Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Semin Cell Dev Biol ; 81: 71-77, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-28923514

RESUMEN

In adult mammalian testes, spermatids, most notably step 17-19 spermatids in stage IV-VIII tubules, are aligned with their heads pointing toward the basement membrane and their tails toward the tubule lumen. On the other hand, these polarized spermatids also align across the plane of seminiferous epithelium, mimicking planar cell polarity (PCP) found in other hair cells in cochlea (inner ear). This orderly alignment of developing spermatids during spermiogenesis is important to support spermatogenesis, such that the maximal number of developing spermatids can be packed and supported by a fixed population of differentiated Sertoli cells in the limited space of the seminiferous epithelium in adult testes. In this review, we provide emerging evidence to demonstrate spermatid PCP in the seminiferous epithelium to support spermatogenesis. We also review findings in the field regarding the biology of spermatid cellular polarity (e.g., head-tail polarity and apico-basal polarity) and its inter-relationship to spermatid PCP. Furthermore, we also provide a hypothetical concept on the importance of PCP proteins in endocytic vesicle-mediated protein trafficking events to support spermatogenesis through protein endocytosis and recycling.


Asunto(s)
Polaridad Celular/fisiología , Transducción de Señal/fisiología , Espermátides/fisiología , Espermatogénesis/fisiología , Animales , Humanos , Masculino , Células de Sertoli/citología , Células de Sertoli/metabolismo , Espermátides/citología , Testículo/citología , Testículo/metabolismo
2.
Semin Cell Dev Biol ; 81: 21-32, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-28965865

RESUMEN

Cell polarity in the adult mammalian testis refers to the polarized alignment of developing spermatids during spermiogenesis and the polarized organization of organelles (e.g., phagosomes, endocytic vesicles, Sertoli cell nuclei, Golgi apparatus) in Sertoli cells and germ cells to support spermatogenesis. Without these distinctive features of cell polarity in the seminiferous epithelium, it is not possible to support the daily production of millions of sperm in the limited space provided by the seminiferous tubules in either rodent or human males through the adulthood. In short, cell polarity provides a novel mean to align spermatids and the supporting organelles (e.g., phagosomes, Golgi apparatus, endocytic vesicles) in a highly organized fashion spatially in the seminiferous epithelium during the epithelial cycle of spermatogenesis. This is analogous to different assembling units in a manufacturing plant such that as developing spermatids move along the "assembly line" conferred by Sertoli cells, different structural/functional components can be added to (or removed from) the developing spermatids during spermiogenesis, so that functional spermatozoa are produced at the end of the assembly line. Herein, we briefly review findings regarding the regulation of cell polarity in the testis with specific emphasis on developing spermatids, supported by an intriguing network of regulatory proteins along a local functional axis. Emerging evidence has suggested that cell cytoskeletons provide the tracks which in turn confer the unique assembly lines in the seminiferous epithelium. We also provide some thought-provoking concepts based on which functional experiments can be designed in future studies.


Asunto(s)
Polaridad Celular , Citoesqueleto/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Animales , Humanos , Masculino , Microtúbulos/metabolismo , Células de Sertoli/citología , Espermátides/citología , Espermatogénesis , Testículo/citología
3.
Semin Cell Dev Biol ; 59: 62-70, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27292315

RESUMEN

When the cross-section of a seminiferous tubule from an adult rat testes is examined microscopically, Sertoli cells and germ cells in the seminiferous epithelium are notably polarized cells. For instance, Sertoli cell nuclei are found near the basement membrane. On the other hand, tight junction (TJ), basal ectoplasmic specialization (basal ES, a testis-specific actin-rich anchoring junction), gap junction (GJ) and desmosome that constitute the blood-testis barrier (BTB) are also located near the basement membrane. The BTB, in turn, divides the epithelium into the basal and the adluminal (apical) compartments. Within the epithelium, undifferentiated spermatogonia and preleptotene spermatocytes restrictively reside in the basal compartment whereas spermatocytes and post-meiotic spermatids reside in the adluminal compartment. Furthermore, the heads of elongating/elongated spermatids point toward the basement membrane with their elongating tails toward the tubule lumen. However, the involvement of polarity proteins in this unique cellular organization, in particular the underlying molecular mechanism(s) by which polarity proteins confer cellular polarity in the seminiferous epithelium is virtually unknown until recent years. Herein, we discuss latest findings regarding the role of different polarity protein complexes or modules and how these protein complexes are working in concert to modulate Sertoli cell and spermatid polarity. These findings also illustrate polarity proteins exert their effects through the actin-based cytoskeleton mediated by actin binding and regulatory proteins, which in turn modulate adhesion protein complexes at the cell-cell interface since TJ, basal ES and GJ utilize F-actin for attachment. We also propose a hypothetical model which illustrates the antagonistic effects of these polarity proteins. This in turn provides a unique mechanism to modulate junction remodeling in the testis to support germ cell transport across the epithelium in particular the BTB during the epithelial cycle of spermatogenesis.


Asunto(s)
Polaridad Celular , Proteínas/metabolismo , Espermatogénesis , Animales , Citoesqueleto/metabolismo , Humanos , Modelos Biológicos , Unión Proteica
4.
Semin Cell Dev Biol ; 59: 141-156, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26779951

RESUMEN

Sertoli cells isolated from rodents or humans and cultured in vitro are known to establish a functional tight junction (TJ)-permeability barrier that mimics the blood-testis barrier (BTB) in vivo. This model has been widely used by investigators to study the biology of the TJ and the BTB. Studies have shown that environmental toxicants (e.g., perfluorooctanesulfonate (PFOS), bisphenol A (BPA) and cadmium) that exert their disruptive effects to induce Sertoli cell injury using this in vitro model are reproducible in studies in vivo. Thus, this in vitro system provides a convenient approach to probe the molecular mechanism(s) underlying toxicant-induced testis injury but also to provide new insights in understanding spermatogenesis, such as the biology of cell adhesion, BTB restructuring that supports preleptotene spermatocyte transport, and others. Herein, we provide a brief and critical review based on studies using this in vitro model of Sertoli cell cultures using primary cells isolated from rodent testes vs. humans to monitor environmental toxicant-mediated Sertoli cell injury. In short, recent findings have shown that environmental toxicants exert their effects on Sertoli cells to induce testis injury through their action on Sertoli cell actin- and/or microtubule-based cytoskeleton. These effects are mediated via their disruptive effects on actin- and/or microtubule-binding proteins. Sertoli cells also utilize differential spatiotemporal expression of these actin binding proteins to confer plasticity to the BTB to regulate germ cell transport across the BTB.


Asunto(s)
Contaminantes Ambientales/toxicidad , Modelos Biológicos , Células de Sertoli/patología , Animales , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Humanos , Masculino , Células de Sertoli/efectos de los fármacos , Espermatogénesis/efectos de los fármacos
5.
Am J Physiol Endocrinol Metab ; 315(5): E924-E948, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30016153

RESUMEN

In the mammalian testis, spermatogenesis is dependent on the microtubule (MT)-specific motor proteins, such as dynein 1, that serve as the engine to support germ cell and organelle transport across the seminiferous epithelium at different stages of the epithelial cycle. Yet the underlying molecular mechanism(s) that support this series of cellular events remain unknown. Herein, we used RNAi to knockdown cytoplasmic dynein 1 heavy chain (Dync1h1) and an inhibitor ciliobrevin D to inactivate dynein in Sertoli cells in vitro and the testis in vivo, thereby probing the role of dynein 1 in spermatogenesis. Both treatments were shown to extensively induce disruption of MT organization across Sertoli cells in vitro and the testis in vivo. These changes also perturbed the transport of spermatids and other organelles (such as phagosomes) across the epithelium. These changes thus led to disruption of spermatogenesis. Interestingly, the knockdown of dynein 1 or its inactivation by ciliobrevin D also perturbed gross disruption of F-actin across the Sertoli cells in vitro and the seminiferous epithelium in vivo, illustrating there are cross talks between the two cytoskeletons in the testis. In summary, these findings confirm the role of cytoplasmic dynein 1 to support the transport of spermatids and organelles across the seminiferous epithelium during the epithelial cycle of spermatogenesis.


Asunto(s)
Dineínas/metabolismo , Espermátides/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Transporte Biológico/fisiología , Dineínas/genética , Masculino , Quinazolinonas/farmacología , Interferencia de ARN , Ratas , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Espermátides/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos
6.
Am J Physiol Endocrinol Metab ; 314(2): E174-E190, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29089336

RESUMEN

The blood-testis barrier (BTB), conferred by Sertoli cells in the mammalian testis, is an important ultrastructure that supports spermatogenesis. Studies using animal models have shown that a disruption of the BTB leads to meiotic arrest, causing defects in spermatogenesis and male infertility. To better understand the regulation of BTB dynamics, we report findings herein to understand the role of ribosomal protein S6 (rpS6), a downstream signaling protein of mammalian target of rapamycin complex 1 (mTORC1), in promoting BTB disruption in the testis in vivo, making the barrier "leaky." Overexpression of wild-type rpS6 (rpS6-WT, the full-length cDNA cloned into the mammalian expression vector pCI-neo) and a constitutively active quadruple phosphomimetic mutant cloned into pCI-neo (p-rpS6-MT) vs. control (empty pCI-neo vector) was achieved by transfecting adult rat testes with the corresponding plasmid DNA using a Polyplus in vivo-jetPEI transfection reagent. On the basis of an in vivo functional BTB integrity assay, p-rpS6-MT was found to induce BTB disruption better than rpS6-WT did (and no effects in empty vector control), leading to defects in spermatogenesis, including loss of spermatid polarity and failure in the transport of cells (e.g., spermatids) and organelles (e.g., phagosomes), to be followed by germ exfoliation. More important, rpS6-WT and p-rpS6-MT exert their disruptive effects through changes in the organization of actin- and microtubule (MT)-based cytoskeletons, which are mediated by changes in the spatiotemporal expression of actin- and MT-based binding and regulatory proteins. In short, mTORC1/rpS6 signaling complex is a regulator of spermatogenesis and BTB by modulating the organization of the actin- and MT-based cytoskeletons.


Asunto(s)
Barrera Hematotesticular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Proteína S6 Ribosómica/fisiología , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Permeabilidad , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Proteína S6 Ribosómica/genética , Transducción de Señal/genética , Espermatogénesis/genética , Testículo/fisiología , Regulación hacia Arriba/genética
7.
Biochim Biophys Acta Biomembr ; 1860(1): 141-153, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28450047

RESUMEN

Signaling pathways that regulate blood-tissue barriers are important for studying the biology of various blood-tissue barriers. This information, if deciphered and better understood, will provide better therapeutic management of diseases particularly in organs that are sealed by the corresponding blood-tissue barriers from systemic circulation, such as the brain and the testis. These barriers block the access of antibiotics and/or chemotherapeutical agents across the corresponding barriers. Studies in the last decade using the blood-testis barrier (BTB) in rats have demonstrated the presence of several signaling pathways that are crucial to modulate BTB function. Herein, we critically evaluate these findings and provide hypothetical models regarding the underlying mechanisms by which these signaling molecules/pathways modulate BTB dynamics. This information should be carefully evaluated to examine their applicability in other tissue barriers which shall benefit future functional studies in the field. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Asunto(s)
Barrera Hematotesticular/metabolismo , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Modelos Cardiovasculares , Transducción de Señal/fisiología , Animales , Humanos , Masculino
8.
FASEB J ; 31(8): 3587-3607, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28487282

RESUMEN

Spermatogenesis takes place in the epithelium of the seminiferous tubules of the testes, producing millions of spermatozoa per day in an adult male in rodents and humans. Thus, multiple cellular events that are regulated by an array of signaling molecules and pathways are tightly coordinated to support spermatogenesis. Here, we report findings of a local regulatory axis between the basement membrane (BM), the blood-testis barrier (BTB), and the apical ectoplasmic specialization (apical ES; a testis-specific, actin-rich adherens junction at the Sertoli cell-spermatid interface) to coordinate cellular events across the seminiferous epithelium during the epithelial cycle. In short, a biologically active fragment, noncollagenous 1 (NC1) domain that is derived from collagen chains in the BM, was found to modulate cell junction dynamics at the BTB and apical ES. NC1 domain from the collagen α3(IV) chain was cloned into a mammalian expression vector, pCI-neo, with and without a collagen signal peptide. We also prepared a specific Ab against the purified recombinant NC1 domain peptide. These reagents were used to examine whether overexpression of NC1 domain with high transfection efficacy would perturb spermatogenesis, in particular, spermatid adhesion (i.e., inducing apical ES degeneration) and BTB function (i.e., basal ES and tight junction disruption, making the barrier leaky), in the testis in vivo We report our findings that NC1 domain derived from collagen α3(IV) chain-a major structural component of the BM-was capable of inducing BTB remodeling, making the BTB leaky in studies in vivo Furthermore, NC1 domain peptide was transported across the epithelium via a microtubule-dependent mechanism and is capable of inducing apical ES degeneration, which leads to germ cell exfoliation from the seminiferous epithelium. Of more importance, we show that NC1 domain peptide exerted its regulatory effect by disorganizing actin microfilaments and microtubules in Sertoli cells so that they failed to support cell adhesion and transport of germ cells and organelles (e.g., residual bodies, phagosomes) across the seminiferous epithelium. This local regulatory axis between the BM, BTB, and the apical ES thus coordinates cellular events that take place across the seminiferous epithelium during the epithelial cycle of spermatogenesis.-Chen, H., Mruk, D. D., Lee, W. M., Cheng, C. Y. Regulation of spermatogenesis by a local functional axis in the testis: role of the basement membrane-derived noncollagenous 1 domain peptide.


Asunto(s)
Membrana Basal/metabolismo , Colágeno Tipo IV/metabolismo , Fragmentos de Péptidos/metabolismo , Células de Sertoli/efectos de los fármacos , Espermatogénesis/fisiología , Actinas/metabolismo , Animales , Masculino , Paclitaxel , Dominios Proteicos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes , Epitelio Seminífero/metabolismo , Uniones Estrechas
9.
FASEB J ; 31(2): 584-597, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27815338

RESUMEN

Laminin α2 is one of the constituent components of the basement membrane (BM) in adult rat testes. Earlier studies that used a mouse genetic model have shown that a deletion of laminin α2 impedes male fertility by disrupting ectoplasmic specialization (ES; a testis-specific, actin-rich anchoring junction) function along the length of Sertoli cell in the testis. This includes ES at the Sertoli cell-elongating/elongated spermatid interface, which is known as apical ES and possibly the Sertoli-Sertoli cell interface, known as basal ES, at the blood-testis barrier (BTB). Studies have also illustrated that there is a local regulatory axis that functionally links cellular events of spermiation that occur near the luminal edge of tubule lumen at the apical ES and the basal ES/BTB remodeling near the BM at opposite ends of the seminiferous epithelium during the epithelial cycle, known as the apical ES-BTB-BM axis. However, the precise role of BM in this axis remains unknown. Here, we show that laminin α2 in the BM serves as the crucial regulator in this axis as laminin α2, likely its 80-kDa fragment from the C terminus, was found to be transported across the seminiferous epithelium at stages VIII-IX of the epithelial cycle, from the BM to the luminal edge of the tubule, possibly being used to modulate apical ES restructuring at these stages. Of more importance, a knockdown of laminin α2 in Sertoli cells was shown to induce the Sertoli cell tight junction permeability barrier disruption via changes in localization of adhesion proteins at the tight junction and basal ES at the Sertoli cell BTB. These changes were found to be mediated by a disruption of F-actin organization that was induced by changes in the spatiotemporal expression of actin binding/regulatory proteins. Furthermore, laminin α2 knockdown also perturbed microtubule (MT) organization by considerable down-regulation of MT polymerization via changes in the spatiotemporal expression of EB1 (end-binding protein 1), a +TIP (MT plus-end tracking protein). In short, laminin α2 in the BM seems to play a crucial role in the BTB-BM axis by modulating BTB dynamics during spermatogenesis.-Gao, Y., Mruk, D., Chen, H., Lui, W.-Y., Lee, W. M., Cheng, C. Y. Regulation of the blood-testis barrier by a local axis in the testis: role of laminin α2 in the basement membrane.


Asunto(s)
Membrana Basal/metabolismo , Barrera Hematotesticular/fisiología , Regulación de la Expresión Génica/fisiología , Laminina/metabolismo , Testículo/fisiología , Animales , Técnicas de Silenciamiento del Gen , Laminina/genética , Masculino , Paclitaxel/farmacología , Ratas , Ratas Sprague-Dawley , Espermatogénesis/fisiología , Testículo/efectos de los fármacos , Moduladores de Tubulina/farmacología
10.
FASEB J ; 30(4): 1436-52, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26678449

RESUMEN

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.


Asunto(s)
Barrera Hematotesticular/metabolismo , Conexina 43/genética , Meiosis/genética , Espermatogénesis/genética , Animales , Barrera Hematotesticular/efectos de los fármacos , Conexina 43/metabolismo , Expresión Génica/efectos de los fármacos , Hidrazinas/farmacología , Immunoblotting , Indazoles/farmacología , Masculino , Ratones Noqueados , Microscopía Fluorescente , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Espermátides/efectos de los fármacos , Espermátides/metabolismo , Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Espermatogonias/metabolismo , Testículo/citología , Testículo/efectos de los fármacos , Testículo/metabolismo
11.
PLoS Genet ; 10(6): e1004447, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24967734

RESUMEN

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.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/genética , Barrera Hematotesticular , Espermatogénesis/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Masculino , Ratones , Epitelio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Testículo/metabolismo
12.
Semin Cell Dev Biol ; 30: 75-85, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24735648

RESUMEN

The transport of germ cells across the seminiferous epithelium is composed of a series of cellular events during the epithelial cycle essential to the completion of spermatogenesis. Without the timely transport of spermatids during spermiogenesis, spermatozoa that are transformed from step 19 spermatids in the rat testis fail to reach the luminal edge of the apical compartment and enter the tubule lumen at spermiation, thereby arriving the epididymis for further maturation. Step 19 spermatids and/or sperms that remain in the epithelium beyond stage VIII of the epithelial cycle will be removed by the Sertoli cell via phagocytosis to form phagosomes and be degraded by lysosomes, leading to subfertility and/or infertility. However, the biology of spermatid transport, in particular the final events that lead to spermiation remain elusive. Based on recent data in the field, we critically evaluate the biology of spermiation herein by focusing on the actin binding proteins (ABPs) that regulate the organization of actin microfilaments at the Sertoli-spermatid interface, which is crucial for spermatid transport during this event. The hypothesis we put forth herein also highlights some specific areas of research that can be pursued by investigators in the years to come.


Asunto(s)
Proteínas de Microfilamentos/fisiología , Espermátides/fisiología , Actinas/fisiología , Animales , Humanos , Masculino , Multimerización de Proteína , Transducción de Señal , Transporte Espermático
13.
FASEB J ; 29(9): 3788-805, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26048141

RESUMEN

Ectoplasmic specialization (ES) is an actin-rich adherens junction in the seminiferous epithelium of adult mammalian testes. ES is restricted to the Sertoli-spermatid (apical ES) interface, as well as the Sertoli cell-cell (basal ES) interface at the blood-testis barrier (BTB). ES is typified by the presence of an array of bundles of actin microfilaments near the Sertoli cell plasma membrane. These actin microfilament bundles require rapid debundling to convert them from a bundled to branched/unbundled configuration and vice versa to confer plasticity to support the transport of 1) spermatids in the adluminal compartment and 2) preleptotene spermatocytes at the BTB while maintaining cell adhesion. Plastin 3 is one of the plastin family members abundantly found in yeast, plant and animal cells that confers actin microfilaments their bundled configuration. Herein, plastin 3 was shown to be a component of the apical and basal ES in the rat testis, displaying spatiotemporal expression during the epithelial cycle. A knockdown (KD) of plastin 3 in Sertoli cells by RNA interference using an in vitro model to study BTB function showed that a transient loss of plastin 3 perturbed the Sertoli cell tight junction-permeability barrier, mediated by changes in the localization of basal ES proteins N-cadherin and ß-catenin. More importantly, these changes were the result of an alteration of the actin microfilaments, converting from their bundled to branched configuration when examined microscopically, and validated by biochemical assays that quantified actin-bundling and polymerization activity. Moreover, these changes were confirmed by studies in vivo by plastin 3 KD in the testis in which mis-localization of N-cadherin and ß-catenin was also detected at the BTB, concomitant with defects in the transport of spermatids and phagosomes and a disruption of cell adhesion most notably in elongated spermatids due to a loss of actin-bundling capability at the apical ES, which in turn affected localization of adhesion protein complexes at the site. In summary, plastin 3 is a regulator of actin microfilament bundles at the ES in which it dictates the configuration of the filamentous actin network by assuming either a bundled or unbundled/branched configuration via changes in its spatiotemporal expression during the epithelial cycle.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/fisiología , Uniones Estrechas/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Cadherinas/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Sertoli/citología , Espermátides/citología , Espermatocitos/citología , beta Catenina/metabolismo
14.
Immunol Cell Biol ; 93(3): 311-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25403570

RESUMEN

The mammalian testis is an immunoprivileged organ where male germ cell autoantigens are immunologically ignored. Both systemic immune tolerance to autoantigens and local immunosuppressive milieu contribute to the testicular immune privilege. Testicular immunosuppression has been intensively studied, but information on systemic immune tolerance to autoantigens is lacking. In the present study, we aimed to determine the role of Axl and Mer receptor tyrosine kinases in maintaining the systemic tolerance to male germ cell antigens using the experimental autoimmune orchitis (EAO) model. Axl and Mer double-knockout (Axl(-/-)Mer(-/-)) mice developed evident EAO after a single immunization with germ cell homogenates emulsified with complete Freund's adjuvant. EAO was characterized by the accumulation of macrophages and T lymphocytes in the testis. Damage to the seminiferous epithelium was also observed. EAO induction was associated with pro-inflammatory cytokine upregulation in the testes, impaired permeability of the blood-testis barrier and generation of autoantibodies against germ cell antigens in Axl(-/-)Mer(-/-) mice. Immunization also induced mild EAO in Axl or Mer single-gene-knockout mice. By contrast, a single immunization failed to induce EAO in wild-type mice. The results indicate that Axl and Mer receptors cooperatively regulate the systemic immune tolerance to male germ cell antigens.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Macrófagos/inmunología , Orquitis/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Linfocitos T/inmunología , Testículo/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/genética , Barrera Hematotesticular/inmunología , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orquitis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
15.
Biol Reprod ; 92(3): 63, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25588509

RESUMEN

The mammalian testis is an immunoprivileged site where male germ cell antigens are immunologically tolerated under physiological conditions. However, some pathological conditions can disrupt the immunoprivileged status and induce autoimmune orchitis, an etiological factor of male infertility. Mechanisms underlying autoimmune orchitis induction are largely unknown. The present study investigated the roles of Toll-like receptor 2 (TLR2) and TLR4 in mediating the induction of experimental autoimmune orchitis (EAO) in mice after immunization with male germ cell antigens emulsified with complete Freund adjuvant. Wild-type mice developed severe EAO after three immunizations, which was characterized by leukocyte infiltration, autoantibody production, and impaired spermatogenesis. Tlr2 or Tlr4 deficient mice showed relatively low susceptibility to EAO induction compared with wild-type mice. Notably, Tlr2 and Tlr4 double knockout mice were almost completely protected from EAO induction. Moreover, we demonstrated that TLR2 was crucial in mediating autoantibody production in response to immunization. The results imply that TLR2 and TLR4 cooperatively mediate EAO induction.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Orquitis/inmunología , Orquitis/fisiopatología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Animales , Autoanticuerpos/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund , Células Germinativas/inmunología , Inmunización , Infertilidad Masculina/etiología , Infertilidad Masculina/inmunología , Infertilidad Masculina/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orquitis/complicaciones , Testículo/inmunología , Testículo/patología , Testículo/fisiopatología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
16.
Proc Natl Acad Sci U S A ; 109(31): 12562-7, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22797892

RESUMEN

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.


Asunto(s)
Barrera Hematotesticular/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Células de Sertoli/metabolismo , Uniones Estrechas/enzimología , Actinas/genética , Actinas/metabolismo , Sustitución de Aminoácidos , Animales , Barrera Hematotesticular/citología , Citoesqueleto/genética , Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación Missense , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/genética , Multimerización de Proteína/fisiología , Ratas , Células de Sertoli/citología , Espermatocitos/citología , Espermatocitos/enzimología , Espermatogénesis/fisiología , Uniones Estrechas/genética , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteína de la Zonula Occludens-1
17.
Am J Physiol Endocrinol Metab ; 307(7): E553-62, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25117412

RESUMEN

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. However, it undergoes cyclic restructuring during the epithelial cycle of spermatogenesis in which the "old" BTB located above the preleptotene spermatocytes being transported across the immunological barrier is "disassembled," whereas the "new" BTB found behind these germ cells is rapidly "reassembled," i.e., mediated by endocytic vesicle-mediated protein trafficking events. Thus, the immunological barrier is maintained when preleptotene spermatocytes connected in clones via intercellular bridges are transported across the BTB. Yet the underlying mechanism(s) in particular the involving regulatory molecules that coordinate these events remains unknown. We hypothesized that c-Src and c-Yes might work in contrasting roles in endocytic vesicle-mediated trafficking, serving as molecular switches, to effectively disassemble and reassemble the old and the new BTB, respectively, to facilitate preleptotene spermatocyte transport across the BTB. Following siRNA-mediated specific knockdown of c-Src or c-Yes in Sertoli cells, we utilized biochemical assays to assess the changes in protein endocytosis, recycling, degradation and phagocytosis. c-Yes was found to promote endocytosed integral membrane BTB proteins to the pathway of transcytosis and recycling so that internalized proteins could be effectively used to assemble new BTB from the disassembling old BTB, whereas c-Src promotes endocytosed Sertoli cell BTB proteins to endosome-mediated protein degradation for the degeneration of the old BTB. By using fluorescence beads mimicking apoptotic germ cells, Sertoli cells were found to engulf beads via c-Src-mediated phagocytosis. A hypothetical model that serves as the framework for future investigation is thus proposed.


Asunto(s)
Barrera Hematotesticular/metabolismo , Proteínas Proto-Oncogénicas c-yes/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Células de Sertoli/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Endocitosis/fisiología , Técnicas de Silenciamiento del Gen , Genes src/genética , Masculino , Proteínas de la Membrana/metabolismo , Fagocitosis/fisiología , Proteínas Proto-Oncogénicas c-yes/genética , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley
18.
Biol Reprod ; 91(1): 8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24876405

RESUMEN

The mammalian testis is an immunoprivileged organ where local tissue-specific cells acquire an effective innate immune function against invading microbial pathogens. The present study demonstrated that mouse Leydig cells had innate antiviral activities in response to viral DNA challenge through p204 activation. The DNA sensor p204 and its signaling adaptor stimulator of interferon (IFN) genes (STING) were constitutively expressed in Leydig cells. Synthetic herpes simplex virus DNA analog (HSV60), a p204 agonist, induced the expression of type I IFNs and various antiviral proteins, including IFN-stimulating gene 15, 2'5'-oligoadenylate synthetase, and Mx glutamyl transpeptidase 1, in Leydig cells. The HSV60-induced innate antiviral response in Leydig cells was significantly reduced by inhibiting p204 signaling using specific small interfering RNAs targeting p204 and Sting. The antiviral response did not affect steroidogenesis in Leydig cells. These results indicated a novel mechanism underlying the testicular innate antiviral response.


Asunto(s)
Inmunidad Innata , Células Intersticiales del Testículo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas Nucleares/genética , Fosfoproteínas/genética , ARN Interferente Pequeño , gamma-Glutamiltransferasa/genética , gamma-Glutamiltransferasa/metabolismo
19.
Hum Reprod ; 29(6): 1279-91, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24532171

RESUMEN

STUDY QUESTION: Can human Sertoli cells cultured in vitro and that have formed an epithelium be used as a model to monitor toxicant-induced junction disruption and to better understand the mechanism(s) by which toxicants disrupt cell adhesion at the Sertoli cell blood-testis barrier (BTB)? SUMMARY ANSWER: Our findings illustrate that human Sertoli cells cultured in vitro serve as a reliable system to monitor the impact of environmental toxicants on the BTB function. WHAT IS KNOWN ALREADY: Suspicions of a declining trend in semen quality and a concomitant increase in exposures to environmental toxicants over the past decades reveal the need of an in vitro system that efficiently and reliably monitors the impact of toxicants on male reproductive function. Furthermore, studies in rodents have confirmed that environmental toxicants impede Sertoli cell BTB function in vitro and in vivo. STUDY DESIGN, SIZE AND DURATION: We examined the effects of two environmental toxicants: cadmium chloride (0.5-20 µM) and bisphenol A (0.4-200 µM) on human Sertoli cell function. Cultured Sertoli cells from three men were used in this study, which spanned an 18-month period. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human Sertoli cells from three subjects were cultured in F12/DMEM containing 5% fetal bovine serum. Changes in protein expression were monitored by immunoblotting using specific antibodies. Immunofluorescence analyses were used to assess changes in the distribution of adhesion proteins, F-actin and actin regulatory proteins following exposure to two toxicants: cadmium chloride and bisphenol A (BPA). MAIN RESULTS AND THE ROLE OF CHANCE: Human Sertoli cells were sensitive to cadmium and BPA toxicity. Changes in the localization of cell adhesion proteins were mediated by an alteration of the actin-based cytoskeleton. This alteration of F-actin network in Sertoli cells as manifested by truncation and depolymerization of actin microfilaments at the Sertoli cell BTB was caused by mislocalization of actin filament barbed end capping and bundling protein Eps8, and branched actin polymerization protein Arp3. Besides impeding actin dynamics, endocytic vesicle-mediated trafficking and the proper localization of actin regulatory proteins c-Src and annexin II in Sertoli cells were also affected. Results of statistical analysis demonstrate that these findings were not obtained by chance. LIMITATIONS, REASONS FOR CAUTION: (i) This study was done in vitro and might not extrapolate to the in vivo state, (ii) conclusions are based on the use of Sertoli cell samples from three men and (iii) it is uncertain if the concentrations of toxicants used in the experiments are reached in vivo. WIDER IMPLICATIONS OF THE FINDINGS: Human Sertoli cells cultured in vitro provide a robust model to monitor environmental toxicant-mediated disruption of Sertoli cell BTB function and to study the mechanism(s) of toxicant-induced testicular dysfunction.


Asunto(s)
Actinas/metabolismo , Compuestos de Bencidrilo/farmacología , Cloruro de Cadmio/farmacología , Adhesión Celular/efectos de los fármacos , Contaminantes Ambientales/farmacología , Fenoles/farmacología , Células de Sertoli/efectos de los fármacos , Células Cultivadas , Humanos , Masculino , Células de Sertoli/metabolismo
20.
FASEB J ; 27(2): 464-77, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23073828

RESUMEN

Development of spermatozoa in adult mammalian testis during spermatogenesis involves extensive cell migration and differentiation. Spermatogonia that reside at the basal compartment of the seminiferous epithelium differentiate into more advanced germ cell types that migrate toward the apical compartment until elongated spermatids are released into the tubule lumen during spermiation. Apical ectoplasmic specialization (ES; a testis-specific anchoring junction) is the only cell junction that anchors and maintains the polarity of elongating/elongated spermatids (step 8-19 spermatids) in the epithelium. Little is known regarding the signaling pathways that trigger the disassembly of the apical ES at spermiation. Here, we show that secreted Frizzled-related protein 1 (sFRP1), a putative tumor suppressor gene that is frequently down-regulated in multiple carcinomas, is a crucial regulatory protein for spermiation. The expression of sFRP1 is tightly regulated in adult rat testis to control spermatid adhesion and sperm release at spermiation. Down-regulation of sFRP1 during testicular development was found to coincide with the onset of the first wave of spermiation at approximately age 45 d postpartum, implying that sFRP1 might be correlated with elongated spermatid adhesion conferred by the apical ES before spermiation. Indeed, administration of sFRP1 recombinant protein to the testis in vivo delayed spermiation, which was accompanied by down-regulation of phosphorylated (p)-focal adhesion kinase (FAK)-Tyr(397) and retention of nectin-3 adhesion protein at the apical ES. To further investigate the functional relationship between p-FAK-Tyr(397) and localization of nectin-3, we overexpressed sFRP1 using lentiviral vectors in the Sertoli-germ cell coculture system. Consistent with the in vivo findings, overexpression of sFRP1 induced down-regulation of p-FAK-Tyr(397), leading to a decline in phosphorylation of nectin-3. In summary, this report highlights the critical role of sFRP1 in regulating spermiation via its effects on the FAK signaling and retention of nectin-3 adhesion complex at the apical ES.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Espermátides/metabolismo , Testículo/metabolismo , Animales , Secuencia de Bases , Adhesión Celular/fisiología , Técnicas de Cocultivo , ADN Complementario/genética , Quinasa 1 de Adhesión Focal/química , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas de la Membrana/genética , Microscopía Electrónica de Transmisión , Complejos Multiproteicos/metabolismo , Nectinas , Fosforilación , Ratas , Ratas Sprague-Dawley , Células de Sertoli/citología , Células de Sertoli/metabolismo , Transducción de Señal , Espermátides/citología , Espermatogénesis/fisiología , Testículo/citología , Testículo/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda