine health, quality, size and general appearance of the cells. Also the efficiency of digestion of the tissue to single cells and the presence of cell clumps is analyzed. Under phase contrast healthy cells maintain a bright and round structure and do not appear dark or fragmented. More detailed information about the intra cellular structures including the nucleus and cytoplasm can be obtained by Hoffmann or Normarski microscopes. The presence of possible loose and floating oocytes can be observed under a binocular microscope. Oocytes are round large cells and depend on their nuclear maturation stage can have nuclear vesicle (GV) or accompanied with one (MI) or two polar bodies (MII).
  • GV nuclear vesicle
  • MI multi-polar bodies
  • follicles in different stages of development from primordial follicle to antral follicle of various sizes.
  • Follicles contain oocytes in the middle and one or several layers of granulosa and theca cells around them.
  • Primordial and primary follicles vary in size from 200-500 ⁇ m and antral follicles can reach to several millimeters and the graffian follicle can reach to 2-3 cm.
  • follicles are embedded in the ovary and are tightly attached to it the chance of finding them without mechanical or enzymatic dissociation is very low.
  • the germline cell flow markers include, but are not limited to, VASA, Oct-4, c-Kit, SSEA-4
  • the oocyte cell IHC markers include, but are not limited to, GDF9, ZP1, ZP4, Scp3.
  • the cortical granule IHC markers include, but are not limited to, PNA. A combination of these markers are used in an assay to assess the presence of different types of ovarian cells, including potential germline cells, and their potential to restore fertility.
  • the germline cell molecular markers include, but are not limited to, VASA, Oct-4, c-Kit, SSEA-4.
  • the oocyte cell molecular markers include, but are not limited to, BicD1, GDF9, ZP1, ZP4, Ybx2, Scp3.
  • the cortical granule molecular markers include, but are not limited to, PNA. A combination of these markers are used in an assay to assess the presence of different types of ovarian cells, including potential germline cells, and their potential to restore fertility.
  • ABO/Rh Typing will be performed to confirm identity of the patient.
  • HLA Typing is performed using DNA based methods to confirm identity of the patient. Blood sample collected at time of tissue collection are sent to an ASHI approved lab for HLA typing and a CLIA approved lab for ABO/Rh typing according to the approved lab's protocol.
  • HIV-1/2, HTLV-I/II, HCV, HBsAg, HBc, Syphilis, CMV, NAT, HIV, HCV and WNV blood samples collected at time of tissue collection are sent to a CLIA approved lab for testing according to the approved lab's protocol.
  • Fungi, aerobic and anaerobic bacteria culture testing, speciation and antibiotic susceptibility testing (if culture is positive) are performed to ensure safety. If culture is positive, then prior to any future autologous transplantation of the ovarian tissue or cells, speciation and antibiotic susceptibility testing is performed to eradicate any microorganisms.
  • Cryopreservation The cells isolated from the tissue processing are suspended in a special media containing cryoprotectants designed for cryopreservation of ovarian cells and tissue. A control rate freezer is then used to slowly freeze the cells at ⁇ 1° C./minute from +4° C. to ⁇ 60° C., and from ⁇ 60° C. to ⁇ 90° C., the freezing rate will be at ⁇ 10° C./minute. For larger cells, follicles, and ovarian tissue strips, other cryopreservation methods such as vitrification are used. The cells are then place in quarantine storage liquid nitrogen vapor phase between ⁇ 150° C. and ⁇ 196° C.
  • Quarantine Storage Cryopreserved products are stored in a temporary quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C.
  • a temporary quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C.
  • the contaminated product is removed to the vapor phase of a permanent quarantine liquid nitrogen tank.
  • the temporary quarantine tank changes status to permanent storage tank between ⁇ 150° C. and ⁇ 196° C.
  • Permanent Storage Products that are free from contamination with aerobic bacteria, anaerobic bacteria, and fungi, and are non-reactive to HIV-1/2, HTLV-I/II, HCV, HBsAG, HBc, and WNV are stored in the vapor phase of a permanent storage liquid nitrogen freezer tank between ⁇ 150° C. and ⁇ 196° C.
  • Products that are found to be contaminated with tumor cells, aerobic bacteria, anaerobic bacteria, or fungi, or reactive to HIV-1/2, HTLV-I/II, HCV, HBsAG, HBc, and WNV, are stored permanently in the vapor phase of a permanent quarantine liquid nitrogen tank between ⁇ 150° C. and ⁇ 196° C. Assent is required to study enrollees for hepatitis and HIV testing. Any positive results are disclosed to the consented parties and referral for appropriate counseling and treatment are provided.
  • Study analysis will include determination of loss of cells and viability for processing and cryopreservation (pre-freeze comparison with post-thaw), determination of product stability in cryogenic storage (post-thaw analysis at different time points after freezing), determination of rate of infertility for various indications, determination of rate of adverse reaction or complication for the surgical collection procedure, determination of the effect of the age of the subjects to viability of the cells after thawing, and determination of the effect of efficient cryopreservation protocol on viability after thaw.
  • Protocol Stoppage Rules The accrual of patients for the trial is suspended and subjected to review by the IRB if there are any deaths or severe adverse reactions directly resulting from the surgical collection procedure. Only if the IRB deems it reasonable to re-initiate the trial will accrual of patients resume.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Reproductive Health (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Endocrinology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Gynecology & Obstetrics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Materials For Medical Uses (AREA)
US12/940,888 2009-11-05 2010-11-05 Germline stem cell banking system Abandoned US20120009156A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/940,888 US20120009156A1 (en) 2009-11-05 2010-11-05 Germline stem cell banking system

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25853509P 2009-11-05 2009-11-05
US12/940,888 US20120009156A1 (en) 2009-11-05 2010-11-05 Germline stem cell banking system

Publications (1)

Publication Number Publication Date
US20120009156A1 true US20120009156A1 (en) 2012-01-12

Family

ID=43478138

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/940,894 Active 2034-01-25 US9657267B2 (en) 2009-11-05 2010-11-05 Ex host maturation of germline stem cells
US12/940,888 Abandoned US20120009156A1 (en) 2009-11-05 2010-11-05 Germline stem cell banking system

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/940,894 Active 2034-01-25 US9657267B2 (en) 2009-11-05 2010-11-05 Ex host maturation of germline stem cells

Country Status (11)

Country Link
US (2) US9657267B2 (fr)
EP (2) EP2496940B1 (fr)
JP (2) JP2013514969A (fr)
KR (2) KR20120120157A (fr)
CN (2) CN103154730B (fr)
AU (2) AU2010314984B2 (fr)
BR (2) BR112012010750A2 (fr)
CA (2) CA2779701A1 (fr)
IL (2) IL219600A0 (fr)
MX (2) MX2012005297A (fr)
WO (2) WO2011057128A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657267B2 (en) 2009-11-05 2017-05-23 Primegen Biotech Llc Ex host maturation of germline stem cells
WO2021046521A1 (fr) * 2019-09-06 2021-03-11 The Brigham And Women's Hospital, Inc. Évaluation automatique d'une mesure d'assurance qualité pour des procédures de reproduction assistée
US20230399608A1 (en) * 2021-01-04 2023-12-14 Paterna Biosciences Inc. Process for establishing a human testicular tissue culture system
RU2810125C1 (ru) * 2019-09-06 2023-12-22 Дэ Бригем Энд Уименс Хоспитал, Инк. Автоматизированная оценка показателей обеспечения качества для процедур вспомогательной репродукции

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA034065B1 (ru) * 2012-02-14 2019-12-24 Вашингтон Стейт Юниверсити Способ культивирования бычьих и свиных сперматогониальных стволовых клеток без использования клеток-фидеров
EP2886644A1 (fr) * 2013-12-20 2015-06-24 Kallistem Procédé de mise en oeuvre de spermatogenèse in vitro et dispositif associé
EP3006560A1 (fr) * 2014-10-09 2016-04-13 European Molecular Biology Laboratory Miwi2 comme marqueur pour des cellules souches spermatogoniales et procédé de diagnostic de cellules souches spermatogoniales basé sur ce marqueur
CN104974982B (zh) * 2015-06-19 2018-07-06 中国农业大学 一种体外培养睾丸组织诱导生成精子细胞的方法
CN106754724A (zh) * 2016-12-09 2017-05-31 西北农林科技大学 一种永生化的牛精原干细胞系及其构建方法
KR101986830B1 (ko) * 2017-09-07 2019-06-07 차의과학대학교 산학협력단 줄기세포 유래 세르톨리유사세포, 그 제조방법, 및 그의 용도
US20200289574A1 (en) * 2017-10-27 2020-09-17 Arteriocyte Medical Systems, Inc. Augmentation of fertility by platelet rich plasma
JP7021828B2 (ja) * 2017-11-10 2022-02-17 一公 川島 卵巣由来幹細胞の減数分裂誘導法
US11845956B2 (en) 2018-09-21 2023-12-19 APstem Therapeutics, Inc. Human pluripotent adult stem cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027329A1 (en) * 2001-05-18 2003-02-06 Lee Dong Ryul In vitro production of haploid germ cells
US20040258673A1 (en) * 2003-04-03 2004-12-23 Hirose Thomas Gordon Elective collection and banking of autologous peripheral blood stem cells

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090622A (en) 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
CN1174091C (zh) * 2002-01-29 2004-11-03 南京医科大学第一附属医院 人成熟卵泡液在未成熟卵体外成熟技术中的应用
US20030215942A1 (en) 2002-02-14 2003-11-20 Stemcyte, Inc. Undesignated allogeneic stem cell bank
US20040091936A1 (en) 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
US7087371B2 (en) 2002-06-28 2006-08-08 The Trustees Of The University Of Pennsylvania Material and methods for the production of sperm and analysis thereof
CN1490400A (zh) * 2002-10-17 2004-04-21 中国科学院力学研究所 毛细管微包被法控制细胞空间分布、形状和大小及其应用
US7425421B2 (en) 2003-06-26 2008-09-16 Litron Laboratories, Ltd. Method for the enumeration of micronucleated erythrocyte populations while distinguishing platelets and/or platelet-associated aggregates
CA2536831A1 (fr) 2003-08-27 2005-03-10 Kyoto University Procede permettant de construire une descendance a partir d'une cellule souche de sperme congele
JP4314372B2 (ja) 2004-03-30 2009-08-12 国立大学法人京都大学 精巣細胞由来多能性幹細胞の製造方法
US20080044395A1 (en) * 2004-04-13 2008-02-21 Sungkwang Educational Foundation In Vitro Method for Isolating, Proliferating and Differentiating Germ-Line Stem Cells
US7955846B2 (en) 2004-05-17 2011-06-07 The General Hospital Corporation Compositions comprising female germline stem cells and methods of use thereof
US20070020759A1 (en) 2004-07-15 2007-01-25 Primegen Biotech Llc Therapeutic reprogramming of germ line stem cells
CN101040042B (zh) 2004-08-16 2013-06-19 细胞研究私人有限公司 自脐带羊膜分离干/祖细胞
CA2607975A1 (fr) * 2005-05-30 2006-12-07 Commonwealth Scientific And Industrial Research Organisation Preparation et utilisation de particules de membrane basale
US7732202B2 (en) 2005-10-21 2010-06-08 International Stem Cell Corporation Oxygen tension for the parthenogenic activation of human oocytes for the production of human embryonic stem cells
WO2007051625A2 (fr) 2005-11-02 2007-05-10 Georg-August-Universität Göttingen Compositions et procedes de production de cellules pluripotentes a partir de testicules adultes
CA2629283A1 (fr) 2005-11-14 2007-05-24 The New England Medical Center Hospitals, Inc. Procedes de preparation de cellules souches matricielles de cordon (cmsc) en vue d'un stockage a long terme et de preparation d'un segment de cordon ombilical en vue d'une cryoconservation
CN1800371A (zh) * 2005-12-19 2006-07-12 浙江大学 一种人体未成熟卵体外成熟培养方法
KR100707985B1 (ko) 2006-03-10 2007-04-16 세원셀론텍(주) 세포치료제를 생산 및 보관하기 위한 모듈 장치
KR100745362B1 (ko) 2006-03-14 2007-08-02 세원셀론텍(주) 세포치료제 설비의 사용방법 및 이를 이용한 네트워크기반의 프랜차이즈 마켓 비즈니스 방법
WO2007115216A1 (fr) 2006-03-30 2007-10-11 Primegen Biotech Llc Reprogrammation de cellules souches testiculaires d'humains adultes en cellules souches pluripotentes de la lignee germinale
US8759090B2 (en) 2006-10-30 2014-06-24 University Of Central Florida Research Foundation, Inc. Stem cell banking system
US20080132803A1 (en) 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
CN100999722B (zh) * 2007-01-08 2011-08-03 安徽省立医院 卵巢组织块内不成熟卵母细胞的体外培养成熟方法
US20080189045A1 (en) 2007-02-06 2008-08-07 Moore Thomas E Method for the Collection And Distribution of Cord Blood Stem Cells
CN100548313C (zh) 2007-06-05 2009-10-14 南京农业大学 细管高密度冷冻猪精液的方法及其产品
EP2176399A1 (fr) * 2007-08-07 2010-04-21 PrimeGen Biotech LLC Isolement, caractérisation et propagation de cellules souches germinales
US20090138354A1 (en) 2007-11-25 2009-05-28 Herbert Zech Methods of Storing and Providing Samples of Cells and Tissue
US8673637B2 (en) * 2008-10-16 2014-03-18 The Board Of Trustees Of The Leland Stanford Junior University Human multipotent germ line stem cells expressing a germ line marker and a pluripotency marker produced by co-culture of testicular tissue
CN101429492B (zh) * 2008-12-12 2010-10-06 深圳华大基因科技有限公司 一种猪卵母细胞体外培养成熟方法
EP2496940B1 (fr) 2009-11-05 2018-08-15 Primegen Biotech, LLC dba Reprocyte Système de classement de cellules souches à lignée germinale

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027329A1 (en) * 2001-05-18 2003-02-06 Lee Dong Ryul In vitro production of haploid germ cells
US20040258673A1 (en) * 2003-04-03 2004-12-23 Hirose Thomas Gordon Elective collection and banking of autologous peripheral blood stem cells

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
Conrad et al., "Generation of pluripotent stem cells from adult human testis," Nature Vol. 456, pp. 344-349 and method supplement; 11/20/2008 (of record). *
Crabbe et al., Human Reproduction, Vol. 13, No. 10, pp. 2791-2796; 1999 (of record). *
Crabbe et al., Human Reproduction, Vol. 13, No. 10, pp. 2791-2796; 1999. *
Geens et al., Human Reproduction Update, Vol. 14, No. 2, pp. 121-129; 1/10/2008 (of record). *
Geens et al., Human Reproduction Update, Vol. 14, No. 2, pp. 121-129; 1/10/2008. *
Hermann et al. (Human Reproduction, Vol. 1, No. 1, pp. 1-13; available online 6/15/2009 *
Hermann et al. (Human Reproduction, Vol. 1, No. 1, pp. 1-13; available online 6/15/2009 (of record). *
Jahnukainen et al., "Clinical Potential and Putative Risks of Fertility Preservation in Children Utilizing Gonadal Tissue of Germline Stem Cells," Pediatric Research, Vol. 59, No. 4, part 2; 2006 (of record). *
Jahnukainen et al., "Clinical Potential and Putative Risks of Fertility Preservation in Children Utilizing Gonadal Tissue ofGermline Stem Cells," Pediatric Research, Vol. 59, No. 4, part 2; 2006 (of record). *
Jahnukainen et al., Pediatric Research, Vol. 59, No. 4, part 2; 2006 (of record). *
Keros et al., "Methods of cryopreservation of testicular tissue with viable spermatogonia in pre-pubertal boys undergoing gonadotoxic cancer treatment," Human Reproduction, Vol. 22, No. 5, pp. 1384-1395; 2007 (of record). *
Keros et al., "Methods of cryopreservation of testicular tissue with viable spermatogonia in pre-pubertal boys undergoinggonadotoxic cancer treatment," Human Reproduction, Vol. 22, No. 5, pp. 1384-1395; 2007 (of record). *
Keros et al., Human Reproduction, Vol. 22, No. 5, pp. 1384-1395; 2007 (of record). *
Wyns et al., "Long-term spermatogonial survival in cryopreserved and xenografted immature human testicular tissue," Human Reproduction Vol. 23, No. 11, pp. 2402-2414, available online 7/28/2008 (of record). *
Wyns et al., "Long-term spermatogonial survival in cryopreserved and xenografted immature human testicular tissue," HumanReproduction Vol. 23, No. 11, pp. 2402-2414, available online 7/28/2008 (of record). *
Wyns et al., Human Reproduction, Vol. 23, No. 11, pp. 2402-2414; 7/28/2008 (of record). *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657267B2 (en) 2009-11-05 2017-05-23 Primegen Biotech Llc Ex host maturation of germline stem cells
WO2021046521A1 (fr) * 2019-09-06 2021-03-11 The Brigham And Women's Hospital, Inc. Évaluation automatique d'une mesure d'assurance qualité pour des procédures de reproduction assistée
RU2810125C1 (ru) * 2019-09-06 2023-12-22 Дэ Бригем Энд Уименс Хоспитал, Инк. Автоматизированная оценка показателей обеспечения качества для процедур вспомогательной репродукции
US20230399608A1 (en) * 2021-01-04 2023-12-14 Paterna Biosciences Inc. Process for establishing a human testicular tissue culture system

Also Published As

Publication number Publication date
WO2011057123A1 (fr) 2011-05-12
AU2010314984B2 (en) 2015-09-17
JP2013514760A (ja) 2013-05-02
US20120083034A1 (en) 2012-04-05
AU2010314989B2 (en) 2013-10-24
MX344716B (es) 2017-01-03
IL219600A0 (en) 2012-06-28
EP2496939B1 (fr) 2017-02-15
BR112012010675A2 (pt) 2016-11-29
MX2012005298A (es) 2012-12-17
WO2011057123A8 (fr) 2012-06-07
AU2010314984A8 (en) 2014-02-27
EP2496939A1 (fr) 2012-09-12
MX2012005297A (es) 2013-02-26
CA2779726C (fr) 2018-03-06
IL219601A (en) 2017-05-29
JP2013514969A (ja) 2013-05-02
CN103154730B (zh) 2015-04-29
CN102713614A (zh) 2012-10-03
BR112012010750A2 (pt) 2019-09-24
EP2496940B1 (fr) 2018-08-15
AU2010314984A1 (en) 2012-05-24
CN103154730A (zh) 2013-06-12
WO2011057128A1 (fr) 2011-05-12
KR20130028041A (ko) 2013-03-18
EP2496940A1 (fr) 2012-09-12
CA2779726A1 (fr) 2011-05-12
IL219601A0 (en) 2012-07-31
US9657267B2 (en) 2017-05-23
AU2010314989A1 (en) 2012-05-24
CA2779701A1 (fr) 2011-05-12
KR20120120157A (ko) 2012-11-01

Similar Documents

Publication Publication Date Title
EP2496940B1 (fr) Système de classement de cellules souches à lignée germinale
US20190300850A1 (en) Compositions and Methods for Autologous Germline Mitochondrial Energy Transfer
AU2014200322A1 (en) Ex host maturation of germline stem cells
AU2017201404B2 (en) Compositions And Methods For Autologous Germline Mitochondrial Energy Transfer
NZ617582B2 (en) Compositions and methods for autologous germline mitochondrial energy transfer

Legal Events

Date Code Title Description
AS Assignment

Owner name: PRIMEGEN BIOTECH LLC DBA REPROCYTE, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:IZADYAR, FARIBORZ;MARH, JOEL;MAKI, CHAD;AND OTHERS;SIGNING DATES FROM 20110117 TO 20110512;REEL/FRAME:026859/0117

AS Assignment

Owner name: HARVARDIANAMD CONSULTING, INC., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHOW, JOHN YUNG-CHIONG;REEL/FRAME:034432/0666

Effective date: 20130808

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION