Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 367(6474): 161-166, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31857493

RESUMO

Tissue homeostasis requires the balance of growth by cell production and regression through cell loss. In the hair cycle, during follicle regression, the niche traverses the skin through an unknown mechanism to reach the stem cell reservoir and trigger new growth. Here, we identify the dermal sheath that lines the follicle as the key driver of tissue regression and niche relocation through the smooth muscle contractile machinery that generates centripetal constriction force. We reveal that the calcium-calmodulin-myosin light chain kinase pathway controls sheath contraction. When this pathway is blocked, sheath contraction is inhibited, impeding follicle regression and niche relocation. Thus, our study identifies the dermal sheath as smooth muscle that drives follicle regression for reuniting niche and stem cells in order to regenerate tissue structure during homeostasis.


Assuntos
Derme/fisiologia , Folículo Piloso/fisiologia , Músculo Liso/fisiologia , Regeneração , Nicho de Células-Tronco/fisiologia , Agrecanas/genética , Animais , Humanos , Camundongos , Camundongos Mutantes , Contração Muscular
2.
Exp Dermatol ; 28(4): 332-344, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30887615

RESUMO

Hair follicle (HF) formation in developing embryonic skin requires stepwise signalling between the epithelial epidermis and mesenchymal dermis, and their specialized derivatives, the placode/germ/peg and dermal condensate/papilla, respectively. Classically, distinct stages of HF morphogenesis have been defined, in the mouse model, based on (a) changes in cell morphology and aggregation; (b) expression of few known molecular markers; (c) the extent of follicle downgrowth; and (d) the presence of differentiating cell types. Refined genetic strategies and recent emerging technologies, such as live imaging and transcriptome analyses of isolated cell populations or single cells, have enabled a closer dissection of the signalling requirements at different stages of HF formation, particularly early on. They have also led to the discovery of precursor cells for placode, dermal condensate and future bulge stem cells that, combined with molecular insights into their fate specification and subsequent formation, serve as novel landmarks for early HF morphogenetic events and studies of the signalling networks mediating these processes. In this review, we integrate the emergence of HF precursor cell states and novel molecular markers of fate and formation to update the widely used 20-year-old seminal classification guide of HF morphogenetic stages by Paus et al. We then temporally describe the latest insights into the early cellular and molecular events and signalling requirements for HF morphogenesis in relation to one another in a holistic manner.


Assuntos
Folículo Piloso/embriologia , Animais , Humanos , Morfogênese , Transdução de Sinais
3.
Dev Cell ; 48(1): 32-48.e5, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30595537

RESUMO

Cell fate transitions are essential for specification of stem cells and their niches, but the precise timing and sequence of molecular events during embryonic development are largely unknown. Here, we identify, with 3D and 4D microscopy, unclustered precursors of dermal condensates (DC), signaling niches for epithelial progenitors in hair placodes. With population-based and single-cell transcriptomics, we define a molecular time-lapse from pre-DC fate specification through DC niche formation and establish the developmental trajectory as the DC lineage emerges from fibroblasts. Co-expression of downregulated fibroblast and upregulated DC genes in niche precursors reveals a transitory molecular state following a proliferation shutdown. Waves of transcription factor and signaling molecule expression then coincide with DC formation. Finally, ablation of epidermal Wnt signaling and placode-derived FGF20 demonstrates their requirement for pre-DC specification. These findings uncover a progenitor-dependent niche precursor fate and the transitory molecular events controlling niche formation and function.


Assuntos
Diferenciação Celular/fisiologia , Derme/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Folículo Piloso/metabolismo , Animais , Fibroblastos/citologia , Folículo Piloso/embriologia , Transdução de Sinais/genética , Pele/metabolismo , Células-Tronco/citologia
4.
Cell Rep ; 25(4): 823-824, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30355488

RESUMO

Compared to mouse models, less is known about human epidermal cell states and differentiation. In this issue of Cell Reports, Cheng et al. (2018) dissect the cell states and heterogeneity in human epidermis with large-scale transcriptomics of 92,889 single epidermal cells from normal and inflamed skin.


Assuntos
Células Epidérmicas , Transcriptoma , Animais , Diferenciação Celular , Epiderme , Humanos , Camundongos , Pele
5.
Cell Rep ; 14(12): 3001-18, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27009580

RESUMO

The hair follicle (HF) is a complex miniorgan that serves as an ideal model system to study stem cell (SC) interactions with the niche during growth and regeneration. Dermal papilla (DP) cells are required for SC activation during the adult hair cycle, but signal exchange between niche and SC precursors/transit-amplifying cell (TAC) progenitors that regulates HF morphogenetic growth is largely unknown. Here we use six transgenic reporters to isolate 14 major skin and HF cell populations. With next-generation RNA sequencing, we characterize their transcriptomes and define unique molecular signatures. SC precursors, TACs, and the DP niche express a plethora of ligands and receptors. Signaling interaction network analysis reveals a bird's-eye view of pathways implicated in epithelial-mesenchymal interactions. Using a systematic tissue-wide approach, this work provides a comprehensive platform, linked to an interactive online database, to identify and further explore the SC/TAC/niche crosstalk regulating HF growth.


Assuntos
Folículo Piloso/metabolismo , Pele/metabolismo , Células-Tronco/citologia , Animais , Citometria de Fluxo , Queratina-14/genética , Queratina-14/metabolismo , Camundongos , Microscopia de Fluorescência , Análise de Componente Principal , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/genética , Pele/citologia , Nicho de Células-Tronco , Células-Tronco/metabolismo , Transcriptoma
6.
FASEB J ; 30(4): 1436-52, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26678449

RESUMO

Earlier studies have shown that rats treated with an acute dose of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide (adjudin, a male contraceptive under development) causes permanent infertility due to irreversible blood-testis barrier (BTB) disruption even though the population of undifferentiated spermatogonia remains similar to normal rat testes, because spermatogonia fail to differentiate into spermatocytes to enter meiosis. Since other studies have illustrated the significance of connexin 43 (Cx43)-based gap junction in maintaining the homeostasis of BTB in the rat testis and the phenotypes of Sertoli cell-conditional Cx43 knockout mice share many of the similarities of the adjudin-treated rats, we sought to examine if overexpression of Cx43 in these adjudin-treated rats would reseal the disrupted BTB and reinitiate spermatogenesis. A full-length Cx43 cloned into mammalian expression vector pCI-neo was used to transfect testes of adjudin-treated ratsversusempty vector. It was found that overexpression of Cx43 indeed resealed the Sertoli cell tight junction-permeability barrier based on a functionalin vivoassay in tubules displaying signs of meiosis as noted by the presence of round spermatids. Thus, these findings suggest that overexpression of Cx43 reinitiated spermatogenesis at least through the steps of meiosis to generate round spermatids in testes of rats treated with an acute dose of adjudin that led to aspermatogenesis. It was also noted that the round spermatids underwent eventual degeneration with the formation of multinucleated cells following Cx43 overexpression due to the failure of spermiogenesis because no elongating/elongated spermatids were detected in any of the tubules examined. The mechanism by which overexpression of Cx43 reboots meiosis and rescues BTB function was also examined. In summary, overexpression of Cx43 in the testis with aspermatogenesis reboots meiosis and reseals toxicant-induced BTB disruption, even though it fails to support round spermatids to enter spermiogenesis.-Li, N., Mruk, D. D., Mok, K.-W., Li, M. W. M., Wong, C. K. C., Lee, W. M., Han, D., Silvestrini, B., Cheng, C. Y. Connexin 43 reboots meiosis and reseals blood-testis barrier following toxicant-mediated aspermatogenesis and barrier disruption.


Assuntos
Barreira Hematotesticular/metabolismo , Conexina 43/genética , Meiose/genética , Espermatogênese/genética , Animais , Barreira Hematotesticular/efeitos dos fármacos , Conexina 43/metabolismo , Expressão Gênica/efeitos dos fármacos , Hidrazinas/farmacologia , Immunoblotting , Indazóis/farmacologia , Masculino , Camundongos Knockout , Microscopia de Fluorescência , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Espermátides/efeitos dos fármacos , Espermátides/metabolismo , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo
7.
Dev Cell ; 34(5): 577-91, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26256211

RESUMO

Defining the unique molecular features of progenitors and their niche requires a genome-wide, whole-tissue approach with cellular resolution. Here, we co-isolate embryonic hair follicle (HF) placode and dermal condensate cells, precursors of adult HF stem cells and the dermal papilla/sheath niche, along with lineage-related keratinocytes and fibroblasts, Schwann cells, melanocytes, and a population inclusive of all remaining skin cells. With next-generation RNA sequencing, we define gene expression patterns in the context of the entire embryonic skin, and through transcriptome cross-comparisons, we uncover hundreds of enriched genes in cell-type-specific signatures. Axon guidance signaling and many other pathway genes are enriched in multiple signatures, implicating these factors in driving the large-scale cellular rearrangements necessary for HF formation. Finally, we share all data in an interactive, searchable companion website. Our study provides an overarching view of signaling within the entire embryonic skin and captures a molecular snapshot of HF progenitors and their niche.


Assuntos
Folículo Piloso/citologia , Folículo Piloso/embriologia , Queratinócitos/citologia , Pele/metabolismo , Células-Tronco/citologia , Transcriptoma/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Camundongos , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Pele/citologia , Pele/embriologia , Nicho de Células-Tronco
8.
Endocrinology ; 156(5): 1900-13, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25714812

RESUMO

In the seminiferous epithelium of rat testes, preleptotene spermatocytes residing in the basal compartment are transported across the blood-testis barrier (BTB) to enter the adluminal compartment at stage VIII of the epithelial cycle. This process involves redistribution of tight junction (TJ) proteins via reorganization of actin cytoskeleton in Sertoli cells that serves as attachment site for adhesion protein complexes. Ribosomal protein S6 (rpS6), a downstream molecule of mTORC1 (mammalian target of rapamycin complex 1), participates in this process via a yet-to-be defined mechanism. Here, we constructed an rpS6 quadruple phosphomimetic mutant by converting Ser residues at 235, 236, 240, and 244 to Glu via site-directed mutagenesis, making this mutant constitutively active. When this rpS6 mutant was overexpressed in Sertoli cells cultured in vitro with an established TJ barrier mimicking the BTB in vivo, it perturbed the TJ permeability by down-regulating and redistributing TJ proteins at the cell-cell interface. These changes are mediated by a reorganization of actin microfilaments, which was triggered by a redistribution of activated actin-related protein 3 (Arp3) as well as changes in Arp3-neuronal Wiskott-Aldrich Syndrome protein (N-WASP) interaction. This in turn induced reorganization of actin microfilaments, converting them from a "bundled" to an "unbundled/branched" configuration, concomitant with a reduced actin bundling activity, thereby destabilizing the TJ-barrier function. These changes were mediated by Akt (transforming oncogene of v-akt), because an Akt knockdown by RNA interference was able to mimic the phenotypes of rpS6 mutant overexpression at the Sertoli cell BTB. In summary, this study illustrates a mechanism by which mTORC1 signal complex regulates BTB function through rpS6 downstream by modulating actin organization via the Arp2/3 complex, which may be applicable to other tissue barriers.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Barreira Hematotesticular/metabolismo , Complexos Multiproteicos/metabolismo , Proteína S6 Ribossômica/genética , Células de Sertoli/metabolismo , Espermatócitos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Junções Íntimas/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Técnicas de Silenciamento de Genes , Técnicas In Vitro , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteína Oncogênica v-akt/genética , Ratos , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
9.
Endocrinology ; 156(2): 680-93, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25456071

RESUMO

During spermatogenesis, developing germ cells are transported across the seminiferous epithelium. Studies propose that because microtubules (MTs) serve as the tracks for transporting cell organelles, they may also serve a similar function in the transport of developing germ cells. Polarized MTs may provide the tracks along which polarized actin microfilaments, which act as vehicles to transport cargo, such as preleptotene spermatocytes through the blood-testis barrier (BTB) and spermatids across the epithelium. Yet the molecular mechanism(s) underlying these events remain unknown. Using an established in vitro Sertoli cell system to study BTB function, we demonstrated herein that a MT regulatory protein end-binding protein 1 (EB1) regulates the MT- and also the actin-based cytoskeleton of the Sertoli cell BTB in the rat. EB1 serves as a coordinator between the two cytoskeletons by regulating MT polymerization and actin filament bundling to modulate germ cell transport at the Sertoli cell BTB. A knockdown of EB1 by RNA interference was found to perturb the tight junction (TJ)-permeability barrier, as evidenced by mislocalization of junctional proteins critical for barrier function to facilitate spermatocyte transport, which was likely achieved by two coordinated events. First, EB1 knockdown resulted in changes in MT polymerization, thereby perturbing MT organization in Sertoli cells in which polarized MT no longer stretched properly across the cell cytosol to serve as the tracks. Second, EB1 knockdown perturbed actin organization via its effects on the branched actin polymerization-inducing protein called Arp3 (actin-related protein 3), perturbing microfilament bundling capability based on a biochemical assay, thereby causing microfilament truncation and misorganization, disrupting the function of the vehicle. This reduced actin microfilament bundling capability thus perturbed TJ-protein distribution and localization at the BTB, destabilizing the TJ barrier, leading to its remodeling to facilitate spermatocyte transport. In summary, EB1 provides a functional link between tubulin- and actin-based cytoskeletons to confer spermatocyte transport at the BTB.


Assuntos
Barreira Hematotesticular , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/fisiologia , Epitélio Seminífero/metabolismo , Espermatogênese , Citoesqueleto de Actina/metabolismo , Animais , Masculino , Interferência de RNA , Ratos Sprague-Dawley , Células de Sertoli/metabolismo , Tubulina (Proteína)/metabolismo
10.
J Cell Sci ; 127(Pt 22): 4870-82, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25217631

RESUMO

Mammalian target of rapamycin complex 1 (mTORC1) is an emerging regulator of blood-tissue barriers that utilizes ribosomal protein S6 (rpS6) as the downstream signaling molecule. To explore the role of rpS6 in blood-testis barrier (BTB) function, a constitutively active quadruple rpS6 phosphomimetic mutant was constructed in mammalian expression vector and overexpressed in Sertoli cells cultured in vitro that mimicked the BTB in vivo. Using this quadruple phosphomimetic mutant, phosphorylated (p)-rpS6 was shown to disrupt IGF-1/insulin signaling, thereby abolishing Akt phosphorylation, which led to an induction of MMP-9. This increase in MMP-9 secretion perturbed the Sertoli cell tight junction permeability barrier by proteolysis-mediated downregulation of tight junction proteins at the BTB. These findings were confirmed by the use of a specific MMP-9 inhibitor that blocked the disruption of the tight junction permeability barrier by the rpS6 mutant. Additionally, RNA interference (RNAi)-mediated Akt silencing was able to mimic the results of rpS6 mutant overexpression in Sertoli cells, further confirming this p-rpS6-Akt-MMP-9 signaling pathway. In conclusion, these data support a new concept of mTORC1-mediated BTB regulation, that is possibly also applicable to other blood-tissue barriers.


Assuntos
Barreira Hematotesticular/metabolismo , Proteína S6 Ribossômica/metabolismo , Testículo/irrigação sanguínea , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Células de Sertoli/metabolismo , Espermatogênese , Junções Íntimas/metabolismo , Transfecção
11.
PLoS Genet ; 10(6): e1004447, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967734

RESUMO

During spermatogenesis, the blood-testis barrier (BTB) segregates the adluminal (apical) and basal compartments in the seminiferous epithelium, thereby creating a privileged adluminal environment that allows post-meiotic spermatid development to proceed without interference of the host immune system. A key feature of the BTB is its continuous remodeling within the Sertoli cells, the major somatic component of the seminiferous epithelium. This remodeling is necessary to allow the transport of germ cells towards the seminiferous tubule interior, while maintaining intact barrier properties. Here we demonstrate that the actin nucleation promoting factor Neuronal Wiskott-Aldrich Syndrome Protein (N-WASP) provides an essential function necessary for BTB restructuring, and for maintaining spermatogenesis. Our data suggests that the N-WASP-Arp2/3 actin polymerization machinery generates branched-actin arrays at an advanced stage of BTB remodeling. These arrays are proposed to mediate the restructuring process through endocytic recycling of BTB components. Disruption of N-WASP in Sertoli cells results in major structural abnormalities to the BTB, including mis-localization of critical junctional and cytoskeletal elements, and leads to disruption of barrier function. These impairments result in a complete arrest of spermatogenesis, underscoring the critical involvement of the somatic compartment of the seminiferous tubules in germ cell maturation.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Barreira Hematotesticular , Espermatogênese/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Masculino , Camundongos , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Testículo/metabolismo
12.
Am J Physiol Endocrinol Metab ; 305(6): E687-99, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23880313

RESUMO

During spermatogenesis, the molecular mechanism that confers spermatid adhesion to the Sertoli cell at the apical ectoplasmic specialization (apical ES), a testis-specific F-actin-rich adherens junction, in the rat testis remains elusive. Herein, the activated form of focal adhesion kinase (FAK), p-FAK-Tyr(397), a component of the apical ES that was expressed predominantly and stage specifically in stage VII-early stage VIII tubules, was found to be a crucial apical ES regulator. Using an FAK-Y397E phosphomimetic mutant cloned in a mammalian expression vector for its transfection vs. FAK and vector alone in adult rat testes in vivo, its overexpression was found to cause defects in spermiation. These defects in spermiation were manifested by entrapment of spermatids in the seminiferous epithelium in late stage VIII-X tubules and were mediated by a disruption on the spatiotemporal expression and/or mislocalization of actin regulatory protein actin-related protein 3, which induces branched actin polymerization, epidermal growth factor receptor pathway substrate 8 (an actin barbed end capping and bundling protein), and palladin (an actin cross-linking and bundling protein). This thus perturbed changes of F-actin organization at the apical ES to facilitate spermiation, which also led to a concomitant alteration in the distribution and upregulation of adhesion proteins nectin-2 and nectin-3 at the apical ES. As such, nectin-2 and -3 remained at the apical ES to anchor step 19 spermatids on to the epithelium, delaying spermiation. These findings illustrate a mechanistic pathway mediated by p-FAK-Tyr(397) that regulates spermatid adhesion at the apical ES in vivo.


Assuntos
Actinas/metabolismo , Adesão Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Espermátides/metabolismo , Espermatogênese/fisiologia , Testículo/fisiologia , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animais , Animais Geneticamente Modificados , Barreira Hematotesticular/metabolismo , Quinase 1 de Adesão Focal/genética , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/citologia , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermátides/citologia , Testículo/metabolismo , Tirosina/metabolismo
13.
FASEB J ; 27(3): 1137-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288930

RESUMO

In the mammalian testis, coexisting tight junctions (TJs), basal ectoplasmic specializations, and gap junctions (GJs), together with desmosomes near the basement membrane, constitute the blood-testis barrier (BTB). The most notable feature of the BTB, however, is the extensive network of actin filament bundles, which makes it one of the tightest blood-tissue barriers. The BTB undergoes restructuring to facilitate the transit of preleptotene spermatocytes at stage VIII-IX of the epithelial cycle. Thus, the F-actin network at the BTB undergoes cyclic reorganization via a yet-to-be explored mechanism. Rictor, the key component of mTORC2 that is known to regulate actin cytoskeleton, was shown to express stage-specifically at the BTB in the seminiferous epithelium. Its expression was down-regulated at the BTB in stage VIII-IX tubules, coinciding with BTB restructuring at these stages. Using an in vivo model, a down-regulation of rictor at the BTB was also detected during adjudin-induced BTB disruption, illustrating rictor expression is positively correlated with the status of the BTB integrity. Indeed, the knockdown of rictor by RNAi was found to perturb the Sertoli cell TJ-barrier function in vitro and the BTB integrity in vivo. This loss of barrier function was accompanied by changes in F-actin organization at the Sertoli cell BTB in vitro and in vivo, associated with a loss of interaction between actin and α-catenin or ZO-1. Rictor knockdown by RNAi was also found to impede Sertoli cell-cell GJ communication, disrupting protein distribution (e.g., occludin, ZO-1) at the BTB, illustrating that rictor is a crucial BTB regulator.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas de Transporte/biossíntese , Comunicação Celular/fisiologia , Junções Comunicantes/metabolismo , Complexos Multiproteicos/metabolismo , Fibras de Estresse/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Barreira Hematotesticular/citologia , Proteínas de Transporte/genética , Comunicação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Junções Comunicantes/genética , Técnicas de Silenciamento de Genes , Hidrazinas/farmacologia , Indazóis/farmacologia , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos/genética , Proteína Companheira de mTOR Insensível à Rapamicina , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/citologia , Epitélio Seminífero/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Fibras de Estresse/genética , Serina-Treonina Quinases TOR/genética , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
14.
Int Rev Cell Mol Biol ; 301: 291-358, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23317821

RESUMO

In mammalian testes, haploid spermatozoa are formed from diploid spermatogonia during spermatogenesis, which is a complicated cellular process. While these cellular events were reported in the 1960s and 1970s, the underlying molecular mechanism(s) that regulates these events remained unexplored until the past ∼10 years. For instance, adhesion proteins were shown to be integrated components at the Sertoli cell-cell interface and/or the Sertoli-spermatid interface in the late 1980s. But only until recently, studies have demonstrated that some of the adhesion proteins serve as the platform for signal transduction that regulates cell adhesion. In this chapter, a brief summary and critical discussion are provided on the latest findings regarding these cell-adhesion proteins in the testis and their relationship to spermatogenesis. Moreover, antagonistic effects of two mammalian target of rapamycin (mTOR) complexes, known as mTORC1 and mTORC2, on cell-adhesion function in the testis are discussed. Finally, a hypothetic model is presented to depict how these two mTOR-signaling complexes having the "yin" and "yang" antagonistic effects on the Sertoli cell tight junction (TJ)-permeability barrier can maintain the blood-testis barrier (BTB) integrity during the epithelial cycle while preleptotene spermatocytes are crossing the BTB.


Assuntos
Barreira Hematotesticular/metabolismo , Complexos Multiproteicos/metabolismo , Espermatogênese , Serina-Treonina Quinases TOR/metabolismo , Actinas/metabolismo , Animais , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Modelos Biológicos
15.
Endocrinology ; 153(10): 5036-48, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22948214

RESUMO

During spermatogenesis, preleptotene spermatocytes residing near the basement membrane of the seminiferous tubule must traverse the blood-testis barrier (BTB) at stage VIII-IX of the epithelial cycle to continue their development in the adluminal compartment. Unlike other blood-tissue barriers (e.g. the blood-brain barrier) that are created by the endothelial tight junction (TJ) barrier of capillaries, the BTB is created by specialized junctions between Sertoli cells in which TJ coexists with basal ectoplasmic specialization (basal ES, a testis-specific adherens junction). The basal ES is typified by the presence of tightly packed actin filament bundles sandwiched between cisternae of endoplasmic reticulum and the apposing plasma membranes of Sertoli cells. These actin filament bundles also confer unusual adhesive strength to the BTB. Yet the mechanisms by which these filamentous actin (F-actin) networks are regulated from the bundled to the debundled state to facilitate the transit of spermatocytes remain elusive. Herein, we provide evidence that ribosomal protein S6 (rpS6), the downstream signaling molecule of the mammalian target of rapamycin complex 1 (mTORC1) pathway, is a major regulator of F-actin organization and adhesion protein recruitment at the BTB. rpS6 is restrictively and spatiotemporally activated at the BTB during the epithelial cycle. An activation of rpS6 led to a disruption of the Sertoli cell TJ barrier and BTB integrity. Its silencing in vitro or in vivo by using small interfering RNA duplexes or short hairpin RNA was found to promote the Sertoli cell TJ permeability barrier by the recruitment of adhesion proteins (e.g. claudin-11 and occludin) to the BTB. Thus, rpS6 in the mTORC1 pathway regulates BTB restructuring via its effects on the F-actin organization and protein recruitment at the BTB.


Assuntos
Actinas/metabolismo , Barreira Hematotesticular/metabolismo , Proteína S6 Ribossômica/metabolismo , Células de Sertoli/metabolismo , Animais , Barreira Hematotesticular/efeitos dos fármacos , Hidrazinas/farmacologia , Indazóis/farmacologia , Masculino , Permeabilidade , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Espermatogênese/efeitos dos fármacos , Espermatogênese/fisiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
16.
Proc Natl Acad Sci U S A ; 109(31): 12562-7, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22797892

RESUMO

Focal adhesion kinase (FAK), a nonreceptor protein tyrosine kinase, displays phosphorylation-dependent localization in the seminiferous epithelium of adult rat testes. FAK is an integrated component of the blood-testis barrier (BTB) involved in regulating Sertoli cell adhesion via its effects on the occludin-zonula occludens-1 complex. Herein, we report that p-FAK-Tyr(407) and p-FAK-Tyr(397) display restricted spatiotemporal and almost mutually exclusive localization in the epithelium, affecting BTB dynamics antagonistically, with the former promoting and the latter disrupting the Sertoli cell tight junction-permeability barrier function. Using primary cultured Sertoli cells as an in vitro model that mimics the BTB in vivo both functionally and ultrastructurally, effects of FAK phosphorylation on BTB function were studied by expressing nonphosphorylatable and phosphomimetic mutants, with tyrosine replaced by phenylalanine (F) and glutamate (E), respectively. Compared with WT FAK, Y407E and Y397F mutations each promoted barrier function, and the promoting effect of the Y407E mutant was abolished in the Y397E-Y407E double mutant, demonstrating antagonism between Tyr(407) and Tyr(397). Furthermore, Y407E mutation induced the recruitment of actin-related protein 3 to the Sertoli cell-cell interface, where it became more tightly associated with neuronal Wiskott-Aldrich syndrome protein, promoting actin-related protein 2/3 complex activity. Conversely, Y407F mutation reduced the rate of actin polymerization at the Sertoli cell BTB. In summary, FAK-Tyr(407) phosphorylation promotes BTB integrity by strengthening the actin filament-based cytoskeleton. FAK serves as a bifunctional molecular "switch" to direct the cyclical disassembly and reassembly of the BTB during the epithelial cycle of spermatogenesis, depending on its phosphorylation status, to facilitate the transit of preleptotene spermatocytes across the BTB.


Assuntos
Barreira Hematotesticular/enzimologia , Quinase 1 de Adesão Focal/metabolismo , Células de Sertoli/metabolismo , Junções Íntimas/enzimologia , Actinas/genética , Actinas/metabolismo , Substituição de Aminoácidos , Animais , Barreira Hematotesticular/citologia , Citoesqueleto/genética , Citoesqueleto/metabolismo , Quinase 1 de Adesão Focal/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação de Sentido Incorreto , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/genética , Multimerização Proteica/fisiologia , Ratos , Células de Sertoli/citologia , Espermatócitos/citologia , Espermatócitos/enzimologia , Espermatogênese/fisiologia , Junções Íntimas/genética , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Proteína da Zônula de Oclusão-1
17.
Spermatogenesis ; 2(2): 117-126, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22670221

RESUMO

During the seminiferous epithelial cycle of spermatogenesis, the ectoplasmic specialization (ES, a testis-specific adherens junction, AJ, type) maintains the polarity of elongating/elongated spermatids and confers adhesion to Sertoli cells in the seminiferous epithelium, and known as the apical ES. On the other hand, the ES is also found at the Sertoli-Sertoli cell interface at the blood-testis barrier (BTB) known as basal ES, which together with the tight junction (TJ), maintains Sertoli cell polarity and adhesion, creating a functional barrier that limits paracellular transport of substances across the BTB. However, the apical and basal ES are segregated and restricted to the adluminal compartment and the BTB, respectively. During the transit of preleptotene spermatocytes across the BTB and the release of sperm at spermiation at stage VIII of the seminiferous epithelial cycle, both the apical and basal ES undergo extensive restructuring to facilitate cell movement at these sites. The regulation of these events, in particular their coordination, remains unclear. Studies in other epithelia have shown that the tubulin cytoskeleton is intimately related to cell movement, and MARK [microtubule-associated protein (MAP)/microtubule affinity-regulating kinase] family kinases are crucial regulators of tubulin cytoskeleton stability. Herein MARK4, the predominant member of the MARK protein family in the testis, was shown to be expressed by both Sertoli and germ cells. MARK4 was also detected at the apical and basal ES, displaying highly restrictive spatiotemporal expression at these sites, as well as co-localizing with markers of the apical and basal ES. The expression of MARK4 was found to be stage-specific during the epithelial cycle, structurally associating with α-tubulin and the desmosomal adaptor plakophilin-2, but not with actin-based BTB proteins occludin, ß-catenin and Eps8 (epidermal growth factor receptor pathway substrate 8, an actin bundling and barbed end capping protein). More importantly, it was shown that the expression of MARK4 tightly associated with the integrity of the apical ES because a diminished expression of MARK4 associated with apical ES disruption that led to the detachment of elongating/elongated spermatids from the epithelium. These findings thus illustrate that the integrity of apical ES, an actin-based and testis-specific AJ, is dependent not only on the actin filament network, but also on the tubulin-based cytoskeleton.

18.
Adv Exp Med Biol ; 763: 318-33, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23397632

RESUMO

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in mammals including rodents and humans. It is used to sequester meiosis I and II, postmeiotic spermatid development via spermiogenesis and the release of sperm at spermiation from the systemic circulation, such that these events take place in an immune-privileged site in the adluminal (apical) compartment behind the BTB, segregated from the host immune system. Additionally, drug transporters, namely efflux (e.g., P-glycoprotein) and influx (e.g., Oatp3) pumps, many of which are integral membrane proteins in Sertoli cells at the BTB also work cooperatively to restrict the entry of drugs, toxicants, chemicals, steroids and other xenobiotics into the adluminal compartment. As such, the BTB that serves as an important physiological and selective barrier to protect germ cell development also poses a "hurdle" in male contraceptive development. For instance, adjudin, 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide, a potential nonhormonal male contraceptive that exerts its effects on germ cell adhesion, most notably at the Sertoli cell-spermatid interface, to induce "premature" germ cell loss from the seminiferous epithelium mimicking spermiation, has a relatively poor bioavailability largely because of the BTB. Since male contraceptives (e.g., adjudin) will be used by healthy men for an extended period of his life span after puberty, a better understanding on the BTB is necessary in order to effectively deliver drugs across this blood-tissue barrier in particular if these compounds exert their effects on developing germ cells in the adluminal compartment. This can also reduce long-term toxicity and health risk if the effective dosing can be lowered in order to widen the margin between its safety and efficacy. Herein, we summarize latest findings in this area of research, we also provide a critical evaluation on research areas that deserve attention in future studies.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Barreira Hematotesticular/metabolismo , Hidrazinas/farmacologia , Indazóis/farmacologia , Testículo/metabolismo , Junções Aderentes/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Transporte Biológico , Barreira Hematotesticular/efeitos dos fármacos , Anticoncepcionais Masculinos/administração & dosagem , Anticoncepcionais Masculinos/farmacologia , Humanos , Hidrazinas/administração & dosagem , Indazóis/administração & dosagem , Masculino , Taxa de Depuração Metabólica , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Espermátides/metabolismo , Testículo/efeitos dos fármacos
19.
Adv Exp Med Biol ; 763: 334-355, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23397633

RESUMO

The blood-testis barrier (BTB), similar to other blood-tissue barriers, such as the blood-brain barrier and the blood-retinal barrier, is used to protect the corresponding organ from harmful substances (e.g., xenobiotics) including drugs and foreign compounds. More importantly, the BTB allows postmeiotic spermatid development to take place in an immune privileged site at the adluminal (or apical) compartment to avoid the production of antibodies against spermatid-specific antigens, many of which express transiently during spermiogenesis and spermiation. The BTB, however, also poses an obstacle in developing nonhormonal-based male contraceptives by sequestering drugs (e.g., adjudin) that exert their effects on germ cells in the adluminal compartment. The effects of these drugs include disruption of germ cell cycle progression and development, apoptosis, cell adhesion, metabolism and others. Recent studies have demonstrated that there is a functional axis that operates locally in the seminiferous epithelium to co-ordinate different cellular events across the Sertoli cell epithelium, such as spermiation and BTB restructuring during the seminiferous epithelial cycle of spermatogenesis. Components of this functional axis, such as the apical ectoplasmic specialization (apical ES, a testis-specific atypical anchoring junction type) and the BTB, in particular their constituent protein complexes, such as alpha6beta1-integrin and occludin at the apical ES and the BTB, respectively, can be the target of male contraception. In this chapter, we highlight recent advances regarding the likely mechanism of action of adjudin in this functional axis with emphasis on the use of molecular modeling technique to facilitate the design of better compounds in male contraceptive development.


Assuntos
Barreira Hematotesticular/efeitos dos fármacos , Anticoncepção/métodos , Espermatogênese/efeitos dos fármacos , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Sequência de Aminoácidos , Animais , Barreira Hematotesticular/metabolismo , Domínio Catalítico , Anticoncepcionais Masculinos/metabolismo , Anticoncepcionais Masculinos/farmacologia , Humanos , Hidrazinas/metabolismo , Hidrazinas/farmacologia , Ligação de Hidrogênio , Indazóis/metabolismo , Indazóis/farmacologia , Integrina alfa6beta1/metabolismo , Masculino , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ocludina/metabolismo , Epitélio Seminífero/efeitos dos fármacos , Epitélio Seminífero/metabolismo , Alinhamento de Sequência , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo
20.
Reproduction ; 141(5): 571-80, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21307270

RESUMO

Adjudin is a derivative of 1H-indazole-3-carboxylic acid that was shown to have potent anti-spermatogenic activity in rats, rabbits, and dogs. It exerts its effects most notably locally in the apical compartment of the seminiferous epithelium, behind the blood-testis barrier, by disrupting adhesion of germ cells, most notably spermatids to the Sertoli cells, thereby inducing release of immature spermatids from the epithelium that leads to infertility. After adjudin is metabolized, the remaining spermatogonial stem cells and spermatogonia repopulate the seminiferous epithelium gradually via spermatogonial self-renewal and differentiation, to be followed by meiosis and spermiogenesis, and thus fertility rebounds. Recent studies in rats have demonstrated unequivocally that the primary and initial cellular target of adjudin in the testis is the apical ectoplasmic specialization, a testis-specific anchoring junction type restricted to the interface between Sertoli cells and elongating spermatids (from step 8 to 19 spermatids). In this review, we highlight some of the recent advances and obstacles regarding the possible use of adjudin as a male contraceptive.


Assuntos
Anticoncepcionais Masculinos/farmacologia , Hidrazinas/farmacologia , Indazóis/farmacologia , Epitélio Seminífero/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Anticoncepcionais Masculinos/toxicidade , Humanos , Hidrazinas/toxicidade , Indazóis/toxicidade , Indenos/farmacologia , Masculino , Piperidinas/farmacologia , Células de Sertoli/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...