Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Development ; 150(14)2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37350382

RESUMO

Retinoic acid (RA) is the proposed mammalian 'meiosis inducing substance'. However, evidence for this role comes from studies in the fetal ovary, where germ cell differentiation and meiotic initiation are temporally inseparable. In the postnatal testis, these events are separated by more than 1 week. Exploiting this difference, we discovered that, although RA is required for spermatogonial differentiation, it is dispensable for the subsequent initiation, progression and completion of meiosis. Indeed, in the absence of RA, the meiotic transcriptome program in both differentiating spermatogonia and spermatocytes entering meiosis was largely unaffected. Instead, transcripts encoding factors required during spermiogenesis were aberrant during preleptonema, and the subsequent spermatid morphogenesis program was disrupted such that no sperm were produced. Taken together, these data reveal a RA-independent model for male meiotic initiation.


Assuntos
Testículo , Tretinoína , Animais , Feminino , Masculino , Tretinoína/farmacologia , Espermatogênese/genética , Espermatogônias , Espermatozoides , Meiose/genética , Mamíferos
2.
Biol Reprod ; 108(5): 822-836, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-36708226

RESUMO

The foundation of mammalian spermatogenesis is provided by undifferentiated spermatogonia, which comprise of spermatogonial stem cells (SSCs) and transit-amplifying progenitors that differentiate in response to retinoic acid (RA) and are committed to enter meiosis. Our laboratory recently reported that the foundational populations of SSCs, undifferentiated progenitors, and differentiating spermatogonia are formed in the neonatal testis in part based on their differential responsiveness to RA. Here, we expand on those findings to define the extent to which RA responsiveness during steady-state spermatogenesis in the adult testis regulates the spermatogonial fate. Our results reveal that both progenitor and differentiating spermatogonia throughout the testis are capable of responding to exogenous RA, but their resulting fates were quite distinct-undifferentiated progenitors precociously differentiated and proceeded into meiosis on a normal timeline, while differentiating spermatogonia were unable to hasten their entry into meiosis. This reveals that the spermatogonia responding to RA must still complete the 8.6 day differentiation program prior to their entry into meiosis. Addition of exogenous RA enriched testes with preleptotene and pachytene spermatocytes one and two seminiferous cycles later, respectively, supporting recent clinical studies reporting increased sperm production and enhanced fertility in subfertile men on long-term RA analog treatment. Collectively, our results reveal that a well-buffered system exists within mammalian testes to regulate spermatogonial RA exposure, that exposed undifferentiated progenitors can precociously differentiate, but must complete a normal-length differentiation program prior to entering meiosis, and that daily RA treatments increased the numbers of advanced germ cells by directing undifferentiated progenitors to continuously differentiate.


Assuntos
Espermatogônias , Tretinoína , Animais , Recém-Nascido , Masculino , Humanos , Tretinoína/farmacologia , Sêmen , Espermatogênese/fisiologia , Testículo , Diferenciação Celular/fisiologia , Mamíferos
3.
Development ; 149(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36250451

RESUMO

In mammalian testes, premeiotic spermatogonia respond to retinoic acid by completing an essential lengthy differentiation program before initiating meiosis. The molecular and cellular changes directing these developmental processes remain largely undefined. This wide gap in knowledge is due to two unresolved technical challenges: (1) lack of robust and reliable in vitro models to study differentiation and meiotic initiation; and (2) lack of methods to isolate large and pure populations of male germ cells at each stage of differentiation and at meiotic initiation. Here, we report a facile in vitro differentiation and meiotic initiation system that can be readily manipulated, including the use of chemical agents that cannot be safely administered to live animals. In addition, we present a transgenic mouse model enabling fluorescence-activated cell sorting-based isolation of millions of spermatogonia at specific developmental stages as well as meiotic spermatocytes.


Assuntos
Espermatogênese , Espermatogônias , Masculino , Camundongos , Animais , Espermatócitos , Testículo , Meiose , Diferenciação Celular , Camundongos Transgênicos , Mamíferos
4.
Development ; 146(12)2019 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-31023878

RESUMO

In the mammalian testis, sustained spermatogenesis relies on spermatogonial stem cells (SSCs); their progeny either remain as stem cells (self-renewal) or proliferate and differentiate to enter meiosis in response to retinoic acid (RA). Here, we sought to uncover elusive mechanisms regulating a key switch fundamental to spermatogonial fate: the capacity of spermatogonia to respond to RA. Using the developing mouse testis as a model, we found that spermatogonia and precursor prospermatogonia exhibit a heterogeneous capacity to respond to RA with at least two underlying causes. First, progenitor spermatogonia are prevented from responding to RA by catabolic activity of cytochrome P450 family 26 enzymes. Second, a smaller subset of undifferentiated spermatogonia enriched for SSCs exhibit catabolism-independent RA insensitivity. Moreover, for the first time, we observed that precursor prospermatogonia are heterogeneous and comprise subpopulations that exhibit the same differential RA responsiveness found in neonatal spermatogonia. We propose a novel model by which mammalian prospermatogonial and spermatogonial fates are regulated by their intrinsic capacity to respond (or not) to the differentiation signal provided by RA before, and concurrent with, the initiation of spermatogenesis.


Assuntos
Regulação da Expressão Gênica , Espermatogênese , Espermatogônias/citologia , Células-Tronco/citologia , Testículo/crescimento & desenvolvimento , Tretinoína/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Família 26 do Citocromo P450/metabolismo , Genômica , Proteínas de Fluorescência Verde/metabolismo , Masculino , Meiose , Camundongos , Células de Sertoli/citologia , Transdução de Sinais , Testículo/embriologia
5.
Biol Reprod ; 100(2): 429-439, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30202948

RESUMO

The self-renewal, proliferation, and differentiation of the spermatogonial populations must be finely coordinated in the mammalian testis, as dysregulation of these processes can lead to subfertility, infertility, or the formation of tumors. There are wide gaps in our understanding of how these spermatogonial populations are formed and maintained, and our laboratory has focused on identifying the molecular and cellular pathways that direct their development. Others and we have shown, using a combination of pharmacologic inhibitors and genetic models, that activation of mTOR complex 1 (mTORC1) is important for spermatogonial differentiation in vivo. Here, we extend those studies to directly test the germ cell-autonomous requirement for mTORC1 in spermatogonial differentiation. We created germ cell conditional knockout mice for "regulatory associated protein of MTOR, complex 1" (Rptor), which encodes an essential component of mTORC1. While germ cell KO mice were viable and healthy, they had smaller testes than littermate controls, and no sperm were present in their cauda epididymides. We found that an initial cohort of Rptor KO spermatogonia proliferated, differentiated, and entered meiosis (which they were unable to complete). However, no self-renewing spermatogonia were formed, and thus the entire germline was lost by adulthood, resulting in Sertoli cell-only testes. These results reveal the cell autonomous requirement for RPTOR in the formation or maintenance of the foundational self-renewing spermatogonial stem cell pool in the mouse testis and underscore complex roles for mTORC1 and its constituent proteins in male germ cell development.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteína Regulatória Associada a mTOR/metabolismo , Espermatozoides/fisiologia , Animais , Epididimo/citologia , Regulação da Expressão Gênica/fisiologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Meiose , Camundongos , Camundongos Knockout , Proteína Regulatória Associada a mTOR/genética , Espermatogênese , Testículo
6.
Cell Rep ; 25(6): 1650-1667.e8, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30404016

RESUMO

Spermatogenesis is a complex and dynamic cellular differentiation process critical to male reproduction and sustained by spermatogonial stem cells (SSCs). Although patterns of gene expression have been described for aggregates of certain spermatogenic cell types, the full continuum of gene expression patterns underlying ongoing spermatogenesis in steady state was previously unclear. Here, we catalog single-cell transcriptomes for >62,000 individual spermatogenic cells from immature (postnatal day 6) and adult male mice and adult men. This allowed us to resolve SSC and progenitor spermatogonia, elucidate the full range of gene expression changes during male meiosis and spermiogenesis, and derive unique gene expression signatures for multiple mouse and human spermatogenic cell types and/or subtypes. These transcriptome datasets provide an information-rich resource for studies of SSCs, male meiosis, testicular cancer, male infertility, or contraceptive development, as well as a gene expression roadmap to be emulated in efforts to achieve spermatogenesis in vitro.


Assuntos
Mamíferos/genética , Análise de Célula Única , Espermátides/citologia , Espermatogênese/genética , Espermatogônias/citologia , Transcriptoma/genética , Adulto , Envelhecimento/genética , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Haploidia , Humanos , Masculino , Meiose , Camundongos Endogâmicos C57BL , Transdução de Sinais , Espermátides/metabolismo , Espermatogônias/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/citologia
7.
Development ; 145(15)2018 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-29980567

RESUMO

Throughout the male reproductive lifespan, spermatogonial stem cells (SSCs) produce committed progenitors that proliferate and then remain physically connected in growing clones via short cylindrical intercellular bridges (ICBs). These ICBs, which enlarge in meiotic spermatocytes, have been demonstrated to provide a conduit for postmeiotic haploid spermatids to share sex chromosome-derived gene products. In addition to ICBs, spermatogonia exhibit multiple thin cytoplasmic projections. Here, we have explored the nature of these projections in mice and find that they are dynamic, span considerable distances from their cell body (≥25 µm), either terminate or physically connect multiple adjacent spermatogonia, and allow for sharing of macromolecules. Our results extend the current model that subsets of spermatogonia exist as isolated cells or clones, and support a model in which spermatogonia of similar developmental fates are functionally connected through a shared dynamic cytoplasm mediated by thin cytoplasmic projections.


Assuntos
Citoplasma/metabolismo , Mamíferos/metabolismo , Espermatogônias/metabolismo , Animais , Diferenciação Celular , Citoplasma/ultraestrutura , Difusão , Proteínas de Fluorescência Verde/metabolismo , Espaço Intracelular/metabolismo , Substâncias Macromoleculares/metabolismo , Masculino , Meiose , Camundongos Transgênicos , Papio , Ratos , Espermatócitos/citologia , Espermatócitos/metabolismo , Espermatogônias/citologia , Espermatogônias/ultraestrutura
8.
Stem Cell Res ; 27: 162-168, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29475796

RESUMO

Mammalian male germ cell development takes place in the testis under the influence of a variety of somatic cells and an incompletely defined paracrine and endocrine influences. Since it is not recapitulated well in vitro, researchers studying spermatogenesis often manipulate the germline by creating transgenic or knockout mice or by administering pharmaceutical agonists/antagonists or inhibitors. The effects of these types of manipulations on germline development can often be determined following microscopic imaging, both of stained and immunostained testis sections. Here, we describe approaches for microscopic analysis of the developing male germline, provide detailed protocols for a variety of immunostaining approaches, and discuss transgenic fluorescent reporter lines for studying the early stages of spermatogenesis.


Assuntos
Espermatogênese/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Espermatogênese/genética , Testículo/citologia , Testículo/metabolismo
9.
Biol Reprod ; 96(4): 816-828, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28379293

RESUMO

Spermatogonial stem cells must balance self-renewal with production of transit-amplifying progenitors that differentiate in response to retinoic acid (RA) before entering meiosis. This self-renewal vs. differentiation fate decision is critical for maintaining tissue homeostasis, as imbalances cause defects that can lead to human testicular cancer or infertility. Little is currently known about the program of differentiation initiated by RA, and the pathways and proteins involved are poorly defined. We recently found that RA stimulation of the Phosphatidylinositol 3-kinase (PI3K)/AKT/Mammalian target of rapamycin (mTOR) kinase signaling pathway is required for differentiation, and that short-term inhibition of mTOR complex 1 (mTORC1) by rapamycin blocked spermatogonial differentiation in vivo and prevented RA-induced translational activation. Since this phenotype resulted from global inhibition of mTORC1, we created conditional germ cell knockout mice to investigate the germ cell-autonomous role of MTOR in spermatogonial differentiation. MTOR germ cell KO mice were viable and healthy, but testes from neonatal (postnatal day (P)8), juvenile (P18), and adult (P > 60) KO mice were smaller than littermate controls, and no sperm were produced in adult testes. Histological and immunostaining analyses revealed that spermatogonial differentiation was blocked, and no spermatocytes were formed at any of the ages examined. Although spermatogonial proliferation was reduced in the neonatal testis, it was blocked altogether in the juvenile and adult testis. Importantly, a small population of self-renewing undifferentiated spermatogonia remained in adult testes. Taken together, these results reveal that MTOR is dispensable for the maintenance of undifferentiated spermatogonia, but is cell autonomously required for their proliferation and differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Espermatogônias/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Masculino , Camundongos , Camundongos Knockout , Espermatogênese , Serina-Treonina Quinases TOR/genética , Testículo/fisiologia
10.
Reproduction ; 152(5): 379-88, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27486269

RESUMO

We previously described a novel germ cell-specific X-linked reproductive homeobox gene (Rhox13) that is upregulated at the level of translation in response to retinoic acid (RA) in differentiating spermatogonia and preleptotene spermatocytes. We hypothesize that RHOX13 plays an essential role in male germ cell differentiation, and have tested this by creating a Rhox13 gene knockout (KO) mouse. Rhox13 KO mice are born in expected Mendelian ratios, and adults have slightly reduced testis weights, yet a full complement of spermatogenic cell types. Young KO mice (at ~7-8 weeks of age) have a ≈50% reduction in epididymal sperm counts, but numbers increased to WT levels as the mice reach ~17 weeks of age. Histological analysis of testes from juvenile KO mice reveals a number of defects during the first wave of spermatogenesis. These include increased apoptosis, delayed appearance of round spermatids and disruption of the precise stage-specific association of germ cells within the seminiferous tubules. Breeding studies reveal that both young and aged KO males produce normal-sized litters. Taken together, our results indicate that RHOX13 is not essential for mouse fertility in a controlled laboratory setting, but that it is required for optimal development of differentiating germ cells and progression of the first wave of spermatogenesis.


Assuntos
Apoptose , Epididimo/citologia , Fertilização/fisiologia , Proteínas de Homeodomínio/fisiologia , Espermatócitos/citologia , Espermatogênese/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Dev Biol ; 407(1): 90-102, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26254600

RESUMO

Spermatogonial stem cells (SSCs) must balance self-renewal with production of transit-amplifying progenitors that differentiate in response to retinoic acid (RA) before entering meiosis. This self-renewal vs. differentiation spermatogonial fate decision is critical for maintaining tissue homeostasis, as imbalances cause spermatogenesis defects that can lead to human testicular cancer or infertility. A great deal of effort has been exerted to understand how the SSC population is maintained. In contrast, little is known about the essential program of differentiation initiated by retinoic acid (RA) that precedes meiosis, and the pathways and proteins involved are poorly defined. We recently reported a novel role for RA in stimulating the PI3/AKT/mTOR kinase signaling pathway to activate translation of repressed mRNAs such as Kit. Here, we examined the requirement for mTOR complex 1 (mTORC1) in mediating the RA signal to direct spermatogonial differentiation in the neonatal testis. We found that in vivo inhibition of mTORC1 by rapamycin blocked spermatogonial differentiation, which led to an accumulation of undifferentiated spermatogonia. In addition, rapamycin also blocked the RA-induced translational activation of mRNAs encoding KIT, SOHLH1, and SOHLH2 without affecting expression of STRA8. These findings highlight dual roles for RA in germ cell development - transcriptional activation of genes, and kinase signaling to stimulate translation of repressed messages required for spermatogonial differentiation.


Assuntos
Complexos Multiproteicos/fisiologia , Espermatogônias/citologia , Serina-Treonina Quinases TOR/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/antagonistas & inibidores , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Testículo/efeitos dos fármacos , Testículo/patologia , Tretinoína/farmacologia
12.
Reproduction ; 149(4): 329-38, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25737569

RESUMO

Prospermatogonia transition to type A spermatogonia, which provide the source for the spermatogonial stem cell (SSC) pool. A percentage of these type A spermatogonia then differentiate to enter meiosis as spermatocytes by ∼P10. It is currently unclear as to when these distinct populations are initially formed in the neonatal testis, and when the expression of markers both characteristic of and required for the adult undifferentiated and differentiating states is established. In this study, we compared expression of known spermatogonial cell fate markers during normal development and in response to the differentiation signal provided by retinoic acid (RA). We found that some markers for the undifferentiated state (ZBTB16/PLZF and CDH1) were expressed in nearly all spermatogonia from P1 through P7. In contrast, differentiation markers (STRA8 and KIT) appeared in a subset of spermatogonia at P4, coincident with the onset of RA signaling. GFRA1, which was present in nearly all prospermatogonia at P1, was only retained in STRA8/KIT- spermatogonia. From P4 through P10, there was a great deal of heterogeneity in the male germ cell population in terms of expression of markers, as markers characteristic of the undifferentiated (except GFRA1) and differentiating states were co-expressed through this interval. After P10, these fate markers diverged to mark distinct populations of undifferentiated and differentiating spermatogonia, and this pattern was maintained in juvenile (P18) and adult (P>60) testes. Taken together, these results reveal that the spermatogonia population is heterogeneous during the first wave of spermatogenesis, and indicate that neonatal spermatogonia may not serve as an ideal substitute for studying the function of adult spermatogonia.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Testículo/citologia , Animais , Animais Recém-Nascidos , Antineoplásicos/farmacologia , Proteínas Cdh1/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Técnicas Imunoenzimáticas , Fatores de Transcrição Kruppel-Like/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Proto-Oncogênicas c-kit/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Tretinoína/farmacologia
13.
Dev Biol ; 397(1): 140-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25446031

RESUMO

In the testis, a subset of spermatogonia retains stem cell potential, while others differentiate to eventually become spermatozoa. This delicate balance must be maintained, as defects can result in testicular cancer or infertility. Currently, little is known about the gene products and signaling pathways directing these critical cell fate decisions. Retinoic acid (RA) is a requisite driver of spermatogonial differentiation and entry into meiosis, yet the mechanisms activated downstream are undefined. Here, we determined a requirement for RA in the expression of KIT, a receptor tyrosine kinase essential for spermatogonial differentiation. We found that RA signaling utilized the PI3K/AKT/mTOR signaling pathway to induce the efficient translation of mRNAs for Kit, which are present but not translated in undifferentiated spermatogonia. Our findings provide an important molecular link between a morphogen (RA) and the expression of KIT protein, which together direct the differentiation of spermatogonia throughout the male reproductive lifespan.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Espermatogênese , Tretinoína/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Espermatogônias/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Testículo/metabolismo
14.
Reproduction ; 148(4): 333-41, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24989903

RESUMO

Sertoli cells undergo terminal differentiation at puberty to support all phases of germ cell development, which occurs in the mouse beginning in the second week of life. By ∼18 days postpartum (dpp), nearly all Sertoli cells have ceased proliferation. This terminal differentiation is accompanied by the development of unique and regionally concentrated filamentous actin (F-actin) structures at the basal and apical aspects of the seminiferous epithelium, and this reorganization is likely to involve the action of actin-binding proteins. Palladin (PALLD) is a widely expressed F-actin-binding and bundling protein recently shown to regulate these structures, yet it is predominantly nuclear in Sertoli cells at puberty. We found that PALLD localized within nuclei of primary Sertoli cells grown in serum-free media but relocalized to the cytoplasm upon serum stimulation. We utilized this system with in vivo relevance to Sertoli cell development to investigate mechanisms regulating nuclear localization of this F-actin-binding protein. Our results indicate that PALLD can be shuttled from the nucleus to the cytoplasm, and that this relocalization occurred following depolymerization of the F-actin cytoskeleton in response to cAMP signaling. Nuclear localization was reduced in Hpg-mutant testes, suggesting the involvement of gonadotropin signaling. We found that PALLD nuclear localization was unaffected in testis tissues from LH receptor and androgen receptor-mutant mice. However, PALLD nuclear localization was reduced in the testes of FSH receptor-mutant mice, suggesting that FSH signaling during Sertoli cell maturation regulates this subcellular localization.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Células de Sertoli/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Citoplasma/metabolismo , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Carioferinas/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Knockout , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína Exportina 1
15.
Mol Reprod Dev ; 80(5): 403-13, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23559268

RESUMO

In the testis, F-actin structures are involved in spermatid nuclear remodeling and cytoplasm reduction, maintenance of the blood-testis barrier, support of the spermatogonial stem cell niche, and release of spermatids into the tubular lumen. To gain a better understanding of actin regulation in Sertoli-germ cell interactions, we investigated the expression of the Palladin (Palld) gene, which encodes a widely expressed phosphoprotein that localizes to actin-rich cytoplasmic structures, including focal adhesions, cell-cell junctions, podosomes, and stress fibers, and serves as a molecular scaffold to bundle actin fibers. In germ cells, PALLD was concentrated along the tubulin- and F-actin-containing cytoplasmic manchette that forms adjacent to the elongating spermatid nucleus during spermiogenesis. To our surprise, PALLD relocated from the cytoplasm to the nucleus of Sertoli cells in the juvenile testis, coincident with the onset of puberty, and this localization was maintained in the adult. We provide evidence that the 140 kDa isoform of PALLD predominates in Sertoli cells, and that it is apparently cleaved, with the C-terminus localizing to the nucleus while the N-terminus remains cytoplasmic. We investigated the nuclear localization of the C-terminus of PALLD and found that it is regulated by a putative nuclear export signal. These results provide the foundation for future work employing Sertoli cell- and spermatid-specific Palld-knockout mice to study diverse roles of PALLD as both a nuclear-actin regulatory protein and as a potential regulator of manchette formation during spermatogenesis.


Assuntos
Núcleo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Proteólise , Células de Sertoli/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Citoplasma/metabolismo , Masculino , Camundongos , Estrutura Terciária de Proteína , Células de Sertoli/citologia , Maturidade Sexual/fisiologia , Espermátides/citologia , Espermátides/metabolismo , Espermatogênese/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...