Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Dev Biol ; 361(2): 301-12, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22056784

RESUMO

Spermatogonial self-renewal and differentiation are essential for male fertility and reproduction. We discovered that germ cell specific genes Sohlh1 and Sohlh2, encode basic helix-loop-helix (bHLH) transcriptional regulators that are essential in spermatogonial differentiation. Sohlh1 and Sohlh2 individual mouse knockouts show remarkably similar phenotypes. Here we show that SOHLH1 and SOHLH2 proteins are co-expressed in the entire spermatogonial population except in the GFRA1(+) spermatogonia, which includes spermatogonial stem cells (SSCs). SOHLH1 and SOHLH2 are expressed in both KIT negative and KIT positive spermatogonia, and overlap Ngn3/EGFP and SOX3 expression. SOHLH1 and SOHLH2 heterodimerize with each other in vivo, as well as homodimerize. The Sohlh1/Sohlh2 double mutant phenocopies single mutants, i.e., spermatogonia continue to proliferate but do not differentiate properly. Further analysis revealed that GFRA1(+) population was increased, while meiosis commenced prematurely in both single and double knockouts. Sohlh1 and Sohlh2 double deficiency has a synergistic effect on gene expression patterns as compared to the single knockouts. SOHLH proteins affect spermatogonial development by directly regulating Gfra1, Sox3 and Kit gene expression. SOHLH1 and SOHLH2 suppress genes involved in SSC maintenance, and induce genes important for spermatogonial differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Espermatogônias/citologia , Espermatogônias/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Masculino , Meiose , Camundongos , Modelos Biológicos , Mutação/genética , Ligação Proteica , Multimerização Proteica , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
3.
Biol Reprod ; 82(5): 888-95, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20107205

RESUMO

Immature testicular tissue of a wide variety of mammalian species continues growth and maturation when ectopically grafted under the dorsal skin of adult nude mouse recipients. Tissues from most donor species fully mature, exhibiting complete spermatogenesis within months. The connection to the recipient's vascular system is mandatory for graft development, and failure of vascularization leads to necrosis in the grafted tissue. In the present study, we analyze to what extent 1) the xenografted immature donor tissue and 2) the recipient's cells and tissues contribute to the functional recovery of a "testicular xenograft." We address whether recipient cells migrate into the testicular parenchyma and whether the circulatory connection between the donor testicular tissue and the recipient is established by ingrowing host or outgrowing donor blood vessels. Although this issue has been repeatedly discussed in previous xenografting studies, so far it has not been possible to unequivocally distinguish between donor and recipient tissues and thus to identify the mechanisms by which the circulatory connection is established. To facilitate the distinction of donor and recipient tissues, herein we used immature green fluorescent protein-positive rat testes as donor tissues and adult nude mice as graft recipients. At the time of graft recovery, donor tissues could be easily identified by the GFP expression in these tissues, allowing us to distinguish donor- and recipient-derived blood vessels. We conclude that the circulatory connection between graft and host is established by a combination of outgrowing small capillaries from the donor tissue and formation of larger vessels by the host, which connect the graft to subcutaneous blood vessels.


Assuntos
Sobrevivência de Enxerto/fisiologia , Neovascularização Fisiológica/fisiologia , Espermatogênese/fisiologia , Testículo/crescimento & desenvolvimento , Animais , Movimento Celular , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Nus , Ratos , Ratos Transgênicos , Recuperação de Função Fisiológica , Especificidade da Espécie , Testículo/irrigação sanguínea , Testículo/metabolismo , Testículo/transplante , Transplante Heterólogo
4.
J Med Primatol ; 39(2): 83-91, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20015158

RESUMO

BACKGROUND: Isolation of spermatogonial stem cells (SSCs) could enable in vitro approaches for exploration of spermatogonial physiology and therapeutic approaches for fertility preservation. SSC isolation from adult testes is difficult due to low cell numbers and lacking cell surface markers. Glial cell-derived neurotrophic factor family receptor alpha-1 (GFRalpha1) plays a crucial role for the maintenance of SSCs in rodents and is expressed in monkey spermatogonia. METHODS: Magnetic activated cell sorting was employed for the enrichment of GFRalpha1+ spermatogonia from adult primate testes. RESULTS: Magnetic activated cell sorting of monkey cells enriched GFRalpha1+ cells threefold. 11.4% of GFRalpha1+ cells were recovered. 42.9% of GFRalpha1+ cells were recovered in sorted fractions of human testicular cells, representing a fivefold enrichment. Interestingly, a high degree of morphological heterogeneity among the GFRalpha1+ cells from human testes was observed. CONCLUSIONS: Magnetic activated cell sorting using anti-GFRalpha1 antibodies provides an enrichment strategy for spermatogonia from monkey and human testes.


Assuntos
Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Macaca mulatta/fisiologia , Espermatogônias/citologia , Testículo/citologia , Adulto , Idoso , Animais , Separação Celular/métodos , Histocitoquímica/veterinária , Humanos , Magnetismo , Masculino , Pessoa de Meia-Idade , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Testículo/metabolismo , Adulto Jovem
5.
Cell Tissue Res ; 337(1): 177-83, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19434428

RESUMO

Spermatogonial stem cells (SSCs) are a documented source for adult multipotent stem cells. Thus, the isolation of SSCs is of great interest. However, the isolation of spermatogonia from mammalian testes is difficult because of their low total numbers and the lack of well-characterized cell surface markers. Glial-cell-derived neurotrophic factor family receptor alpha-1 (GFRalpha1) is expressed on undifferentiated mouse spermatogonia (including SSCs) and plays a crucial role, in rodents, for the maintenance of SSCs mediated by the Sertoli cell product GDNF. The present study has aimed to optimize the sorting efficiency and total cell yield of magnetic activated cell sorting (MACS) with anti-GFRalpha1 antibodies. Because of the technical limitations intrinsic to the magnetic columns, various sorting setups and strategies were compared. Use of Mini-MACS (MS) columns for single cell suspensions from 7-day-old rat testes resulted in a three-fold enrichment of GFRalpha1-positive cells in sorted fractions versus presorted fractions. However, with this method, only 1.77% of cells loaded onto the column were recovered in the sorted fraction. A sequential two-step sorting approach did not improve this poor yield. We therefore evaluated cell separation by using larger volume Midi-MACS (LS) columns. Enrichment of GFRalpha1-positive cells in sorted fractions was four-fold, and 14.5% of cells loaded onto the column were directed to the sorted fraction. With this method, approximately half of all GFRalpha1-positive cells present in the sample were found in the sorted fraction. We conclude that GFRalpha1 serves as a suitable surface marker for the enrichment of rat spermatogonia, and that the large-volume Midi-MACS separation system is superior to the routinely used small-volume Mini-MACS separation system.


Assuntos
Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Separação Imunomagnética/métodos , Espermatogônias/citologia , Testículo/citologia , Animais , Antígenos de Superfície/metabolismo , Diferenciação Celular , Masculino , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Ratos , Kit de Reagentes para Diagnóstico , Espermatogônias/metabolismo , Testículo/metabolismo
6.
Tissue Eng Part A ; 25(7-8): 663-676, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30311859

RESUMO

IMPACT STATEMENT: This study developed and characterized human testis extracellular matrix (htECM) and porcine testis ECM (ptECM) for testing in human spermatogonial stem cell (hSSC) culture. Results confirmed the hypothesis that ECM from the homologous species (human) and homologous tissue (testis) is optimal for maintaining hSSCs. We describe a simplified feeder-free, serum-free condition for future iterative testing to achieve the long-term goal of stable hSSC cultures. To facilitate analysis and understand the fate of hSSCs in culture, we describe a multiparameter, high-throughput, quantitative flow cytometry approach to rapidly count undifferentiated spermatogonia, differentiated spermatogonia, apoptotic spermatogonia, and proliferative spermatogonia in hSSC cultures.


Assuntos
Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Espermatogônias/citologia , Testículo/citologia , Testículo/metabolismo , Engenharia Tecidual/métodos , Células Cultivadas , Matriz Extracelular/química , Humanos , Masculino
7.
Reproduction ; 136(4): 459-69, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18660385

RESUMO

The first morphological sign of testicular differentiation is the formation of testis cords. Prior to cord formation, newly specified Sertoli cells establish adhesive junctions, and condensation of somatic cells along the surface epithelium of the genital ridge occurs. Here, we show that Sertoli cell aggregation is necessary for subsequent testis cord formation, and that neurotrophic tyrosine kinase receptors (NTRKs) regulate this process. In a three-dimensional cell culture assay, immature rat Sertoli cells aggregate to form large spherical aggregates (81.36+/-7.34 microm in diameter) in a highly organized, hexagonal arrangement (376.95+/-21.93 microm average distance between spherical aggregates). Exposure to NTRK inhibitors K252a and AG879 significantly disrupted Sertoli cell aggregation in a dose-dependent manner. Sertoli cells were prevented from establishing cell-cell contacts and from forming spherical aggregates. In vitro-derived spherical aggregates were xenografted into immunodeficient nude mice to investigate their developmental potential. In controls, seminiferous tubule-like structures showing polarized single-layered Sertoli cell epithelia, basement membranes, peritubular myoid cells surrounding the tubules, and lumen were observed in histological sections. By contrast, grafts from treatment groups were devoid of tubules and only few single Sertoli cells were present in xenografts after 4 weeks. Furthermore, the grafts were significantly smaller when Sertoli cell aggregation was disrupted by K252a in vitro (20.87 vs 6.63 mg; P<0.05). We conclude from these results that NTRK-regulated Sertoli-Sertoli cell contact is essential to the period of extensive growth and remodeling that occurs during testicular tubulogenesis, and our data indicate its potential function in fetal and prepubertal testis differentiation.


Assuntos
Receptor trkA/metabolismo , Túbulos Seminíferos/citologia , Células de Sertoli/citologia , Testículo/embriologia , Animais , Carbazóis/farmacologia , Agregação Celular/efeitos dos fármacos , Comunicação Celular , Técnicas de Cultura de Células , Relação Dose-Resposta a Droga , Alcaloides Indólicos/farmacologia , Masculino , Camundongos , Camundongos Nus , Morfogênese/fisiologia , Ratos , Ratos Endogâmicos , Receptor trkA/antagonistas & inibidores , Células de Sertoli/transplante , Testículo/crescimento & desenvolvimento , Transplante Heterólogo , Tirfostinas/farmacologia
8.
Ann N Y Acad Sci ; 1120: 152-67, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18184913

RESUMO

The organogenesis of a functional testis is the basis for male fertility and perpetuation of each species. In mammals, testicular development is dependent on two crucial events during embryonic and pubertal development. First, primary sex determination is initiated by expression of the Sry gene on the Y chromosome and directs the primordial gonad toward testicular development rather than ovarian differentiation. The male pathway comprises highly regulated cell differentiation of somatic cells within the gonadal primordium, as well as migration of mesonephric cells and primordial germ cells, ultimately leading to the formation of testis cords. These cords present the earliest visible sign of male gonadal differentiation. Second, during puberty immature Sertoli cells cease to proliferate and differentiate into their postmitotic, adult phenotype. The maturation of the Sertoli cells is pivotal for initiation and maintenance of spermatogenesis. The regulation of the two separate functions of Sertoli cells-during testis development and in spermatogenesis-are poorly understood. In this review, different models that have been used to study embryonic gonadal development and testicular maturation are compared. In vivo models, organ, and cell culture systems are discussed as regards their applicability to study testicular organogenesis. Then, a new tissue engineering approach is presented that mimics male embryonic gonadogenesis and that offers novel ways to study early testicular differentiation, as well as Sertoli cell maturation and spermatogonial stem-cell niche formation.


Assuntos
Morfogênese/fisiologia , Testículo/embriologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Masculino , Modelos Animais , Técnicas de Cultura de Órgãos , Células de Sertoli/fisiologia , Espermatogênese/fisiologia , Engenharia Tecidual/métodos , Transplante Heterólogo/métodos
9.
J Androl ; 27(4): 611-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16582408

RESUMO

Testicular development is initiated with the differentiation of Sertoli cells in the embryonic gonad. The aggregation of Sertoli cells is crucial for the generation of testicular cords and thus for the first sign of male gonadal development. To date, functional testicular tissue has not yet been generated in vitro. The objective of this study was to explore the de novo morphogenesis of testicular tissue from isolated postnatal rat testicular cells using a combination of in vitro culture and ectopic xenografting. Immature rat testicular cells were cultured in either a 2-dimensional (laminin-coated coverglass) or a 3-dimensional (extracellular matrix gel) culture system. Whereas testicular cells cultured on laminin showed a slow morphogenetic cascade resulting in cord formation after about 10 days of culture, cells cultured on extracellular matrix gel assembled to a network of cordlike structures within several hours after plating and formed spherical cell aggregates at day 3. Further progression of the morphogenetic cascade was not obtained in either the 2- or the 3-dimensional culture system. In contrast, structures resembling immature testicular tissue were obtained after xenografting of extracellular matrix gel-enclosed spherical testicular cell aggregates. The grafts were vascularized and contained elongated seminiferous tubules. Histologic analysis revealed the presence of a basement membrane, a histologically normal interstitium containing putative Leydig cells, the establishment of tubule lumen, and the integration of few putative spermatogonia into the seminiferous epithelium. We conclude that immature rat testicular cells carry the full potential to generate all somatic components of a testis in xenografts, thus opening fascinating pathways to study testicular organogenesis.


Assuntos
Morfogênese/fisiologia , Túbulos Seminíferos/crescimento & desenvolvimento , Células de Sertoli/transplante , Animais , Diferenciação Celular/fisiologia , Masculino , Camundongos , Camundongos Nus , Ratos , Transplante Heterólogo
10.
Fertil Steril ; 105(2): 256-66, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26746133

RESUMO

Infertility is a prevalent condition that has insidious impacts on the infertile individuals, their families, and society, which extend far beyond the inability to have a biological child. Lifestyle changes, fertility treatments, and assisted reproductive technology (ART) are available to help many infertile couples achieve their reproductive goals. All of these technologies require that the infertile individual is able to produce at least a small number of functional gametes (eggs or sperm). It is not possible for a person who does not produce gametes to have a biological child. This review focuses on the infertile man and describes several stem cell-based methods and gene therapy approaches that are in the research pipeline and may lead to new fertility treatment options for men with azoospermia.


Assuntos
Criopreservação , Preservação da Fertilidade/métodos , Fertilidade , Infertilidade Masculina/terapia , Preservação do Sêmen/métodos , Transplante de Células-Tronco/métodos , Testículo/transplante , Fatores Etários , Animais , Preservação da Fertilidade/efeitos adversos , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Infertilidade Masculina/etiologia , Infertilidade Masculina/fisiopatologia , Masculino , Fatores de Risco , Preservação do Sêmen/efeitos adversos , Espermatogênese , Transplante de Células-Tronco/efeitos adversos , Testículo/patologia , Testículo/fisiopatologia
11.
Methods Mol Biol ; 1210: 193-202, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25173170

RESUMO

Spermatogonial stem cells (SSCs) and undifferentiated progenitor spermatogonia in mammalian seminiferous tubules are organized in chains, connected by intracellular bridges. Clone size is generally related to stem cell potential, with shorter chains containing the majority of the stem cell population. Immunofluorescence detection of spermatogonia-specific proteins in whole-mount seminiferous tubule preparations is the only method that allows researchers to relate clone size with the molecular phenotype in spermatogenic lineage development. Here we describe in detail the method used to detect nuclear, cytoplasmic, and cell surface molecules in seminiferous tubules isolated from mouse, monkey, and human testes.


Assuntos
Diferenciação Celular , Espermatogênese , Espermatogônias/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Linhagem da Célula , Haplorrinos , Humanos , Imuno-Histoquímica/métodos , Masculino , Camundongos , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Espermatogônias/metabolismo
12.
PLoS One ; 8(1): e53976, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326552

RESUMO

The spermatogenic lineage is established after birth when gonocytes migrate to the basement membrane of seminiferous tubules and give rise to spermatogonial stem cells (SSC). In adults, SSCs reside within the population of undifferentiated spermatogonia (A(undiff)) that expands clonally from single cells (A(single)) to form pairs (A(paired)) and chains of 4, 8 and 16 A(aligned) spermatogonia. Although stem cell activity is thought to reside in the population of A(single) spermatogonia, new research suggests that clone size alone does not define the stem cell pool. The mechanisms that regulate self-renewal and differentiation fate decisions are poorly understood due to limited availability of experimental tools that distinguish the products of those fate decisions. The pluripotency factor SALL4 (sal-like protein 4) is implicated in stem cell maintenance and patterning in many organs during embryonic development, but expression becomes restricted to the gonads after birth. We analyzed the expression of SALL4 in the mouse testis during the first weeks after birth and in adult seminiferous tubules. In newborn mice, the isoform SALL4B is expressed in quiescent gonocytes at postnatal day 0 (PND0) and SALL4A is upregulated at PND7 when gonocytes have colonized the basement membrane and given rise to spermatogonia. During steady-state spermatogenesis in adult testes, SALL4 expression overlapped substantially with PLZF and LIN28 in A(single), A(paired) and A(aligned) spermatogonia and therefore appears to be a marker of undifferentiated spermatogonia in mice. In contrast, co-expression of SALL4 with GFRα1 and cKIT identified distinct subpopulations of A(undiff) in all clone sizes that might provide clues about SSC regulation. Collectively, these results indicate that 1) SALL4 isoforms are differentially expressed at the initiation of spermatogenesis, 2) SALL4 is expressed in undifferentiated spermatogonia in adult testes and 3) SALL4 co-staining with GFRα1 and cKIT reveals distinct subpopulations of A(undiff) spermatogonia that merit further investigation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Espermatogônias/crescimento & desenvolvimento , Espermatogônias/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Isoformas de Proteínas , Espermatogênese/genética , Espermatogônias/citologia , Testículo/citologia
13.
Theriogenology ; 77(2): 299-310, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21958640

RESUMO

The loss of genetic diversity due to premature death of valuable individuals is a significant problem in animal conservation programs, including endangered felids. Testis tissue xenografting has emerged as a system to obtain spermatozoa from dead immature animals, however protocols to store this tissue before xenografting are still lacking. This study focused on testis tissue cryopreservation and storage from the domestic cat (Felis catus) classified as "pre-pubertal" and "pubertal" according to spermatogenesis development. Grafts from testis tissue cryopreserved with DMSO 1.4M, recovered after 10 weeks xenografting, presented seminiferous tubules with no germ cells. On the contrary, testis tissue from pre-pubertal animals preserved in ice-cold medium for 2 to 5 days presented no loss of viability or spermatogenic potential, while the number of grafts of pubertal cat testis tissue with germ cells after 10 weeks of xenografting decreased with increasing storage time. Nevertheless, even grafts from pre-pubertal cat testis tissue presented lower anti-DDX4 and anti-BOULE staining (proteins necessary for the meiosis completion), when compared with adult cat testis. Finally, a strong correlation found between testis weight and xenograft outcome may help choose good candidates for xenografting.


Assuntos
Gatos , Espécies em Perigo de Extinção , Espermatogênese , Testículo/transplante , Preservação de Tecido/veterinária , Transplante Heterólogo/veterinária , Animais , Temperatura Baixa , Criopreservação/métodos , Criopreservação/veterinária , Felidae , Masculino , Camundongos , Camundongos Nus , Tamanho do Órgão , Maturidade Sexual , Testículo/anatomia & histologia , Testículo/crescimento & desenvolvimento , Fatores de Tempo , Preservação de Tecido/métodos , Resultado do Tratamento
14.
Endocrinology ; 151(8): 4018-23, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20555023

RESUMO

The hypothalamic-pituitary-gonadal (HPG) axis is involved in both the regulation of growth of the developing testis and in controlling spermatogenic and steroidogenic activity in the adult testis. Here, we develop a novel testicular xenografting model to examine to which degree testicular growth and function are controlled by intra- and extratesticular factors. Two or eight halves of neonatal Djungarian hamster testes were implanted into intact, hemicastrated, or castrated nude mouse recipients, and the development of the grafts under reduced or increased competition of testicular tissue was monitored and analyzed. We hypothesized that the outgrowth of the testicular grafts is influenced by the total amount of testicular tissue present in a host and that less testicular tissue in a host would result in more extended outgrowth of the grafts. Our results reveal that the hypothesis is wrong, because implanted hamster testis tissue irrespectively of the grafting condition grows to a similar size revealing an intrinsic mechanism for testicular growth. In contrast, similar size of seminal vesicle as bio-indicator of androgen levels in all hosts revealed that the steroidogenic activity is independent from the mass of testicular tissue and that steroid levels are extrinsically regulated by the recipient's HPG axis. We propose that the model of testicular xenografting provides highly valuable options to explore testicular growth and endocrine regulation of the HPG axis.


Assuntos
Modelos Teóricos , Hormônios Testiculares/fisiologia , Testículo/anatomia & histologia , Testículo/crescimento & desenvolvimento , Testículo/transplante , Animais , Animais Recém-Nascidos , Peso Corporal , Células Cultivadas , Cricetinae , Sistema Endócrino/fisiologia , Masculino , Camundongos , Camundongos Nus , Orquiectomia , Tamanho do Órgão/fisiologia , Phodopus , Hormônios Testiculares/metabolismo , Testículo/fisiologia , Transplante Heterólogo
15.
Philos Trans R Soc Lond B Biol Sci ; 365(1546): 1663-78, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20403877

RESUMO

This article will provide an updated review of spermatogonial stem cells and their role in maintaining the spermatogenic lineage. Experimental tools used to study spermatogonial stem cells (SSCs) will be described, along with research using these tools to enhance our understanding of stem cell biology and spermatogenesis. Increased knowledge about the biology of SSCs improves our capacity to manipulate these cells for practical application. The chapter concludes with a discussion of future directions for fundamental investigation and practical applications of SSCs.


Assuntos
Espermatogênese/fisiologia , Espermatogônias/fisiologia , Células-Tronco/fisiologia , Animais , Ciclo Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem da Célula/fisiologia , Humanos , Masculino , Espermatogônias/citologia , Células-Tronco/citologia
16.
J Exp Zool A Ecol Genet Physiol ; 309(5): 278-87, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18412097

RESUMO

The mechanisms behind testicular aging are poorly understood. Previous studies suggest that the testicular microenvironment is more affected by age than the male germ cell lineage. Here we analyze male reproductive aging using a unique xenografting approach. By exposing young and aged mice to newborn hamster testicular tissue, we can explore (a) whether the development and endocrine activity of hamster testicular grafts and the initiation of stem cell activity within them are affected by age of the recipients and (b) whether the endocrine response to the xenografted hamster tissue varies with recipient age. Newborn Djungarian hamster (Phodopus sungorus) testes were grafted into young (12 weeks) and aged (1 year) adult castrated nude mice. We also analyzed intact and castrated young and old control groups. After 13 weeks, 100 grafts were recovered from a total of 15 recipients and were histologically analyzed. Anatomical and endocrine parameters were recorded for each recipient as well as for the controls. Xenografted recipients responded with a normalization of their endocrine and anatomical parameters to an extent typical for their age. Although recipient age did not significantly affect graft survival and size, histopathological changes as well as spermatogenic damage within the grafts were more pronounced in the young recipients (56% Sertoli-cell-only tubules vs. 32% in the old recipients). We conclude from our data that the androgen-related changes associated with male reproductive aging are not primarily controlled by the testis. We speculate that the better development of testicular grafts in aged recipients may be owing to immunosenescence.


Assuntos
Animais Recém-Nascidos/fisiologia , Espermatogênese/fisiologia , Testículo/fisiologia , Testículo/transplante , Transplante Heterólogo/fisiologia , Envelhecimento/fisiologia , Animais , Cricetinae , Masculino , Orquiectomia , Phodopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA