Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Tissue Eng ; 14: 20417314231197282, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38029018

RESUMEN

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.

2.
Stem Cell Res ; 73: 103257, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38000347

RESUMEN

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.


Asunto(s)
Anemia de Células Falciformes , Células Madre Pluripotentes Inducidas , Humanos , Masculino , Niño , Células Madre Pluripotentes Inducidas/patología , Semen , Trasplante de Médula Ósea , Anemia de Células Falciformes/patología , Fibroblastos/patología
3.
Dev Cell ; 57(9): 1160-1176.e5, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35504286

RESUMEN

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.


Asunto(s)
Análisis de la Célula Individual , Testículo , Anciano , Envejecimiento , Índice de Masa Corporal , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Células de Sertoli , Espermatogénesis/genética , Testículo/metabolismo
4.
Nat Commun ; 12(1): 3876, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162856

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Homeostasis/genética , Células Madre Mesenquimatosas/metabolismo , Regeneración/genética , Testículo/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula/genética , Células Cultivadas , Femenino , Perfilación de la Expresión Génica/métodos , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de la Célula Individual/métodos , Testículo/citología
5.
Proc Natl Acad Sci U S A ; 117(30): 17832-17841, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32661178

RESUMEN

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.


Asunto(s)
Transducción de Señal , Espermatogonias/citología , Espermatogonias/metabolismo , Transcriptoma , Biomarcadores , Separación Celular , Células Cultivadas , Biología Computacional , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Masculino , Fosfatidato Fosfatasa/genética , Fosfatidato Fosfatasa/metabolismo
6.
Dev Cell ; 54(4): 529-547.e12, 2020 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-32504559

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Análisis de la Célula Individual , Espermatogénesis/genética , Testículo/crecimiento & desarrollo , Animales , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Macaca/genética , Macaca/crecimiento & desarrollo , Masculino , Meiosis/genética , Ratones , Análisis de Secuencia de ARN , Espermatogonias/citología , Testículo/metabolismo
7.
Science ; 363(6433): 1314-1319, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30898927

RESUMEN

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.


Asunto(s)
Preservación de la Fertilidad/métodos , Fertilización , Espermatogénesis , Espermatozoides/crecimiento & desarrollo , Testículo/fisiología , Testículo/trasplante , Animales , Autoinjertos , Criopreservación , Macaca mulatta , Masculino , Reproducción , Maduración Sexual , Trasplante Autólogo
8.
Nat Commun ; 9(1): 5339, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30559363

RESUMEN

A major challenge in stem cell differentiation is the availability of bioassays to prove cell types generated in vitro are equivalent to cells in vivo. In the mouse, differentiation of primordial germ cell-like cells (PGCLCs) from pluripotent cells was validated by transplantation, leading to the generation of spermatogenesis and to the birth of offspring. Here we report the use of xenotransplantation (monkey to mouse) and homologous transplantation (monkey to monkey) to validate our in vitro protocol for differentiating male rhesus (r) macaque PGCLCs (rPGCLCs) from induced pluripotent stem cells (riPSCs). Specifically, transplantation of aggregates containing rPGCLCs into mouse and nonhuman primate testicles overcomes a major bottleneck in rPGCLC differentiation. These findings suggest that immature rPGCLCs once transplanted into an adult gonadal niche commit to differentiate towards late rPGCs that initiate epigenetic reprogramming but do not complete the conversion into ENO2-positive spermatogonia.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Espermatocitos/citología , Espermatogénesis/fisiología , Espermatogonias/citología , Testículo/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Trasplante Heterólogo , Trasplante Homólogo
9.
Nat Cell Biol ; 20(6): 655-665, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29713018

RESUMEN

Dysregulation of genetic pathways during human germ cell development leads to infertility. Here, we analysed bona fide human primordial germ cells (hPGCs) to probe the developmental genetics of human germ cell specification and differentiation. We examined the distribution of OCT4 occupancy in hPGCs relative to human embryonic stem cells (hESCs). We demonstrated that development, from pluripotent stem cells to germ cells, is driven by switching partners with OCT4 from SOX2 to PAX5 and PRDM1. Gain- and loss-of-function studies revealed that PAX5 encodes a critical regulator of hPGC development. Moreover, an epistasis analysis indicated that PAX5 acts upstream of OCT4 and PRDM1. The PAX5-OCT4-PRDM1 proteins form a core transcriptional network that activates germline and represses somatic programmes during human germ cell differentiation. These findings illustrate the power of combined genome editing, cell differentiation and engraftment for probing human developmental genetics that have historically been difficult to study.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias Humanas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factor de Transcripción PAX5/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Edición Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias Humanas/trasplante , Humanos , Masculino , Ratones Desnudos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor de Transcripción PAX5/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Unión Proteica , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Testículo/embriología , Factores de Tiempo , Transcripción Genética
10.
Stem Cell Reports ; 10(2): 553-567, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29337115

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Espermatogénesis/genética , Espermatogonias/citología , Animales , Antígeno CD146/genética , Linaje de la Célula/genética , Factores de Crecimiento de Fibroblastos/genética , Citometría de Flujo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Ratones , Transducción de Señal/genética , Espermatogonias/crecimiento & desarrollo , Nicho de Células Madre/genética , Células Madre/citología , Testículo/citología
11.
Stem Cell Reports ; 9(1): 329-341, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28579394

RESUMEN

Primordial germ cells (PGCs) are the earliest embryonic progenitors in the germline. Correct formation of PGCs is critical to reproductive health as an adult. Recent work has shown that primate PGCs can be differentiated from pluripotent stem cells; however, a bioassay that supports their identity as transplantable germ cells has not been reported. Here, we adopted a xenotransplantation assay by transplanting single-cell suspensions of human and nonhuman primate embryonic Macaca mulatta (rhesus macaque) testes containing PGCs into the seminiferous tubules of adult busulfan-treated nude mice. We discovered that both human and nonhuman primate embryonic testis are xenotransplantable, generating colonies while not generating tumors. Taken together, this work provides two critical references (molecular and functional) for defining transplantable primate PGCs. These results provide a blueprint for differentiating pluripotent stem cells to transplantable PGC-like cells in a species that is amenable to transplantation and fertility studies.


Asunto(s)
Células Germinativas/trasplante , Túbulos Seminíferos/cirugía , Testículo/embriología , Testículo/trasplante , Animales , Busulfano/uso terapéutico , Femenino , Humanos , Inmunosupresores/uso terapéutico , Macaca mulatta , Masculino , Ratones Desnudos , Trasplante Heterólogo/métodos
12.
Biol Reprod ; 96(3): 707-719, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28339678

RESUMEN

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.


Asunto(s)
ARN Helicasas DEAD-box/genética , Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Modelos Animales , Espermatogénesis , Células Madre Germinales Adultas , Animales , Células Cultivadas , Femenino , Genes Reporteros , Masculino , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Ratas Transgénicas
13.
PLoS One ; 11(10): e0165268, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27768780

RESUMEN

The mechanisms underlying human germ cell development are largely unknown, partly due to the scarcity of primordial germ cells and the inaccessibility of the human germline to genetic analysis. Human embryonic stem cells can differentiate to germ cells in vitro and can be genetically modified to study the genetic requirements for germ cell development. Here, we studied NANOS3 and DAZL, which have critical roles in germ cell development in several species, via their over expression in human embryonic stem cells using global transcriptional analysis, in vitro germ cell differentiation, and in vivo germ cell formation assay by xenotransplantation. We found that NANOS3 over expression prolonged pluripotency and delayed differentiation. In addition, we observed a possible connection of NANOS3 with inhibition of apoptosis. For DAZL, our results suggest a post-transcriptional regulation mechanism in hES cells. In addition, we found that DAZL suppressed the translation of OCT4, and affected the transcription of several genes associated with germ cells, cell cycle arrest, and cell migration. Furthermore, DAZL over expressed cells formed spermatogonia-like colonies in a rare instance upon xenotransplantation. These data can be used to further elucidate the role of NANOS3 and DAZL in germ cell development both in vitro and in vivo.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Unión al ARN/metabolismo , Diferenciación Celular , Femenino , Expresión Génica , Células Germinativas/citología , Xenoinjertos , Humanos , Masculino , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Transcripción Genética
14.
Cell Rep ; 17(1): 149-164, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681428

RESUMEN

The developmental origins of most adult stem cells are poorly understood. Here, we report the identification of a transcription factor-RHOX10-critical for the initial establishment of spermatogonial stem cells (SSCs). Conditional loss of the entire 33-gene X-linked homeobox gene cluster that includes Rhox10 causes progressive spermatogenic decline, a phenotype indistinguishable from that caused by loss of only Rhox10. We demonstrate that this phenotype results from dramatically reduced SSC generation. By using a battery of approaches, including single-cell-RNA sequencing (scRNA-seq) analysis, we show that Rhox10 drives SSC generation by promoting pro-spermatogonia differentiation. Rhox10 also regulates batteries of migration genes and promotes the migration of pro-spermatogonia into the SSC niche. The identification of an X-linked homeobox gene that drives the initial generation of SSCs has implications for the evolution of X-linked gene clusters and sheds light on regulatory mechanisms influencing adult stem cell generation in general.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Ligados a X , Proteínas de Homeodominio/genética , Espermatogénesis/genética , Espermatogonias/metabolismo , Células Madre Germinales Adultas/citología , Animales , Genes del Desarrollo , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Noqueados , Familia de Multigenes , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Espermatogonias/citología
15.
Biol Reprod ; 94(1): 11, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26607719

RESUMEN

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.


Asunto(s)
Transducción de Señal/fisiología , Espermatogénesis/fisiología , Testosterona/fisiología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Fertilidad/efectos de los fármacos , Fertilidad/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Proteínas Proto-Oncogénicas c-kit/genética , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Testículo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Genes Dev ; 29(23): 2420-34, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26584619

RESUMEN

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.


Asunto(s)
Células Madre Adultas/enzimología , Regulación Enzimológica de la Expresión Génica , Espermatogonias/enzimología , Telomerasa/genética , Telomerasa/metabolismo , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/enzimología , Citometría de Flujo , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética
17.
Sci Rep ; 5: 15041, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26456624

RESUMEN

Deletions of the AZFa region (AZoospermia Factor-a) region of the human Y chromosome cause irreversible spermatogenic failure that presents clinically in men as Sertoli-cell only (SCO) pathology of the testis. Deletions of the AZFa region typically encompass two genes: DDX3Y and USP9Y. However, human genetic evidence indicates that SCO is most tightly linked to deletion of DDX3Y and that deletions/mutations of USP9Y can be transmitted from one generation to the next. Here, we generated stable iPSC lines with AZFa deletions, tested complementation via introduction of DDX3Y, and assessed ability to form germ cells in vivo in a xenotransplantation model. We observed a quantifiable improvement in formation of germ cell like cells (GCLCs) from complemented donor iPSCs. Moreover, expression of UTF1, a prospermatogonial protein, was restored in cells complemented by introduction of DDX3Y on the AZFa background. Whole-genome RNA sequencing of purified GCLCs revealed an enrichment of genes involved in translational suppression and transcriptional control in DDX3Y-rescued GCLCs over mutant GCLCs, which maintained a molecular phenotype more similar to undifferentiated iPSCs. This study demonstrates the ability to probe fundamental genetics of human germ cell formation by complementation and indicates that DDX3Y functions in the earliest stages of human germ cell development.


Asunto(s)
Cromosomas Humanos Y/metabolismo , ARN Helicasas DEAD-box/genética , Células Madre Pluripotentes Inducidas/citología , Espermatogénesis/genética , Espermatozoides/metabolismo , Transcripción Genética , Animales , Busulfano/farmacología , Diferenciación Celular , Cromosomas Humanos Y/química , ARN Helicasas DEAD-box/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Prueba de Complementación Genética , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Desnudos , Antígenos de Histocompatibilidad Menor , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Piel/citología , Piel/metabolismo , Espermatozoides/citología , Testículo/citología , Testículo/efectos de los fármacos , Testículo/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Trasplante Heterólogo , Proteína Fluorescente Roja
18.
Sci Rep ; 4: 6432, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25242416

RESUMEN

Turner syndrome is caused by complete or partial loss of the second sex chromosome and is characterized by spontaneous fetal loss in >90% of conceptions. Survivors possess an array of somatic and germline clinical characteristics. Induced pluripotent stem cells (iPSCs) offer an opportunity for insight into genetic requirements of the X chromosome linked to Turner syndrome. We derived iPSCs from Turner syndrome and control individuals and examined germ cell development as a function of X chromosome composition. We demonstrate that two X chromosomes are not necessary for reprogramming or maintenance of pluripotency and that there are minimal differences in gene expression, at the single cell level, linked to X chromosome aneuploidies. Formation of germ cells, as assessed in vivo through a murine xenotransplantation model, indicated that undifferentiated iPSCs, independent of X chromosome composition, are capable of forming germ-cell-like cells (GCLCs) in vivo. In combination with clinical data regarding infertility in women with X chromosome aneuploidies, results suggest that two intact X chromosomes are not required for human germ cell formation, qualitatively or quantitatively, but rather are likely to be required for maintenance of human germ cells to adulthood.


Asunto(s)
Cromosomas Humanos X/genética , Células Germinativas/patología , Células Madre Pluripotentes Inducidas/patología , Síndrome de Turner/genética , Aneuploidia , Animales , Femenino , Expresión Génica , Células Germinativas/crecimiento & desarrollo , Humanos , Ratones , Análisis de la Célula Individual , Trasplante Heterólogo , Síndrome de Turner/patología
19.
Fertil Steril ; 102(4): e11-2, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25150391

RESUMEN

OBJECTIVE: To illustrate the step-by-step protocol followed to assay germ cell transplantation into the seminiferous epithelium of mouse testes. DESIGN: Video presentation of an animal model for research in reproductive and regenerative medicine. SETTING: Research laboratory. ANIMAL(S): Male nude mice (NU-Foxn1(nu)). INTERVENTION(S): Mice were chemically sterilized with alkylant compounds (busulfan) followed by gonadal microsurgery to inject donor germ cells. MAIN OUTCOME MEASURE(S): Donor cells should be labeled with reporter genes, such as green fluorescent protein (GFP), lactose operon (LacZ), or alternatively design an effective strategy with specific antibodies to track them within the recipient testes. Sperm detection in the ejaculate can also be used as a read out. However, in this case detection of the donor genotype in the sperm is mandatory to elucidate their origin. RESULT(S): In the present study we describe the complete protocol for germ cell transplant by efferent duct injection, including the preparation of recipient mice, surgery for the germ cell transplant, and analysis of recipient testes. The main strength of this technique is that it constitutes the gold standard for a functional test of the germ cell potential as only spermatogonial stem cells are able to properly colonize the seminal lumen. Both fresh and frozen/thawed testicular cells are suitable for this technique as donor germ cells. Also, enrichment of living spermatogonial stem cells, previous to the transplant, seems to improve the efficiency of colonization. For proper colonization of germ cells, the niche should be available and thus mouse strains that lack endogenous spermatogenesis such as W/W(v) mutant mice are usually used. In the case of nonmatched donor cells, seminiferous epithelium of immune-suppressed recipient mice should be germ cell depleted before the transplant. One limitation of this technique is that the procedure can take up to 3 months. Also, in contrast to the full recovery of spermatogenesis in mouse-to-mouse transplants, xenotransplantation of germ cells from phylogenetically distant species, such as humans into mouse recipients, results in colonization of donor cells and spermatogonial expansion, but fail in their spermatogenic progression due to evolutive incompatibilities with the recipient niche. Xenografting of pieces of donor testis tissue under the skin of mouse hosts is an alternative approach that is currently being investigated to try to solve this limitation. CONCLUSION(S): Transplantation of spermatogonial stem cells into the seminal lumen of mouse testes is a functional assay that defines this cellular subpopulation by its ability to colonize it. This technique can be used as a model to elucidate the insights of spermatogonial stem cells, to produce transgenic animals by genetically manipulating donor cells before transplantation, but also it has potential applications in fertility preservation in cattle and humans as it is feasible in large animals, as recent reports have demonstrated with rhesus monkeys, that recovered spermatogenesis after allogenic transplantation, and even from human cadaver testes. Therefore spermatogonial stem cells isolated from prepuberal boys, who are treated with alkylant chemotherapy, could be returned to their testis to regenerate spermatogenesis in the future.


Asunto(s)
Epitelio Seminífero/cirugía , Espermatogonias/trasplante , Procedimientos Quirúrgicos Urológicos Masculinos/métodos , Animales , Rastreo Celular , Criopreservación , Genes Reporteros , Genotipo , Masculino , Ratones Desnudos , Microcirugia , Epitelio Seminífero/fisiopatología , Espermatogénesis , Espermatogonias/fisiología , Esterilización Reproductiva , Transfección
20.
Fertil Steril ; 102(2): 566-580.e7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24890267

RESUMEN

OBJECTIVE: To determine the molecular characteristics of human spermatogonia and optimize methods to enrich spermatogonial stem cells (SSCs). DESIGN: Laboratory study using human tissues. SETTING: Research institute. PATIENT(S): Healthy adult human testicular tissue. INTERVENTION(S): Human testicular tissue was fixed or digested with enzymes to produce a cell suspension. Human testis cells were fractionated by fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS). MAIN OUTCOME MEASURE(S): Immunostaining for selected markers, human-to-nude mouse xenotransplantation assay. RESULT(S): Immunohistochemistry costaining revealed the relative expression patterns of SALL4, UTF1, ZBTB16, UCHL1, and ENO2 in human undifferentiated spermatogonia as well as the extent of overlap with the differentiation marker KIT. Whole mount analyses revealed that human undifferentiated spermatogonia (UCHL1+) were typically arranged in clones of one to four cells whereas differentiated spermatogonia (KIT+) were typically arranged in clones of eight or more cells. The ratio of undifferentiated-to-differentiated spermatogonia is greater in humans than in rodents. The SSC colonizing activity was enriched in the THY1dim and ITGA6+ fractions of human testes sorted by FACS. ITGA6 was effective for sorting human SSCs by MACS; THY1 and EPCAM were not. CONCLUSION(S): Human spermatogonial differentiation correlates with increased clone size and onset of KIT expression, similar to rodents. The undifferentiated-to-differentiated developmental dynamics in human spermatogonia is different than rodents. THY1, ITGA6, and EPCAM can be used to enrich human SSC colonizing activity by FACS, but only ITGA6 is amenable to high throughput sorting by MACS.


Asunto(s)
Células Madre Adultas/metabolismo , Separación Celular/métodos , Citometría de Flujo , Separación Inmunomagnética , Espermatogonias/metabolismo , Testículo/metabolismo , Células Madre Adultas/trasplante , Animales , Antígenos de Neoplasias/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Molécula de Adhesión Celular Epitelial , Humanos , Integrina alfa6/metabolismo , Masculino , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Espermatogonias/trasplante , Testículo/citología , Testículo/trasplante , Antígenos Thy-1/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA