Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Mol Reprod Dev ; 87(2): 223-230, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32011766

RESUMO

Sertoli cells (SCs) are presumed to be the center of testis differentiation because they provide both structural support and biological regulation for spermatogenesis. Previous studies suggest that SCs control germ cell (GC) count and Leydig cell (LC) development in mouse testes. However, the regulatory role of SCs on peritubular myoid (PTM) cell fate in fetal testis has not been clearly reported. Here, we employed Amh-Cre; diphtheria toxin fragment A (DTA) mouse model to selectively ablate SCs from embryonic day (E) 14.5. Results found that SC ablation in the fetal stage caused the disruption of testis cords and the massive loss of GCs. Furthermore, the number of α-smooth muscle actin-labeled PTM cells was gradually decreased from E14.5 and almost lost at E18.5 in SC ablation testis. Interestingly, some Ki67 and 3ß-HSD double-positive fetal LCs could be observed in Amh-Cre; DTA testes at E16.5 and E18.5. Consistent with this phenomenon, the messenger RNA levels of Hsd3b1, Cyp11a1, Lhr, Star and the protein levels of 3ß-HSD and P450Scc were significantly elevated by SC ablation. SC ablation appears to induce ectopic proliferation of fetal LCs although the total LC number appeared reduced. Together, these findings bring us a better understanding of SCs' central role in fetal testis development.


Assuntos
Diferenciação Celular/genética , Toxina Diftérica/genética , Maturidade dos Órgãos Fetais , Integrases/genética , Fragmentos de Peptídeos/genética , Túbulos Seminíferos/embriologia , Células de Sertoli/metabolismo , Animais , Proliferação de Células/genética , Toxina Diftérica/metabolismo , Células Germinativas/metabolismo , Integrases/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Modelos Animais , Fragmentos de Peptídeos/metabolismo , Ratos Transgênicos , Espermatogênese
2.
FASEB J ; 33(8): 9075-9086, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31084574

RESUMO

As the major somatic cell type, Sertoli cells undergo active proliferation and play essential roles to establish testis cord at fetal stage. They also function to maintain germ cell development throughout the life of testicular development. However, the significance of Sertoli cell number for testis cord development and gonocyte fate is still unclear. Nuclear protein ataxia-telangiectasia (NPAT, also known as p220), a substrate of cyclin E/cyclin-dependent kinase 2, is well known as a regulator of cell proliferation through regulating histone expression. To study the role of NPAT during Sertoli cell development, we generated a mouse strain carrying conditional floxed Npat alleles, when crossing with anti-Müllerian hormone-cre, leading to the specific deletion of Npat in Sertoli cells. Npat disruption in Sertoli cells inhibited the programmed proliferation of fetal Sertoli cells resulting in disruption of developing testis cords, and subsequent postnatal mutant testes were severely hypoplastic. Germ cells, which are presumed to be in quiescent status during perinatal stage, exited G0 phase arrest and re-enter mitotic cell cycle prematurely. Of particular note, some germ cells possessed the meiotic signal in Npat-deficient testes. Our data thus indicates that the function of Npat-dependent Sertoli cells is essential at multiple steps in testis development, and this study also identifies Sertoli cells as a major regulator of germ cell development, which are required to maintain a local growth niche to repress premature mitosis and meiosis of gonocytes.-Jiang, X., Yin, S., Fan, S., Bao, J., Jiao, Y., Ali, A., Iqbal, F., Xu, J., Zhang, Y., Shi, Q. Npat-dependent programmed Sertoli cell proliferation is indispensable for testis cord development and germ cell mitotic arrest.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Testículo/embriologia , Testículo/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Feminino , Masculino , Meiose/genética , Meiose/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose/genética , Mitose/fisiologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Gravidez , Túbulos Seminíferos/anormalidades , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Espermatogênese/genética , Espermatogênese/fisiologia , Espermatozoides/citologia , Espermatozoides/metabolismo , Testículo/citologia
3.
Biol Reprod ; 94(2): 42, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26792939

RESUMO

Testis cords, embryonic precursors of the seminiferous tubules, are fundamental for testis structure and function. Delay or disruption of testis cord formation could result in gonadal dysgenesis. Although mechanisms regulating testis cord formation during sex determination have been well-studied, the genes and signaling pathways involving in testis cord maintenance after the cords have formed are not well characterized. It is now clear that the maintenance of cord structure is an active process. In this review, we summarize the recent findings regarding the regulation of testis cord integrity by a series of Sertoli cell transcription factors, including the WT1-SOX8/SOX9-beta-CATENIN-DHH network, GPR56, STIM1, and NR0B1 (also known as DAX1). In particularly, we emphasize the underappreciated role of peritubular myoid cells in testis cord maintenance and their cooperation with Sertoli cells. The regulation of the size, shape, and number of testis cords by Sertoli cell proliferation (e.g., SMAD4, GATA4, and TGF-beta signaling), Leydig cell products (e.g., ACTIVIN A), vascular development (a lesson learned from PDGF signaling), and available gonad space (as observed in Ift144 mutant mice) is also addressed. Further efforts and new genetic models are needed to unveil the gene networks and underlying mechanisms regulating testis cord integrity and morphology after sex determination.


Assuntos
Desenvolvimento Embrionário/genética , Túbulos Seminíferos/embriologia , Transdução de Sinais/genética , Cordão Espermático/embriologia , Animais , Masculino , Camundongos , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOX9/metabolismo , Túbulos Seminíferos/metabolismo , Células de Sertoli/metabolismo , Cordão Espermático/metabolismo , Proteínas WT1 , beta Catenina/metabolismo
4.
Biol Reprod ; 93(4): 83, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26269506

RESUMO

The nuclear receptor steroidogenic factor 1 (SF-1, AD4BP, NR5A1) is a key regulator of the endocrine axes and is essential for adrenal and gonad development. Partial rescue of Nr5a1(-/-) mice with an SF-1-expressing transgene caused a hypomorphic phenotype that revealed its roles in Leydig cell development. In contrast to controls, all male rescue mice (Nr5a1(-/-);tg(+/0)) showed varying signs of androgen deficiency, including spermatogenic arrest, cryptorchidism, and poor virilization. Expression of various Leydig cell markers measured by immunohistochemistry, Western blot analysis, and RT-PCR indicated fetal and adult Leydig cell development were differentially impaired. Whereas fetal Leydig cell development was delayed in Nr5a1(-/-);tg(+/0) embryos, it recovered to control levels by birth. In contrast, Sult1e1, Vcam1, and Hsd3b6 transcript levels in adult rescue testes indicated complete blockage in adult Leydig cell development. In addition, between Postnatal Days 8 and 12, peritubular cells expressing PTCH1, SF-1, and CYP11A1 were observed in control testes but not in rescue testes, indicating SF-1 is needed for either survival or differentiation of adult Leydig cell progenitors. Cultured prepubertal rat peritubular cells also expressed SF-1 and PTCH1, but Cyp11a1 was expressed only after treatment with cAMP and retinoic acid. Together, data show SF-1 is needed for proper development of fetal and adult Leydig cells but with distinct primary functions; in fetal Leydig cells, it regulates differentiation, whereas in adult Leydig cells it regulates progenitor cell formation and/or survival.


Assuntos
Células Intersticiais do Testículo/fisiologia , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/fisiologia , Testículo/crescimento & desenvolvimento , Androgênios/deficiência , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Hormônios Esteroides Gonadais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ratos , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/crescimento & desenvolvimento , Túbulos Seminíferos/metabolismo , Células-Tronco , Fator Esteroidogênico 1/biossíntese , Testículo/embriologia , Testículo/metabolismo
5.
Biol Reprod ; 86(3): 80, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22133695

RESUMO

The signaling molecule DHH, secreted by Sertoli cells, has essential regulatory functions in testicular differentiation. DHH is required for the differentiation of peritubular myoid cells that line the seminiferous cords and steroidogenic Leydig cells. The testicular cords in Dhh-null male mice lack a basal lamina and develop abnormally. To date, the DHH-signaling pathway has never been examined outside of any eutherian mammals. This study examined the effects of inhibition of DHH signaling in a marsupial mammal, the tammar wallaby, by culturing gonads in vitro in the presence of the hedgehog-signaling inhibitors cyclopamine and forskolin. Disruption of hedgehog signaling in the tammar testes caused highly disorganized cord formation. SOX9 protein remained strongly expressed in Sertoli cells, laminin distribution was highly fragmented, and germ cells were distributed around the cortical regions of treated testes in an ovarianlike morphology. This suggests that hedgehog signaling regulates cord formation in the tammar wallaby testis as it does in eutherian mammals. These data demonstrate that the hedgehog pathway has been highly conserved in mammals for at least 160 million years.


Assuntos
Proteínas Hedgehog/fisiologia , Marsupiais/embriologia , Marsupiais/fisiologia , Túbulos Seminíferos/embriologia , Transdução de Sinais/fisiologia , Testículo/embriologia , Animais , Colforsina/farmacologia , Desenvolvimento Fetal/fisiologia , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/efeitos dos fármacos , Masculino , Técnicas de Cultura de Órgãos , Fatores de Transcrição SOX9/metabolismo , Túbulos Seminíferos/citologia , Túbulos Seminíferos/efeitos dos fármacos , Células de Sertoli/metabolismo , Testículo/citologia , Testículo/efeitos dos fármacos , Alcaloides de Veratrum/farmacologia
6.
Mymensingh Med J ; 21(1): 34-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22314451

RESUMO

This cross sectional descriptive study was done to observe the mean diameter of seminiferous tubules and to determine its differences between different age groups in Bangladeshi male. Thirty human testes of different age groups were collected by purposive sampling technique. Among them 22 specimens were collected from cadavers during routine postmortem examination and 8 specimens were from dead fetuses from Gynaecology & Obstetrics Department. The specimens were grouped into three categories Group A (28 to 42 weeks of gestational age), Group B (Up to 14 years) and Group C (15 to 70 years). The mean diameter of seminiferous tubules was measured and significant differences of the dimensions between different age groups were determined. The mean±SD diameter was 85.37±15.51 µm in Group A, 144.04±63.34 µm in Group B and 227.92±22.47 µm in Group C. Statistically, differences between age groups were calculated by using Unpaired Students 't' test. The present study revealed that the diameter increased with age and mean differences were statistically significant between Groups A&C, B&C and A&B.


Assuntos
Túbulos Seminíferos/anatomia & histologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Estudos Transversais , Feto/anatomia & histologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Túbulos Seminíferos/embriologia , Adulto Jovem
7.
Okajimas Folia Anat Jpn ; 87(3): 103-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21174939

RESUMO

The development of the testicular cord structure was investigated in 4 human fetuses between 70 and 90 days of gestation, in which the testicular cords are differentiating into the seminiferous tubules. Histological examinations were performed using stains with haematoxylin-eosin (HE), Masson's trichrome (MT), periodic acid schiff (PAS), anti-proliferating cell nuclear antigen (PCNA) monoclonal antibodies, and TUNEL methods. It was found that the testicular cords structures were indefinitely observed in HE-stained sections of four fetuses. However, the basement membranes of the testicular cord were clearly stained with MT, showing the tubular structure. Furthermore, cells in the testicular cords were positive with PAS, but the interstitial tissues outside the testicular cords were negative. PCNA-positive cells were detected not only inside but also outside the testicular cords, however, TUNEL positive cells are not detected throughout all testicular tissues.


Assuntos
Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Cordão Espermático/citologia , Cordão Espermático/embriologia , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Idade Gestacional , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Antígeno Nuclear de Célula em Proliferação/metabolismo , Túbulos Seminíferos/metabolismo , Cordão Espermático/metabolismo , Coloração e Rotulagem
8.
Reproduction ; 137(1): 79-93, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18801836

RESUMO

Lunatic fringe belongs to a family of beta1-3 N-acetyltransferases that modulate the affinity of the Notch receptors for their ligands through the elongation of O-fucose moieties on their extracellular domain. A role for Notch signaling in vertebrate fertility has been predicted by the intricate expression of the Notch receptors and their ligands in the oocyte and granulosa cells of the ovary and the spermatozoa and Sertoli cells of the testis. It has been demonstrated that disruption of Notch signaling by inactivation of lunatic fringe led to infertility associated with pleiotropic defects in follicle development and meiotic maturation of oocytes. Lunatic fringe null males were found to be subfertile. Here, we report that gene expression data demonstrate that fringe and Notch signaling genes are expressed in the developing testis and the intratesticular ductal tract, predicting roles for this pathway during embryonic gonadogenesis and spermatogenesis. Spermatogenesis was not impaired in the majority of the lunatic fringe null males; however, spermatozoa were unilaterally absent in the epididymis of many mice. Histological and immunohistochemical analysis of these testes revealed the development of unilateral cystic dilation of the rete testis. Tracer dye experiments confirm a block in the connection between the rete testis and the efferent ducts. Further, the dye studies demonstrated that many lunatic fringe mutant males had partial blocks of the connection between the rete testis and the efferent ducts bilaterally.


Assuntos
Cistos/patologia , Glicosiltransferases/deficiência , Rede do Testículo/patologia , Animais , Cruzamento , Cistos/genética , Cistos/metabolismo , Dilatação Patológica , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Imuno-Histoquímica , Hibridização In Situ/métodos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Notch/genética , Receptores Notch/metabolismo , Rede do Testículo/embriologia , Rede do Testículo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/patologia , Coloração e Rotulagem
9.
Mol Cell Endocrinol ; 294(1-2): 70-80, 2008 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-18790002

RESUMO

The role transforming growth factor beta (TGFb) isoforms TGFb1, TGFb2 and TGFb3 have in the regulation of embryonic gonadal development was investigated with the use of null-mutant (i.e. knockout) mice for each of the TGFb isoforms. Late embryonic gonadal development was investigated because homozygote TGFb null-mutant mice generally die around birth, with some embryonic loss as well. In the testis, the TGFb1 null-mutant mice had a decrease in the number of germ cells at birth, postnatal day 0 (P0). In the testis, the TGFb2 null-mutant mice had a decrease in the number of seminiferous cords at embryonic day 15 (E15). In the ovary, the TGFb2 null-mutant mice had an increase in the number of germ cells at P0. TGFb isoforms appear to have a role in gonadal development, but interactions between the isoforms is speculated to compensate in the different TGFb isoform null-mutant mice.


Assuntos
Desenvolvimento Embrionário/genética , Mutação/genética , Ovário/embriologia , Ovário/metabolismo , Testículo/embriologia , Testículo/metabolismo , Fator de Crescimento Transformador beta/deficiência , Animais , Antígenos Nucleares/metabolismo , Apoptose , Contagem de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Genótipo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/citologia , Fenótipo , Ratos , Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Razão de Masculinidade , Testículo/citologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/deficiência , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/deficiência , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo
10.
Iran Biomed J ; 22(4): 258-63, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29397043

RESUMO

Background: In vitro spermatogenesis has a long research history beginning in the early 20th century. This organ culture method was therefore abandoned, and alternative cell culture methods were chosen by many researchers. Here, whether Tnp1, Tekt1, and Plzf, which play a crucial role in spermatogenesis, can be expressed during testis organ culture was assessed. Methods: Testes of 10 mouse pups were first removed, and the testis tissue was then separated into smaller pieces of seminiferous tubules. The size of the pieces was arbitrary; approximately 1 mg in weight or 1 mm3 in size when compacted. Afterwards, the testis tissue fragments (1­3) were transferred to the hexahedrons, incubated in a culture incubator and cultured for 12 weeks. Histological assessment and molecular evaluation were carried out at the end of the study. Results: The results showed that the expression of Tekt1 as a mitotic gene in mouse pups decreased significantly (p ≤ 0.05) in comparison to adult mouse testis. Meanwhile, the expression of Tnp1 as a meiotic gene increased significantly (p ≤ 0.05) as compared to neonate mouse testis at the beginning of the culture. The expression of Plzf showed no significant difference during the 12 weeks of culture (p ≥ 0.05). Based on histological study, different types of spermatocytes and post-meiotic stages of germ cells could not be detected. Conclusion: This kind of three-dimensional culture can induce expression of post-meiotic gene, Tnp1, but only at the molecular level and not beyond meiosis.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas dos Microtúbulos/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Espermatogênese , Testículo/embriologia , Animais , Perfilação da Expressão Gênica , Masculino , Meiose , Camundongos , Mitose , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase em Tempo Real , Túbulos Seminíferos/embriologia , Espermatócitos/citologia
11.
Life Sci ; 79(22): 2112-8, 2006 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-16884740

RESUMO

More than half of ADAM (a disintegrin and metalloprotease) family members are expressed in mammalian male reproductive organs such as testis and epididymis. The ADAM19 gene identified in mouse is a member of the ADAM family and is highly enriched in testes of a newborn mouse. The present study was performed to determine its expression pattern in whole mouse testes in vivo as well as its in vitro action and regulation in testis cells from 2-day-old mice. Reverse transcriptase polymerase chain reaction (RT-PCR) detected ADAM19 mRNA from 15.5 days postcoitum (dpc) to 21 days postpartum (dpp), with high expression during the perinatal period. Immunohistochemistry demonstrated ADAM19 protein localization to the seminiferous cords at both embryonic and postnatal ages examined (from 15.5-19.5 dpc to 2 dpp). In particular, we obtained new evidence that a neutralizing antibody to ADAM19 had no influence on the proliferation of 2 dpp testis cells cultured in serum-free medium when compared to controls. Interestingly, it inhibited the 2 dpp testis cell proliferation elicited by stimulation with 10% FCS (P<0.01) or FSH (P<0.05). Lastly, using a model of 2 dpp testis cell cultures and RT-PCR procedures, we demonstrated that follicle stimulating-hormone (FSH) reduced the levels of ADAM19 mRNA in a time-dependent manner. Taken together, these results indicate that the expression of ADAM19 may be subject to regulation by FSH during mouse testis development. Furthermore, ADAM19 can act to regulate the proliferation of perinatal testis cells in the perinatal period.


Assuntos
Proteínas ADAM/genética , Envelhecimento/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Testículo/embriologia , Animais , Primers do DNA , Masculino , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/crescimento & desenvolvimento , Suínos , Testículo/citologia , Testículo/crescimento & desenvolvimento
12.
Results Probl Cell Differ ; 58: 225-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27300181

RESUMO

A healthy man typically produces between 50 × 10(6) and 200 × 10(6) spermatozoa per day by spermatogenesis; in the absence of Sertoli cells in the male gonad, this individual would be infertile. In the adult testis, Sertoli cells are sustentacular cells that support germ cell development by secreting proteins and other important biomolecules that are essential for germ cell survival and maturation, establishing the blood-testis barrier, and facilitating spermatozoa detachment at spermiation. In the fetal testis, on the other hand, pre-Sertoli cells form the testis cords, the future seminiferous tubules. However, the role of pre-Sertoli cells in this process is much less clear than the function of Sertoli cells in the adult testis. Within this framework, we provide an overview of the biology of the fetal, pubertal, and adult Sertoli cell, highlighting relevant cell biology studies that have expanded our understanding of mammalian spermatogenesis.


Assuntos
Células de Sertoli/citologia , Espermatogênese , Espermatozoides/citologia , Testículo/citologia , Animais , Barreira Hematotesticular/citologia , Barreira Hematotesticular/embriologia , Barreira Hematotesticular/crescimento & desenvolvimento , Diferenciação Celular , Humanos , Masculino , Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/crescimento & desenvolvimento , Testículo/embriologia , Testículo/crescimento & desenvolvimento
13.
PLoS One ; 11(1): e0145398, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26765744

RESUMO

A previous study indicated that genetic information encoded on the mouse Y chromosome short arm (Yp) is required for efficient completion of the second meiotic division (that generates haploid round spermatids), restructuring of the sperm head, and development of the sperm tail. Using mouse models lacking a Y chromosome but with varying Yp gene complements provided by Yp chromosomal derivatives or transgenes, we recently identified the Y-encoded zinc finger transcription factors Zfy1 and Zfy2 as the Yp genes promoting the second meiotic division. Using the same mouse models we here show that Zfy2 (but not Zfy1) contributes to the restructuring of the sperm head and is required for the development of the sperm tail. The preferential involvement of Zfy2 is consistent with the presence of an additional strong spermatid-specific promotor that has been acquired by this gene. This is further supported by the fact that promotion of sperm morphogenesis is also seen in one of the two markedly Yp gene deficient models in which a Yp deletion has created a Zfy2/1 fusion gene that is driven by the strong Zfy2 spermatid-specific promotor, but encodes a protein almost identical to that encoded by Zfy1. Our results point to there being further genetic information on Yp that also has a role in restructuring the sperm head.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Cabeça do Espermatozoide/metabolismo , Cauda do Espermatozoide/metabolismo , Espermatogênese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cromossomo Y/genética , Animais , Masculino , Camundongos , Modelos Animais , Morfogênese/genética , Mapeamento Físico do Cromossomo , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Cauda do Espermatozoide/ultraestrutura
14.
Endocrinology ; 157(3): 1276-88, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26727108

RESUMO

Timely and appropriate levels of thyroid hormone (TH) signaling are necessary to ensure normal developmental outcomes in many tissues. Studies using pharmacological models of altered TH status have revealed an influence of these hormones on testis development and size, but little is known about the role of endogenous determinants of TH action in the developing male gonads. Using a genetic approach, we demonstrate that the type 3 deiodinase (D3), which inactivates TH and protects developing tissues from undue TH action, is a key factor. D3 is highly expressed in the developing testis, and D3-deficient (D3KO) mice exhibit thyrotoxicosis and cell proliferation arrest in the neonatal testis, resulting in an approximately 75% reduction in testis size. This is accompanied by larger seminiferous tubules, impaired spermatogenesis, and a hormonal profile indicative of primary hypogonadism. A deficiency in the TH receptor-α fully normalizes testis size and adult testis gene expression in D3KO mice, indicating that the effects of D3 deficiency are mediated through this type of receptor. Similarly, genetic deficiencies in the D2 or in the monocarboxylate transporter 8 partially rescue the abnormalities in testis size and gonadal axis gene expression featured in the D3KO mice. Our study highlights the testis as an important tissue in which determinants of TH action coordinately converge to ensure normal development and identifies D3 as a critical factor in testis development and in testicular protection from thyrotoxicosis.


Assuntos
Hipogonadismo/genética , Iodeto Peroxidase/genética , RNA Mensageiro/metabolismo , Testículo/metabolismo , Tireotoxicose/genética , Tiroxina/metabolismo , Animais , Animais Recém-Nascidos , Imuno-Histoquímica , Iodeto Peroxidase/metabolismo , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/patologia , Espermatogênese/genética , Simportadores , Testículo/embriologia , Receptores alfa dos Hormônios Tireóideos/genética , Transcriptoma , Iodotironina Desiodinase Tipo II
15.
Int J Dev Biol ; 41(5): 699-703, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9415489

RESUMO

Cloning and characterization of H3.3A variant histone expression has recently been reported to be associated with meiotic development in mouse testis and ovary. Using Northern analysis and in situ hybridization, the pattern of H3.3A expression was studied during the development of different tissues. In addition to the differential expression detected in male and female meiosis, H3.3A was found to be highly expressed in preantral follicles of adult ovaries and in the basal regions of seminiferous epithelium corresponding to spermatogonia. Different patterns of expression were observed in somatic tissues, which also differed with respect to the developmental stage of the tissue. The lowest expression was detected in adult skeletal muscle. High expressions were found in foetal liver and spinal cord. These different expressions might reflect a possible function of H3.3A in cell differentiation as detected in MEL cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Ovário/embriologia , RNA Mensageiro/análise , Testículo/embriologia , Animais , Northern Blotting , Diferenciação Celular , Clonagem Molecular , Feminino , Histocitoquímica , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Fígado/embriologia , Fígado/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Ovário/metabolismo , RNA Mensageiro/genética , Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Medula Espinal/embriologia , Medula Espinal/metabolismo , Testículo/metabolismo
16.
Int J Dev Biol ; 39(2): 335-44, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7669546

RESUMO

The distribution of laminin chains and basement membranes (BMs) in the ontogenesis and sex differentiation of male and female mouse gonads and mesonephros was studied by conventional and immunocytochemical light and electron microscopy. The alpha 1 (synonymous to A) chain was recognized with MAbs against fragment E3, and three chains of laminin with PAbs raised against EHS-laminin. BMs, which formed around the mesonephric duct, the mesonephric tubules, and the paramesonephric duct, contained the laminin alpha 1 chain. The alpha 1 chain appeared with epithelial differentiation in the developing gonads in both sexes. The alpha 1 chain was first evident around the embryonic gonadal cords and remained, after development, in the BMs of the testicular cords and ovarian follicles. The laminin alpha 1 chain was also detected in BMs of the myoid cells around the epithelial rete cords, and transiently in the surface epithelium and in the corpus luteum. Laminin beta-gamma chains were found in many locations where the alpha 1 chain was not detected. These included the mesenchyme of the early mesonephros, the BMs of blood vessels and surface epithelium in the differentiated testis and ovary, between the theca cells in the ovary, and in some corpora lutea. The morphological differentiation of the BMs of the embryonic testicular cords proceeded rapidly. In contrast, the BM of the ovarian cords remained relatively poorly differentiated during the prenatal phases, and developed concomitantly with the differentiation of the follicles. The results show that BMs in the differentiating internal genitalia are heterogeneous with respect to their laminin chains, and suggest that all known laminin chains must be analyzed in the differentiation of gonadal epithelia for a complete role of the BMs in gonadal sex differentiation.


Assuntos
Genitália/química , Genitália/embriologia , Laminina/análise , Diferenciação Sexual , Animais , Membrana Basal/química , Corpo Lúteo/química , Corpo Lúteo/embriologia , Epitélio/química , Epitélio/embriologia , Feminino , Immunoblotting , Masculino , Mesonefro/química , Mesonefro/crescimento & desenvolvimento , Camundongos , Folículo Ovariano/química , Folículo Ovariano/embriologia , Ovário/química , Ovário/embriologia , Túbulos Seminíferos/química , Túbulos Seminíferos/embriologia , Testículo/química , Testículo/embriologia , Fatores de Tempo
17.
Mol Endocrinol ; 29(9): 1334-46, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26258622

RESUMO

Defects in spermatogenesis, a process that produces spermatozoa inside seminiferous tubules of the testis, result in male infertility. Spermatogenic progression is highly dependent on a microenvironment provided by Sertoli cells, the only somatic cells and epithelium of seminiferous tubules. However, genes that regulate such an important activity of Sertoli cells are poorly understood. Here, we found that AT-rich interactive domain 4B (ARID4B), is essential for the function of Sertoli cells to regulate spermatogenesis. Specifically, we generated Sertoli cell-specific Arid4b knockout (Arid4bSCKO) mice, and showed that the Arid4bSCKO male mice were completely infertile with impaired testis development and significantly reduced testis size. Importantly, severe structural defects accompanied by loss of germ cells and Sertoli cell-only phenotype were found in many seminiferous tubules of the Arid4bSCKO testes. In addition, maturation of Sertoli cells was significantly delayed in the Arid4bSCKO mice, associated with delayed onset of spermatogenesis. Spermatogenic progression was also defective, showing an arrest at the round spermatid stage in the Arid4bSCKO testes. Interestingly, we showed that ARID4B functions as a "coactivator" of androgen receptor and is required for optimal transcriptional activation of reproductive homeobox 5, an androgen receptor target gene specifically expressed in Sertoli cells and critical for spermatogenesis. Together, our study identified ARID4B to be a key regulator of Sertoli cell function important for male germ cell development.


Assuntos
Proteínas de Ligação a DNA/genética , Túbulos Seminíferos/embriologia , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Espermatozoides/crescimento & desenvolvimento , Animais , Apoptose/genética , Linhagem Celular , Claudina-3/biossíntese , Regulação para Baixo/genética , Epididimo/crescimento & desenvolvimento , Proteínas de Homeodomínio/biossíntese , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/metabolismo , Túbulos Seminíferos/citologia , Túbulos Seminíferos/fisiopatologia , Células de Sertoli/citologia , Espermatogênese/genética , Espermatozoides/citologia , Fatores de Transcrição/biossíntese
18.
J Endocrinol ; 145(1): 35-42, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7798028

RESUMO

In order to investigate the ontogeny of gonadal inhibin production in the male fetal sheep, testes were collected from male fetuses at days 70, 100, 130 and 140 of gestation (term = 145 days). The expression and localization of inhibin alpha- and inhibin beta A-subunit mRNA and protein were evaluated using in situ hybridization and immunocytochemistry. The expression of inhibin alpha-subunit mRNA was localized within the seminiferous cords of the developing fetal testis and progressively increased with gestational age. Immunostaining corresponding to immunoreactive inhibin alpha-subunit was detected in Sertoli cells within the seminiferous cords at days 100, 130 and 140 of gestation. In addition, immunostaining was detectable in a small proportion of Leydig cells. No expression of inhibin beta A-subunit mRNA or immunoreactivity was detected in any testicular tissue at any stage of gestation. These data show that the Sertoli cells of the developing fetal sheep testis have the capacity to produce inhibin alpha-subunit by day 100 of gestation and that production increases during late gestation.


Assuntos
Inibinas/biossíntese , Biossíntese Peptídica , Proteínas Secretadas pela Próstata , Ovinos/embriologia , Testículo/embriologia , Animais , Idade Gestacional , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Inibinas/análise , Inibinas/genética , Masculino , Peptídeos/análise , Peptídeos/genética , RNA Mensageiro/análise , Túbulos Seminíferos/química , Túbulos Seminíferos/embriologia , Células de Sertoli/química , Ovinos/metabolismo , Testículo/química , Testículo/metabolismo
19.
Anat Embryol (Berl) ; 202(6): 475-90, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11131015

RESUMO

The development of the intragonadal rete testis and the establishment of the connection between seminiferous and straight testicular tubules was studied using ultrastructural and histochemical methods in 60 bovine embryos and fetuses ranging from day 39 through day 225 post conceptionem. The methodology included a modified acetylcholinesterase (AChE) reaction as a selective marker for pre-Sertoli cells and a modified microsomal aminopeptidase (MAP) reaction as a selective marker for the epithelia of rete testis and straight testicular tubules. Between 40 and 45 days, the rete testis is predominantly an extratesticular rete situated in the cranial peduncle of the gonadal fold and in broad contact with the pro/mesonephric giant corpuscle. During this period, the intragonadal rete enters the gonad proper from its craniodorsal pole and extends into the cranial fourth of the testis. Between 60 and 110 days the rete testis attains its definitive position, extending into the central longitudinal axis as far as to the caudal fourth of the testis. For the caudal expansion of the rete testis the preceding proliferation of the mediastinal stroma is an important prerequisite. In the 40 to 45-day-old embryo the area of the testicular cords may be divided into two zones. A narrow outer zone contains plate-like cords with a thick diameter, and a larger central zone is filled with a network of thinner cords. Only the thick outer cords transform into the permanent seminiferous tubules, whereas the thinner cords in the central zone are transitory structures that disappear between 45 and 110 days. One important function of these transitory cords is to establish a continuous system of basal laminae that allows a direct connection between the central ends of the growing seminiferous tubules and the peripheral extensions of the rete testis (future straight testicular tubules). The first true straight testicular tubules become visible between 85 and 110 days. Due to a strong proliferation of the tubulus rectus-cells the straight testicular tubules elongate continuously, and the border between the rete system and the seminiferous tubules is slowly shifted towards the testicular periphery. This shift is not restricted to the prenatal period, but proceeds until after birth. At the cytological level, the formation and elongation of the straight testicular tubules is effected by proliferating cells that advance along the continuous basal lamina into the area of the seminiferous tubules. The pre-Sertoli and germ cells in this zone of invasion are separated from each other and overgrown by the tubulus rectus-cells. Exposed to the special milieu of the straight testicular tubules, pre-Sertoli and germ cells apparently cannot survive and finally disappear.


Assuntos
Rede do Testículo/embriologia , Túbulos Seminíferos/embriologia , Acetilcolinesterase/análise , Animais , Bovinos , Desenvolvimento Embrionário e Fetal , Células Epiteliais/ultraestrutura , Imuno-Histoquímica , Leucil Aminopeptidase/análise , Masculino , Rede do Testículo/enzimologia , Túbulos Seminíferos/enzimologia
20.
Anat Embryol (Berl) ; 160(1): 11-27, 1980.
Artigo em Inglês | MEDLINE | ID: mdl-7469033

RESUMO

Sertoli cells have various functions: mechanical (creation of two compartments in the seminiferous tubules, migration of germinal cells), secretory (secretion of anti-Müllerian hormone, inhibin, androgen-binding-protein and estrogen) and phagocytic. We report an ultrastructural study of the rat Sertoli cell during maturation and consider possible correlations between the acquisition of certain morphological characteristics and certain functions. During fetal life, the Sertoli cell possesses differentiated zones of junction with the gonocytes and seems to have a role in the migration of the gonocytes towards the periphery of the seminiferous tubule. The Sertoli cell performs the phagocytosis of the gonocytes which degenerate during their migration, and seems to be the site of production of protein granules, whose presence can be related to the synthesis of anti-Müllerian hormone. After birth and before puberty, when the inclusions resembling secretory granules disappear, the Sertoli cell membranes in contact with spermatocytes II and spermatids differentiate, forming, through the differentiated junctional complexes, two compartments (adluminal and luminal) in the seminiferous tubules. Finally, they acquire the characteristics of active secretory cells, capable, in particular, of steroid synthesis.


Assuntos
Ratos/embriologia , Túbulos Seminíferos/embriologia , Células de Sertoli/ultraestrutura , Testículo/embriologia , Animais , Masculino , Microscopia Eletrônica , Fagocitose , Ratos/anatomia & histologia , Túbulos Seminíferos/crescimento & desenvolvimento , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA