Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell Tissue Res ; 392(2): 581-604, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36627392

RESUMEN

Experimental autoimmune orchitis (EAO) is a chronic inflammatory disorder that causes progressive spermatogenic impairment. EAO is characterized by high intratesticular levels of nitric oxide (NO) and tumor necrosis factor alpha (TNFα) causing germ cell apoptosis and Sertoli cell dysfunction. However, the impact of this inflammatory milieu on the spermatogenic wave is unknown. Therefore, we studied the effect of inflammation on spermatogonia and preleptotene spermatocyte cell cycle progression in an EAO context and through the intratesticular DETA-NO and TNFα injection in the normal rat testes. In EAO, premeiotic germ cell proliferation is limited as a consequence of the undifferentiated spermatogonia (CD9+) cell cycle arrest in G2/M and the reduced number of differentiated spermatogonia (c-kit+) and preleptotene spermatocytes that enter in the meiotic S-phase. Although inflammation disrupts spermatogenesis in EAO, it is maintained in some seminiferous tubules at XIV and VII-VIII stages of the epithelial cell cycle, thereby guaranteeing sperm production. We found that DETA-NO (2 mM) injected in normal testes arrests spermatogonia and preleptotene spermatocyte cell cycle; this effect reduces the number of proliferative spermatogonia and the number of preleptotene spermatocytes in meiosis S-phase (36 h after). The temporal inhibition of spermatogonia clonal amplification delayed progression of the spermatogenic wave (5 days after) finally altering spermatogenesis. TNFα (0.5 and 1 µg) exposure did not affect premeiotic germ cell cycle or spermatogenic wave. Our results show that in EAO the inflammatory microenvironment altered spermatogenesis kinetics through premeiotic germ cell cycle arrest and that NO is a sufficient factor contributing to this phenomenon.


Asunto(s)
Orquitis , Factor de Necrosis Tumoral alfa , Ratas , Humanos , Animales , Masculino , Factor de Necrosis Tumoral alfa/farmacología , Semen , Espermatogénesis/fisiología , Espermatogonias , Testículo , Espermatocitos , Células de Sertoli/fisiología , Inflamación/patología
2.
Mol Hum Reprod ; 27(2)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33313783

RESUMEN

Experimental autoimmune orchitis (EAO) is a useful model to study organ-specific autoimmunity and chronic testicular inflammation. This model reflects testicular pathological changes reported in immunological infertility in men. Progression of EAO in rodents is associated with a significantly increased percentage of testicular endothelial cells and interstitial testicular blood vessels, indicating an ongoing angiogenic process. Vascular endothelial growth factor A (VEGFA), the main regulator of physiological and pathological angiogenesis, can stimulate endothelial cell proliferation, chemotaxis and vascular permeability. The aim of this study was to explore the role of VEGFA in the pathogenesis of testicular inflammation. Our results found VEGFA expression in Leydig cells, endothelial cells and macrophages in testis of rats with autoimmune orchitis. VEGFA level was significantly higher in testicular fluid and serum of rats at the end of the immunization period, preceding testicular damage. VEGF receptor (VEGFR) 1 is expressed mainly in testicular endothelial cells, whereas VEGFR2 was detected in germ cells and vascular smooth muscle cells. Both receptors were expressed in testicular interstitial cells. VEGFR2 increased after the immunization period in the testicular interstitium and VEGFR1 was downregulated in EAO testis. In-vivo-specific VEGFA inhibition by Bevacizumab prevented the increase in blood vessel number and reduced EAO incidence and severity. Our results unveil relevance of VEGFA-VEGFR axis during orchitis development, suggesting that VEGFA might be an early marker of testicular inflammation and Bevacizumab a therapeutic tool for treatment of testicular inflammation associated with subfertility and infertility.


Asunto(s)
Enfermedades Autoinmunes/patología , Neovascularización Patológica , Testículo/irrigación sanguínea , Testículo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/prevención & control , Bevacizumab/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Orquitis/inmunología , Orquitis/metabolismo , Orquitis/prevención & control , Codorniz/embriología , Ratas Wistar , Transducción de Señal , Testículo/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Reproduction ; 158(1): 109-122, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31042673

RESUMEN

Perigestational alcohol consumption up to early organogenesis can produce abnormal maternal vascularization via altered decidual VEGF/receptor expression. CF-1 female mice were administered with 10% ethanol in drinking water for 17 days prior to and up to day 10 of gestation. Control females received water without ethanol. Treated females had reduced frequency of implantation sites with expanded vascular lumen (P < 0.05), α-SMA-immunoreactive spiral arteries in proximal mesometrial decidua, reduced PCNA-positive endothelial cells (P < 0.01) and diminished uterine NK cell numbers (P < 0.05) in proximal decidua compared to controls. The VEGF expression (laser capture microscopy, RT-PCR, western blot and immunohistochemistry) was reduced in decidual tissue after perigestational alcohol consumption (P < 0.05). The uNK-DBA+ cells of treated females had reduced VEGF immunoexpression compared to controls (P < 0.01). Very low decidual and endothelial cell KDR immunoreactivity and reduced decidual gene and protein KDR expression was found in treated females compared to controls (P < 0.001). Instead, strong FLT-1 immunoexpression was detected in decidual and uNK cells (P < 0.05) in the proximal decidua from treated females compared to controls. In conclusion, perigestational alcohol ingestion induces the reduction of lumen expansion of spiral arteries, concomitant with reduced endothelial cell proliferation and uNK cell population, and uncompleted remodeling of the artery smooth muscle. These effects were supported by low decidual VEGF and KDR gene and protein expression and increased FLT-1 expression, suggesting that VEGF and KDR reduction may contribute, in part, to mechanisms involved in deficient decidual angiogenesis after perigestational alcohol consumption in mouse.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Decidua/irrigación sanguínea , Endotelio Vascular/patología , Exposición Materna/efectos adversos , Neovascularización Patológica/patología , Organogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Células Cultivadas , Decidua/efectos de los fármacos , Decidua/metabolismo , Decidua/patología , Implantación del Embrión/efectos de los fármacos , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Desarrollo Embrionario , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Ratones , Neovascularización Patológica/inducido químicamente , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
4.
J Cell Physiol ; 233(12): 9640-9651, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30054911

RESUMEN

In previous studies, we described the presence of fibroblast growth factor 2 (FGF-2) and its receptors (FGFRs) in human testis and sperm, which are involved in spermatogenesis and in motility regulation. The aim of the present study was to analyze the role of FGF-2 in the maintenance of sperm physiology using FGF-2 knockout (KO) mice. Our results showed that in wild-type (WT) animals, FGF-2 is expressed in germ cells of the seminiferous epithelium, in epithelial cells of the epididymis, and in the flagellum and acrosomal region of epididymal sperm. In the FGF-2 KO mice, we found alterations in spermatogenesis kinetics, higher numbers of spermatids per testis, and enhanced daily sperm production compared with the WT males. No difference in the percentage of sperm motility was detected, but a significant increase in sperm concentration and in sperm head abnormalities was observed in FGF-2 KO animals. Sperm from KO mice depicted reduced phosphorylation on tyrosine residues (a phenomenon that was associated with sperm capacitation) and increased acrosomal loss after incubation under capacitating conditions. However, the FGF-2 KO males displayed no apparent fertility defects, since their mating with WT females showed no differences in the time to delivery, litter size, and pup weight in comparison with WT males. Overall, our findings suggest that FGF-2 exerts a role in mammalian spermatogenesis and that the lack of FGF-2 leads to dysregulated sperm production and altered sperm morphology and function. FGF-2-deficient mice constitute a model for the study of the complex mechanisms underlying mammalian spermatogenesis.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/deficiencia , Espermatogénesis , Espermatozoides/fisiología , Animales , Peso Corporal , Epidídimo/metabolismo , Femenino , Fertilidad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Espermatozoides/ultraestructura , Testículo/metabolismo
5.
Reproduction ; 155(6): 529-541, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29626105

RESUMEN

Male chronic alcohol abuse causes testicular failure and infertility. We analyzed the effects of moderate sub-chronic alcohol intake on sperm morphology, capacitation, fertilization and sperm head decondensation. CF-1 male mice were administered 15% ethanol in drinking water for 15 days; control mice received ethanol-free water. Similar patterns of tyrosine phosphorylation were observed in capacitated spermatozoa of control and treated males. Percentage of hyperactivation (H) and spontaneous (SAR) and progesterone-induced (IAR) acrosome reaction significantly decreased at 120 and 150 min of capacitation in treated males compared to controls (H: 14.1 ± 2.5 vs 23.7 ± 2.6, P < 0.05; SAR-T120 min: 17.9 ± 2.5 vs 32.9 ± 4.1, P < 0.01; IAR-150 min: 43.3 ± 3.5 vs 73.1 ± 1.1, P < 0.001, n = 6). During in vitro fertilization (2.5, 3.5 and 4.5 h post-insemination), there was an increased percentage of fertilized oocytes (with a decondensed sperm head and one or two pronuclei) in treated males (P < 0.001, n = 7). After 60 min of in vitro decondensation with glutathione plus heparin, the percentage of decondensed sperm heads was significantly higher in treated males than in controls (mean ± s.d.: 57.1 ± 5.6 vs 48.3 ± 4.5, P < 0.05, n = 5). The percentage of morphologically normal sperm heads was significantly decreased in treated males with respect to controls (P < 0.001, n = 9). These results show that short-term moderate alcohol consumption in outbred mice affect sperm morphology, hyperactivation, acrosomal exocytosis, and the dynamics of in vitro fertilization and in vitro sperm nuclear decondensation.


Asunto(s)
Reacción Acrosómica/efectos de los fármacos , Etanol/administración & dosificación , Oocitos/efectos de los fármacos , Capacitación Espermática/efectos de los fármacos , Interacciones Espermatozoide-Óvulo/efectos de los fármacos , Animales , Antiinfecciosos Locales/administración & dosificación , Fertilización In Vitro , Masculino , Ratones , Oocitos/patología
6.
Reproduction ; 156(2): 163-172, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866768

RESUMEN

Fibroblast growth factor 2 (FGF2) and its receptors (FGFRs) have been described in several tissues, where they regulate cellular proliferation, differentiation, motility and apoptosis. Although FGF2/FGFRs expression in the male reproductive tract has been reported, there is scarce evidence on their presence in the female reproductive tract and their involvement in the modulation of sperm function. Therefore, the objective of this study was to determine the expression of FGF2 in the female reproductive tract and to assess the role of the FGF2/FGFRs system in the regulation of sperm physiology using the murine model. FGF2 was detected in uterus and oviduct protein extracts, and it was immunolocalized in epithelial cells of the uterus, isthmus and ampulla, as well as in the cumulus oophorus-oocyte complex. The receptors FGFR1, FGFR2, FGFR3 and FGFR4 were immunodetected in the flagellum and acrosomal region of sperm recovered from the cauda epididymis. Analysis of testis sections showed the expression of FGFRs in germ cells at different stages of the spermatogenesis, suggesting the testicular origin of the sperm FGFRs. Sperm incubation with recombinant FGF2 (rFGF2) led to increased sperm motility and velocity and to enhanced intracellular Ca2+ levels and acrosomal loss compared to the control. In conclusion, this study shows that FGF2 is expressed in tissues of the female reproductive tract. Also, the fact that functional FGFRs are present in mouse sperm and that rFGF2 affects sperm motility and acrosomal exocytosis, suggests the involvement of this system in the in vivo regulation of sperm function.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Genitales Femeninos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Espermatozoides/fisiología , Animales , Femenino , Masculino , Ratones , Testículo/metabolismo
7.
Mol Reprod Dev ; 84(10): 1086-1099, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28708332

RESUMEN

Perigestational alcohol consumption by CF-1 mouse, from before mating up to the period of embryo organogenesis, leads to retarded early embryo development and neural tube defects. Here, we addressed if perigestational alcohol ingestion up to Day 10 of pregnancy induces oxidative stress and changes in macromolecules and organ tissues of early organogenic embryos. Adult CF-1 female mice were administered 10% ethanol in their drinking water for 17 days prior to mating and until Day 10 of gestation, whereas control females were administered ethanol-free water. Our results demonstrated significantly reduced Catalase abundance and activity and increased glutathione content in the embryos of ethanol-treated females. The nitrite level was significantly reduced, but TBARS (thiobarbituric acid reactive substances) content, an index of lipid peroxidation, did not change. Embryos derived from ethanol-treated females also showed higher abundance of 3-nitrotyrosine (3-NT)-containing proteins in all tissues, compared to the control group. Apoptosis was significantly increased in the ectoderm and mesoderm, but not in the heart-although this organ did contain more cleaved Caspase-3-positive cardiomyocytes per area of ventricular myocardium than controls. In sum, moderate perigestational alcohol ingestion up to Day 10 of gestation in mice induces oxidative stress by altering radical nitrogen species and antioxidant enzymatic and non-enzymatic mechanisms in embryos. Further, generalized protein nitration, due to unbalanced nitric oxide levels associated with tissue-specific apoptosis, was detected in embryos, suggesting that oxidative mechanisms may play an important role in the perigestational alcohol-induced malformation of organogenic embryos exposed to ethanol.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Apoptosis/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Etanol/farmacología , Estrés Oxidativo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Animales , Animales no Consanguíneos , Antioxidantes/metabolismo , Daño del ADN/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Etanol/efectos adversos , Femenino , Ratones , Organogénesis/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología
8.
Biology (Basel) ; 13(5)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38785812

RESUMEN

Sertoli cells (SCs) are essential to maintaining germ cell development. Metformin, the main pharmacologic treatment for pediatric type 2 diabetes, is administered to children during SC maturation. The present study aimed to analyze whether metformin affects SC energy metabolism and blood-testis barrier (BTB) integrity. Primary SC cultures were used for the in vitro studies. In vivo effects were studied in Sprague-Dawley rats treated with 200 mg/kg metformin from Pnd14 to Pnd30. Metformin decreased fatty acid oxidation and increased 3-hydroxybutyrate production in vitro. Moreover, it decreased the transepithelial electrical resistance across the monolayer and induced ZO-1 redistribution, suggesting an alteration of cell junctions. In vivo, a mild but significant increase in BTB permeability and ZO-1 expression was observed in the metformin group, without changes in testicular histology and meiosis progression. Additionally, adult rats that received metformin treatment during the juvenile period showed no alteration in BTB permeability or daily sperm production. In conclusion, metformin exposure may affect BTB permeability in juvenile rats, but this seems not to influence spermatogenesis progression. Considering the results obtained in adult animals, it is possible to speculate that metformin treatment during the juvenile period does not affect testicular function in adulthood.

9.
Biology (Basel) ; 13(4)2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38666890

RESUMEN

Experimental autoimmune orchitis (EAO) is a well-established rodent model of organ-specific autoimmunity associated with infertility in which the testis immunohistopathology has been extensively studied. In contrast, analysis of testis biopsies from infertile patients associated with inflammation has been more limited. In this work, testicular biopsies from patients with idiopathic non-obstructive azoospermia diagnosed with hypospermatogenesis (HypoSp) [mild: n = 9, and severe: n = 11], with obstructive azoospermia and complete Sp (spermatogenesis) (control group, C, n = 9), and from Sertoli cell-only syndrome (SCOS, n = 9) were analyzed for the presence of immune cells, spermatogonia and Sertoli cell (SCs) alterations, and reproductive hormones levels. These parameters were compared with those obtained in rats with EAO. The presence of increased CD45+ cells in the seminiferous tubules (STs) wall and lumen in severe HypoSp is associated with increased numbers of apoptotic meiotic germ cells and decreased populations of undifferentiated and differentiated spermatogonia. The SCs showed an immature profile with the highest expression of AMH in patients with SCOS and severe HypoSp. In SCOS patients, the amount of SCs/ST and Ki67+ SCs/ST increased and correlated with high serum FSH levels and CD45+ cells. In the severe phase of EAO, immune cell infiltration and apoptosis of meiotic germ cells increased and the number of undifferentiated and differentiated spermatogonia was lowest, as previously reported. Here, we found that orchitis leads to reduced sperm number, viability, and motility. SCs were mature (AMH-) but increased in number, with Ki67+ observed in severely damaged STs and associated with the highest levels of FSH and inflammatory cells. Our findings demonstrate that in a scenario where a chronic inflammatory process is underway, FSH levels, immune cell infiltration, and immature phenotypes of SCs are associated with severe changes in spermatogenesis, leading to azoospermia. Furthermore, AMH and Ki67 expression in SCs is a distinctive marker of severe alterations of STs in human orchitis.

10.
Biol Reprod ; 87(5): 122, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23018187

RESUMEN

Inflammation of the male reproductive tract is accepted as being an important etiological factor of infertility. Experimental autoimmune orchitis (EAO) is characterized by interstitial lymphomononuclear cell infiltration and severe damage of seminiferous tubules with germ cells that undergo apoptosis and sloughing. Because the blood-testis barrier (BTB) is relevant for the protection of haploid germ cells against immune attack, the aim of this study was to analyze BTB permeability and the expression of tight junction proteins (occludin, claudin 11, and tight junction protein 1 [TJP1]) in rats during development of autoimmune orchitis. The role of IL6 as modulator of tight junction dynamics was also evaluated because intratesticular content of this cytokine is increased in EAO rats. Orchitis was induced in Sprague-Dawley adult rats by active immunization with testicular homogenate and adjuvants. Control rats (C) were injected with saline solution and adjuvants. Untreated (N) rats were also studied. Concomitant with early signs of germ cell sloughing, a reduced expression of occludin and delocalization of claudin 11 and TJP1 were detected in the testes of rats with EAO compared to C and N groups. The use of tracers showed increased BTB permeability in EAO rats. Intratesticular injection of IL6 induced focal testicular inflammation, which is associated with damaged seminiferous tubules. Rat Sertoli cells cultured in the presence of IL6 exhibited a redistribution of tight junction proteins and reduced transepithelial electrical resistance. These data indicate the possibility that IL6 might be involved in the downregulation of occludin expression and in the modulation of BTB permeability that occur in rats undergoing autoimmune orchitis.


Asunto(s)
Barrera Hematotesticular/metabolismo , Interleucina-6/farmacología , Interleucina-6/fisiología , Ocludina/metabolismo , Orquitis/inmunología , Células de Sertoli/ultraestructura , Uniones Estrechas/fisiología , Animales , Enfermedades Autoinmunes/metabolismo , Permeabilidad de la Membrana Celular , Proliferación Celular , Células Cultivadas , Masculino , Orquitis/metabolismo , Ratas , Ratas Sprague-Dawley , Testículo/química , Testículo/efectos de los fármacos , Testículo/inmunología , Uniones Estrechas/efectos de los fármacos
11.
Birth Defects Res ; 114(12): 611-630, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35775613

RESUMEN

BACKGROUND: Gestation alcohol consumption produces fetal growth restriction and malformations by affecting the embryo-fetal development. Recently a relationship between abnormal placentation and fetal malformation and intrauterine growth retardation has been suggested. However, the effects of perigestational alcohol ingestion up to early pregnancy on the placenta at term and its association with fetal abnormalities are little known. METHODS: In female mice, ethanol 10% in water was administered for 15 days previous and up to days 4 (D4), 8 (D8), or 10 (D10) of gestation (TF), and gestation continues without ethanol exposure. Control females (CF) received ethanol-free water. At day 18, feto-placental units and implantation sites were studied. RESULTS: TF had increased resorptions and only fetuses from D8-TF and D10-TF had significantly increased weights versus CF. D4 and D10-TF-placentas had significantly reduced weights. All TF had increased junctional zone (JZ) and reduced labyrinth (Lab) areas (PAS-histology and morphometry) compared with CF. Fetuses with mainly with craniofacial abnormalities and skeletal defects (Alizarin red staining), significantly increase; while the fetal bone density (alizarin color intensity, ImageJ) was reduced in D4, D8 and D10-TF versus CF. Although all TF-placentas were histo-structural affected, TF-abnormal fetuses had the most severe placental anomalies, with junctional abundant glycogenic cells into the labyrinth, disorganized labyrinthine vascularization with signs of leukocyte infiltrates and feto-maternal blood mix. CONCLUSIONS: Perigestational alcohol consumption up to early gestation induces at term fetal growth alterations, dysmorphology and defective skeleton, linked to deficient growth and abnormal morphogenesis of placenta, highlighting insight into the prenatal etiology of FASD.


Asunto(s)
Placenta , Placentación , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Etanol/efectos adversos , Femenino , Desarrollo Fetal , Retardo del Crecimiento Fetal/etiología , Humanos , Ratones , Placenta/patología , Embarazo , Agua
12.
Birth Defects Res B Dev Reprod Toxicol ; 92(6): 560-74, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21922637

RESUMEN

The aim was to study the control females (CF)-1 mouse embryo differentiation, growth, morphology on embryonic E- and N-cadherin expression at midgestation after periconceptional moderate alcohol ingestion. Adult female mice were exposed to 10% ethanol in drinking water for 17 days previous to and up to day 10 of gestation (ethanol-exposed females, EF) and were compared with nonexposed CF. EF presented reduced quantities of E10 to E10.5 embryos, greater percentage of embryos at stages less than E7.5, reduced implantation site numbers/female, and increased resorptions compared with CF. EF-embryo growth was significantly affected as evidenced by reduced cephalic and body sizes of E10 and E10.5 embryos (scanning electron microscopy) and decreased protein content of E10.5 embryos vs. CF embryos. A significantly higher percentage of EF-E10-10.5 embryos presented abnormal neural tube (NT) closure vs. the percentage of CF. E10 embryos from EF presented elevated tissue disorganization, pyknosis and nuclear condensation in somites, mesenchymal and neuroepithelial tissue. Immunohistochemical E- and N-cadherin distribution patterns were similar in organic structures of E10 embryos between groups. However, western blot revealed that E- and N-cadherin expression levels were significantly increased in EF-derived embryos vs. controls. Perigestational ethanol consumption by CF-1 mice induced significant damage in the organogenic embryogenesis by producing delayed differentiation, growth deficiencies, and increasing the frequency of NT defects. Ethanol exposure may disrupt cell-cell adhesion leading to upregulation of E- and N-cadherin expression suggesting that deregulation of cell adhesion molecules could be involved in the disruption of embryo development at organogenesis in CF-1 mouse.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Embrión de Mamíferos/efectos de los fármacos , Exposición Materna/efectos adversos , Defectos del Tubo Neural/inducido químicamente , Organogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Western Blotting , Cadherinas/metabolismo , Agua Potable , Embrión de Mamíferos/metabolismo , Femenino , Técnicas para Inmunoenzimas , Ratones , Defectos del Tubo Neural/metabolismo , Embarazo
13.
Int J Dev Biol ; 65(4-5-6): 289-299, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32930361

RESUMEN

Shaping the vertebrate eye requires evagination of the optic vesicles. These vesicles subsequently fold into optic cups prior to undergoing neurogenesis and allocating a population of late progenitors at the margin of the eye. mab21l2 encodes a protein of unknown biological function expressed in the developing optic vesicles, and loss of mab21l2 function results in malformed eyes. The bases of these defects are, however, poorly understood. To further study mab21l2 we used CRISPR/Cas9 to generate a new zebrafish mutant allele (mab21l2u517). We characterized eye morphogenesis and neurogenesis upon loss of mab21l2 function using tissue/cell-type-specific transgenes and immunostaining, in situ hybridization and bromodeoxyuridine incorporation. mab21l2u517 eyes fail to grow properly and display an excess of progenitors in the ciliary marginal zone. The expression of a transgene reporter for the vsx2 gene -a conserved marker for retinal progenitors- was delayed in mutant eyes and accompanied by disruptions in the epithelial folding that fuels optic cup morphogenesis. Mutants also displayed nasal-temporal malformations suggesting asynchronous development along that axis. Consistently, nasal retinal neurogenesis initiated but did not propagate in a timely fashion to the temporal retina. Later in development, mutant retinas did laminate and differentiate. Thus, mab21l2u517 mutants present a complex eye morphogenesis phenotype characterized by an organ-specific developmental delay. We propose that mab21l2 facilitates optic cup development with consequences both for timely neurogenesis and allocation of progenitors to the zebrafish ciliary marginal zone. These results confirm and extend previous analyses supporting the role of mab21l2 in coordinating morphogenesis and differentiation in developing eyes.


Asunto(s)
Ojo/embriología , Proteínas de Homeodominio/genética , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Neurogénesis/genética , Retina/embriología , Pez Cebra/embriología , Pez Cebra/genética
14.
Front Endocrinol (Lausanne) ; 12: 615678, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33776912

RESUMEN

It has been postulated that glyphosate (G) or its commercial formulation Roundup (R) might lead to male fertility impairment. In this study, we investigated the possible effects of G or R treatment of juvenile male rats on blood-testis barrier function and on adult male sperm production. Pups were randomly assigned to the following groups: control group (C), receiving water; G2 and G50 groups, receiving 2 and 50 mg/kg/day G respectively; and R2 and R50 groups receiving 2 and 50 mg/kg/day R respectively. Treatments were performed orally from postnatal day (PND) 14 to 30, period of life that is essential to complete a functional blood-testis barrier. Evaluation was done on PND 31. No differences in body and testis weight were observed between groups. Testis histological analysis showed disorganized seminiferous epithelium, with apparent low cellular adhesion in treated animals. Blood-testis barrier permeability to a biotin tracer was examined. A significant increase in permeable tubules was observed in treated groups. To evaluate possible mechanisms that could explain the effects on blood-testis barrier permeability, intratesticular testosterone levels, androgen receptor expression, thiobarbituric acid reactive substances (TBARS) and the expression of intercellular junction proteins (claudin11, occludin, ZO-1, connexin43, 46, and 50 which are components of the blood-testis barrier) were examined. No modifications in the above-mentioned parameters were detected. To evaluate whether juvenile exposure to G and R could have consequences during adulthood, a set of animals of the R50 group was allowed to grow up until PND 90. Histological analysis showed that control and R50 groups had normal cellular associations and complete spermatogenesis. Also, blood-testis barrier function was recovered and testicular weight, daily sperm production, and epididymal sperm motility and morphology did not seem to be modified by juvenile treatment. In conclusion, the results presented herein show that continuous exposure to low doses of G or R alters blood-testis barrier permeability in juvenile rats. However, considering that adult animals treated during the juvenile stage showed no differences in daily sperm production compared with control animals, it is feasible to think that blood-testis barrier impairment is a reversible phenomenon. More studies are needed to determine possible damage in the reproductive function of human juvenile populations exposed to low doses of G or R.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Glicina/análogos & derivados , Herbicidas/administración & dosificación , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Barrera Hematotesticular/metabolismo , Claudinas/metabolismo , Conexinas/metabolismo , Glicina/administración & dosificación , Masculino , Ocludina/metabolismo , Ratas , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Glifosato
15.
Asian J Androl ; 22(1): 112-117, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31115365

RESUMEN

The aim of this work was to study effects of ketotifen fumarate (KF) on prevention of tissue damage in testes of rats with experimental autoimmune orchitis (EAO) and on the contralateral testis in a model of prolonged testicular cord torsion (TCT). Rats with EAO or TCT were injected intraperitoneally once daily with KF or saline solution (vehicle group). Incidence and severity of testicular damage were evaluated by histopathology using an EAO score or a Johnsen score. Mast cells (MC) were identified by histochemistry and quantified. In EAO model, KF significantly reduced severity of histopathological testicular damage compared to rats in the vehicle group. KF also reduced the number of testicular MC compared to vehicle group. Similarly, in TCT model, multifocal damage of the contralateral testis was observed 30 days after testicular torsion characterized by sloughing of the germinal epithelium, seminiferous tubule atrophy, and interstitial edema. Focal signs of inflammation and fibrosis of seminiferous tubular walls were also observed. In contrast, sections of contralateral testis of rats injected with KF and killed 30 days after surgery showed normal histological features. A significant decrease in the number of MC was observed in rats treated with KF compared to untreated animals. In conclusion, we demonstrated that treatment with KF reduced testicular inflammatory process and MC infiltrates in both EAO and TCT models. The results suggest a promising treatment for infertile male patients with testicular pathologies associated with inflammation and germ cell loss.


Asunto(s)
Enfermedades Autoinmunes/patología , Epididimitis/patología , Antagonistas de los Receptores Histamínicos H1/farmacología , Cetotifen/farmacología , Mastocitos/efectos de los fármacos , Orquitis/patología , Torsión del Cordón Espermático/patología , Testículo/efectos de los fármacos , Animales , Enfermedades Autoinmunes/inmunología , Recuento de Células , Epidídimo/efectos de los fármacos , Epidídimo/inmunología , Epidídimo/patología , Epididimitis/inmunología , Hipersensibilidad Tardía , Inmunidad Celular/efectos de los fármacos , Masculino , Mastocitos/inmunología , Mastocitos/patología , Orquitis/inmunología , Ratas , Índice de Severidad de la Enfermedad , Torsión del Cordón Espermático/inmunología , Testículo/inmunología , Testículo/patología , Vacunación
16.
Toxicol In Vitro ; 62: 104682, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31626902

RESUMEN

Roundup (R), a formulation that contains glyphosate (G) as the active ingredient, is a commonly used nonselective herbicide that has been proposed to affect male fertility. It is well known that an adequate Sertoli cell function is essential to maintain germ cell development. The aim of the present study was to analyze whether G and R are able to affect Sertoli cell functions, such as energy metabolism and blood-testis barrier (BTB) integrity. Sertoli cell cultures from 20-day-old rats were exposed to 10 and 100 ppm of G or R, doses which do not decrease cell viability. Neither G nor R caused impairment in lactate production or fatty acid oxidation. G and R decreased Transepithelial Electrical Resistance, which indicates the establishment of a Sertoli cell junction barrier. However, neither G nor R modified the expression of claudin11, ZO1 and occludin, proteins that constitute the BTB. Analysis of cellular distribution of claudin11 by immunofluorescence showed that G and R induced a delocalization of the signal from membrane to the cytoplasm. The results suggest that G and R could alter an important function of Sertoli cell such as BTB integrity and thus they could compromise the normal development of spermatogenesis.


Asunto(s)
Glicina/análogos & derivados , Herbicidas/toxicidad , Células de Sertoli/efectos de los fármacos , Animales , Barrera Hematotesticular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Claudinas/biosíntesis , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Glicina/toxicidad , Uniones Intercelulares/efectos de los fármacos , Ácido Láctico/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Espermatogénesis/efectos de los fármacos , Glifosato
17.
Sci Rep ; 9(1): 15919, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685866

RESUMEN

Male meiotic germ cell including the spermatozoa represent a great challenge to the immune system, as they appear long after the establishment of normal immune tolerance mechanisms. The capacity of the testes to tolerate autoantigenic germ cells as well as survival of allogeneic organ engrafted in the testicular interstitium have led to consider the testis an immunologically privileged site. Disruption of this immune privilege following trauma, tumor, or autoimmune orchitis often results in male infertility. Strong evidence indicates that indoleamine 2,3-dioxygenase (IDO) has been implicated in fetal and allograft tolerance, tumor immune resistance, and regulation of autoimmune diseases. IDO and tryptophan 2,3-dioxygenase (TDO) catalyze the same rate-limiting step of tryptophan metabolism along a common pathway, which leads to tryptophan starvation and generation of catabolites collectively known as kynurenines. However, the relevance of tryptophan metabolism in testis pathophysiology has not yet been explored. Here we assessed the in vivo role of IDO/TDO in experimental autoimmune orchitis (EAO), a model of autoimmune testicular inflammation and immunologically impaired spermatogenesis. EAO was induced in adult Wistar rats with testicular homogenate and adjuvants. Control (C) rats injected with saline and adjuvants and normal untreated rats (N) were also studied. mRNA expression of IDO decreased in whole testes and in isolated Sertoli cells during EAO. TDO and IDO localization and level of expression in the testis were analyzed by immunostaining and Western blot. TDO is expressed in granulomas from EAO rats, and similar protein levels were observed in N, C, and EAO groups. IDO was detected in mononuclear and endothelial cells and reduced IDO expression was detected in EAO group compared to N and C rats. This phenomenon was concomitant with a significant reduction of IDO activity in EAO testis measured by tryptophan and kynurenine concentrations (HPLC). Finally, in vivo inhibition of IDO with 1-methyl-tryptophan increased severity of the disease, demonstrating down regulation of IDO-based tolerance when testicular immune regulation was disrupted. We present evidence that an IDO-based mechanism is involved in testicular immune privilege.


Asunto(s)
Privilegio Inmunológico , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Testículo/enzimología , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Epidídimo/patología , Privilegio Inmunológico/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Quinurenina/análisis , Ganglios Linfáticos/enzimología , Ganglios Linfáticos/metabolismo , Masculino , Orquitis/metabolismo , Orquitis/patología , Ratas , Ratas Wistar , Células de Sertoli/citología , Células de Sertoli/metabolismo , Índice de Severidad de la Enfermedad , Testículo/metabolismo , Testículo/patología , Triptófano/análogos & derivados , Triptófano/análisis , Triptófano/metabolismo , Triptófano/farmacología , Triptófano Oxigenasa/genética , Triptófano Oxigenasa/metabolismo
18.
Reprod Biol ; 19(4): 329-339, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31757605

RESUMEN

During an inflammatory process of the testis, the network of somatic, immune, and germ cell interactions is altered leading to organ dysfunction. In testicular biopsies of infertile men, spermatogenesis impairment is associated with reduced spermatogonia proliferation, increased number of immune cells, and content of pro-inflammatory cytokines. TNFα-TNFR and nitric oxide (NO)-NO synthase systems are up-regulated in models of testicular damage and in human testis with maturation arrest. The purpose of this study was to test the hypothesis that TNFα-TNFR system and NO alter the function of spermatogonia in the inflamed testis. We studied the effect of TNFα and NO on GC-1 spermatogonia cell cycle progression and death by flow cytometry. GC-1 cells expressed TNFR1 and TNFR2 (immunofluorescence). TNFα (10 and 50 ng/ml) and DETA-Nonoate (0.5 and 2 mM), a NO releaser, increased the percentage of cells in S-phase of the cell cycle and reduced the percentage in G1, inducing also cell apoptosis. TNFα effect was not mediated by oxidative stress unlike NO, since the presence of N-acetyl-l-cysteine (2.5 and 5.0 mM) prevented NO induced cell cycle arrest and death. GC-1 spermatogonia overpass NO induced cell cycle arrest but no TNFα, since after removal of NO, spermatogonia progressed through the cell cycle. We propose TNFα and NO might contribute to impairment of spermatogenesis by preventing adequate functioning of the spermatogonia population. Our results showed that TNFα and NO impaired spermatogonia cell cycle, inducing GC-1 arrest in the S phase.


Asunto(s)
Inflamación/fisiopatología , Óxido Nítrico/fisiología , Espermatogonias/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Apoptosis , Ciclo Celular , Línea Celular , Humanos , Masculino , Estrés Oxidativo , Receptores del Factor de Necrosis Tumoral/metabolismo , Espermatogénesis
19.
Reprod Toxicol ; 22(1): 77-86, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16678996

RESUMEN

This study investigated the effect of DEHP exposure on N-cadherin and alpha-, beta- and p120-catenin immunoreactivities in the rat testis. DEHP was administered by daily gavage to 25-day-old male Sprague-Dawley rats at a dose of 2 g DEHP/5 ml corn oil/kg body weight for 2 days or 7 days. Control rats were treated with corn oil vehicle under the same conditions. Animals were killed at 24h after the last treatment. Another group of rats treated with DEHP or corn oil vehicle (control group) for 2 days were held for 30 days without treatment to observe recovery. Testes were analyzed for histopathology, TUNEL staining, immunofluorescence (IF) and Western blot analyses. Animals exposed to DEHP for 2 days or 7 days showed severe alterations of seminiferous tubules characterized by germ cell sloughing. Animals from the longer term recovery group treated with DEHP showed foci of delayed spermatogenesis. A linear and continuous pattern of N-cadherin was observed in the basal compartment of the seminiferous tubules. A similar pattern but with higher IF intensity was observed for N-cadherin in rats treated with DEHP for 2 days or 7 days, compared to control animals. The alpha-, beta- and p120-catenins were detected in the basal compartment of seminiferous tubules in similar localization and IF pattern for DEHP and control groups. A significant increase in testicular N-cadherin and alpha-catenin levels was detected by Western blot analysis in DEHP-exposed versus control rats. No variations in N-cadherin or catenin expression were detected in the recovery groups. These findings demonstrate that DEHP induces an up-regulation of N-cadherin and alpha-catenin expression and may perturb cell-cell adhesion phenomena in the seminiferous tubule.


Asunto(s)
Cadherinas/metabolismo , Cateninas/metabolismo , Dietilhexil Ftalato/toxicidad , Células de Sertoli/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Peso Corporal/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Dietilhexil Ftalato/administración & dosificación , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Intubación Gastrointestinal , Masculino , Microscopía Electrónica de Transmisión , Tamaño de los Órganos/efectos de los fármacos , Fosfoproteínas/metabolismo , Plastificantes/administración & dosificación , Plastificantes/toxicidad , Ratas , Ratas Sprague-Dawley , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/patología , Células de Sertoli/efectos de los fármacos , Células de Sertoli/ultraestructura , Espermatocitos/efectos de los fármacos , Espermatocitos/patología , Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Espermatogonias/patología , Testículo/efectos de los fármacos , Testículo/patología , Testículo/ultraestructura , Catenina delta
20.
Reprod Toxicol ; 58: 203-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26498383

RESUMEN

We analyzed the potential role of oxidative stress induced by mono (2-ethylhexyl) phthalate (MEHP) in adherent cell junction protein expression of prepubertal rat Sertoli cells (SC) in vitro. Five-day SC cultures were treated with MEHP (200µM) for 24h and compared to cells in basal conditions. Western blot and immunofluorescent (IF) analyses showed that MEHP induced increase of N-cadherin and catenin expression, modifying its distribution. Concomitantly, Cx-43 expression decreased significantly and delocalization of the IF signal for tight junction proteins (occludin, claudin-11 and ZO-1) occurred. Indicative of oxidative stress, MEHP induced in SC an increase of lipoperoxides, a decrease in glutathione (GSH) levels and a concomitant increase in Glutathione S-Transferases (GST) activity. Antioxidant N-acetyl-cysteine (1mM) treatment prevented GSH decrease and N-cadherin and α-catenin up-regulation induced by MEHP. Our data suggest that oxidative stress signaling is a mechanism involved in adherent cell junctions disruption induced by MEHP in SC cultures.


Asunto(s)
Dietilhexil Ftalato/análogos & derivados , Uniones Intercelulares/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células de Sertoli/efectos de los fármacos , Animales , Antioxidantes/farmacología , Cadherinas/metabolismo , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Claudina-1/metabolismo , Conexina 43/metabolismo , Citoprotección , Dietilhexil Ftalato/toxicidad , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Uniones Intercelulares/metabolismo , Uniones Intercelulares/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/metabolismo , Ocludina/metabolismo , Ratas Sprague-Dawley , Células de Sertoli/metabolismo , Células de Sertoli/patología , Proteína de la Zonula Occludens-1/metabolismo , alfa Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA