Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 30(16): 47349-47365, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36737566

RESUMEN

Deca brominated diphenyl ether (BDE-209) is a widely used flame retardant with endocrine-disrupting activity which reportedly caused sperm quality decline and damaged blood-testis barrier (BTB). However, whether BDE-209 exposure led to BTB integrity dysfunction through affecting microtubule cytoskeletal organization and junctions was not well-elucidated. This study aimed to investigate the role of estrogen receptor α (ERα) in BDE-209-mediated perturbation of BTB integrity. Male rats and primary culture Sertoli cells were co-treated with BDE-209 and propylpyrazoletriol (PPT). The data demonstrated that BDE-209 impaired BTB integrity by reducing crucial tight junction-related proteins with ZO-1 and Occludin. Furthermore, the data suggested that BDE-209 diminished the apical ectoplasmic specialization markers with Eps8 and Formin1. In addition, BDE-209 damaged BTB ultrastructure including tight junctions and ectoplasmic specialization structures with broken tight junctions and the absence of actin microfilaments. Further experiments revealed that ERα was triggered in BDE-209-treated Sertoli cells. Unexpectedly, we found that PPT rescued BDE-209-mediated disruption of BTB integrity including tight junction and apical ectoplasmic specialization by activating ERα in Sertoli cells. Taken together, these findings indicated that intratesticular BDE-209 exposure perturbed BTB integrity and destroyed BTB structure by blocking ERα pathway. Our findings provide a new therapeutic target for male reproductive dysfunction.


Asunto(s)
Receptor alfa de Estrógeno , Éteres Difenilos Halogenados , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Éteres Difenilos Halogenados/toxicidad , Éteres Difenilos Halogenados/metabolismo , Receptor alfa de Estrógeno/metabolismo , Espermatogénesis/fisiología , Barrera Hematotesticular/fisiología , Semen , Células de Sertoli/metabolismo , Transducción de Señal
2.
Cells ; 11(3)2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35159162

RESUMEN

Spermatogenesis is a complex process occurring in mammalian testes, and constant sperm production depends on the exact regulation of the microenvironment in the testes. Many studies have indicated the crucial role of blood-testis barrier (BTB) junctions and retinoic acid (RA) signaling in the spermatogenesis process. The BTB consists of junctions between adjacent Sertoli cells, comprised mainly of tight junctions and gap junctions. In vitamin A-deficient mice, halted spermatogenesis could be rebooted by RA or vitamin A administration, indicating that RA is absolutely required for spermatogenesis. Accordingly, this manuscript will review and discuss how RA and the BTB regulate spermatogenesis and the interaction between RA signaling and BTB function.


Asunto(s)
Barrera Hematotesticular , Tretinoina , Animales , Barrera Hematotesticular/fisiología , Masculino , Mamíferos , Ratones , Células de Sertoli , Espermatogénesis/fisiología , Tretinoina/farmacología , Vitamina A
3.
Cells ; 10(9)2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34572053

RESUMEN

A protocol for the isolation and long-term propagation of adult rat Sertoli cells (SCs) using conditional reprogramming (CR) was developed and the formation of tight junctions as an in vitro model for the blood testis barrier (BTB) was studied. Three pure primary SC lines were isolated successfully and maintained for several months without significant changes in expression levels of SC-typical markers such as SRY-box transcription factor 9 (SOX9), transferrin, clusterin, androgen receptor (AR), and GATA binding protein 1 (GATA1). In addition to AR expression, the tight junction proteins, zonula occludens-1 (ZO-1) and the junctional adhesion molecule-3 (JAM-3), were upregulated and the SC barrier integrity was enhanced by testosterone. Peritubular/myoid cells did not increase the tightness of the SC. The cytokines, interleukin-6 (IL-6), bone morphogenetic protein-2 (BMP2), and transforming growth factor beta-3 (TGF-ß3), negatively affected the tightness of the SC barrier. We have established a protocol for the isolation and long-term propagation of highly pure primary adult rat SCs, which are able to respond to androgen treatments, to form tight junctions and to maintain the mRNA expression of SC-specific genes. By applying this new method, adult SCs can now be analyzed in more detail and might serve as an in vitro model for the study of many SC functions.


Asunto(s)
Andrógenos/farmacología , Biomarcadores/metabolismo , Barrera Hematotesticular/fisiología , Regulación de la Expresión Génica , Células de Sertoli/citología , Testículo/citología , Animales , Barrera Hematotesticular/efectos de los fármacos , Citocinas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/fisiología
4.
Endocrinology ; 162(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34343260

RESUMEN

Studies suggest that HIV-1 invades the testis through initial permeation of the blood-testis barrier (BTB). The selectivity of the BTB to antiretroviral drugs makes this site a sanctuary for the virus. Little is known about how HIV-1 crosses the BTB and invades the testis. Herein, we used 2 approaches to examine the underlying mechanism(s) by which HIV-1 permeates the BTB and gains entry into the seminiferous epithelium. First, we examined if recombinant Tat protein was capable of perturbing the BTB and making the barrier leaky, using the primary rat Sertoli cell in vitro model that mimics the BTB in vivo. Second, we used HIV-1-infected Sup-T1 cells to investigate the activity of HIV-1 infection on cocultured Sertoli cells. Using both approaches, we found that the Sertoli cell tight junction permeability barrier was considerably perturbed and that HIV-1 effectively permeates the BTB by inducing actin-, microtubule-, vimentin-, and septin-based cytoskeletal changes in Sertoli cells. These studies suggest that HIV-1 directly perturbs BTB function, potentially through the activity of the Tat protein.


Asunto(s)
Barrera Hematotesticular/fisiología , Citoesqueleto/metabolismo , VIH-1/fisiología , Evasión Inmune/fisiología , Testículo/virología , Animales , Animales Recién Nacidos , Barrera Hematotesticular/ultraestructura , Permeabilidad de la Membrana Celular , Células Cultivadas , Citoesqueleto/ultraestructura , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Masculino , Ratas , Ratas Sprague-Dawley , Testículo/inmunología , Testículo/metabolismo , Testículo/ultraestructura
5.
Asian J Androl ; 23(2): 123-128, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32896837

RESUMEN

Collagen α3 (IV) chains are one of the major constituent components of the basement membrane in the mammalian testis. Studies have shown that biologically active fragments, such as noncollagenase domain (NC1)-peptide, can be released from the C-terminal region of collagen α3 (IV) chains, possibly through the proteolytic action of metalloproteinase 9 (MMP9). NC1-peptide was shown to promote blood-testis barrier (BTB) remodeling and fully developed spermatid (e.g., sperm) release from the seminiferous epithelium because this bioactive peptide was capable of perturbing the organization of both actin- and microtubule (MT)-based cytoskeletons at the Sertoli cell-cell and also Sertoli-spermatid interface, the ultrastructure known as the basal ectoplasmic specialization (ES) and apical ES, respectively. More importantly, recent studies have shown that this NC1-peptide-induced effects on cytoskeletal organization in the testis are mediated through an activation of mammalian target of rapamycin complex 1/ribosomal protein S6/transforming retrovirus Akt1/2 protein (mTORC1/rpS6/Akt1/2) signaling cascade, involving an activation of cell division control protein 42 homolog (Cdc42) GTPase, but not Ras homolog family member A GTPase (RhoA), and the participation of end-binding protein 1 (EB1), a microtubule plus (+) end tracking protein (+TIP), downstream. Herein, we critically evaluate these findings, providing a critical discussion by which the basement membrane modulates spermatogenesis through one of its locally generated regulatory peptides in the testis.


Asunto(s)
Membrana Basal/metabolismo , Barrera Hematotesticular/metabolismo , Colágeno Tipo IV/metabolismo , Fragmentos de Péptidos/metabolismo , Epitelio Seminífero/metabolismo , Espermatogénesis/fisiología , Citoesqueleto de Actina , Animales , Membrana Basal/fisiología , Barrera Hematotesticular/fisiología , Comunicación Celular , Colágeno Tipo IV/fisiología , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos , Fragmentos de Péptidos/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína S6 Ribosómica/metabolismo , Epitelio Seminífero/fisiología , Células de Sertoli/metabolismo , Células de Sertoli/fisiología , Transducción de Señal , Espermátides/metabolismo , Espermátides/fisiología , Testículo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Biol Reprod ; 103(4): 880-891, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32678429

RESUMEN

Spermatogenesis is a complex process that establishes male fertility and involves proper communication between the germline (spermatozoa) and the somatic tissue (Sertoli cells). Many factors that are important for spermatozoa production are also required for Sertoli cell function. Recently, we showed that the transcriptional cofactor ubiquitously expressed transcript (UXT) encodes a protein that is essential in germ cells for spermatogenesis and fertility. However, the role of UXT within Sertoli cells and how it affects Sertoli cell function was still unclear. Here we describe a novel role for UXT in the Sertoli cell's ability to support spermatogenesis. We find that the conditional deletion of Uxt in Sertoli cells results in smaller testis size and weight, which coincided with a loss of germ cells in a subset of seminiferous tubules. In addition, the deletion of Uxt has no impact on Sertoli cell abundance or maturity, as they express markers of mature Sertoli cells. Gene expression analysis reveals that the deletion of Uxt in Sertoli cells reduces the transcription of genes involved in the tight junctions of the blood-testis barrier (BTB). Furthermore, tracer experiments and electron microscopy reveal that the BTB is permeable in UXT KO animals. These findings broaden our understanding of UXT's role in Sertoli cells and its contribution to the structural integrity of the BTB.


Asunto(s)
Barrera Hematotesticular/fisiología , Proteínas de Ciclo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Células de Sertoli/metabolismo , Animales , Adhesión Celular , Proteínas de Ciclo Celular/genética , Regulación hacia Abajo , Eliminación de Gen , Regulación de la Expresión Génica , Células Germinativas/fisiología , Masculino , Ratones , Chaperonas Moleculares/genética , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
7.
Trends Mol Med ; 26(2): 215-231, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31727542

RESUMEN

Spermatogenesis is supported by intricate crosstalk between Sertoli cells and germ cells including spermatogonia, spermatocytes, haploid spermatids, and spermatozoa, which takes place in the epithelium of seminiferous tubules. Sertoli cells, also known as 'mother' or 'nurse' cells, provide nutrients, paracrine factors, cytokines, and other biomolecules to support germ cell development. Sertoli cells facilitate the generation of several biologically active peptides, which include F5-, noncollagenous 1 (NC1)-, and laminin globular (LG)3/4/5-peptide, to modulate cellular events across the epithelium. Here, we critically evaluate the involvement of these peptides in facilitating crosstalk between Sertoli and germ cells to support spermatogenesis and thus fertility. Modulating or mimicking the activity of F5-, NC1-, and LG3/4/5-peptide could be used to enhance the transport across the blood-testis barrier (BTB) of contraceptive drugs or to treat male infertility.


Asunto(s)
Fertilidad/fisiología , Células Germinativas/fisiología , Células de Sertoli/fisiología , Animales , Barrera Hematotesticular/fisiología , Humanos , Infertilidad Masculina/fisiopatología , Masculino , Espermatogénesis/fisiología
8.
FASEB J ; 33(12): 14461-14478, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31682474

RESUMEN

Noncollagenous domain 1 (NC1)-peptide is a biologically active peptide derived from the C-terminal region of collagen α3(IV) chain, a structural constituent protein at the basement membrane in the rat testis, likely via proteolytic cleavage of matrix metalloproteinase 9. Studies have shown that this NC1 peptide regulates testis function by inducing Sertoli cell blood-testis barrier (BTB) remodeling and is also capable of inducing elongate spermatid exfoliation through its disruptive effects on the organization of actin- and microtubule (MT)-based cytoskeletons at these cell adhesion sites. However, the underlying molecular mechanism remains unknown. NC1 peptide was found to exert its biologic effects through an activation of small GTPase cell division control protein 42 homolog (Cdc42) because cooverexpression of the dominant negative mutant of Cdc42 [namely, Cdc42-T17N (via a single mutation of amino acid residue 17 from the N terminus from Thr to Asn by site-directed mutagenesis, making it constitutively inactive)] and NC1 peptide was able to block the NC1 peptide-induced Sertoli cell tight junction-permeability barrier disruption. Their cooverexpression also blocked the NC1 peptide-induced misdistribution of BTB-associated proteins at the cell-cell interface and also disruptive cytoskeletal organization of F-actin and MTs through changes in spatial expression of the corresponding actin and MT regulatory proteins. Interestingly, NC1 peptide was also found to induce an up-regulation of phosphorylated (p)-ribosomal protein S6 (rpS6) (namely, p-rpS6-S235/S236) and a concomitant down-regulation of p-Akt1/2 (namely, p-Akt1-S473 and p-Akt2-S474), but these changes could not be blocked by overexpression of Cdc42-T17N. More importantly, NC1 peptide-induced Cdc42 activation was effectively blocked by treatment of Sertoli cell epithelium with a p-Akt1/2 activator SC79, which is also capable of blocking NC1 peptide-induced down-regulation of p-Akt1-S473 and p-Akt2/S474, but not p-rpS6-S235/S236 up-regulation. In summary, these findings illustrate that Cdc42 is working downstream of the mammalian target of rapamycin complex 1/rpS6/Akt1/2 signaling pathway to support NC1 peptide-mediated effects on Sertoli cell function in the testis using the rat as an animal model.-Su, W., Cheng, C. Y. Cdc42 is involved in NC1 peptide-regulated BTB dynamics through actin and microtubule cytoskeletal reorganization.


Asunto(s)
Actinas/metabolismo , Barrera Hematotesticular/fisiología , Microtúbulos/fisiología , Células de Sertoli/efectos de los fármacos , Proteína de Unión al GTP cdc42/metabolismo , Acetatos/farmacología , Actinas/genética , Animales , Benzopiranos/farmacología , Citoesqueleto/fisiología , ADN/genética , ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Plásmidos , Dominios Proteicos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Sertoli/fisiología , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
9.
Mol Nutr Food Res ; 63(10): e1800843, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30924608

RESUMEN

SCOPE: Luteolin, a natural flavonoid, displays protective activities to testicular tissue. However, the molecular mechanisms are still unclear. In this study, the aim is to identify the protective effects and underlying mechanisms of luteolin against triptolide (TP)-induced damage of testicular tissue. METHODS AND RESULTS: Pre-incubation of Sertoli cells (SCs) with luteolin results in a significant reduction of TP-induced apoptotic cells, which occurs concomitantly with the effective inhibition of reactive oxygen species accumulation. Luteolin results in a significant reduction in testicular damage and spermatogenesis dysfunction in a mouse model of testicular damage. Mechanistic studies reveal that luteolin significantly triggers Nrf2 translocation, increases antioxidant response element-luciferase reporter activity, and induces antioxidant enzyme expression. Nrf2 siRNA reduces luteolin-induced protection in SCs. Besides inhibiting apoptosis, luteolin recovers the blood-testis barrier (BTB) integrity by upregulating connexin43 (Cx43) expression. Moreover, specifically blocked Cx43 activity completely blocks repairmen of luteolin to BTB values. In accordance with in vitro results, luteolin suppresses testicular injury and spermatogenesis dysfunction by activation of Nrf2 and Cx43 in a testicular injury model. CONCLUSION: Luteolin is identified as a novel active ingredient that contributes to the protective activity in testicular damage through activating the Nrf2 signaling pathway and by upregulating Cx43.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Conexina 43/metabolismo , Luteolina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Testículo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Barrera Hematotesticular/fisiología , Diterpenos/toxicidad , Compuestos Epoxi/toxicidad , Masculino , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , Fenantrenos/toxicidad , Sustancias Protectoras/farmacología , Células de Sertoli/efectos de los fármacos , Células de Sertoli/patología , Transducción de Señal/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Regulación hacia Arriba/efectos de los fármacos
10.
Environ Sci Pollut Res Int ; 26(5): 4801-4820, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30565106

RESUMEN

Deca-bromodiphenyl ether (BDE-209) regulates various aspects of spermatogenesis and male fertility through its effect on estrogen receptor α (ERα), but the underlying mechanism remains unclear. Because molecular mechanisms such as remodeling of the blood-testis barrier (BTB) play crucial roles in spermatogenesis, we investigated the disruptive effects of ERα agonists on the BTB in spermatogenesis. In this study, 0, 300, and 500 mg/kg/day of BDE-209 were administered to pregnant adult mice by oral gavage from gestation day 7 to postnatal day 21. SerW3 cells were treated with methylpiperidino pyrazole (MPP) for 30 min before being treated with 50 µg/mL of BDE-209. BDE-209 increases ERα in time- and dose-dependent manners and decreases formin 1 and BTB-associated protein in F1 male mice. Furthermore, BDE-209 impairs the structure and function of the BTB. Activation of ERα signaling could disrupt the BTB, leading to spermatogenesis dysfunction. The results identified the role of ERα in BTB disruption during spermatogenesis and suggested that BTB disruption occurs because of exposure to BDE-209, which could potentially affect spermatogenesis. In conclusion, Sertoli cells seem to be the primary target of BDE-209 in the perinatal period, and this period constitutes a critical window of susceptibility to BDE-209. Also, the SerW3 cell model may not be a particularly useful cell model for studying the function of the cytoskeleton.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Éteres Difenilos Halogenados/toxicidad , Células de Sertoli/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Animales , Barrera Hematotesticular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Proteínas Fetales/metabolismo , Forminas , Éteres Difenilos Halogenados/administración & dosificación , Éteres Difenilos Halogenados/farmacocinética , Masculino , Ratones Endogámicos ICR , Proteínas de Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Embarazo , Transducción de Señal/efectos de los fármacos , Espermatogénesis/fisiología , Testículo/efectos de los fármacos
11.
J Vis Exp ; (142)2018 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-30582592

RESUMEN

Spermatogenesis is the development of spermatogonia into mature spermatozoa in the seminiferous tubules of the testis. This process is supported by Sertoli cell junctions at the blood-testis barrier (BTB), which is the tightest tissue barrier in the mammalian body and segregates the seminiferous epithelium into two compartments, a basal and an adluminal. The BTB creates a unique microenvironment for germ cells in meiosis I/II and for the development of postmeiotic spermatids into spermatozoa via spermiogenesis. Here, we describe a reliable assay to monitor BTB integrity of mouse testis in vivo. An intact BTB blocks the diffusion of FITC-conjugated inulin from the basal to the apical compartment of the seminiferous tubules. This technique is suitable for studying gene candidates, viruses, or environmental toxicants that may affect BTB function or integrity, with an easy procedure and a minimal requirement of surgical skills compared to alternative methods.


Asunto(s)
Barrera Hematotesticular/fisiología , Animales , Barrera Hematotesticular/metabolismo , Barrera Hematotesticular/fisiopatología , Masculino , Ratones
12.
PLoS One ; 13(6): e0198100, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856785

RESUMEN

Connexin43 (Cx43) is the predominant testicular gap junction protein and in cases of impaired spermatogenesis, Cx43 expression has been shown to be altered in several mammals. Amongst other functions, Cx43 is supposed to regulate junction formation of the blood-testis barrier (BTB). The aim of the present study was to investigate the expression pattern of different tight junction (TJ) proteins of the murine BTB using SC-specific Cx43 knockout mice (SCCx43KO). Adult homozygous male SCCx43KO mice (SCCx43KO-/-) predominantly show an arrest of spermatogenesis and SC-only tubules that might have been caused by an altered BTB assembly, composition or regulation. TJ molecules claudin-3, -5 and -11 were examined in adult wild type (WT) and SCCx43KO-/- mice using immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR). In this context, investigation of single tubules with residual spermatogenesis in SCCx43KO-/- mice was particularly interesting to identify a potential Cx43-independent influence of germ cells (GC) on BTB composition and dynamics. In tubules without residual spermatogenesis, a diffuse cytoplasmic distribution pattern for claudin-11 protein could be demonstrated in mutant mice. Nevertheless, claudin-11 seems to form functional TJ. Claudin-3 and -5 could not be detected immunohistochemically in the seminiferous epithelium of those tubules. Correspondingly, claudin-3 and -5 mRNA expression was decreased, providing evidence of generally impaired BTB dynamics in adult KO mice. Observations of tubules with residual spermatogenesis suggested a Cx43-independent regulation of TJ proteins by GC populations. To determine initial BTB formation in peripubertal SCCx43KO-/- mice, immunohistochemical staining and qRT-PCR of claudin-11 were carried out in adolescent SCCx43KO-/- and WT mice. Additionally, BTB integrity was functionally analysed using a hypertonic glucose fixative. These analyses revealed that SCCx43KO-/- mice formed an intact BTB during puberty in the same time period as WT mice, which however seemed to be accelerated.


Asunto(s)
Barrera Hematotesticular/fisiología , Conexina 43/deficiencia , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Uniones Estrechas/fisiología , Animales , Claudinas/genética , Claudinas/metabolismo , Conexina 43/fisiología , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/ultraestructura , Células de Sertoli/ultraestructura , Maduración Sexual , beta-Galactosidasa/metabolismo
13.
Methods Mol Biol ; 1748: 245-252, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29453576

RESUMEN

The blood-testis barrier is a unique ultrastructure in the mammalian testis, located near the basement membrane of the seminiferous tubule that segregates the seminiferous epithelium into the basal and the adluminal (apical) compartment. Besides restricting paracellular and transcellular passage of biomolecules (e.g., paracrine factors, hormones), water, electrolytes, and other substances including toxicants and/or drugs to enter the adluminal compartment of the epithelium, the BTB is an important ultrastructure that supports spermatogenesis. As such, a sensitive and reliable assay to monitor its integrity in vivo is helpful for studying testis biology. This assay is based on the ability of an intact BTB to exclude the diffusion of a small molecule such as sulfo-NHS-LC-biotin (C20H29N4NaO9S2, Mr. 556.59, a water-soluble and membrane-impermeable biotinylation reagent) from the basal to the apical compartment of the seminiferous epithelium. Herein, we summarize the detailed procedures on performing the assay and to obtain semiquantitative data to assess the extent of BTB damage when compared to positive controls, such as treatment of rats with cadmium chloride (CdCl2) which is known to compromise the BTB integrity.


Asunto(s)
Biotina/metabolismo , Barrera Hematotesticular/fisiología , Epitelio Seminífero/metabolismo , Células de Sertoli/metabolismo , Uniones Estrechas/metabolismo , Animales , Barrera Hematotesticular/efectos de los fármacos , Células Cultivadas , Masculino , Ratas , Ratas Sprague-Dawley , Epitelio Seminífero/citología , Células de Sertoli/citología
14.
Methods Mol Biol ; 1748: 229-243, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29453575

RESUMEN

The blood-testis barrier (BTB) is an important ultrastructure in the testis that supports meiosis and postmeiotic spermatid development since a delay in the establishment of a functional Sertoli cell barrier during postnatal development in rats or mice by 17-20 day postpartum (dpp) would lead to a delay of the first wave of meiosis. Furthermore, irreversible disruption of the BTB by toxicants also induces infertility in rodents. Herein, we summarize recent findings that BTB dynamics (i.e., disassembly, reassembly, and stabilization) are supported by the concerted efforts of the actin- and microtubule (MT)-based cytoskeletons. We focus on the role of two actin nucleation protein complexes, namely, the Arp2/3 (actin-related protein 2/3) complex and formin 1 (or the formin 1/spire 1 complex) known to induce actin nucleation, respectively, by conferring plasticity to actin cytoskeleton. We also focus on the MT plus (+)-end tracking protein (+TIP) EB1 (end-binding protein 1) which is known to confer MT stabilization. Furthermore, we discuss in particular how the interactions of these proteins modulate BTB dynamics during spermatogenesis. These findings also yield a novel hypothetical concept regarding the molecular mechanism that modulates BTB function.


Asunto(s)
Actinas/metabolismo , Barrera Hematotesticular/fisiología , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratas , Células de Sertoli/citología
15.
J Biol Chem ; 292(52): 21490-21503, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29123028

RESUMEN

The Ig-like cell adhesion molecule (IgCAM) BT-IgSF (brain- and testis-specific Ig superfamily protein) plays a major role in male fertility in mice. However, the molecular mechanism by which BT-IgSF supports fertility is unclear. Here, we found that it is localized in Sertoli cells at the blood-testis barrier (BTB) and at the apical ectoplasmic specialization. The absence of BT-IgSF in Sertoli cells in both global and conditional mouse mutants (i.e. AMHCre and Rosa26CreERT2 lines) resulted in male infertility, atrophic testes with vacuolation, azoospermia, and spermatogenesis arrest. Although transcripts of junctional proteins such as connexin43, ZO-1, occludin, and claudin11 were up-regulated in the absence of BT-IgSF, the functional integrity of the BTB was impaired, as revealed by injection of a BTB-impermeable component into the testes under in vivo conditions. Disruption of the BTB coincided with mislocalization of connexin43, which was present throughout the seminiferous epithelium and not restricted to the BTB as in wild-type tissues, suggesting impaired cell-cell communication in the BT-IgSF-KO mice. Because EM images revealed a normal BTB structure between Sertoli cells in the BT-IgSF-KO mice, we conclude that infertility in these mice is most likely caused by a functionally impaired BTB. In summary, our results indicate that BT-IgSF is expressed at the BTB and is required for male fertility by supporting the functional integrity of the BTB.


Asunto(s)
Barrera Hematotesticular/fisiología , Inmunoglobulinas/fisiología , Espermatogénesis/genética , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/metabolismo , Conexina 43/metabolismo , Fertilidad/fisiología , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Noqueados , Ocludina/metabolismo , Epitelio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
16.
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
17.
Biochim Biophys Acta ; 1863(8): 1996-2005, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27113856

RESUMEN

Actin remodeling is a vital process for signaling, movement and survival in all cells. In the testes, extensive actin reorganization occurs at spermatid-Sertoli cell junctions during sperm release (spermiation) and at inter Sertoli cell junctions during restructuring of the blood testis barrier (BTB). During spermiation, tubulobulbar complexes (TBCs), rich in branched actin networks, ensure recycling of spermatid-Sertoli cell junctional molecules. Similar recycling occurs during BTB restructuring around the same time as spermiation occurs. Actin related protein 2/3 complex is an essential actin nucleation and branching protein. One of its subunits, Arpc1b, was earlier found to be down-regulated in an estrogen-induced rat model of spermiation failure. Also, Arpc1b was found to be estrogen responsive through estrogen receptor beta in seminiferous tubule culture. Here, knockdown of Arpc1b by siRNA in adult rat testis led to defects in spermiation caused by failure in TBC formation. Knockdown also compromised BTB integrity and caused polarity defects of mature spermatids. Apart from these effects pertaining to Sertoli cells, Arpc1b reduction perturbed ability of germ cells to enter G2/M phase thus hindering cell division. In summary, Arpc1b, an estrogen responsive gene, is a regulator of spermiation, mature spermatid polarity, BTB integrity and cell division during adult spermatogenesis.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/fisiología , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Espermatozoides/metabolismo , Citoesqueleto de Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/antagonistas & inhibidores , Complejo 2-3 Proteico Relacionado con la Actina/genética , Animales , Barrera Hematotesticular/fisiología , Estradiol/fisiología , Humanos , Masculino , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Células de Sertoli/ultraestructura , Recuento de Espermatozoides , Espermátides/metabolismo , Espermátides/ultraestructura , Espermatocitos/metabolismo , Espermatocitos/ultraestructura , Espermatozoides/ultraestructura
18.
Arch Toxicol ; 90(4): 971-83, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25743374

RESUMEN

Perfluorooctanoic acid (PFOA) is correlated with male reproductive dysfunction in animals and humans, but the underlying mechanisms for this remain unknown. To explore the potential reproductive toxicity of PFOA, we studied blood-testis barrier (BTB) damage using in vivo and in vitro models. Male mice were gavage-administered PFOA (0-20 mg/kg/d) for 28 consecutive days, and breeding capacity and permeability of the Sertoli cell-based BTB were estimated. Primary Sertoli cells (SCs) were exposed to PFOA (0-500 µM) for 48 h, and transepithelial electrical resistance (TER) was assessed. Furthermore, BTB-associated protein expression, TNFα content, and phosphorylation and protein levels of the mitogen-activated protein kinase (MAPK) pathway were detected. An apparent decrease in mated and pregnant females per male mouse as well as litter weight was observed. Marked BTB damage was evidenced by increased red biotin fluorescence in the lumen tubular of the testes and the decrease in TER in SCs in vitro. The protein levels of claudin-11, connexin-43, N-cadherin, ß-catenin, and occludin were significantly decreased in the testes and also in the SCs in vitro except for N-cadherin and ß-catenin. TNFα content showed a dose-dependent increase in the testes and a dose- and time-dependent increase in the SCs, with the p-p38/p38 MAPK ratio also increasing in testes and SCs after PFOA exposure. Moreover, PFOA altered expressions of claudin-11, connexin-43, TNFα, and p-p38 MAPK were recovered 48 h after PFOA removal in the SCs. The SCs appeared to be target to PFOA, and the disruption of the BTB may be crucial to PFOA-induced reproductive dysfunction in mice.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Caprilatos/toxicidad , Fluorocarburos/toxicidad , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Barrera Hematotesticular/fisiología , Femenino , Fertilidad/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Embarazo , Células de Sertoli/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/inmunología
19.
Reproduction ; 151(3): R29-41, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26628556

RESUMEN

The blood-testis barrier (BTB) is an important ultrastructure in the testis, since the onset of meiosis and spermiogenesis coincides with the establishment of a functional barrier in rodents and humans. It is also noted that a delay in the assembly of a functional BTB following treatment of neonatal rats with drugs such as diethylstilbestrol or adjudin also delays the first wave of spermiation. While the BTB is one of the tightest blood-tissue barriers, it undergoes extensive remodeling, in particular, at stage VIII of the epithelial cycle to facilitate the transport of preleptotene spermatocytes connected in clones across the immunological barrier. Without this timely transport of preleptotene spermatocytes derived from type B spermatogonia, meiosis will be arrested, causing aspermatogenesis. Yet the biology and regulation of the BTB remains largely unexplored since the morphological studies in the 1970s. Recent studies, however, have shed new light on the biology of the BTB. Herein, we critically evaluate some of these findings, illustrating that the Sertoli cell BTB is regulated by actin-binding proteins (ABPs), likely supported by non-receptor protein kinases, to modulate the organization of actin microfilament bundles at the site. Furthermore, microtubule-based cytoskeleton is also working in concert with the actin-based cytoskeleton to confer BTB dynamics. This timely review provides an update on the unique biology and regulation of the BTB based on the latest findings in the field, focusing on the role of ABPs and non-receptor protein kinases.


Asunto(s)
Citoesqueleto de Actina/fisiología , Barrera Hematotesticular/fisiología , Proteínas de Microfilamentos/fisiología , Proteínas Quinasas/metabolismo , Animales , Membrana Basal/fisiología , Barrera Hematotesticular/anatomía & histología , Humanos
20.
Asian J Androl ; 17(4): 591-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25966624

RESUMEN

The onslaught of foreign antigens carried by spermatozoa into the epididymis, an organ that has not demonstrated immune privilege, a decade or more after the establishment of central immune tolerance presents a unique biological challenge. Historically, the physical confinement of spermatozoa to the epididymal tubule enforced by a tightly interwoven wall of epithelial cells was considered sufficient enough to prevent cross talk between gametes and the immune system and, ultimately, autoimmune destruction. The discovery of an intricate arrangement of mononuclear phagocytes (MPs) comprising dendritic cells and macrophages in the murine epididymis suggests that we may have underestimated the existence of a sophisticated mucosal immune system in the posttesticular environment. This review consolidates our current knowledge of the physiology of MPs in the steady state epididymis and speculates on possible interactions between auto-antigenic spermatozoa, pathogens and the immune system by drawing on what is known about the immune system in the intestinal mucosa. Ultimately, further investigation will provide valuable information regarding the origins of pathologies arising as a result of autoimmune or inflammatory responses in the epididymis, including epididymitis and infertility.


Asunto(s)
Epidídimo/fisiología , Monocitos/fisiología , Fagocitos/fisiología , Animales , Barrera Hematotesticular/fisiología , Epidídimo/irrigación sanguínea , Humanos , Intestino Delgado/citología , Intestino Delgado/fisiología , Masculino , Flujo Sanguíneo Regional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA