Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 147(13)2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32554530

RESUMEN

In mammalian testis, contractile peritubular myoid cells (PMCs) regulate the transport of sperm and luminal fluid, while secreting growth factors and extracellular matrix proteins to support the spermatogonial stem cell niche. However, little is known about the role of testicular smooth muscle cells during postnatal testicular development. Here we report age-dependent expression of hypermethylated in cancer 1 (Hic1; also known as ZBTB29) in testicular smooth muscle cells, including PMCs and vascular smooth muscle cells, in the mouse. Postnatal deletion of Hic1 in smooth muscle cells led to their increased proliferation and resulted in dilatation of seminiferous tubules, with increased numbers of PMCs. These seminiferous tubules contained fewer Sertoli cells and more spermatogonia, and fibronectin was not detected in their basement membrane. The expression levels of genes encoding smooth muscle contractile proteins, Acta2 and Cnn1, were downregulated in the smooth muscle cells lacking Hic1, and the seminiferous tubules appeared to have reduced contractility. These data imply a role for Hic1 in determining the size of seminiferous tubules by regulating postnatal smooth muscle cell proliferation, subsequently affecting spermatogenesis in adulthood.


Asunto(s)
Fibronectinas/metabolismo , Miocitos del Músculo Liso/metabolismo , Testículo/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Fibronectinas/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo
2.
FASEB J ; 35(5): e21513, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811704

RESUMEN

Human male reproductive development has a prolonged prepubertal period characterized by juvenile quiescence of germ cells with immature spermatogonial stem cell (SSC) precursors (gonocytes) present in the testis for an extended period of time. The metabolism of gonocytes is not defined. We demonstrate with mitochondrial ultrastructure studies via TEM and IHC and metabolic flux studies with UHPLC-MS that a distinct metabolic transition occurs during the maturation to SSCs. The mitochondrial ultrastructure of prepubertal human spermatogonia is shared with prepubertal pig spermatogonia. The metabolism of early prepubertal porcine spermatogonia (gonocytes) is characterized by the reliance on OXPHOS fuelled by oxidative decarboxylation of pyruvate. Interestingly, at the same time, a high amount of the consumed pyruvate is also reduced and excreted as lactate. With maturation, prepubertal spermatogonia show a metabolic shift with decreased OXHPOS and upregulation of the anaerobic metabolism-associated uncoupling protein 2 (UCP2). This shift is accompanied with stem cell specific promyelocytic leukemia zinc finger protein (PLZF) protein expression and glial cell-derived neurotropic factor (GDNF) pathway activation. Our results demonstrate that gonocytes differently from mature spermatogonia exhibit unique metabolic demands that must be attained to enable their maintenance and growth in vitro.


Asunto(s)
Regulación de la Expresión Génica , Células Germinativas/metabolismo , Estrés Oxidativo , Células Madre/metabolismo , Testículo/metabolismo , Animales , Células Germinativas/citología , Glucólisis , Humanos , Masculino , Potencial de la Membrana Mitocondrial , Fenotipo , Células Madre/citología , Porcinos , Testículo/citología
3.
Proc Natl Acad Sci U S A ; 116(51): 25677-25687, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31754036

RESUMEN

Mammalian primordial germ cells (PGCs) are induced in the embryonic epiblast, before migrating to the nascent gonads. In fish, frogs, and birds, the germline segregates even earlier, through the action of maternally inherited germ plasm. Across vertebrates, migrating PGCs retain a broad developmental potential, regardless of whether they were induced or maternally segregated. In mammals, this potential is indicated by expression of pluripotency factors, and the ability to generate teratomas and pluripotent cell lines. How the germline loses this developmental potential remains unknown. Our genome-wide analyses of embryonic human and mouse germlines reveal a conserved transcriptional program, initiated in PGCs after gonadal colonization, that differentiates germ cells from their germline precursors and from somatic lineages. Through genetic studies in mice and pigs, we demonstrate that one such gonad-induced factor, the RNA-binding protein DAZL, is necessary in vivo to restrict the developmental potential of the germline; DAZL's absence prolongs expression of a Nanog pluripotency reporter, facilitates derivation of pluripotent cell lines, and causes spontaneous gonadal teratomas. Based on these observations in humans, mice, and pigs, we propose that germ cells are determined after gonadal colonization in mammals. We suggest that germ cell determination was induced late in embryogenesis-after organogenesis has begun-in the common ancestor of all vertebrates, as in modern mammals, where this transition is induced by somatic cells of the gonad. We suggest that failure of this process of germ cell determination likely accounts for the origin of human testis cancer.


Asunto(s)
Diferenciación Celular/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Células Germinativas , Gónadas , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Femenino , Células Germinativas/metabolismo , Células Germinativas/fisiología , Gónadas/citología , Gónadas/fisiología , Masculino , Ratones , Neoplasias Ováricas/genética , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Porcinos , Teratoma/genética , Neoplasias Testiculares/genética
4.
Int J Mol Sci ; 23(9)2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35562927

RESUMEN

Spermatogonial stem cells (SSCs) provide the basis for lifelong male fertility through self-renewal and differentiation. Prepubertal male cancer patients may be rendered infertile by gonadotoxic chemotherapy and, unlike sexually mature men, cannot store sperm. Alternatively, testicular biopsies taken prior to treatment may be used to restore fertility in adulthood. Testicular SSC populations are limited, and in vitro culture systems are required to increase numbers of SSCs for treatment, demanding culture systems for SSC propagation. Using the pig as a non-rodent model, we developed culture systems to expand spermatogonia from immature testis tissue, comparing different feeders (Sertoli cells, peritubular myoid cells (PMCs) and pig fetal fibroblasts (PFFs)). Spermatogonia co-cultured with Sertoli cells, PMCs and PFFs had comparable rates of proliferation and apoptosis. To elucidate the mechanism behind the beneficial nature of feeder layers, we investigated the role of extracellular vesicles in crosstalk between spermatogonia and feeder cells. Sertoli cell-released exosomes are incorporated by spermatogonia, and inhibition of exosomal release reduces spermatogonial proliferation. Together, these results show that PMCs, PFFs and Sertoli cells promote spermatogonial proliferation in co-culture, with exosomal exchange representing one possible mechanism. Further characterization of exosomal cargo may ultimately allow the development of feeder-free culture systems for clinical use.


Asunto(s)
Vesículas Extracelulares , Espermatogonias , Adulto , Animales , Células Cultivadas , Técnicas de Cocultivo , Humanos , Masculino , Células de Sertoli , Porcinos , Testículo
5.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670439

RESUMEN

The spermatogonial stem cell (SSC) is a unique adult stem cell that requires tight physiological regulation during development and adulthood. As the foundation of spermatogenesis, SSCs are a potential tool for the treatment of infertility. Understanding the factors that are necessary for lifelong maintenance of a SSC pool in vivo is essential for successful in vitro expansion and safe downstream clinical usage. This review focused on the current knowledge of prepubertal testicular development and germ cell metabolism in different species, and implications for translational medicine. The significance of metabolism for cell biology, stem cell integrity, and fate decisions is discussed in general and in the context of SSC in vivo maintenance, differentiation, and in vitro expansion.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Espermatogénesis/fisiología , Espermatogonias/fisiología , Adulto , Células Madre Germinales Adultas/citología , Animales , Células Cultivadas , Humanos , Masculino , Espermatogonias/citología
6.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34948348

RESUMEN

Male survivors of childhood cancer are at risk of suffering from infertility in adulthood because of gonadotoxic chemotherapies. For adult men, sperm collection and preservation are routine procedures prior to treatment; however, this is not an option for pre-pubertal children. From young boys, a small biopsy may be taken before chemotherapy, and spermatogonia may be propagated in vitro for future transplantation to restore fertility. A robust system that allows for scalable expansion of spermatogonia within a controlled environment is therefore required. Stirred suspension culture has been applied to different types of stem cells but has so far not been explored for spermatogonia. Here, we report that pre-pubertal porcine spermatogonia proliferate more in bioreactor suspension culture, compared with static culture. Interestingly, oxygen tension provides an avenue to modulate spermatogonia status, with culture under 10% oxygen retaining a more undifferentiated state and reducing proliferation in comparison with the conventional approach of culturing under ambient oxygen levels. Spermatogonia grown in bioreactors upregulate the Wnt/ ß-catenin pathway, which, along with enhanced gas and nutrient exchange observed in bioreactor culture, may synergistically account for higher spermatogonia proliferation. Therefore, stirred suspension bioreactors provide novel platforms to culture spermatogonia in a scalable manner and with minimal handling.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Espermatogonias/fisiología , Suspensiones , Vía de Señalización Wnt , Animales , Masculino , Espermatogonias/metabolismo , Sus scrofa
7.
Cell Tissue Res ; 380(1): 191-200, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31900662

RESUMEN

Most mammalian cells possess a single, non-motile primary cilium that plays an important role in mediating cellular signaling pathways, such as Hedgehog (Hh) signaling. Primary cilia are present on testicular somatic cells and demonstrate a temporal expression during development; however, their role in testicular morphogenesis is not well characterized. To investigate the role of primary cilia and Hh signaling in Sertoli cells on morphogenesis, we inhibited assembly of primary cilia through CRISPR Cas9-mediated gene editing of ODF2, a structural component of primary cilia and siRNA-mediated gene silencing of IFT88, a functional component of the intraflagellar transport system. Knockdown of ODF2 and IFT88 resulted in a 50% reduction in the number of cells with primary cilia and significant shortening of the remaining cilia. The expression of GLI1, a downstream target of Hh signaling, was significantly reduced when IFT88 but not ODF2, was downregulated. When morphogenesis was examined using tubule formation in vitro and a novel testicular organoid system, loss of cilia after knockdown of both targets affected cellular assembly and organization. While the Hh pathway was found to be active during morphogenesis in vitro, addition of the Hh antagonist cyclopamine did not affect morphogenesis in either in vitro system. These results indicate that primary cilia are important for morphogenesis in vitro but Hh signaling is not the cilia-mediated pathway responsible for orchestrating morphogenic organization.


Asunto(s)
Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Organoides/metabolismo , Animales , Masculino , Morfogénesis , Transducción de Señal , Porcinos , Testículo , Transfección
8.
Biol Reprod ; 100(6): 1648-1660, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30927418

RESUMEN

Three-dimensional (3D) organoids can serve as an in vitro platform to study cell-cell interactions, tissue development, and toxicology. Development of organoids with tissue architecture similar to testis in vivo has remained a challenge. Here, we present a microwell aggregation approach to establish multicellular 3D testicular organoids from pig, mouse, macaque, and human. The organoids consist of germ cells, Sertoli cells, Leydig cells, and peritubular myoid cells forming a distinct seminiferous epithelium and interstitial compartment separated by a basement membrane. Sertoli cells in the organoids express tight junction proteins claudin 11 and occludin. Germ cells in organoids showed an attenuated response to retinoic acid compared to germ cells in 2D culture indicating that the tissue architecture of the organoid modulates response to retinoic acid similar to in vivo. Germ cells maintaining physiological cell-cell interactions in organoids also had lower levels of autophagy indicating lower levels of cellular stress. When organoids were treated with mono(2-ethylhexyl) phthalate (MEHP), levels of germ cell autophagy increased in a dose-dependent manner, indicating the utility of the organoids for toxicity screening. Ablation of primary cilia on testicular somatic cells inhibited the formation of organoids demonstrating an application to screen for factors affecting testicular morphogenesis. Organoids can be generated from cryopreserved testis cells and preserved by vitrification. Taken together, the testicular organoid system recapitulates the 3D organization of the mammalian testis and provides an in vitro platform for studying germ cell function, testicular development, and drug toxicity in a cellular context representative of the testis in vivo.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/citología , Testículo/citología , Andamios del Tejido , Animales , Recuento de Células , Técnicas de Cultivo de Célula/instrumentación , Preescolar , Dietilhexil Ftalato/análogos & derivados , Dietilhexil Ftalato/farmacología , Humanos , Lactante , Macaca mulatta , Masculino , Ratones , Organoides/fisiología , Espermatogénesis/efectos de los fármacos , Espermatogénesis/fisiología , Espermatogonias/citología , Espermatogonias/efectos de los fármacos , Espermatogonias/fisiología , Porcinos , Andamios del Tejido/química , Tretinoina/farmacología
9.
Mol Reprod Dev ; 85(3): 250-261, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29393557

RESUMEN

Spermatogonia represent a diploid germ cell population that includes spermatogonial stem cells. In this report, we describe new methods for isolation of highly enriched porcine spermatogonia based on light scatter properties, and for targeted mutagenesis in porcine spermatogonia using nucleofection and TALENs. We optimized a nucleofection protocol to deliver TALENs specifically targeting the DMD locus in porcine spermatogonia. We also validated specific sorting of porcine spermatogonia based on light scatter properties. We were able to obtain a highly enriched germ cell population with over 90% of cells being UCH-L1 positive undifferentiated spermatogonia. After gene targeting in porcine spermatogonia, indel (insertion or deletion) mutations as a result of non-homologous end joining (NHEJ) were detected in up to 18% of transfected cells. Our report demonstrates for the first time an approach to obtain a live cell population highly enriched in undifferentiated spermatogonia from immature porcine testes, and that gene targeting can be achieved in porcine spermatogonia which will enable germ line modification.


Asunto(s)
Marcación de Gen/veterinaria , Espermatogonias/metabolismo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Animales , Edición Génica/veterinaria , Masculino , Espermatogénesis , Espermatogonias/citología , Porcinos , Testículo/metabolismo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo
10.
Cell Tissue Res ; 368(1): 215-223, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27841005

RESUMEN

The primary cilium is a microtubule-based sensory organelle found on nearly all eukaryotic cells but little is understood about its function in the testis. We investigate the role of primary cilia on testis cells in vitro by inhibiting formation of the primary cilium with Ciliobrevin D, a cell-permeable, reversible chemical inhibitor of ATPase motor cytoplasmic dynein. We analyzed cultured cells for the presence of primary cilia and their involvement in hedgehog signaling. Primary cilia were present on 89.3 ± 2.3 % of untreated testicular somatic cells compared to 3.1 ± 2.5 % cells with primary cilia for Ciliobrevin D-treated cells. Protein levels of Gli-2 and Smoothened were lower on Western blots after suppression of cilia with Ciliobrevin D. The inhibitor did not affect centrosome localization or cell proliferation, indicating that changes were due to ablation of the primary cilium. Testicular somatic cells have the ability to form three-dimensional tubules in vitro. In vitro-formed tubules were significantly longer and wider in the control group than in the Ciliobrevin D-treated group (9.91 ± 0.35 vs. 5.540 ± 1.08 mm and 339.8 ± 55.78 vs. 127.2 ± 11.9 µm, respectively) indicating that primary cilia play a role in tubule formation. Our results establish that the inhibition of ATPase motor cytoplasmic dynein perturbs formation of primary cilia in testicular somatic cells, affects the hedgehog signaling pathway and impairs tubule formation in vitro. These findings provide evidence for a role of cilia in the testis in cell signaling and tubular morphogenesis in vitro.


Asunto(s)
Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Morfogénesis , Transducción de Señal , Testículo/citología , Animales , Proliferación Celular/efectos de los fármacos , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Cilios/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Masculino , Morfogénesis/efectos de los fármacos , Quinazolinonas/farmacología , Transducción de Señal/efectos de los fármacos , Sus scrofa , Testículo/efectos de los fármacos , Testículo/metabolismo
11.
Cell Tissue Res ; 358(2): 597-605, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25107611

RESUMEN

In vertebrates, a variety of cell types generate a primary cilium. Cilia are implicated in determination and differentiation of a wide variety of organs and during embryonic development. However, there is little information on the presence or function of primary cilia in the mammalian testis. Therefore, the objective of this study was to characterize expression of primary cilia in the developing pig testis. Testicular tissue from pigs at 2-10 weeks of age was analyzed for primary cilia by immunocytochemistry. Expression of primary cilia was also analyzed in testicular tissue formed de novo from a single cell suspension ectopically grafted into a mouse host. Functionality of primary cilia was monitored based on cilia elongation after exposure to lithium. Analysis showed that the primary cilium is present in testis cords as well as in the interstitium of the developing pig testis. Germ cells did not express primary cilia. However, we identified Sertoli cells as one of the somatic cell types that produce a primary cilium within the developing testis. Primary cilium expression was reduced from the second to the third week of pig testis development in situ and during de novo morphogenesis of testis tissue from a single cell suspension after xenotransplantation. In vitro, primary cilia were elongated in response to lithium treatment. These results indicate that primary cilia on Sertoli cells may function during testicular development. De novo morphogenesis of testis tissue from single cell suspensions may provide an accessible platform to study and manipulate expression and function of primary cilia.


Asunto(s)
Cilios/metabolismo , Sus scrofa/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Inmunohistoquímica , Masculino , Ratones Desnudos , Células de Sertoli/citología , Células de Sertoli/metabolismo , Testículo/citología , Trasplante Heterólogo
12.
Reproduction ; 148(5): R71-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25150043

RESUMEN

Spermatogenesis is a dynamic and complex process that involves endocrine and testicular factors. During xenotransplantation of testicular tissue fragments into immunodecifient mice, a functional communication between host brain and donor testis is established. This interaction allows for the progression of spermatogenesis and recovery of fertilisation-competent spermatozoa from a broad range of mammalian species. In the last few years, significant progress has been achieved in testis tissue xenografting that improves our knowledge about the factors determining the success of grafting. The goal of this review is to provide up to date information about the role of factors such as donor age, donor species, testis tissue preservation or type of recipient mouse on the efficiency of this technique. Applications are described and compared with other techniques with similar purposes. Recent work has demonstrated that testicular tissue xenografting is used as a model to study gonadotoxicity of drugs and to obtain sperm from valuable young males.


Asunto(s)
Espermatogénesis , Espermatozoides/trasplante , Testículo/trasplante , Factores de Edad , Animales , Supervivencia de Injerto , Humanos , Masculino , Ratones SCID , Recuperación de la Función , Especificidad de la Especie , Espermatozoides/inmunología , Espermatozoides/metabolismo , Testículo/crecimiento & desarrollo , Testículo/inmunología , Testículo/metabolismo , Tolerancia al Trasplante , Trasplante Heterólogo
13.
Reproduction ; 148(1): 109-17, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24803491

RESUMEN

De novo formation of testis tissue from single-cell suspensions allows manipulation of different testicular compartments before grafting to study testicular development and the spermatogonial stem cell niche. However, the low percentages of newly formed seminiferous tubules supporting complete spermatogenesis and lack of a defined protocol have limited the use of this bioassay. Low spermatogenic efficiency in de novo formed tissue could result from the scarcity of germ cells in the donor cell suspension, cell damage caused by handling or from hypoxia during tissue formation in the host environment. In this study, we compared different proportions of spermatogonia in the donor cell suspension and the use of Matrigel as a scaffold to support de novo tissue formation and spermatogenesis. Then, we used the system to investigate the role of vascular endothelial growth factor 165 (VEGF165) during testicular morphogenesis on blood vessel and seminiferous tubule formation, and on presence of germ cells in the de novo developed tubules. Our results show that donor cell pellets with 10×10(6) porcine neonatal testicular cells in Matrigel efficiently formed testis tissue de novo. Contrary to what was expected, the enrichment of the cell suspension with germ cells did not result in higher numbers of tubules supporting spermatogenesis. The addition of VEGF165 did not improve blood vessel or tubule formation, but it enhanced the number of tubules containing spermatogonia. These results indicate that spermatogenic efficiency was improved by the addition of Matrigel, and that VEGF165 may have a protective role supporting germ cell establishment in their niche.


Asunto(s)
Transducción de Señal/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Espermatogonias/trasplante , Testículo/efectos de los fármacos , Testículo/trasplante , Ingeniería de Tejidos/métodos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Animales Recién Nacidos , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Colágeno/metabolismo , Combinación de Medicamentos , Supervivencia de Injerto/efectos de los fármacos , Laminina/metabolismo , Masculino , Ratones SCID , Morfogénesis/efectos de los fármacos , Orquiectomía , Proteoglicanos/metabolismo , Proteínas Recombinantes/farmacología , Espermatogénesis/efectos de los fármacos , Espermatogonias/metabolismo , Sus scrofa , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Tiempo , Andamios del Tejido
14.
Stem Cells ; 31(10): 2205-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836497

RESUMEN

Studies on spermatogonial stem cells (SSCs) are of unusual significance because they are the unique stem cells that transmit genetic information to subsequent generations and they can acquire pluripotency to become embryonic stem-like cells that have therapeutic applications in human diseases. MicroRNAs (miRNAs) have recently emerged as critical endogenous regulators in mammalian cells. However, the function and mechanisms of individual miRNAs in regulating SSC fate remain unknown. Here, we report for the first time that miRNA-20 and miRNA-106a are preferentially expressed in mouse SSCs. Functional assays in vitro and in vivo using miRNA mimics and inhibitors reveal that miRNA-20 and miRNA-106a are essential for renewal of SSCs. We further demonstrate that these two miRNAs promote renewal at the post-transcriptional level via targeting STAT3 and Ccnd1 and that knockdown of STAT3, Fos, and Ccnd1 results in renewal of SSCs. This study thus provides novel insights into molecular mechanisms regulating renewal and differentiation of SSCs and may have important implications for regulating male reproduction.


Asunto(s)
Células Madre Adultas/fisiología , Ciclina D1/genética , MicroARNs/fisiología , Interferencia de ARN , Factor de Transcripción STAT3/genética , Animales , Secuencia de Bases , Proliferación Celular , Células Cultivadas , Ciclina D1/metabolismo , Femenino , Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Familia de Multigenes , Fenotipo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Factor de Transcripción STAT3/metabolismo , Espermatogénesis/genética , Testículo/citología
15.
Reprod Fertil Dev ; 26(6): 817-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23763851

RESUMEN

The use of assisted reproductive techniques for endangered species is a major goal for conservation. One of these techniques, testis tissue xenografting, allows for the development of spermatozoa from animals that die before reaching sexual maturity. To assess the potential use of this technique with endangered species, testis tissue from six Iberian lynxes (one fetus, two perinatal cubs, two 6-month-old and one 2-year-old lynx), two Cuvier's gazelle fetuses and one 8-month-old Mohor gazelle were transplanted ectopically into nude mice. Tissue from the lynx fetus, perinatal cubs and 2-year-old donors degenerated, whereas spermatogonia were present in 15% of seminiferous tubules more than 70 weeks after grafting in transplanted testis tissue from 6-month-old donors. Seminal vesicle weights (indicative of testosterone production) increased over time in mice transplanted with tissue from 6-month-old lynxes. Progression of spermatogenesis was observed in xenografts from gazelles and was donor age dependent. Tissue from Cuvier's gazelle fetuses contained spermatocytes 40 weeks after grafting. Finally, round spermatids were found 28 weeks after transplantation in grafts from the 8-month-old Mohor gazelle. This is the first time that xenotransplantation of testicular tissue has been performed with an endangered felid and the first successful xenotransplantation in an endangered species. Our results open important options for the preservation of biological diversity.


Asunto(s)
Antílopes , Especies en Peligro de Extinción , Células Germinativas/fisiología , Supervivencia de Injerto/fisiología , Lynx , Espermatogénesis , Espermatozoides/fisiología , Testículo/trasplante , Animales , Diferenciación Celular , Supervivencia Celular , Conservación de los Recursos Naturales/métodos , Xenoinjertos , Masculino , Ratones , Ratones Desnudos , Recuperación de la Esperma/veterinaria , Testículo/citología
16.
Biol Reprod ; 88(1): 27, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23221397

RESUMEN

Genetic modification of germline stem cells (GSCs) is an alternative approach to generate large transgenic animals where transgenic GSCs are transplanted into a recipient testis to generate donor-derived transgenic sperm. The objective of the present study was to explore the application of viral vectors in delivering an enhanced green fluorescent protein (EGFP) transgene into GSCs for production of transgenic gametes through germ cell transplantation. Both adeno-associated virus (AAV)- and lentivirus (LV)-based vectors were effective in transducing pig GSCs, resulting in the production of transgenic sperm in recipient boars. Twenty-one boars treated with busulfan to deplete endogenous GSCs and nine nontreated boars received germ cell transplantation at 12 wk of age. Semen was collected from recipient boars from 5 to 7 mo posttransplantation when boars became sexually mature, and semen collection continued for as long as 5 yr for some boars. The percentage of ejaculates that were positive for the EGFP transgene ranged from 0% to 54.8% for recipients of AAV vector-transduced germ cells (n = 17) and from 0% to 25% for recipients of LV vector-transduced germ cells (n = 5). When semen from two AAV recipients was used for in vitro fertilization (IVF), 9.09% and 64.3% of embryos were transgenic. Semen collected from two LV-vector recipients produced 7.7% and 26.3% transgenic IVF embryos. Here, we not only demonstrated AAV-mediated GSC transduction in another large animal model (pigs) but also showed, to our knowledge for the first time, that LV-mediated GSC transduction resulted in transgene transmission in pigs.


Asunto(s)
Células Germinativas/trasplante , Proteínas Fluorescentes Verdes/metabolismo , Porcinos/genética , Transducción Genética/veterinaria , Animales , Animales Modificados Genéticamente , Dependovirus , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/genética , Lentivirus , Masculino , Espermatozoides
17.
Nat Cell Biol ; 8(12): 1448-54, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17115026

RESUMEN

Lymphoid specific helicase (Lsh) is a major epigenetic regulator that is essential for DNA methylation and transcriptional silencing of parasitic elements in the mammalian genome. However, whether Lsh is involved in the regulation of chromatin-mediated processes during meiosis is not known. Here, we show that Lsh is essential for the completion of meiosis and transcriptional repression of repetitive elements in the female gonad. Oocytes from Lsh knockout mice exhibit demethylation of transposable elements and tandem repeats at pericentric heterochromatin, as well as incomplete chromosome synapsis associated with persistent RAD51 foci and gammaH2AX phosphorylation. Failure to load crossover-associated foci results in the generation of non-exchange chromosomes. The severe oocyte loss observed and lack of ovarian follicle formation, together with the patterns of Lsh nuclear compartmentalization in the germ line, demonstrate that Lsh has a critical and previously unidentified role in epigenetic gene silencing and maintenance of genomic stability during female meiosis.


Asunto(s)
Emparejamiento Cromosómico , Cromosomas de los Mamíferos/metabolismo , ADN Helicasas/metabolismo , Silenciador del Gen , Meiosis , Oocitos/citología , Retroelementos/genética , Animales , ADN Helicasas/genética , Metilación de ADN , Femenino , Regulación de la Expresión Génica , Heterocromatina/metabolismo , Ratones , Ovario/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recombinación Genética , Secuencias Repetitivas de Ácidos Nucleicos , Cromosoma X/metabolismo
18.
Andrology ; 11(6): 1132-1146, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36690000

RESUMEN

BACKGROUND: Survivors of childhood cancer often suffer from infertility. While sperm cryopreservation is not feasible before puberty, the patient's own spermatogonial stem cells could serve as a germ cell reservoir, enabling these patients to father their own children in adulthood through the isolation, in vitro expansion, and subsequent transplantation of spermatogonial stem cells. However, this approach requires large numbers of stem cells, and methods for successfully propagating spermatogonial stem cells in the laboratory are yet to be established for higher mammals and humans. The improvement of spermatogonial stem cell culture requires deeper understanding of their metabolic requirements and the mechanisms that regulate metabolic homeostasis. AIM: This review gives a summary on our knowledge of spermatogonial stem cell metabolism during maintenance and differentiation and highlights the potential influence of Sertoli cell and stem cell niche maturation on spermatogonial stem cell metabolic requirements during development. RESULTS AND CONCLUSIONS: Fetal human spermatogonial stem cell precursors, or gonocytes, migrate into the seminiferous cords and supposedly mature to adult stem cells within the first year of human development. However, the spermatogonial stem cell niche does not fully differentiate until puberty, when Sertoli cells dramatically rearrange the architecture and microenvironment within the seminiferous epithelium. Consequently, pre-pubertal and adult spermatogonial stem cells experience two distinct niche environments potentially affecting spermatogonial stem cell metabolism and maturation. Indeed, the metabolic requirements of mouse primordial germ cells and pig gonocytes are distinct from their adult counterparts, and novel single-cell RNA sequencing analysis of human and porcine spermatogonial stem cells during development confirms this metabolic transition. Knowledge of the metabolic requirements and their changes and regulation during spermatogonial stem cell maturation is necessary to implement laboratory-based techniques and enable clinical use of spermatogonial stem cells. Based on the advancement in our understanding of germline metabolism circuits and maturation events of niche cells within the testis, we propose a new definition of spermatogonial stem cell maturation and its amendment in the light of metabolic change.


Asunto(s)
Nicho de Células Madre , Testículo , Niño , Humanos , Masculino , Adulto , Animales , Porcinos , Ratones , Testículo/metabolismo , Espermatogénesis/fisiología , Semen , Espermatogonias/metabolismo , Células Madre/metabolismo , Mamíferos
19.
Cells ; 12(21)2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37947660

RESUMEN

Spermatogonial stem cell (SSC) transplantation into the testis of a germ cell (GC)-depleted surrogate allows transmission of donor genotype via donor-derived sperm produced by the recipient. Transplantation of gene-edited SSCs provides an approach to propagate gene-edited large animal models. DAZL is a conserved RNA-binding protein important for GC development, and DAZL knockout (KO) causes defects in GC commitment and differentiation. We characterized DAZL-KO pigs as SSC transplantation recipients. While there were GCs in 1-week-old (wko) KO, complete GC depletion was observed by 10 wko. Donor GCs were transplanted into 18 DAZL-KO recipients at 10-13 wko. At sexual maturity, semen and testes were evaluated for transplantation efficiency and spermatogenesis. Approximately 22% of recipient seminiferous tubules contained GCs, including elongated spermatids and proliferating spermatogonia. The ejaculate of 89% of recipients contained sperm, exclusively from donor origin. However, sperm concentration was lower than the wild-type range. Testicular protein expression and serum hormonal levels were comparable between DAZL-KO and wild-type. Intratesticular testosterone and Leydig cell volume were increased, and Leydig cell number decreased in transplanted DAZL-KO testis compared to wild-type. In summary, DAZL-KO pigs support donor-derived spermatogenesis following SSC transplantation, but low spermatogenic efficiency currently limits their use for the production of offspring.


Asunto(s)
Semen , Espermatogonias , Masculino , Animales , Porcinos , Espermatogonias/metabolismo , Testículo , Espermatozoides , Trasplante de Células Madre
20.
Genesis ; 50(7): 517-24, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22266914

RESUMEN

Basonuclin (BNC1) is a zinc finger protein expressed primarily in gametogenic cells and proliferative keratinocytes. Our previous work suggested that BNC1 is present in spermatogonia, spermatocytes, and spermatids, but absent in the Sertoli cells. BNC1's role in spermatogenesis is unknown. Here, we show that BNC1 is required for the maintenance of spermatogenesis. Bnc1-null male mice were sub-fertile, losing germ cells progressively with age. The Bnc1-null seminiferous epithelia began to degenerate before 8 weeks of age and eventually became Sertoli cell-only. Sperm count and motility also declined with age. Furthermore, Bnc1 heterozygotes, although fertile, showed a significant drop in sperm count and in testis weight by 24 weeks of age, suggesting a dosage effect of Bnc1 on testis development. In conclusion, our data demonstrate for the first time BNC1's essential role in maintaining mouse spermatogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Epitelio Seminífero/metabolismo , Túbulos Seminíferos/fisiología , Espermatogénesis/fisiología , Factores de Transcripción/genética , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Fertilidad/fisiología , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Homocigoto , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Epitelio Seminífero/citología , Células de Sertoli/fisiología , Recuento de Espermatozoides , Motilidad Espermática/fisiología , Espermatozoides/fisiología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA