Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Int J Mol Sci ; 24(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36674801

RESUMEN

Canonical coxsackievirus and adenovirus receptor (CXADR) is a transmembrane component of cell junctions that is crucial for cardiac and testicular functions via its homophilic and heterophilic interaction. CXADR is expressed in both Sertoli cells and germ cells and is localized mainly at the interface between Sertoli-Sertoli cells and Sertoli-germ cells. Knockout of CXADR in mouse Sertoli cells specifically impairs male reproductive functions, including a compromised blood-testis barrier, apoptosis of germ cells, and premature loss of spermatids. Apart from serving as an important component for cell junctions, recent progress has showed the potential roles of CXADR as a signaling mediator in spermatogenesis. This review summarizes current research progress related to the regulation and role of CXADR in spermatogenesis as well as in pathological conditions. We hope this review provides some future directions and a blueprint to promote the further study on the roles of CXADR.


Asunto(s)
Receptores Virales , Espermatogénesis , Animales , Masculino , Ratones , Infecciones por Coxsackievirus , Enterovirus , Ratones Noqueados , Receptores Virales/metabolismo , Células de Sertoli/fisiología , Espermátides , Testículo
2.
Adv Exp Med Biol ; 1288: 95-109, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34453733

RESUMEN

Coxsackievirus and adenovirus receptor (CXADR) belongs to immunoglobulin superfamily of cell adhesion molecules. It expresses in most tissues, but displays unique and indispensable functions in some tissues such as heart and testis. CXADR is a multifunctional protein that can serve as a viral receptor, a junction structural protein and a signalling molecule. Thus, it exerts a wide range of functions such as facilitating leukocyte transmigration, regulating barrier function and cell adhesion, promoting EMT transition, and mediating spermatogenesis. This review aims to provide an overview and highlights some recent findings on CXADR in the field with emphasis on studies in the testis, upon which future studies can be designed to delineate the roles and regulation of CXADR in spermatogenesis.


Asunto(s)
Receptores Virales , Espermatogénesis , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Humanos , Masculino , Receptores Virales/genética , Transducción de Señal
3.
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
4.
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
5.
FASEB J ; 33(6): 7588-7602, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30892947

RESUMEN

Blood-testis barrier (BTB) and apical ectoplasmic specialization (ES) serve as structural supports for germ cell (GC) development. We demonstrated that the Sertoli cell (SC)-specific coxsackievirus and adenovirus receptor (CXADR) knockout (SC-CXADR-/-), but not the GC-specific knockout, impaired spermatogenesis. An increase in GC apoptosis and premature loss of elongated spermatids were observed in SC-CXADR-/- testes. The BTB function was compromised in SC-CXADR-/- testes with dysregulation of oocludin and zonula occludens-1 expression at the basal compartment of the seminiferous epithelium. An integrated omics analyses confirmed that altered gene ontology terms identified in SC-CXADR-/- testes are highly associated with spermatid development and differentiation, spermatogenesis, and sperm motility and are considered as unique testicular function terms. Leptin, Nasp, Tektin3, Larp 7, and acrosin, which are highly associated with male fertility, were found to be down-regulated in SC-CXADR-/- testes. Based on the data from the omics analyses, we employed the CXADR-deficient SC model to further investigate the molecular mechanisms involved. We unraveled that SC-CXADRs are required for ß-catenin inactivation and cell division cycle protein 42 (Cdc42) activation, resulting in maintaining the integrity and function of the BTB and apical ES as well as inhibiting gene transcription, such as the Myc gene, in the testes. We demonstrated for the first time that CXADR is an important mediator governing ß-catenin and Cdc42 signaling that is essential for spermatogenesis. The molecular mechanisms identified herein may provide new insights to unravel the novel functions and signaling cascades of CXADR in other key CXADR-expressing tissues.-Huang, K., Ru, B., Zhang, Y., Chan, W.-L., Chow, S.-C., Zhang, J., Lo, C., Lui, W.-Y. Sertoli cell-specific coxsackievirus and adenovirus receptor knockout regulates cell adhesion and gene transcription via ß-catenin inactivation and Cdc42 activation.


Asunto(s)
Adenoviridae/metabolismo , Adhesión Celular/fisiología , Enterovirus/metabolismo , Receptores Virales/fisiología , Transcripción Genética/fisiología , beta Catenina/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/metabolismo , Animales , Barrera Hematotesticular/metabolismo , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Proteómica , Receptores Virales/genética , Epitelio Seminífero/citología , Transducción de Señal , Transcriptoma
6.
Semin Cell Dev Biol ; 59: 54-61, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26851625

RESUMEN

Nectins and nectin-like molecules (Necls) belong to Ca2+-independent immunoglobulin superfamily of cell adhesion molecules and they are ubiquitously expressed in most tissues. They not only interact in trans-homo and trans-hetero with others, but also interact in cis with growth factor receptors and integrins. Owning to these properties, nectins and Necls exert their role far beyond cell-cell adhesion. Nectins and Necls are crucial for various cellular processes not limited to cell movement, cell proliferation and polarization. They are involved in organogenesis and developmental processes such as spermatogenesis. This review aims to provide an overview and highlight some recent findings on nectins and Necls in the field with emphasis on studies in the testis, upon which future studies can be designed to delineate the role and regulation of nectins and Necls in spermatogenesis.


Asunto(s)
Nectinas/metabolismo , Espermatogénesis , Animales , Fenómenos Fisiológicos Celulares , Humanos , Modelos Biológicos , Nectinas/química
7.
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
8.
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
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.
Biochim Biophys Acta ; 1849(6): 601-11, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25817991

RESUMEN

Junctional adhesion molecule-B (JAM-B) is found between Sertoli cells at the blood-testis barrier (BTB) as well as between Sertoli and germ cells at the apical ectoplasmic specializations (ES) in the testis. The expression of JAM-B is tightly regulated to modulate the passage of spermatocytes across the BTB as well as the release of mature spermatozoa from the seminiferous epithelium. Transforming growth factor beta (TGF-ß) family is implicated in the regulation of testicular cell junction dynamics during spermatogenesis. This study aims to investigate the effects of TGF-ß3 on the expression of JAM-B as well as the underlying mechanisms on how TGF-ß3 regulates JAM-B expression to facilitate the disassembly of the BTB and apical ES. Our results revealed that TGF-ß3 suppresses JAM-B at post-transcriptional and post-translational levels. Inhibitor, siRNA knockdown and co-immunoprecipitation have shown that TGF-ß3 induces JAM-B protein degradation via ubiquitin-proteasome pathway. Immunofluorescence staining further confirmed that blockage of ubiquitin-proteasome pathway could abrogate TGF-ß3-induced loss of JAM-B at the cell-cell interface. siRNA knockdown and immunofluorescence staining also demonstrated that activation of Smad signaling is required for TGF-ß3-induced JAM-B protein degradation. In addition, TGF-ß3 reduces JAM-B mRNA levels, at least in part, via post-transcriptional regulation. mRNA stability assay has confirmed that TGF-ß3 promotes the degradation of JAM-B transcript and TGF-ß3-mediated mRNA destabilization requires the activation of ERK1/2 and p54 JNK signal cascades. Taken together, TGF-ß3 significantly downregulates JAM-B expression via post-transcriptional and post-translational modulation and results in the disruption of BTB and apical ES.


Asunto(s)
Barrera Hematotesticular , Uniones Intercelulares/genética , Molécula B de Adhesión de Unión/genética , Espermatogénesis/genética , Factor de Crecimiento Transformador beta3/biosíntesis , Endocitosis/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Uniones Intercelulares/metabolismo , Molécula B de Adhesión de Unión/biosíntesis , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteolisis , Estabilidad del ARN/genética , Células de Sertoli/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factor de Crecimiento Transformador beta3/genética
11.
Biochim Biophys Acta ; 1839(9): 873-84, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25046863

RESUMEN

Nectin-2, a junction molecule, is found at the basal and apical ectoplasmic specializations (ES) for the formation of the blood-testis barrier (BTB) (constituted by tight junctions and basal ES) and Sertoli-spermatid adhesion. Loss of nectin-2 causes male infertility, suggesting nectin-2-based ES is crucial for spermatogenesis. Cadmium (Cd) has been known to induce severe testicular injury. Recent evidence has shown that the basal ES at the BTB and apical ES are the targets of Cd, suggesting that unique junction protein at the ES may explain why testis is more susceptible than other tissues. Since nectin-2 is expressed exclusively at the ES, it is highly possible that nectin-2 is the direct target of Cd. In this study, we investigate if nectin-2 is the target protein of Cd toxicity and the mechanism on how Cd down-regulates nectin-2 to achieve ES disruption. Our results revealed that Cd suppresses nectin-2 at transcriptional and post-translational levels. Inhibitor and shRNA knockdown have shown that Cd induces nectin-2 protein degradation via clathrin-dependent endocytosis. Immunofluorescence staining and endocytosis assays further confirmed that nectin-2 internalization is promoted upon Cd treatment. Besides, Cd directly represses nectin-2 transcription. EMSA and ChIP assays showed that Cd inhibits the binding of positive regulators to nectin-2 promoter. siRNA and overexpression analyses have demonstrated that Cd reduces the expression and binding affinity of positive regulators for transcription. Taken together, nectin-2 is the direct molecular target of Cd and its disruptive effects are mediated via direct repressing nectin-2 transcription and endocytosis of nectin-2 for degradation.


Asunto(s)
Cadmio/toxicidad , Moléculas de Adhesión Celular/genética , Infertilidad Masculina/inducido químicamente , Testículo/metabolismo , Animales , Células Cultivadas , Clatrina/fisiología , Endocitosis , Masculino , Ratones , Nectinas , Procesamiento Proteico-Postraduccional , Testículo/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
12.
Biol Reprod ; 90(3): 59, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24478392

RESUMEN

Coxsackievirus and adenovirus receptor (CAR) is a junction molecule that expresses on Sertoli and germ cells. It mediates Sertoli-germ cell adhesion and facilitates migration of preleptotene/leptotene spermatocytes across the blood-testis barrier, suggesting that CAR-based cell adhesion and migration are crucial for spermatogenesis. Interferon-gamma (IFNG) and tumor necrosis factor alpha (TNF) are two major cytokines that are elevated during testicular inflammation and cause reduced fertility. We investigated the mechanism by which IFNG and TNF exert their disruptive effects on testicular cell adhesion. We have demonstrated that combined treatment with IFNG and TNF (IFNG+TNF) exerts a synergistic effect by downregulating CAR mRNA and protein levels. Immunofluorescence staining revealed that IFNG+TNF treatment effectively removes CAR from the site of cell-cell contact. Using inhibitor and co-immunoprecipitation, we confirmed that IFNG+TNF mediates CAR protein degradation via ubiquitin-proteasome and NFKB pathways. Blockage of ubiquitin-proteasome pathway significantly inhibits CAR degradation, as indicated by the reappearance of CAR at the site of cell-cell contact. Additionally, IFNG+TNF reduces CAR mRNA via transcriptional regulation. Mutational studies have shown that IFNG+TNF-induced CAR repression is achieved by suppression of the basal transcription. Electrophoretic mobility shift assay and chromatin immunoprecipitation assays further confirmed that IFNG+TNF treament not only inhibits binding of the basal transcription factors but also promotes binding of NFKB subunits and Sp1 (negative regulators) to the CAR promoter region. Taken together, IFNG+TNF treatment significantly downregulates CAR expression, which provides an explanation of how cell sloughing in the epithelium mediates, by loss of CAR-based cell adhesion, during testicular inflammation.


Asunto(s)
Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/efectos de los fármacos , Inflamación/patología , Interferón gamma/farmacología , Enfermedades Testiculares/patología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Ensayo de Cambio de Movilidad Electroforética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Complejo de la Endopetidasa Proteasomal , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Túbulos Seminíferos/citología , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/fisiología , Testículo/patología , Transfección , Ubiquitina/genética
13.
Proc Natl Acad Sci U S A ; 108(49): 19623-8, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22106313

RESUMEN

The blood-testis barrier (BTB), one of the tightest blood-tissue barriers in the mammalian body, creates an immune-privileged site for postmeiotic spermatid development to avoid the production of antibodies against spermatid-specific antigens, many of which express transiently during spermiogenesis and spermiation. However, the BTB undergoes extensive restructuring at stage VIII of the epithelial cycle to facilitate the transit of preleptotene spermatocytes and to prepare for meiosis. This action thus prompted us to investigate whether this stage can be a physiological window for the delivery of therapeutic and/or contraceptive drugs across the BTB to exert their effects at the immune-privileged site. Herein, we report findings that P-glycoprotein, an ATP-dependent efflux drug transporter and an integrated component of the occludin/zonula occludens 1 (ZO-1) adhesion complex at the BTB, structurally interacted with focal adhesion kinase (FAK), creating the occludin/ZO-1/FAK/P-glycoprotein regulatory complex. Interestingly, a knockdown of P-glycoprotein by RNAi was found to impede Sertoli cell BTB function, making the tight junction (TJ) barrier "leaky." This effect was mediated by changes in the protein phosphorylation status of occludin via the action of FAK, thereby affecting the endocytic vesicle-mediated protein trafficking events that destabilized the TJ barrier. However, the silencing of P-glycoprotein, although capable of impeding drug transport across the BTB and TJ permeability barrier function, was not able to induce the BTB to be "freely" permeable to adjudin. These findings indicate that P-glycoprotein is involved in BTB restructuring during spermatogenesis but that P-glycoprotein-mediated restructuring does not "open up" the BTB to make it freely permeable to drugs.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Barrera Hematotesticular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Barrera Hematotesticular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Hidrazinas/farmacología , Immunoblotting , Indazoles/farmacología , Cinética , Masculino , Microscopía Confocal , Ocludina , Cultivo Primario de Células , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Proteína de la Zonula Occludens-1
14.
Mar Pollut Bull ; 203: 116422, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38749155

RESUMEN

The COVID-19 pandemic has resulted in unprecedented plastic pollution from single-used personal protective equipment (PPE), especially face masks, in coastal and marine environments. The secondary pollutants, microplastics from face masks (mask MP), rise concern about their detrimental effects on marine organisms, terrestrial organisms and even human. Using a mouse model, oral exposure to mask MP at two doses, 0.1 and 1 mg MP/day for 21 days, caused no change in animal locomotion, total weight, or sperm counts, but caused damage to sperm motility with increased curvilinear velocity (VCL). The high-dose mask MP exposure caused a significant decrease in linearity (LIN) of sperm motility. Further testicular transcriptomic analysis revealed perturbed pathways related to spermatogenesis, oxidative stress, inflammation, metabolism and energy production. Collectively, our findings substantiate that microplastics from face masks yield adverse effects on mammalian reproductive capacity, highlighting the need for improved plastic waste management and development of environmentally friendly materials.


Asunto(s)
Máscaras , Microplásticos , Motilidad Espermática , Animales , Masculino , Microplásticos/toxicidad , Ratones , Motilidad Espermática/efectos de los fármacos , COVID-19 , Testículo/efectos de los fármacos
15.
Toxicology ; 506: 153834, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38763425

RESUMEN

INTRODUCTION: Growing concerns regarding the reproductive toxicity associated with daily life exposure to micro-/nano-plastics (abbreviated as MNPs) have become increasingly prevalent. In reality, MNPs exposure involves a heterogeneous mixture of MNPs of different sizes rather than a single size. METHODS: In this study, an oral exposure mouse model was used to evaluate the effects of MNPs of four size ranges: 25-30 nm, 1-5 µm, 20-27 µm, and 125-150 µm. Adult male C57BL/6 J mice were administered environmentally relevant concentrations of 0.1 mg MNPs/day for 21 days. After that, open field test and computer assisted sperm assessment (CASA) were conducted. Immunohistochemical analyses of organ and cell type localization of MNPs were evaluated. Testicular transcriptome analysis was carried out to understand the molecular mechanisms. RESULTS: Our result showed that MNPs of different size ranges all impaired sperm motility, with a decrease in progressive sperm motility, linearity and straight-line velocity of sperm movement. Alterations did not manifest in animal locomotion, body weight, or sperm count. Noteworthy effects were most pronounced in the smaller MNPs size ranges (25-30 nm and 1-5 µm). Linear regression analysis substantiated a negative correlation between the size of MNPs and sperm curvilinear activity. Immunohistochemical analysis unveiled the intrusions of 1-5 µm MNPs, but not 20-27 µm and 125-150 µm MNPs, into Leydig cells and testicular macrophages. Further testicular transcriptomic analysis revealed perturbations in pathways related to spermatogenesis, oxidative stress, and inflammation. Particularly within the 1-5 µm MNPs group, a heightened perturbation in pathways linked to spermatogenesis and oxidative stress was observed. CONCLUSIONS: Our data support the size-dependent impairment of MNPs on sperm functionality, underscoring the pressing need for apprehensions about and interventions against the escalation of environmental micro-/nano-plastics contamination. This urgency is especially pertinent to small-sized MNPs.

16.
Toxicology ; 484: 153413, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36581016

RESUMEN

A great variety of endocrine-disrupting chemicals (EDCs) have been used extensively and become widespread in the environment nowadays. Limited mammalian studies have shown that certain EDCs may target chromosome and epigenome of the germline, leading to adverse effects in subsequent generations, despite these progenies having never been exposed to the EDC before. However, the underlying mechanisms of chromosomal changes induced by these pollutants remain poorly known. Using the human ovarian granulosa tumor cell line COV434 as a model, we investigated and compared the transcriptomic changes induced by nine EDCs with diverse chemical structures (i.e. BDE-47, BPA, BP-3, DEHP, DHP, EE2, TCS, TDCPP and NP), to inquire if there is any common epigenetic modification associated with reproductive functions induced by these EDCs. Our results showed that COV434 cells were more responsive to BP-3, NP, DEHP and EE2, and more importantly, these four EDCs altered the expression of gene clusters related to DNA damage response, cell cycle, proliferation, and chromatin remodeling, which can potentially lead to epigenetic modifications and transgenerational inheritance. Furthermore, dysregulation of similar gene clusters was common in DEHP and NP treatments. Bioinformatics analysis further revealed that BP-3 disturbed signaling pathways associated with reproductive functions, whereas alterations in telomere-related pathways were highlighted upon EE2 exposure. Overall, this study highlighted chromatin modifications caused by a class of chemicals which that may potentially lead to epigenetic changes and transgenerational reproductive impairments.


Asunto(s)
Dietilhexil Ftalato , Disruptores Endocrinos , Contaminantes Ambientales , Animales , Humanos , Transcriptoma , Epigénesis Genética , Disruptores Endocrinos/toxicidad , Cromatina , Mamíferos/genética
17.
Adv Exp Med Biol ; 763: 281-94, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23397630

RESUMEN

Spermatogenesis involves precise co-ordination of multiple cellular events that take place in the seminiferous epithelium composed of Sertoli cells and developing germ cells during the seminiferous epithelial cycle. Given the cyclic and co-ordinated nature of spermatogenesis, temporal and spatial expression of certain genes pertinent to a specific cellular event are essential. As such, transcriptional regulation is one of the major regulatory machineries in controlling the cell type- and stage-specific gene expression, some of which are under the influence of gonadotropins (e.g., FSH and LH) and sex steroids (e.g., testosterone and estradiol-17beta). Recent findings regarding transcriptional control of spermatogenesis, most notably target genes at the Sertoli-Sertoli and Sertoli-spermatid interface at the site of the blood-testis barrier (BTB) and apical ectoplasmic specialization (apical ES), respectively, involving in cell adhesion are reviewed and discussed herein. This is a much neglected area of research and a concerted effort by investigators is needed to understand transcriptional regulation of cell adhesion function in the testis particularly at the BTB during spermatogenesis.


Asunto(s)
Barrera Hematotesticular/metabolismo , Adhesión Celular , Espermatogénesis , Testículo/metabolismo , Transcripción Genética , Animales , Barrera Hematotesticular/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Masculino , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Epitelio Seminífero/citología , Epitelio Seminífero/metabolismo , Células de Sertoli/citología , Células de Sertoli/metabolismo , Nicho de Células Madre , Testículo/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Adv Exp Med Biol ; 763: 149-70, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23397623

RESUMEN

The Scribble polarity complex or module is one of the three polarity modules that regulate cell polarity in multiple epithelia including blood-tissue barriers. This protein complex is composed of Scribble, Lethal giant larvae (Lgl) and Discs large (Dlg), which are well conserved across species from fruitflies and worms to mammals. Originally identified in Drosophila and C. elegans where the Scribble complex was found to work with the Par-based and Crumbs-based polarity modules to regulate apicobasal polarity and asymmetry in cells and tissues during embryogenesis, their mammalian homologs have all been identified in recent years. Components of the Scribble complex are known to regulate multiple cellular functions besides cell polarity, which include cell proliferation, assembly and maintenance of adherens junction (AJ) and tight junction (TJ), and they are also tumor suppressors. Herein, we provide an update on the Scribble polarity complex and how this protein complex modulates cell adhesion with some emphasis on its role in Sertoli cell blood-testis barrier (BTB) function. It should be noted that this is a rapidly developing field, in particular the role of this protein module in blood-tissue barriers, and this short chapter attempts to provide the information necessary for investigators studying reproductive biology and blood-tissue barriers to design future studies. We also include results of recent studies from flies and worms since this information will be helpful in planning experiments for future functional studies in the testis to understand how Scribble-based proteins regulate BTB dynamics and spermatogenesis.


Asunto(s)
Barrera Hematotesticular/metabolismo , Polaridad Celular , Proteínas de Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Barrera Hematotesticular/citología , Adhesión Celular , Proliferación Celular , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Mucosa Intestinal/citología , Mamíferos/metabolismo , Proteínas de la Membrana/genética , Complejos Multiproteicos/genética , Mutación , Estructura Terciaria de Proteína , Transporte de Proteínas , Transducción de Señal , Espermatogénesis , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , Proteínas Supresoras de Tumor/genética
20.
Reproduction ; 141(5): 571-80, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307270

RESUMEN

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.


Asunto(s)
Anticonceptivos Masculinos/farmacología , Hidrazinas/farmacología , Indazoles/farmacología , Epitelio Seminífero/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Animales , Adhesión Celular/efectos de los fármacos , Anticonceptivos Masculinos/toxicidad , Humanos , Hidrazinas/toxicidad , Indazoles/toxicidad , Indenos/farmacología , Masculino , Piperidinas/farmacología , Células de Sertoli/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda