Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Andrology ; 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37415418

RESUMEN

BACKGROUND: The mammalian epididymis is a specialized duct system that serves a critical role in sperm maturation and storage. Its distinctive, highly coiled tissue morphology provides a unique opportunity to investigate the link between form and function in reproductive biology. Although recent genetic studies have identified key genes and signaling pathways involved in the development and physiological functions of the epididymis, there has been limited discussion about the underlying dynamic and mechanical processes that govern these phenomena. AIMS: In this review, we aim to address this gap by examining two key aspects of the epididymis across its developmental and physiological phases. RESULTS AND DISCUSSION: First, we discuss how the complex morphology of the Wolffian/epididymal duct emerges through collective cell dynamics, including duct elongation, cell proliferation, and arrangement during embryonic development. Second, we highlight dynamic aspects of luminal fluid flow in the epididymis, essential for regulating the microenvironment for sperm maturation and motility, and discuss how this phenomenon emerges and interplays with epididymal epithelial cells. CONCLUSION: This review not only aims to summarize current knowledge but also to provide a starting point for further exploration of mechanobiological aspects related to the cellular and extracellular fluid dynamics in the epididymis.

2.
Biol Reprod ; 108(2): 241-257, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36525341

RESUMEN

Primary cilia play pivotal roles in embryonic patterning and organogenesis through transduction of the Hedgehog signaling pathway (Hh). Although mutations in Hh morphogens impair the development of the gonads and trigger male infertility, the contribution of Hh and primary cilia in the development of male reproductive ductules, including the epididymis, remains unknown. From a Pax2Cre; IFT88fl/fl knock-out mouse model, we found that primary cilia deletion is associated with imbalanced Hh signaling and morphometric changes in the Wolffian duct (WD), the embryonic precursor of the epididymis. Similar effects were observed following pharmacological blockade of primary cilia formation and Hh modulation on WD organotypic cultures. The expression of genes involved in extracellular matrix, mesenchymal-epithelial transition, canonical Hh and WD development was significantly altered after treatments. Altogether, we identified the primary cilia-dependent Hh signaling as a master regulator of genes involved in WD development. This provides new insights regarding the etiology of sexual differentiation and male infertility issues.


Asunto(s)
Cilios , Proteínas Hedgehog , Animales , Ratones , Masculino , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Cilios/fisiología , Conductos Mesonéfricos/metabolismo , Transducción de Señal/fisiología , Organogénesis , Ratones Noqueados
3.
Life Sci ; 315: 121329, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36584913

RESUMEN

Depressive disorders (DD) have affected millions of people worldwide. Venlafaxine, antidepressant of the class of serotonin and norepinephrine reuptake inhibitors, has been prescribed for the treatment of DD. In rat testes, venlafaxine induces testosterone (T) aromatization and increases estrogen levels. Aromatase is a key enzyme for the formation of estrogen in the epididymis, an essential organ for male fertility. We investigated the impact of serotonergic/noradrenergic venlafaxine effect on the epididymal cauda region, focusing on aromatase, V-ATPase and EGF epithelial immunoexpression, smooth muscle (SM) integrity and mast cells number (MCN). Male rats were distributed into control (CG; n = 10) and venlafaxine (VFG, n = 10) groups. VFG received 30 mg/kg b.w. of venlafaxine for 35 days. The epididymal cauda was processed for light and transmission electron microscopy (TEM). The expression of connexin 43 (Cx43) and estrogen alpha (Esr1), adrenergic (Adra1a) and serotonergic (Htr1b) receptors were analyzed. Clear cells (CCs) area, SM thickness, viable spermatozoa (VS) and MCN were evaluated. Apoptosis was confirmed by TUNEL and TEM. The following immunoreactions were performed: T, aromatase, T/aromatase co-localization, V-ATPase, EGF, Cx43 and PCNA. The increased Adra1a and reduced Htr1b expressions confirmed the noradrenergic and serotonergic venlafaxine effects, respectively, corroborating the increased MCN, apoptosis and atrophy of SM. In VFG, the epithelial EGF increased, explaining Cx43 overexpression and basal cells mitotic activity. T aromatization and Esr1 downregulation indicate high estrogen levels, explaining CCs hypertrophy and changes in the V-ATPase localization, corroborating VS reduction. Thus, in addition to serotonergic/noradrenergic effects, T/estrogen imbalance, induced by venlafaxine, impairs epididymal structure and function.


Asunto(s)
Epidídimo , ATPasas de Translocación de Protón Vacuolares , Ratas , Masculino , Animales , Clorhidrato de Venlafaxina/farmacología , Clorhidrato de Venlafaxina/metabolismo , Aromatasa , Conexina 43/metabolismo , Mastocitos/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , ATPasas de Translocación de Protón Vacuolares/farmacología , Estrógenos/farmacología , Miocitos del Músculo Liso/metabolismo
4.
Andrology ; 10(1): 190-201, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34415685

RESUMEN

BACKGROUND: Immunoregulatory genes encoding activin A (Inhba) and B (Inhbb), and indolamine 2,3-dioxygenase-1 (Ido1) are highly expressed in the murine caput epididymidis, which also has a network of intraepithelial mononuclear phagocytes. This environment is postulated to promote immunological tolerance to epididymal sperm. The factors regulating the immunoregulatory agents in the epididymal caput are poorly understood. OBJECTIVES: This study aimed to investigate the potential role of testicular lumicrine factors in regulating activin and other immune-related genes in the caput epididymidis. MATERIALS AND METHODS: The efferent ducts in adult C57/Bl6 mice were exposed and ligated bilaterally. Serum and tissues were collected seven days later. Animals with bilateral sham ligation and animals with no ligations (collectively referred to as the "intact" group) were used as controls. RESULTS: Pressure-induced seminiferous epithelial damage due to intratubular fluid accumulation was observed in all ligated testes. Testicular inhibin was significantly increased and testosterone was elevated in some animals following bilateral ligation, but serum testosterone, serum LH, and serum inhibin were normal. Ligation caused epithelial regression in the initial segment, with similar but less severe effects in other caput segments. Activin A staining by immunohistochemistry in the epithelium was reduced in bilateral ligation, particularly in the initial segment, with moderately reduced staining intensity in the rest of the caput. Inhba expression within the caput was not significantly affected by bilateral ligation, but Inhbb was reduced by more than 60%. Transcripts encoding the macrophage-specific receptor Cx3cr1 were significantly reduced following bilateral ligation, but other immune cell markers, Ido1, and inflammatory genes were unaffected. CONCLUSION: These data indicate that testicular lumicrine secretion regulates several genes that are preferentially expressed in the initial segment, but has marginal effects on genes such as those encoding activin A and IDO1, which are expressed more widely in the caput.


Asunto(s)
Activinas/inmunología , Epidídimo/inmunología , Tolerancia Inmunológica/genética , Inhibinas/inmunología , Testículo/inmunología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Espermatozoides/inmunología
5.
J Reprod Immunol ; 149: 103456, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34915277

RESUMEN

The Wolffian duct (WD) is an embryonic tissue that undergoes androgen-induced morphological changes to become the epididymis. Toll-like receptor 4 (TLR4)- and nuclear factor kB (NFKB)-induced effectors are expressed in the adult epididymis and represent important players in epididymal innate immune responses. TLR4/NFKB signaling pathway is evolutionarily conserved and plays a critical morphogenetic role in several species; however, its function during WD morphogenesis is unknown. We hypothesized that TLR4/NFKB pathway plays a role during WD development. Here we examined TLR4 expression and regulation of TLR4-target genes during rat WD morphogenesis between embryonic days (e) 17.5-20.5. The functionality of TLR4/NFKB signaling was examined using WD organotypic cultures treated with lipopolysaccharide (LPS) from E. coli (TLR4 agonist) and PDTC (NFKB inhibitor). TLR4 was detected at mRNA level in e17.5 (uncoiled duct) and e20.5 (coiled duct) WDs, and spatio-temporal changes in TLR4 immunoreactivity were observed between these two time points. Expression level analysis of a subset of TLR4-regulated genes showed that TLR4/NFKB pathway was activated after exposure of cultured WD to LPS (4 h), an event that was abrogated by PDTC. Long-term exposure of cultured WDs to LPS (96 h) resulted in dysregulations of morphogenetic events and LAMA1 immunodistribution changes, suggesting the extracellular matrix at the intersection between WD morphogenesis and balance of innate immune components. Our results unveil the epididymal morphogenesis as an event equipped with TLR4/NFKB signaling components that may serve developmental functions, and eventually transition to host defense function when the fetus is exposed to an infectious or noninfectious threat.


Asunto(s)
Epidídimo/fisiología , Morfogénesis/fisiología , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Conductos Mesonéfricos/fisiología , Animales , Células Cultivadas , Desarrollo Embrionario , Femenino , Inmunidad Innata , Lipopolisacáridos/inmunología , Masculino , Técnicas de Cultivo de Órganos , Embarazo , Ratas , Ratas Wistar , Transducción de Señal
6.
Differentiation ; 118: 41-71, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33441255

RESUMEN

Estrogen has always been considered the female hormone and testosterone the male hormone. However, estrogen's presence in the testis and deleterious effects of estrogen treatment during development have been known for nearly 90 years, long before estrogen receptors (ESRs) were discovered. Eventually it was learned that testes actually synthesize high levels of estradiol (E2) and sequester high concentrations in the reproductive tract lumen, which seems contradictory to the overwhelming number of studies showing reproductive pathology following exogenous estrogen exposures. For too long, the developmental pathology of estrogen has dominated our thinking, even resulting in the "estrogen hypothesis" as related to the testicular dysgenesis syndrome. However, these early studies and the development of an Esr1 knockout mouse led to a deluge of research into estrogen's potential role in and disruption of development and function of the male reproductive system. What is new is that estrogen action in the male cannot be divorced from that of androgen. This paper presents what is known about components of the estrogen pathway, including its synthesis and target receptors, and the need to achieve a balance between androgen- and estrogen-action in male reproductive tract differentiation and adult functions. The review focuses on what is known regarding development of the male reproductive tract, from the rete testis to the vas deferens, and examines the expression of estrogen receptors and presence of aromatase in the male reproductive system, traces the evidence provided by estrogen-associated knockout and transgenic animal models and discusses the effects of fetal and postnatal exposures to estrogens. Hopefully, there will be enough here to stimulate discussions and new investigations of the androgen:estrogen balance that seems to be essential for development of the male reproductive tract.


Asunto(s)
Andrógenos/metabolismo , Receptor alfa de Estrógeno/genética , Estrógenos/metabolismo , Testosterona/metabolismo , Andrógenos/genética , Animales , Embrión de Mamíferos , Desarrollo Embrionario/genética , Epidídimo/crecimiento & desarrollo , Epidídimo/metabolismo , Estradiol/metabolismo , Estrógenos/genética , Femenino , Genitales Masculinos , Masculino , Ratones , Ratones Noqueados/genética , Red Testicular/crecimiento & desarrollo , Red Testicular/metabolismo , Testosterona/genética
7.
Cell Death Differ ; 27(10): 2797-2809, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32332916

RESUMEN

Although the roles of the Hippo pathway in organogenesis and tumorigenesis have been well studied in multiple organs, its role in sperm maturation and male fertility has not been investigated. The initial segment (IS) of the epididymis plays a critical role in sperm maturation. IS differentiation is governed by ERK1/2, but the mechanisms of ERK1/2 activation in IS are not fully understood. Here we show that double knockout (dKO) of mammalian sterile 20-like kinases 1 and 2 (Mst1 and Mst2), homologs of Hippo in Drosophila, in the epididymal epithelium led to male infertility in mice. Sperm in the cauda epididymides of mutant mice were immotile with flagellar angulation and severely disorganized structures. Loss of Mst1/2 activated YAP and increased proliferation and cell death in all the segments of epididymis. The mutant mice showed substantially suppressed MEK/ERK signaling in the IS and failed IS differentiation. Deletion of Yap restored the reduced MEK/ERK signaling, and partially rescued the defective IS differentiation and fertility in Mst1/2 dKO mice. Our results demonstrate that YAP inhibits the MEK/ERK pathway in IS epithelial cells, and MST1/2 control IS differentiation and fertility at least partially by repressing YAP. Taken together, the Hippo pathway is essential for sperm maturation and male fertility.


Asunto(s)
Epidídimo , Células Epiteliales , Infertilidad Masculina/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Diferenciación Celular , Epidídimo/citología , Epidídimo/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Serina-Treonina Quinasa 3
8.
J Gerontol A Biol Sci Med Sci ; 74(6): 751-759, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29762647

RESUMEN

Carcinogenesis is frequently linked to genetic background, however, exposure to environmental risk factors has gained attention as the etiologic agent for several types of cancer, including prostate. The intrauterine microenvironment has been described as a preponderant factor for offspring health; and maternal exposure to insult has been linked to chronic disease in older offspring. Using a model of maternal exposure to low-protein diet (LPD; 6% protein), we demonstrated that impairment of offspring rat prostatic growth on postnatal day (PND) 21 was associated with prostate carcinogenesis in older offspring (PND 540). One explanation is that maternal LPD consumption exposed offspring to an estrogenic intrauterine microenvironment, which potentially sensitized prostate cells early during glandular morphogenesis, increasing cellular response to estrogen in older rats. The onset of accelerated prostatic growth, observed on PND 21, associated with an unbalanced estrogen/testosterone ratio and increased circulating IGF-1 in older offspring appears to contribute to the development of prostate carcinoma in groups on gestational low protein and gestational and lactational low protein diets (33 and 50%, respectively). Our study strongly indicated maternal exposure to LPD as a potential risk factor for induction of slow-growing prostate carcinogenesis in rat offspring later in life.


Asunto(s)
Carcinogénesis , Dieta con Restricción de Proteínas , Próstata/crecimiento & desarrollo , Neoplasias de la Próstata/patología , Factores de Edad , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Femenino , Hormonas/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley
9.
Reproduction ; 155(6): 467-479, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29748247

RESUMEN

The cauda epididymidis is the major sperm storage region whose androgenic supply, essential for the sperm viability, is provided by the vasculature and is dependent upon testosterone diffusion through the stromal tissue to reach the epithelial cells. We have focused our efforts on examining the regulation of this important epididymal region by evaluating the impact of the androgen disrupter cimetidine on the epithelial-stromal androgenic microenvironment. Male rats received 100 mg/kg cimetidine (CMTG) or saline (CG) for 50 days, serum testosterone levels were measured and the epididymal cauda region was processed for light and transmission electron microscopy. In the proximal cauda region, the duct diameter was measured and birefringent collagen in the stroma was quantified. TUNEL-labeled epithelial cells were quantified, and androgen receptor (AR), karyopherin alpha (KPNA) and sex hormone-binding globulin (SHBG) levels were analyzed by immunofluorescence and Western blot. CMTG showed reduced duct diameter and high number of apoptotic epithelial cells. In the epithelium, the total AR concentration and the KPNA immunoreactivity were reduced, and a weak/absent AR nuclear immunofluorescence was observed in contrast to the enhanced AR immunolabeling observed in the cytoplasm of the epithelial cells. A significant reduction of collagen and SHBG levels in the stroma was also observed. Cimetidine treatment impairs AR nuclear import in the epithelium, causing androgenic dysfunction and subsequent epithelial cell apoptosis and duct atrophy. The connective tissue atrophy and reduction of SHBG stromal levels associated with epithelial androgenic dysfunction indicate a possible role of stromal SHBG in the androgenic supply of the sperm storage region of the epididymis.


Asunto(s)
Epidídimo/metabolismo , Epidídimo/patología , Células Epiteliales/metabolismo , Receptores Androgénicos/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Espermatozoides/fisiología , Células del Estroma/metabolismo , Animales , Células Cultivadas , Células Epiteliales/patología , Masculino , Ratas , Espermatozoides/citología , Células del Estroma/patología , Testosterona/metabolismo
10.
Biol Reprod ; 99(3): 536-545, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29590317

RESUMEN

A fully functional initial segment, the most proximal region of the epididymis, is important for male fertility. Our previous study generated a mouse model to investigate the importance of initial segment function in male fertility. In that model, phosphatase and tensin homolog (Pten) was conditionally removed from the initial segment epithelium, which resulted in epithelial de-differentiation. When spermatozoa progressed through the de-differentiated epithelial duct, they developed angled flagella, suggesting compromised sperm maturation, which eventually resulted in male infertility. To understand the molecular mechanisms, by which PTEN regulates epididymal sperm maturation, we compared the transcriptome profile of the initial segment between controls and initial segment-specific Pten knockouts and revealed that water, ion, and organic solute transporter activities were one of the top molecular and cellular functions altered following loss of Pten. Alteration in protein levels and localization of several transporters following loss of Pten were also observed by immunofluorescence analysis. Epithelial cells of the initial segment from knockouts were more permeable to fluorescein isothiocyanate-dextran (4000 Da) compared to controls. Interestingly, conditional deletion of Pten from other organs also resulted in changes in transporter activity, suggesting a common role of PTEN in regulation of transporter activity. Taken together, our data support the hypothesis that loss of Pten from the initial segment epithelium results in changes in the transporting and permeability characteristics of the epithelium, which in turn altered the luminal fluid microenvironment that is so important for sperm maturation and male fertility.


Asunto(s)
Epidídimo/fisiopatología , Infertilidad Masculina/fisiopatología , Proteínas de Transporte de Membrana/fisiología , Fosfohidrolasa PTEN/deficiencia , Animales , Diferenciación Celular , Permeabilidad de la Membrana Celular/fisiología , Células Epiteliales/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Técnica del Anticuerpo Fluorescente , Masculino , Proteínas de Transporte de Membrana/análisis , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Maduración del Esperma/fisiología
11.
Dev Biol ; 438(1): 33-43, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29580943

RESUMEN

Wolffian duct morphogenesis must be highly coordinated with its specialized function of providing an optimal microenvironment for sperm maturation. Without normal Wolffian duct morphogenesis, male infertility will result. Our previous study showed that mediolateral and radial intercalation of epithelial and mesenchymal cells respectively, were major drivers of ductal elongation and were regulated by protein tyrosine kinase 7 (PTK7), a member of the planar cell polarity (PCP) non-canonical Wnt pathway. To understand the mechanism by which PTK7 regulates cell rearrangement/intercalation, we investigated the integrity of the extracellular matrix (ECM) and the activity of intracellular cytoskeleton mediators following loss of Ptk7. Abnormal assembly of nephronectin, laminin, and collagen IV at the basement membrane and fibrosis-like deposition of fibrilla collagen in the interstitium were observed in Ptk7 knockout Wolffian ducts. Further, the activity levels of RAC1 and myosin II, two cytoskeleton mediators, decreased in the Ptk7 knockout mesenchyme compared to controls. In addition, in-vitro experiments suggested that alterations of ECM and cytoskeleton mediators resulted in changes in Wolffian duct morphogenesis. When in-vitro-cultured Wolffian ducts were treated with collagenase IV, the degree of cross-linked fibrilla collagen was reduced, Wolffian duct elongation and coiling were significantly reduced, and an expanded cyst-like duct was observed. When Wolffian ducts were treated with RAC1 inhibitor NSC23766, mesenchymal fibrilla collagen was disassembled, and Wolffian duct elongation was significantly reduced. Our findings provide evidence that PTK7 regulates ECM integrity and the activity levels of RAC1 and myosin II, which in turn regulates Wolffian duct morphogenesis and therefore, epididymal function.


Asunto(s)
Morfogénesis/genética , Miosina Tipo II/metabolismo , Neuropéptidos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Conductos Mesonéfricos/embriología , Proteína de Unión al GTP rac1/metabolismo , Animales , Polaridad Celular/genética , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Laminina/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Análisis de Secuencia de ARN , Transducción de Señal
12.
Mol Cell Endocrinol ; 442: 142-152, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27989506

RESUMEN

The Wolffian duct (WD) undergoes morphological changes induced by androgens to form the epididymis, which is an organ essential for sperm maturation. Androgen action in WD epithelium involves paracrine factors of mesenchymal origin that function by still poorly understood mechanisms. Here we studied the antimicrobial ß-defensin SPAG11C as a new player in duct morphogenesis, localized prenatally in the WD mesenchyme. Organotypic culture of rat WDs and tissues from Androgen Receptor (AR) knockout mice (ARKO) were used. Our results show that androgen/AR signaling differentially regulated SPAG11C expression at mRNA and protein levels in the developing WD. WDs incubated with recombinant human SPAG11C were shorter and less coiled as a result of reduced epithelial cell proliferation, but not increased apoptosis. Our results suggested ß-defensin SPAG11C as an androgen-target required for WD morphogenesis. This highlights the multifunctional repertoire of the ß-defensin protein family and their potential contribution to the in utero environment that determines male reproductive success.


Asunto(s)
Andrógenos/farmacología , Antiinfecciosos/farmacología , Morfogénesis/efectos de los fármacos , Conductos Mesonéfricos/efectos de los fármacos , beta-Defensinas/farmacología , Animales , Antígenos de Superficie/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Epidídimo/efectos de los fármacos , Epidídimo/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Noqueados , Organogénesis/efectos de los fármacos , Ratas , Ratas Wistar , Receptores Androgénicos/metabolismo , Conductos Mesonéfricos/metabolismo
13.
Biol Reprod ; 95(1): 15, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27281706

RESUMEN

Without a fully developed and functioning initial segment, the most proximal region of the epididymis, male infertility results. Therefore, it is important to understand the development of the initial segment. During postnatal development of the epididymis, many cellular processes of the initial segment are regulated by lumicrine factors, which are produced by the testis and enter the epididymis with testicular luminal fluid. In this report, we showed that prior to Postnatal Day 15 (P15), the initial segment was lumicrine factor independent in the mouse. However, from P19 onward, lumicrine factors were essential for the proliferation and survival of initial segment epithelial cells. Therefore, P15 to P19 was a critical window that established the dependency of lumicrine factors in the initial segment epithelium. The initial segment-specific kinase activity profile, a marker of initial segment differentiation, was also established during this window. The SFK (SRC proto-oncogene family kinases), ERK pathway (known as the RAF/MEK/ERK pathway) components, and AMPK (AMP-activated protein kinases) pathway components had increased activities from P15 to P19, suggesting that lumicrine factors regulated SFK/ERK/AMPK signaling to initiate differentiation of the initial segment from P15 to P19. Compared with litter mate controls, juvenile Src null mice displayed lower levels of MAPK3/1 (mitogen-activated protein kinase 3/1) activity and a reduced level of differentiation in the initial segment epithelium, a similar phenotype resulting from inhibition of SRC activity within the window of P15 to P19. Therefore, lumicrine factor-dependent SRC activity signaling through MAPK3/1 is important for the initiation of initial segment differentiation during a critical window of development.


Asunto(s)
Epidídimo/crecimiento & desarrollo , Células Epiteliales/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal/fisiología , Testículo/crecimiento & desarrollo , Animales , Diferenciación Celular , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Epidídimo/metabolismo , Células Epiteliales/citología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Testículo/metabolismo
14.
Biol Reprod ; 95(1): 10, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27122633

RESUMEN

The mammalian epididymis is more than a highly convoluted tube divided into four regions: initial segment, caput, corpus and cauda. It is a highly segmented structure with each segment expressing its own and overlapping genes, proteins, and signal transduction pathways. Therefore, the epididymis may be viewed as a series of organs placed side by side. In this review we discuss the contributions of septa that divide the epididymis into segments and present hypotheses as to the mechanism by which septa form. The mechanisms of Wolffian duct segmentation are likened to the mechanisms of segmentation of the renal nephron and somites. The renal nephron may provide valuable clues as to how the Wolffian duct is patterned during development, whereas somitogenesis may provide clues as to the timing of the development of each segment. Emphasis is also placed upon how segments are differentially regulated, in support of the idea that the epididymis can be considered a series of multiple organs placed side by side. One region in particular, the initial segment, which consists of 2 or 4 segments in mice and rats, respectively, is unique with respect to its regulation and vascularity compared to other segments; loss of development of these segments leads to male infertility. Different ways of thinking about how the epididymis functions may provide new directions and ideas as to how sperm maturation takes place.


Asunto(s)
Epidídimo/anatomía & histología , Epidídimo/fisiología , Fertilidad/fisiología , Maduración del Esperma/fisiología , Espermatogénesis/fisiología , Animales , Humanos , Masculino
15.
Dev Biol ; 412(2): 219-33, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944093

RESUMEN

The Wolffian duct, the proximal end of the mesonephric duct, undergoes non-branching morphogenesis to achieve an optimal length and size for sperm maturation. It is important to examine the mechanisms by which the developing mouse Wolffian duct elongates and coils for without proper morphogenesis, male infertility will result. Here we show that highly proliferative epithelial cells divide in a random orientation relative to the elongation axis in the developing Wolffian duct. Convergent extension (CE)-like of cell rearrangements is required for elongating the duct while maintaining a relatively unchanged duct diameter. The Wolffian duct epithelium is planar polarized, which is characterized by oriented cell elongation, oriented cell rearrangements, and polarized activity of regulatory light chain of myosin II. Conditional deletion of protein tyrosine kinase 7 (PTK7), a regulator of planar cell polarity (PCP), from mesoderm results in loss of the PCP characteristics in the Wolffian duct epithelium. Although loss of Ptk7 does not alter cell proliferation or division orientation, it affects CE and leads to the duct with significantly shortened length, increased diameter, and reduced coiling, which eventually results in loss of sperm motility, a key component of sperm maturation. In vitro experiments utilizing inhibitors of myosin II results in reduced elongation and coiling, similar to the phenotype of Ptk7 knockout. This data suggest that PTK7 signaling through myosin II regulates PCP, which in turn ensures CE-like of cell rearrangements to drive elongation and coiling of the Wolffian duct. Therefore, PTK7 is essential for Wolffian duct morphogenesis and male fertility.


Asunto(s)
Embrión de Mamíferos/metabolismo , Morfogénesis/genética , Proteínas Tirosina Quinasas Receptoras/genética , Conductos Mesonéfricos/metabolismo , Amidas/farmacología , Animales , Embrión de Mamíferos/embriología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Femenino , Fertilidad/genética , Masculino , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Morfogénesis/efectos de los fármacos , Miosina Tipo II/metabolismo , Piridinas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Motilidad Espermática/genética , Conductos Mesonéfricos/citología , Conductos Mesonéfricos/embriología
16.
Asian J Androl ; 18(2): 323-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26763543

RESUMEN

ß-defensins are components of host defense, with antimicrobial and pleiotropic immuno-modulatory properties. Research over the last 15 years has demonstrated abundant expression of a variety of ß-defensins in the postnatal epididymis of different species. A gradient of region- and cell-specific expression of these proteins is observed in the epithelium of the postnatal epididymis. Their secretion into the luminal fluid and binding to spermatozoa as they travel along the epididymis has suggested their involvement in reproduction-specific tasks. Therefore, continuous attention has been given to various ß-defensins for their role in sperm function and fertility. Although ß-defensins are largely dependent on androgens, the underlying mechanisms regulating their expression and function in the epididymis are not well understood. Recent investigation has pointed out to a new and interesting scenario where ß-defensins emerge with a different expression pattern in the Wolffian duct, the embryonic precursor of the epididymis, as opposed to the adult epididymis, thereby redefining the concept concerning the multifunctional roles of ß-defensins in the developing epididymis. In this review, we summarize some current views of ß-defensins in the epididymis highlighting our most recent data and speculations on their role in the developing epididymis during the prenatal-to-postnatal transition, bringing attention to the many unanswered questions in this research area that may contribute to a better understanding of epididymal biology and male fertility.


Asunto(s)
Epidídimo/fisiología , beta-Defensinas/fisiología , Envejecimiento/fisiología , Animales , Epidídimo/embriología , Epidídimo/metabolismo , Feto/fisiología , Regulación de la Expresión Génica , Humanos , Masculino , Espermatozoides/fisiología , beta-Defensinas/metabolismo
17.
Asian J Androl ; 17(5): 749-55, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26112482

RESUMEN

The development of the Wolffian/epididymal duct is crucial for proper function and, therefore, male fertility. The development of the epididymis is complex; the initial stages form as a transient embryonic kidney; then the mesonephros is formed, which in turn undergoes extensive morphogenesis under the influence of androgens and growth factors. Thus, understanding of its full development requires a wide and multidisciplinary view. This review focuses on mouse models that display abnormalities of the Wolffian duct and mesonephric development, the importance of these mouse models toward understanding male reproductive tract development, and how these models contribute to our understanding of clinical abnormalities in humans such as congenital anomalies of the kidney and urinary tract (CAKUT).


Asunto(s)
Epidídimo/embriología , Mesonefro/embriología , Morfogénesis/genética , Conductos Mesonéfricos/embriología , Animales , Masculino , Ratones , Ratones Transgénicos
18.
Proc Natl Acad Sci U S A ; 111(52): 18643-8, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512490

RESUMEN

Without a fully developed initial segment, the most proximal region of the epididymis, male infertility results. Therefore, it is important to understand the development and regulation of this crucial region. In addition to distinctively high activity levels of the components of the ERK pathway, which are essential for initial-segment differentiation, the initial segment exhibits high protein and activity levels of phosphatase and tensin homolog (PTEN). To understand the role of PTEN in the regulation of the initial segment, we generated a mouse model with a conditional deletion of Pten from the epithelial cells of the proximal epididymis from postnatal day 17 (P17) onward. Shortly after Pten deletion, hypertrophy of the proximal epididymis became evident. Loss of Pten resulted in activation of the AKT (protein kinase B) pathway components from P28 onward, which in turn gradually suppressed RAF1 proto-oncogene serine/threonine kinase (RAF1)/ERK signaling through the interaction between AKT and RAF1. Consistent with progressive changes in RAF1/ERK signaling, loss of Pten progressively altered cell shape, size, organization, proliferation, and survival in the initial-segment epithelium and resulted in dedifferentiation and extensive epithelial folding. Most importantly, knockout males progressively lost fertility and became infertile from 6 to 12 mo. Spermatozoa from older knockout mice showed a lower percentage of motility and a higher percentage of flagellar angulation compared with controls, suggesting compromised sperm maturation. Therefore, under normal physiological conditions, PTEN suppresses AKT activity to maintain activation of the RAF1/ERK signaling pathway, which in turn maintains normal function of the initial segment and therefore, normal sperm maturation.


Asunto(s)
Epidídimo/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fertilidad/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Fosfohidrolasa PTEN/metabolismo , Quinasas raf/metabolismo , Animales , Proliferación Celular/genética , Forma de la Célula/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Eliminación de Gen , Hipertrofia/enzimología , Hipertrofia/genética , Hipertrofia/patología , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinasas raf/genética
19.
Biol Reprod ; 89(2): 48, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23782834

RESUMEN

The components of the extracellular signal-regulated kinase (ERK) pathway are involved in the regulation of epididymal cellular processes. Interestingly, our previous studies showed that there are two different activity levels of the ERK pathway components in the epididymal epithelium: a basal level in most regions and a higher level in the differentiated initial segment (IS). In this study we analyzed the role of fibroblast growth factor receptor substrate 2 (FRS2) in the regulation of these two levels. Two mouse models were generated. In the first model, Frs2 was deleted from epithelial cells of most epididymal regions except for the IS from the embryonic period onward. Loss of Frs2 dampened the basal activity level of the ERK pathway components, which resulted in an increase in apoptosis along the epididymal duct. This was observed during the period when FRS2 expression level was highest in wild-type epididymides. In the second model, Frs2 was deleted from the proximal epididymal epithelium from Postnatal Day 17 onward. Most of the epididymides in this model exhibited normal morphology. Loss of Frs2 in these epididymides did not affect the high activity level of the ERK pathway components in the IS. However, a subgroup of epididymides in the second model showed increased apoptosis which resulted in an abnormally shaped proximal region or development of granulomas. Therefore, data from these two models showed that FRS2 played different roles in the regulation of two activity levels of the ERK pathway components in the epididymis.


Asunto(s)
Epidídimo/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/metabolismo , Testículo/metabolismo , Animales , Epidídimo/citología , Células Epiteliales/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Testículo/citología
20.
Adv Exp Med Biol ; 763: 237-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23397628

RESUMEN

The blood-testis barrier (BTB) is known for its ability to create an immune privilege site in the seminiferous epithelium, but less is known of the blood-epididymal barrier (BEB). It is already established that the fully functional BTB and BEB are much more complex and consist of anatomical/physical (tight junctions, basolateral and apical membranes), physiological and immunological components, which are all necessary to make a functioning barrier in the testis and epididymis. However, comparative data for metazoans suggest that an effective Sertoli cell barrier is not entirely necessary for the development of germ cells during spermatogenesis or that our knowledge about the barrier structure/function in metazoans is still immature. This chapter compares the unique barrier formed by the Sertoli cells of the testis to that formed by the apical junctional complexes of the epididymal epithelium.


Asunto(s)
Barrera Hematotesticular/inmunología , Epidídimo/inmunología , Testículo/inmunología , Animales , Barrera Hematotesticular/anatomía & histología , Barrera Hematotesticular/fisiología , Diferenciación Celular , Permeabilidad de la Membrana Celular , Epidídimo/anatomía & histología , Epidídimo/fisiología , Supervivencia de Injerto/inmunología , Humanos , Tolerancia Inmunológica , Masculino , Filogenia , Epitelio Seminífero/inmunología , Epitelio Seminífero/fisiología , Células de Sertoli/inmunología , Células de Sertoli/fisiología , Células de Sertoli/ultraestructura , Maduración del Esperma , Espermatogénesis , Espermatozoides/inmunología , Espermatozoides/fisiología , Testículo/anatomía & histología , Testículo/fisiología , Uniones Estrechas/inmunología , Uniones Estrechas/fisiología , Uniones Estrechas/ultraestructura , Inmunología del Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...