Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(30): 17832-17841, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32661178

RESUMO

Spermatogonial stem cells (SSCs) are essential for the generation of sperm and have potential therapeutic value for treating male infertility, which afflicts >100 million men world-wide. While much has been learned about rodent SSCs, human SSCs remain poorly understood. Here, we molecularly characterize human SSCs and define conditions favoring their culture. To achieve this, we first identified a cell-surface protein, PLPPR3, that allowed purification of human primitive undifferentiated spermatogonia (uSPG) highly enriched for SSCs. Comparative RNA-sequencing analysis of these enriched SSCs with differentiating SPG (KIT+ cells) revealed the full complement of genes that shift expression during this developmental transition, including genes encoding key components in the TGF-ß, GDNF, AKT, and JAK-STAT signaling pathways. We examined the effect of manipulating these signaling pathways on cultured human SPG using both conventional approaches and single-cell RNA-sequencing analysis. This revealed that GDNF and BMP8B broadly support human SPG culture, while activin A selectively supports more advanced human SPG. One condition-AKT pathway inhibition-had the unique ability to selectively support the culture of primitive human uSPG. This raises the possibility that supplementation with an AKT inhibitor could be used to culture human SSCs in vitro for therapeutic applications.


Assuntos
Transdução de Sinais , Espermatogônias/citologia , Espermatogônias/metabolismo , Transcriptoma , Biomarcadores , Separação Celular , Células Cultivadas , Biologia Computacional , Imunofluorescência , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imunofenotipagem , Masculino , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/metabolismo
2.
Genes Dev ; 29(23): 2420-34, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26584619

RESUMO

Telomerase inactivation causes loss of the male germline in worms, fish, and mice, indicating a conserved dependence on telomere maintenance in this cell lineage. Here, using telomerase reverse transcriptase (Tert) reporter mice, we found that very high telomerase expression is a hallmark of undifferentiated spermatogonia, the mitotic population where germline stem cells reside. We exploited these high telomerase levels as a basis for purifying undifferentiated spermatogonia using fluorescence-activated cell sorting. Telomerase levels in undifferentiated spermatogonia and embryonic stem cells are comparable and much greater than in somatic progenitor compartments. Within the germline, we uncovered an unanticipated gradient of telomerase activity that also enables isolation of more mature populations. Transcriptomic comparisons of Tert(High) undifferentiated spermatogonia and Tert(Low) differentiated spermatogonia by RNA sequencing reveals marked differences in cell cycle and key molecular features of each compartment. Transplantation studies show that germline stem cell activity is confined to the Tert(High) cKit(-) population. Telomere shortening in telomerase knockout strains causes depletion of undifferentiated spermatogonia and eventual loss of all germ cells after undifferentiated spermatogonia drop below a critical threshold. These data reveal that high telomerase expression is a fundamental characteristic of germline stem cells, thus explaining the broad dependence on telomerase for germline immortality in metazoans.


Assuntos
Células-Tronco Adultas/enzimologia , Regulação Enzimológica da Expressão Gênica , Espermatogônias/enzimologia , Telomerase/genética , Telomerase/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/enzimologia , Citometria de Fluxo , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética
3.
Biol Reprod ; 96(3): 707-719, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28339678

RESUMO

Spermatogonial stem cells (SSC) are essential for spermatogenesis and male fertility. In addition, these adult tissue stem cells can be used as vehicles for germline modification in animal models and may have application for treating male infertility. To facilitate the investigation of SSCs and germ lineage development in rats, we generated a DEAD-box helicase 4 (DDX4) (VASA) promoter-enhanced green fluorescent protein (EGFP) reporter transgenic rat. Quantitative real-time polymerase chain reaction and immunofluorescence confirmed that EGFP was expressed in the germ cells of the ovaries and testes and was absent in somatic cells and tissues. Germ cell transplantation demonstrated that the EGFP-positive germ cell population from DDX4-EGFP rat testes contained SSCs capable of establishing spermatogenesis in experimentally infertile mouse recipient testes. EGFP-positive germ cells could be easily isolated by fluorescence-activated cells sorting, while simultaneously removing testicular somatic cells from DDX4-EGFP rat pup testes. The EGFP-positive fraction provided an optimal cell suspension to establish rat SSC cultures that maintained long-term expression of zinc finger and BTB domain containing 16 (ZBTB16) and spalt-like transcription factor 4 (SALL4), two markers of mouse SSCs that are conserved in rats. The novel DDX4-EGFP germ cell reporter rat described here combined with previously described GCS-EGFP rats, rat SSC culture and gene editing tools will improve the utility of the rat model for studying stem cells and germ lineage development.


Assuntos
RNA Helicases DEAD-box/genética , Células Germinativas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Modelos Animais , Espermatogênese , Células-Tronco Germinativas Adultas , Animais , Células Cultivadas , Feminino , Genes Reporter , Masculino , Regiões Promotoras Genéticas , Ratos Sprague-Dawley , Ratos Transgênicos
4.
Hum Mol Genet ; 23(12): 3071-84, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24449759

RESUMO

Studies of human germ cell development are limited in large part by inaccessibility of germ cells during development. Moreover, although several studies have reported differentiation of mouse and human germ cells from pluripotent stem cells (PSCs) in vitro, differentiation of human germ cells from PSCs in vivo has not been reported. Here, we tested whether mRNA reprogramming in combination with xeno-transplantation may provide a viable system to probe the genetics of human germ cell development via use of induced pluripotent stem cells (iPSCs). For this purpose, we derived integration-free iPSCs via mRNA-based reprogramming with OCT3/4, SOX2, KLF4 and cMYC alone (OSKM) or in combination with the germ cell-specific mRNA, VASA (OSKMV). All iPSC lines met classic criteria of pluripotency. Moreover, global gene expression profiling did not distinguish large differences between undifferentiated OSKM and OSKMV iPSCs; however, some differences were observed in expression of pluripotency factors and germ cell-specific genes, and in epigenetic profiles and in vitro differentiation studies. In contrast, transplantation of undifferentiated iPSCs directly into the seminiferous tubules of germ cell-depleted immunodeficient mice revealed divergent fates of iPSCs produced with different factors. Transplantation resulted in morphologically and immunohistochemically recognizable germ cells in vivo, particularly in the case of OSKMV cells. Significantly, OSKMV cells also did not form tumors while OSKM cells that remained outside the seminiferous tubule proliferated extensively and formed tumors. Results indicate that mRNA reprogramming in combination with transplantation may contribute to tools for genetic analysis of human germ cell development.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Túbulos Seminíferos/metabolismo , Espermatozoides/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Epigênese Genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Nus , Espermatozoides/citologia , Transplante Heterólogo/métodos
5.
Biol Reprod ; 94(1): 11, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26607719

RESUMO

Testosterone acts though the androgen receptor in Sertoli cells to support germ cell development (spermatogenesis) and male fertility, but the molecular and cellular mechanisms by which testosterone acts are not well understood. Previously, we found that in addition to acting through androgen receptor to directly regulate gene expression (classical testosterone signaling pathway), testosterone acts through a nonclassical pathway via the androgen receptor to rapidly activate kinases that are known to regulate spermatogenesis. In this study, we provide the first evidence that nonclassical testosterone signaling occurs in vivo as the MAP kinase cascade is rapidly activated in Sertoli cells within the testis by increasing testosterone levels in the rat. We find that either classical or nonclassical signaling regulates testosterone-mediated Rhox5 gene expression in Sertoli cells within testis explants. The selective activation of classical or nonclassical signaling pathways in Sertoli cells within testis explants also resulted in the differential activation of the Zbtb16 and c-Kit genes in adjacent spermatogonia germ cells. Delivery of an inhibitor of either pathway to Sertoli cells of mouse testes disrupted the blood-testis barrier that is essential for spermatogenesis. Furthermore, an inhibitor of nonclassical testosterone signaling blocked meiosis in pubertal mice and caused the loss of meiotic and postmeiotic germ cells in adult mouse testes. An inhibitor of the classical pathway caused the premature release of immature germ cells. Collectively, these observations indicate that classical and nonclassical testosterone signaling regulate overlapping and distinct functions that are required for the maintenance of spermatogenesis and male fertility.


Assuntos
Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , Testosterona/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Fertilidade/efeitos dos fármacos , Fertilidade/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Proto-Oncogênicas c-kit/biossíntese , Proteínas Proto-Oncogênicas c-kit/genética , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/biossíntese , Receptores Androgênicos/genética , Células de Sertoli/metabolismo , Transdução de Sinais/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Nat Genet ; 36(6): 653-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15156143

RESUMO

Little is known of the molecular mechanisms whereby spermatogonia, mitotic germ cells of the testis, self-renew and differentiate into sperm. Here we show that Zfp145, encoding the transcriptional repressor Plzf, has a crucial role in spermatogenesis. Zfp145 expression was restricted to gonocytes and undifferentiated spermatogonia and was absent in tubules of W/W(v) mutants that lack these cells. Mice lacking Zfp145 underwent a progressive loss of spermatogonia with age, associated with increases in apoptosis and subsequent loss of tubule structure but without overt differentiation defects or loss of the supporting Sertoli cells. Spermatogonial transplantation experiments revealed a depletion of spermatogonial stem cells in the adult. Microarray analysis of isolated spermatogonia from Zfp145-null mice before testis degeneration showed alterations in the expression profile of genes associated with spermatogenesis. These results identify Plzf as a spermatogonia-specific transcription factor in the testis that is required to regulate self-renewal and maintenance of the stem cell pool.


Assuntos
Proteínas de Ligação a DNA/genética , Espermatogônias/citologia , Células-Tronco/citologia , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Expressão Gênica , Fatores de Transcrição Kruppel-Like , Masculino , Camundongos , Camundongos Knockout , Proteína com Dedos de Zinco da Leucemia Promielocítica , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/transplante , Células-Tronco/metabolismo , Testículo/citologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo
7.
Stem Cell Res ; 73: 103257, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38000347

RESUMO

Curative bone marrow transplantation (BMT) therapies for sickle cell disease (SCD) can cause infertility. The Fertility Preservation Program (FPP) in Pittsburgh cryopreserves testicular tissues for SCD patients prior to BMT in anticipation that those tissues can be thawed in the future and matured to produce sperm. Here, we generated and validated two isogenic patient-derived induced pluripotent stem cell (iPSC) lines from testicular biopsy fibroblasts of a 12-year-old SCD patient.


Assuntos
Anemia Falciforme , Células-Tronco Pluripotentes Induzidas , Humanos , Masculino , Criança , Células-Tronco Pluripotentes Induzidas/patologia , Sêmen , Transplante de Medula Óssea , Anemia Falciforme/patologia , Fibroblastos/patologia
8.
J Tissue Eng ; 14: 20417314231197282, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38029018

RESUMO

Female cancer patients who have undergone chemotherapy have an elevated risk of developing ovarian dysfunction and failure. Experimental approaches to treat iatrogenic infertility are evolving rapidly; however, challenges and risks remain that hinder clinical translation. Biomaterials have improved in vitro follicle maturation and in vivo transplantation in mice, but there has only been marginal success for early-stage human follicles. Here, we developed methods to obtain an ovarian-specific extracellular matrix hydrogel to facilitate follicle delivery and establish an in situ ovary (ISO), which offers a permissive environment to enhance follicle survival. We demonstrate sustainable follicle engraftment, natural pregnancy, and the birth of healthy pups after intraovarian microinjection of isolated exogenous follicles into chemotherapy-treated (CTx) mice. Our results confirm that hydrogel-based follicle microinjection could offer a minimally invasive delivery platform to enhance follicle integration for patients post-chemotherapy.

9.
Dev Cell ; 57(9): 1160-1176.e5, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35504286

RESUMO

Aging men display reduced reproductive health; however, testis aging is poorly understood at the molecular and genomic levels. Here, we utilized single-cell RNA-seq to profile over 44,000 cells from both young and older men and examined age-related changes in germline development and in the testicular somatic cells. Age-related changes in spermatogonial stem cells appeared modest, whereas age-related dysregulation of spermatogenesis and somatic cells ranged from moderate to severe. Altered pathways included signaling and inflammation in multiple cell types, metabolic signaling in Sertoli cells, hedgehog signaling and testosterone production in Leydig cells, cell death and growth in testicular peritubular cells, and possible developmental regression in both Leydig and peritubular cells. Remarkably, the extent of dysregulation correlated with body mass index in older but not in younger men. Collectively, we reveal candidate molecular mechanisms underlying the complex testicular changes conferred by aging and their possible exacerbation by concurrent chronic conditions such as obesity.


Assuntos
Análise de Célula Única , Testículo , Idoso , Envelhecimento , Índice de Massa Corporal , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Células de Sertoli , Espermatogênese/genética , Testículo/metabolismo
10.
Hum Reprod ; 26(12): 3222-31, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22016413

RESUMO

BACKGROUND: Chemotherapy and radiation treatments for cancer and other diseases can cause male infertility. There are currently no options to preserve the fertility of prepubertal boys who are not yet making sperm. Cryopreservation of spermatogonial stem cells (SSCs, obtained via testicular biopsy) followed by autologous transplantation back into the testes at a later date may restore fertility in these patients. However, this approach carries an inherent risk of reintroducing cancer. METHODS: To address this aspect of SSC transplantation safety, prepubertal non-human primate testis cell suspensions were inoculated with MOLT4 T-lymphoblastic leukemia cells and subsequently sorted for cell surface markers CD90 (THY-1) and CD45. RESULTS: Cancer cells segregated to the CD90-/CD45+ fraction and produced tumors in nude mice. Nearly all sorted DEAD box polypeptide 4-positive (VASA+) spermatogonia segregated to the CD90+/CD45- fraction. In a preliminary experiment, a purity check of the sorted putative stem cell fraction (CD90+/CD45-) revealed 0.1% contamination with cancer cells, which was sufficient to produce tumors in nude mice. We hypothesized that the contamination resulted from mis-sorting due to cell clumping and employed singlet discrimination (SD) in four subsequent experiments. Purity checks revealed no cancer cell contamination in the CD90+/CD45- fraction from three of the four SD replicates and these fractions produced no tumors when transplanted into nude mouse testes. CONCLUSIONS: We conclude that spermatogonia can be separated from contaminating malignant cells by fluorescence-activated cell sorting prior to SSC transplantation and that post-sorting purity checks are required to confirm elimination of malignant cells.


Assuntos
Separação Celular/métodos , Preservação da Fertilidade/métodos , Citometria de Fluxo/métodos , Espermatogônias/citologia , Testículo/citologia , Animais , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Macaca mulatta , Masculino , Maturidade Sexual , Espermatogônias/transplante , Preservação de Tecido
11.
Nat Commun ; 12(1): 3876, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162856

RESUMO

Testicular development and function rely on interactions between somatic cells and the germline, but similar to other organs, regenerative capacity declines in aging and disease. Whether the adult testis maintains a reserve progenitor population remains uncertain. Here, we characterize a recently identified mouse testis interstitial population expressing the transcription factor Tcf21. We found that TCF21lin cells are bipotential somatic progenitors present in fetal testis and ovary, maintain adult testis homeostasis during aging, and act as potential reserve somatic progenitors following injury. In vitro, TCF21lin cells are multipotent mesenchymal progenitors which form multiple somatic lineages including Leydig and myoid cells. Additionally, TCF21+ cells resemble resident fibroblast populations reported in other organs having roles in tissue homeostasis, fibrosis, and regeneration. Our findings reveal that the testis, like other organs, maintains multipotent mesenchymal progenitors that can be potentially leveraged in development of future therapies for hypoandrogenism and/or infertility.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Homeostase/genética , Células-Tronco Mesenquimais/metabolismo , Regeneração/genética , Testículo/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula/genética , Células Cultivadas , Feminino , Perfilação da Expressão Gênica/métodos , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Célula Única/métodos , Testículo/citologia
12.
BMC Dev Biol ; 10: 17, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20158911

RESUMO

BACKGROUND: The tetracycline-inducible gene regulation system is a powerful tool that allows temporal and dose-dependent regulation of target transgene expression in vitro and in vivo. Several tetracycline-inducible transgenic mouse models have been described with ubiquitous or tissue-specific expression of tetracycline-transactivator (tTA), reverse tetracycline-transactivator (rtTA) or Tet repressor (TetR). Here we describe a Tet-On transgenic rat that ubiquitously expresses rtTA-M2 driven by the murine ROSA 26 promoter. RESULTS: The homozygous rat line (ROSA-rtTA-M2) generated by lentiviral vector injection, has a single integration site and was derived from the offspring of a genetic mosaic founder with multiple transgene integrations. The rtTA-M2 transgene integrated into an intron of a putative gene on chromosome 2 and does not appear to affect the tissue-specificity or expression of that gene. Fibroblasts from the ROSA-rtTA-M2 rats were transduced with a TetO7/CMV-EGFP lentivirus and exhibited doxycycline dose-dependent expression of the EGFP reporter transgene, in vitro. In addition, doxycycline-inducible EGFP expression was observed, in vivo, when the TetO7/CMV-EGFP lentivirus was injected into testis, kidney and muscle tissues of ROSA-rtTA-M2 rats. CONCLUSIONS: This conditional expression rat model may have application for transgenic overexpression or knockdown studies of gene function in development, disease and gene therapy.


Assuntos
Técnicas Genéticas , Ratos Transgênicos , Ratos/genética , Animais , Linhagem Celular , Fibroblastos , Camundongos , Regiões Promotoras Genéticas , Tetraciclina , Transativadores/genética
13.
Reproduction ; 139(3): 479-93, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19880674

RESUMO

Spermatogonial stem cells (SSCs) maintain spermatogenesis throughout the reproductive life of mammals. While A(single) spermatogonia comprise the rodent SSC pool, the identity of the stem cell pool in the primate spermatogenic lineage is not well established. The prevailing model is that primate spermatogenesis arises from A(dark) and A(pale) spermatogonia, which are considered to represent reserve and active stem cells respectively. However, there is limited information about how the A(dark) and A(pale) descriptions of nuclear morphology correlate with the clonal (A(single), A(paired), and A(aligned)), molecular (e.g. GFRalpha1 (GFRA1) and PLZF), and functional (SSC transplantation) descriptions of rodent SSCs. Thus, there is a need to investigate primate SSCs using criteria, tools, and approaches that have been used to investigate rodent SSCs over the past two decades. SSCs have potential clinical application for treating some cases of male infertility, providing impetus for characterizing and learning to manipulate these adult tissue stem cells in primates (nonhuman and human). This review recounts the development of a xenotransplant assay for functional identification of primate SSCs and progress dissecting the molecular and clonal characteristics of the primate spermatogenic lineage. These observations highlight the similarities and potential differences between rodents and primates regarding the SSC pool and the kinetics of spermatogonial self-renewal and clonal expansion. With new tools and reagents for studying primate spermatogonia, the field is poised to develop and test new hypotheses about the biology and regenerative capacity of primate SSCs.


Assuntos
Primatas/fisiologia , Roedores/fisiologia , Espermatogônias/fisiologia , Células-Tronco/fisiologia , Adulto , Animais , Proliferação de Células , Humanos , Masculino , Modelos Biológicos , Espermatogênese/fisiologia , Espermatogônias/citologia , Testículo/citologia , Testículo/fisiologia
14.
Dev Cell ; 54(4): 529-547.e12, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32504559

RESUMO

Spermatogenesis is a highly regulated process that produces sperm to transmit genetic information to the next generation. Although extensively studied in mice, our current understanding of primate spermatogenesis is limited to populations defined by state-specific markers from rodent data. As between-species differences have been reported in the duration and differentiation hierarchy of this process, it remains unclear how molecular markers and cell states are conserved or have diverged from mice to man. To address this challenge, we employ single-cell RNA sequencing to identify transcriptional signatures of major germ and somatic cell types of the testes in human, macaque, and mice. This approach reveals similarities and differences in expression throughout spermatogenesis, including the stem/progenitor pool of spermatogonia, markers of differentiation, potential regulators of meiosis, RNA turnover during spermatid differentiation, and germ cell-soma communication. These datasets provide a rich foundation for future targeted mechanistic studies of primate germ cell development and in vitro gametogenesis.


Assuntos
Diferenciação Celular/genética , Análise de Célula Única , Espermatogênese/genética , Testículo/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Macaca/genética , Macaca/crescimento & desenvolvimento , Masculino , Meiose/genética , Camundongos , Análise de Sequência de RNA , Espermatogônias/citologia , Testículo/metabolismo
15.
Hum Reprod ; 24(7): 1704-16, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19336441

RESUMO

BACKGROUND: The spermatogonial stem cell (SSC) pool in the testes of non-human primates is poorly defined. METHODS: To begin characterizing SSCs in rhesus macaque testes, we employed fluorescence-activated cell sorting (FACS), a xenotransplant bioassay and immunohistochemical methods and correlated our findings with classical descriptions of germ cell nuclear morphology (i.e. A(dark) and A(pale) spermatogonia). RESULTS: FACS analysis identified a THY-1+ fraction of rhesus testis cells that was enriched for consensus SSC markers (i.e. PLZF, GFRalpha1) and exhibited enhanced colonizing activity upon transplantation to nude mouse testes. We observed a substantial conservation of spermatogonial markers from mice to monkeys [PLZF, GFRalpha1, Neurogenin 3 (NGN3), cKIT]. Assuming that molecular characteristics correlate with function, the pool of putative SSCs (THY-1+, PLZF+, GFRalpha1+, NGN3+/-, cKIT-) comprises most A(dark) and A(pale) and is considerably larger in primates than in rodents. It is noteworthy that the majority of A(dark) and A(pale) share a common molecular phenotype, considering their distinct functional classifications as reserve and renewing stem cells, respectively. NGN3 is absent from A(dark), but is expressed by some A(pale) and may mark the transition from undifferentiated (cKIT-) to differentiating (cKIT+) spermatogonia. Finally, the pool of transit-amplifying progenitor spermatogonia (PLZF+, GFRalpha1+, NGN3+, cKIT+/-) is smaller in primates than in rodents. CONCLUSIONS These results provide an in-depth analysis of molecular characteristics of primate spermatogonia, including SSCs, and lay a foundation for future studies investigating the kinetics of spermatogonial renewal, clonal expansion and differentiation during primate spermatogenesis.


Assuntos
Linhagem da Célula , Células Germinativas/citologia , Espermatogônias/citologia , Espermatogônias/patologia , Animais , Bioensaio , Diferenciação Celular , Separação Celular , Citometria de Fluxo , Imuno-Histoquímica/métodos , Cinética , Macaca mulatta , Masculino , Fenótipo , Testículo/metabolismo , Testículo/patologia , Transplante Heterólogo
16.
Science ; 363(6433): 1314-1319, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30898927

RESUMO

Testicular tissue cryopreservation is an experimental method to preserve the fertility of prepubertal patients before they initiate gonadotoxic therapies for cancer or other conditions. Here we provide the proof of principle that cryopreserved prepubertal testicular tissues can be autologously grafted under the back skin or scrotal skin of castrated pubertal rhesus macaques and matured to produce functional sperm. During the 8- to 12-month observation period, grafts grew and produced testosterone. Complete spermatogenesis was confirmed in all grafts at the time of recovery. Graft-derived sperm were competent to fertilize rhesus oocytes, leading to preimplantation embryo development, pregnancy, and the birth of a healthy female baby. Pending the demonstration that similar results are obtained in noncastrated recipients, testicular tissue grafting may be applied in the clinic.


Assuntos
Preservação da Fertilidade/métodos , Fertilização , Espermatogênese , Espermatozoides/crescimento & desenvolvimento , Testículo/fisiologia , Testículo/transplante , Animais , Autoenxertos , Criopreservação , Macaca mulatta , Masculino , Reprodução , Maturidade Sexual , Transplante Autólogo
17.
BMC Biotechnol ; 8: 52, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18522747

RESUMO

BACKGROUND: Establishing a suitable level of exogenous gene expression in mammalian cells in general, and embryonic stem (ES) cells in particular, is an important aspect of understanding pathways of cell differentiation, signal transduction and cell physiology. Despite its importance, this process remains challenging because of the poor correlation between the presence of introduced exogenous DNA and its transcription. Consequently, many transfected cells must be screened to identify those with an appropriate level of expression. To improve the screening process, we investigated the utility of the human interleukin 12 (IL-12) p40 cDNA as a reporter gene for studies of mammalian gene expression and for high-throughput screening of engineered mouse embryonic stem cells. RESULTS: A series of expression plasmids were used to study the utility of IL-12 p40 as an accurate reporter of gene activity. These studies included a characterization of the IL-12 p40 expression system in terms of: (i) a time course of IL-12 p40 accumulation in the medium of transfected cells; (ii) the dose-response relationship between the input DNA and IL-12 p40 mRNA levels and IL-12 p40 protein secretion; (iii) the utility of IL-12 p40 as a reporter gene for analyzing the activity of cis-acting genetic elements; (iv) expression of the IL-12 p40 reporter protein driven by an IRES element in a bicistronic mRNA; (v) utility of IL-12 p40 as a reporter gene in a high-throughput screening strategy to identify successful transformed mouse embryonic stem cells; (vi) demonstration of pluripotency of IL-12 p40 expressing ES cells in vitro and in vivo; and (vii) germline transmission of the IL-12 p40 reporter gene. CONCLUSION: IL-12 p40 showed several advantages as a reporter gene in terms of sensitivity and ease of the detection procedure. The IL-12 p40 assay was rapid and simple, in as much as the reporter protein secreted from the transfected cells was accurately measured by ELISA using a small aliquot of the culture medium. Remarkably, expression of Il-12 p40 does not affect the pluripotency of mouse ES cells. To our knowledge, human IL-12 p40 is the first secreted reporter protein suitable for high-throughput screening of mouse ES cells. In comparison to other secreted reporters, such as the widely used alkaline phosphatase (SEAP) reporter, the IL-12 p40 reporter system offers other real advantages.


Assuntos
Bioensaio/métodos , Células-Tronco Embrionárias/fisiologia , Perfilação da Expressão Gênica/métodos , Genes Reporter/genética , Interleucina-12/genética , Interleucina-12/metabolismo , Engenharia de Proteínas/métodos , Animais , Linhagem Celular , Humanos , Camundongos , Proteínas Recombinantes/metabolismo
18.
Stem Cells ; 25(11): 2695-2704, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17641389

RESUMO

Human embryonic stem cells (hESCs) hold great biomedical promise, but experiments comparing them produce heterogeneous results, raising concerns regarding their reliability and utility, although these variations may result from their disparate and anonymous origins. To determine whether primate ESCs have intrinsic biological limitations compared with mouse ESCs, we examined expression profiles and pluripotency of newly established nonhuman primate ESC (nhpESCs). Ten pedigreed nhpESC lines, seven full siblings (fraternal quadruplets and fraternal triplets), and nine half siblings were derived from 41 rhesus embryos; derivation success correlated with embryo quality. Each line has been growing continuously for approximately 1 year with stable diploid karyotype (except for one stable trisomy) and expresses in vitro pluripotency markers, and eight have already formed teratomas. Unlike the heterogeneous gene expression profiles found among hESCs, these nhpESCs display remarkably homogeneous profiles (>97%), with full-sibling lines nearly identical (>98.2%). Female nhpESCs express genes distinct from their brother lines; these sensitive analyses are enabled because of the very low background differences. Experimental comparisons among these primate ESCs may prove more reliable than currently available hESCs, since they are akin to inbred mouse strains in which genetic variables are also nearly eliminated. Finally, contrasting the biological similarities among these lines with the heterogeneous hESCs might suggest that additional, more uniform hESC lines are justified. Taken together, pedigreed primate ESCs display homogeneous and reliable expression profiles. These similarities to mouse ESCs suggest that heterogeneities found among hESCs likely result from their disparate origins rather than intrinsic biological limitations with primate embryonic stem cells.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Linhagem , Animais , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Feminino , Macaca mulatta , Masculino
19.
Stem Cells ; 25(9): 2330-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17585169

RESUMO

Spermatogonial stem cells (SSCs) are at the foundation of mammalian spermatogenesis. Whereas rare A(single) spermatogonia comprise the rodent SSC pool, primate spermatogenesis arises from more abundant A(dark) and A(pale) spermatogonia, and the identity of the stem cell is subject to debate. The fundamental differences between these models highlight the need to investigate the biology of primate SSCs, which have greater relevance to human physiology. The alkylating chemotherapeutic agent, busulfan, ablates spermatogenesis in rodents and causes infertility in humans. We treated adult rhesus macaques with busulfan to gain insights about its effects on SSCs and spermatogenesis. Busulfan treatment caused acute declines in testis volume and sperm counts, indicating a disruption of spermatogenesis. One year following high-dose busulfan treatment, sperm counts remained undetectable, and testes were depleted of germ cells. Similar to rodents, rhesus spermatogonia expressed markers of germ cells (VASA, DAZL) and stem/progenitor spermatogonia (PLZF and GFRalpha1), and cells expressing these markers were depleted following high-dose busulfan treatment. Furthermore, fresh or cryopreserved germ cells from normal rhesus testes produced colonies of spermatogonia, which persisted as chains on the basement membrane of mouse seminiferous tubules in the primate to nude mouse xenotransplant assay. In contrast, testis cells from animals that received high-dose busulfan produced no colonies. These studies provide basic information about rhesus SSC activity and the impact of busulfan on the stem cell pool. In addition, the germ cell-depleted testis model will enable autologous/homologous transplantation to study stem cell/niche interactions in nonhuman primate testes.


Assuntos
Bussulfano/efeitos adversos , Criopreservação , Infertilidade Masculina/induzido quimicamente , Preservação do Sêmen/efeitos adversos , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Animais , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacologia , Bussulfano/farmacologia , Relação Dose-Resposta a Droga , Macaca mulatta , Masculino , Camundongos , Camundongos Nus , Modelos Biológicos , Contagem de Espermatozoides , Espermatogênese/efeitos dos fármacos , Espermatogônias/transplante , Transplante de Células-Tronco , Células-Tronco/citologia , Testículo/anatomia & histologia , Testículo/efeitos dos fármacos , Transplante Heterólogo
20.
Stem Cell Reports ; 10(2): 553-567, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29337115

RESUMO

Undifferentiated spermatogonia comprise a pool of stem cells and progenitor cells that show heterogeneous expression of markers, including the cell surface receptor GFRα1. Technical challenges in isolation of GFRα1+ versus GFRα1- undifferentiated spermatogonia have precluded the comparative molecular characterization of these subpopulations and their functional evaluation as stem cells. Here, we develop a method to purify these subpopulations by fluorescence-activated cell sorting and show that GFRα1+ and GFRα1- undifferentiated spermatogonia both demonstrate elevated transplantation activity, while differing principally in receptor tyrosine kinase signaling and cell cycle. We identify the cell surface molecule melanocyte cell adhesion molecule (MCAM) as differentially expressed in these populations and show that antibodies to MCAM allow isolation of highly enriched populations of GFRα1+ and GFRα1- spermatogonia from adult, wild-type mice. In germ cell culture, GFRα1- cells upregulate MCAM expression in response to glial cell line-derived neurotrophic factor (GDNF)/fibroblast growth factor (FGF) stimulation. In transplanted hosts, GFRα1- spermatogonia yield GFRα1+ spermatogonia and restore spermatogenesis, albeit at lower rates than their GFRα1+ counterparts. Together, these data provide support for a model of a stem cell pool in which the GFRα1+ and GFRα1- cells are closely related but show key cell-intrinsic differences and can interconvert between the two states based, in part, on access to niche factors.


Assuntos
Diferenciação Celular/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Espermatogênese/genética , Espermatogônias/citologia , Animais , Antígeno CD146/genética , Linhagem da Célula/genética , Fatores de Crescimento de Fibroblastos/genética , Citometria de Fluxo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Transdução de Sinais/genética , Espermatogônias/crescimento & desenvolvimento , Nicho de Células-Tronco/genética , Células-Tronco/citologia , Testículo/citologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa