Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 702
Filtrar
1.
Vet Res Commun ; 47(2): 849-859, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36454511

RESUMEN

Under field conditions, sedation may be required for a full assessment of the reproductive potential of farm animals. The present study aimed to investigate the effect of xylazine sedation on testicular hemodynamics (TBF), echotexture, testicular volume (TV), and circulating hormones in goats. Sixteen male Shiba goats were sedated using the recommended dose of xylazine (0.05 mg/Kg BW). Testicular hemodynamics were evaluated using color-pulsed Doppler ultrasonography before and after sedation. Echotexture of the testicular parenchyma and TV were assessed using computerized image analysis. Concentrations of testosterone, estradiol (E2), inhibin, cortisol, follicle-stimulating hormone (FSH), and luteinizing hormone (LH) were measured using radioimmunoassay. There were no effects of xylazine sedation in TBF, TV, testicular parenchyma parameters, and concentrations of testosterone, inhibin, FSH, and LH (P ˃ 0.05). However, after sedation, there was significantly (P ˂ 0.05) lower cortisol and E2 concentration (42.88 ± 6.79 ng/ml and 2.47 ± 0.58 pg/ml, respectively) than before sedation (94.89 ± 13.74 ng/ml and 8.65 ± 1.79 pg/ml, respectively). The required time to perform the full scanning of the testis was significantly lower (8.50 ± 0.38 min) after xylazine sedation compared to the non-sedated goats (25.75 ± 1.14 min). In conclusion, xylazine sedation may be practically recommended for the evaluation of TBF in goats because it did not significantly alter velocities parameters and Doppler indices of blood flow within the testicular arteries. Most plasma hormones did not significantly change; however, E2 and cortisol were significantly reduced after xylazine administration.


Asunto(s)
Testículo , Xilazina , Masculino , Animales , Testículo/diagnóstico por imagen , Testículo/irrigación sanguínea , Xilazina/farmacología , Cabras , Hidrocortisona , Hormona Luteinizante , Hormona Folículo Estimulante/farmacología , Testosterona/farmacología , Hemodinámica , Inhibinas/fisiología
2.
Mol Cell Endocrinol ; 517: 110963, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32745576

RESUMEN

Anti-Müllerian hormone (AMH/Amh) plays a role in gonadal differentiation and function across vertebrates. In zebrafish we demonstrated that Amh deficiency caused severe gonadal dysgenesis and dysfunction. The mutant gonads showed extreme hypertrophy with accumulation of early germ cells in both sexes, namely spermatogonia in the testis and primary growth oocytes in the ovary. In amh mutant females, the folliculogenesis was normal in young fish but receded progressively in adults, which was accompanied by progressive decrease in follicle-stimulating hormone (fshb) expression. Interestingly the expression of fshb increased in the pituitary of juvenile amh mutant males but decreased in adults. The upregulation of fshb in mutant male juveniles was likely one of the mechanisms for triggering gonadal hypergrowth, whereas the downregulation of fshb in adults might involve a negative feedback by gonadal inhibin. Further analysis using mutants of fshb and growth differentiation factor 9 (gdf9) provided evidence for a role of FSH in triggering ovarian hypertrophy in young female amh mutant as well. In summary, the present study provided comprehensive genetic evidence for dual roles of Amh in controlling zebrafish gonadal homeostasis and gametogenesis in both sexes. Amh suppresses proliferation or accumulation of early germ cells (spermatogonia in testis and primary growth oocytes in ovary) while promoting their exit to advanced stages, and its action may involve both endocrine and paracrine pathways.


Asunto(s)
Hormona Antimülleriana/fisiología , Gametogénesis/fisiología , Homeostasis/fisiología , Proteínas de Pez Cebra/fisiología , Activinas/fisiología , Animales , Hormona Antimülleriana/deficiencia , Hormona Antimülleriana/genética , Secuencia de Bases , Sistemas CRISPR-Cas , Retroalimentación Fisiológica , Femenino , Hormona Folículo Estimulante/biosíntesis , Hormona Folículo Estimulante/genética , Técnicas de Inactivación de Genes , Factor 9 de Diferenciación de Crecimiento/genética , Hipertrofia , Infertilidad Femenina/genética , Infertilidad Masculina/genética , Inhibinas/fisiología , Masculino , Ovario/metabolismo , Ovario/patología , Comunicación Paracrina , Adenohipófisis/metabolismo , Maduración Sexual/genética , Testículo/metabolismo , Testículo/patología , Pez Cebra
3.
Geroscience ; 41(3): 267-273, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31243699

RESUMEN

Women live longer than men in virtually all circumstances. However, a more common pattern among animals is that one sex lives longer under some conditions, the other lives longer under other conditions. In laboratory mice, interventions that extend longevity are surprisingly often sex-specific in their effects. Understanding these conditional sex differences could provide mechanistic insight into how longevity could be modulated in humans. One way that longevity can be consistently enhanced is by inhibiting reproduction or eliminating the capacity to reproduce. Thus, there appears to be a mechanistic link between gonadal activity and longevity. There also appears to be a mechanistic link between some types of neuroendocrine signaling and longevity. Combining these two observations suggest that communication between the brain and gonad is a ripe avenue for further exploring longevity-assurance mechanisms. Also, because the timing and activity of specific brain-gonad endocrine differs between the sexes, neuroendocrine linkages between the brain and gonad, particularly among the less obvious hormones such as activin and inhibin, could provide additional insight into mechanisms of sex differences in aging.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/fisiopatología , Gónadas/fisiopatología , Caracteres Sexuales , Activinas/fisiología , Animales , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Inhibinas/fisiología , Longevidad/fisiología , Masculino , Sistemas Neurosecretores/fisiología , Reproducción/fisiología
4.
Calcif Tissue Int ; 102(2): 251-264, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29353450

RESUMEN

Considerable advances in oncology over recent decades have led to improved survival, while raising concerns about long-term consequences of anticancer treatments. In patients with breast or prostate malignancies, bone health is a major issue due to the high risk of bone metastases and the frequent prolonged use of hormone therapies that alter physiological bone turnover, leading to increased fracture risk. Thus, the onset of cancer treatment-induced bone loss (CTIBL) should be considered by clinicians and recent guidelines should be routinely applied to these patients. In particular, baseline and periodic follow-up evaluations of bone health parameters enable the identification of patients at high risk of osteoporosis and fractures, which can be prevented by the use of bone-targeting agents (BTAs), calcium and vitamin D supplementation and modifications of lifestyle. This review will focus upon the pathophysiology of breast and prostate cancer treatment-induced bone loss and the most recent evidence about effective preventive and therapeutic strategies.


Asunto(s)
Antineoplásicos/efectos adversos , Huesos/efectos de los fármacos , Neoplasias de la Mama/complicaciones , Osteoporosis/inducido químicamente , Neoplasias de la Próstata/complicaciones , Activinas/fisiología , Andrógenos/fisiología , Antineoplásicos/uso terapéutico , Huesos/fisiología , Huesos/fisiopatología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/fisiopatología , Estrógenos/fisiología , Femenino , Hormona Liberadora de Gonadotropina/agonistas , Humanos , Inhibinas/fisiología , Masculino , Osteoporosis/fisiopatología , Guías de Práctica Clínica como Asunto , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/fisiopatología
5.
PLoS One ; 11(12): e0167813, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936218

RESUMEN

Inhibins are members of the TGFß superfamily, which regulate many cellular processes including differentiation, proliferation, survival and apoptosis. Although initially described as hormones regulating the hypothalamus-pituitary-gonadal axis, based on their ability to antagonize Activins, our group has recently reported that they play a role in thymocyte differentiation and survival, as well as in thymic stromal cell maturation and nTreg generation. Here, we used Inhibin knock out mice (Inhα-/-) to investigate the role of Inhibins in peripheral dendritic cell maturation and function. We first demonstrated that LPS treated Inhα+/+ bone marrow derived dendritic cells (BMDC) were capable to produce significant levels of Inhibin A. Interestingly, Inhα-/- BMDC showed reduced MHCII and CD86 upregulation and increased PD-L1 expression in response to LPS compared to Inhα+/+, which correlated with reduced ability to induce proliferation of allogeneic T cells. The "semi-mature" phenotype displayed by Inhα-/- mBMDC correlated with increased levels of IL-10 and slightly decreased IL-6 production after LPS stimulation. In addition, Inhα-/- mBMDC showed impaired migration towards CCL19 and CCL21, assessed by in vitro chemotaxis and in vivo competitive homing experiments, despite their normal CCR7 expression. Furthermore, in vivo LPS-induced DC maturation was also diminished in Inhα-/- mice, specially within the LC (CD207+ CD11b+ CD103-) subpopulation. Finally, analysis of delayed type hypersensitivity responses in Inhα-/- mice, showed reduced ear swelling as a result of reduced cellular infiltration in the skin, correlating with impaired homing of CD207+ DCs to the draining lymph nodes. In summary, our data demonstrate for the first time that Inhibins play a key role in peripheral DC maturation and function, regulating the balance between immunity and tolerance.


Asunto(s)
Diferenciación Celular/fisiología , Células Dendríticas/citología , Inhibinas/fisiología , Animales , Células Dendríticas/metabolismo , Hipersensibilidad Tardía , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Linfocitos T/citología
6.
Artículo en Inglés | MEDLINE | ID: mdl-27328872

RESUMEN

Since their original discovery as regulators of follicle-stimulating hormone (FSH) secretion and erythropoiesis, the TGF-ß family members activin and inhibin have been shown to participate in a variety of biological processes, from the earliest stages of embryonic development to highly specialized functions in terminally differentiated cells and tissues. Herein, we present the history, structures, signaling mechanisms, regulation, and biological processes in which activins and inhibins participate, including several recently discovered biological activities and functional antagonists. The potential therapeutic relevance of these advances is also discussed.


Asunto(s)
Activinas/fisiología , Inhibinas/fisiología , Activinas/química , Animales , Femenino , Humanos , Inhibinas/química , Ligandos , Conformación Proteica , Transducción de Señal
7.
Hum Reprod Update ; 22(3): 342-57, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26884470

RESUMEN

BACKGROUND: Activins are members of the pleiotrophic family of the transforming growth factor-beta (TGF-ß) superfamily of cytokines, initially isolated for their capacity to induce the release of FSH from pituitary extracts. Subsequent research has demonstrated that activins are involved in multiple biological functions including the control of inflammation, fibrosis, developmental biology and tumourigenesis. This review summarizes the current knowledge on the roles of activin in reproductive and developmental biology. It also discusses interesting advances in the field of modulating the bioactivity of activins as a therapeutic target, which would undoubtedly be beneficial for patients with reproductive pathology. METHODS: A comprehensive literature search was carried out using PUBMED and Google Scholar databases to identify studies in the English language which have contributed to the advancement of the field of activin biology, since its initial isolation in 1987 until July 2015. 'Activin', 'testis', 'ovary', 'embryonic development' and 'therapeutic targets' were used as the keywords in combination with other search phrases relevant to the topic of activin biology. RESULTS: Activins, which are dimers of inhibin ß subunits, act via a classical TGF-ß signalling pathway. The bioactivity of activin is regulated by two endogenous inhibitors, inhibin and follistatin. Activin is a major regulator of testicular and ovarian development. In the ovary, activin A promotes oocyte maturation and regulates granulosa cell steroidogenesis. It is also essential in endometrial repair following menstruation, decidualization and maintaining pregnancy. Dysregulation of the activin-follistatin-inhibin system leads to disorders of female reproduction and pregnancy, including polycystic ovary syndrome, ectopic pregnancy, miscarriage, fetal growth restriction, gestational diabetes, pre-eclampsia and pre-term birth. Moreover, a rise in serum activin A, accompanied by elevated FSH, is characteristic of female reproductive aging. In the male, activin A is an autocrine and paracrine modulator of germ cell development and Sertoli cell proliferation. Disruption of normal activin signalling is characteristic of many tumours affecting reproductive organs, including endometrial carcinoma, cervical cancer, testicular and ovarian cancer as well as prostate cancer. While activin A and B aid the progression of many tumours of the reproductive organs, activin C acts as a tumour suppressor. Activins are important in embryonic induction, morphogenesis of branched glandular organs, development of limbs and nervous system, craniofacial and dental development and morphogenesis of the Wolffian duct. CONCLUSIONS: The field of activin biology has advanced considerably since its initial discovery as an FSH stimulating agent. Now, activin is well known as a growth factor and cytokine that regulates many aspects of reproductive biology, developmental biology and also inflammation and immunological mechanisms. Current research provides evidence for novel roles of activins in maintaining the structure and function of reproductive and other organ systems. The fact that activin A is elevated both locally as well as systemically in major disorders of the reproductive system makes it an important biomarker. Given the established role of activin A as a pro-inflammatory and pro-fibrotic agent, studies of its involvement in disorders of reproduction resulting from these processes should be examined. Follistatin, as a key regulator of the biological actions of activin, should be evaluated as a therapeutic agent in conditions where activin A overexpression is established as a contributing factor.


Asunto(s)
Activinas/fisiología , Ovario/fisiología , Reproducción/fisiología , Testículo/fisiología , Activinas/química , Animales , Femenino , Folistatina/química , Folistatina/fisiología , Glicoproteínas , Humanos , Subunidades beta de Inhibinas , Inhibinas/química , Inhibinas/fisiología , Masculino , Neoplasias Ováricas , Preeclampsia , Embarazo
8.
Endocr Relat Cancer ; 22(6): R339-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26373571

RESUMEN

Epithelial ovarian cancer comprises ∼85% of all ovarian cancer cases. Despite acceptance regarding the influence of reproductive hormones on ovarian cancer risk and considerable advances in the understanding of epithelial ovarian carcinogenesis on a molecular level, complete understanding of the biologic processes underlying malignant transformation of ovarian surface epithelium is lacking. Various hypotheses have been proposed over the past several decades to explain the etiology of the disease. The role of reproductive hormones in epithelial ovarian carcinogenesis remains a key topic of research. Primary questions in the field of ovarian cancer biology center on its developmental cell of origin, the positive and negative effects of each class of hormones on ovarian cancer initiation and progression, and the role of the immune system in the ovarian cancer microenvironment. The development of the female reproductive tract is dictated by the hormonal milieu during embryogenesis. Intensive research efforts have revealed that ovarian cancer is a heterogenous disease that may develop from multiple extra-ovarian tissues, including both Müllerian (fallopian tubes, endometrium) and non-Müllerian structures (gastrointestinal tissue), contributing to its heterogeneity and distinct histologic subtypes. The mechanism underlying ovarian localization, however, remains unclear. Here, we discuss the role of reproductive hormones in influencing the immune system and tipping the balance against or in favor of developing ovarian cancer. We comment on animal models that are critical for experimentally validating existing hypotheses in key areas of endocrine research and useful for preclinical drug development. Finally, we address emerging therapeutic trends directed against ovarian cancer.


Asunto(s)
Carcinoma/etiología , Transformación Celular Neoplásica , Hormonas Esteroides Gonadales/fisiología , Gonadotropinas Hipofisarias/fisiología , Neoplasias Ováricas/etiología , Activinas/fisiología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Carcinoma/embriología , Carcinoma/inmunología , Carcinoma/fisiopatología , Carcinoma/terapia , Diferenciación Celular , Pollos , Ensayos de Selección de Medicamentos Antitumorales , Células Epiteliales/patología , Trompas Uterinas/patología , Femenino , Genes Relacionados con las Neoplasias , Genitales Femeninos/embriología , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Inmunológico/fisiopatología , Inmunoterapia , Inhibinas/fisiología , Ratones , Modelos Animales , Modelos Biológicos , Mutación , FN-kappa B/fisiología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Neoplasias Ováricas/embriología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/fisiopatología , Neoplasias Ováricas/terapia , Primates , Células del Estroma/patología , Microambiente Tumoral
9.
Reprod Biol Endocrinol ; 13: 26, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25889399

RESUMEN

BACKGROUND: This study was conducted to clarify the effect of the inhibiting action of inhibin on porcine granulosa cell proliferation and function, and to investigate the underlying intracellular regulatory molecular mechanisms. METHODS: Porcine granulosa cells were cultured in vitro, and were treated with an anti-inhibin alpha subunit antibody, with or without co-treatment of follicle-stimulating hormone (FSH) in the culture medium. RESULTS: Treatment with anti-inhibin alpha subunit antibody led to a significant increase in estradiol (E2) secretion and cell proliferation. Anti-inhibin alpha subunit antibody worked synergistically with FSH at low concentrations (25 microg/mL) to stimulate E2 secretion, but attenuated FSH action at high concentrations (50 microg/mL). Immunoneutralization of inhibin bioactivity increased FOXL2, Smad3, and PKA phosphorylation, and mRNA expression of the transcription factors CEBP and c-FOS. The expression of genes encoding gonadotropin receptors, FSHR and LHR, and of those involved in steroidogenesis, as well as IGFs and IGFBPs, the cell cycle progression factors cyclinD1 and cyclinD2, and the anti-apoptosis and anti-atresia factors Bcl2, TIMP, and ADAMTS were upregulated following anti-inhibin alpha-subunit treatment. Treatment with anti-inhibin alpha subunit down regulated expression of the pro-apoptotic gene encoding caspase3. Although expression of the pro-angiogenesis genes FN1, FGF2, and VEGF was upregulated, expression of the angiogenesis-inhibiting factor THBS1 was downregulated following anti-inhibin alpha subunit treatment. CONCLUSIONS: These results suggest that immunoneutralization of inhibin bioactivity, through augmentation of the activin and gonadotropin receptor signaling pathways and regulation of gene expression, permits the development of healthy and viable granulosa cells. These molecular mechanisms help to explain the enhanced ovarian follicular development observed following inhibin immunization in animal models.


Asunto(s)
Anticuerpos/farmacología , Proliferación Celular/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Inhibinas/fisiología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Estradiol/metabolismo , Femenino , Hormona Folículo Estimulante/farmacología , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Inhibinas/antagonistas & inhibidores , Inhibinas/inmunología , Fosforilación , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Proteína smad3/metabolismo , Porcinos/fisiología
10.
Endocr Rev ; 35(5): 747-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25051334

RESUMEN

When it was initially discovered in 1923, inhibin was characterized as a hypophysiotropic hormone that acts on pituitary cells to regulate pituitary hormone secretion. Ninety years later, what we know about inhibin stretches far beyond its well-established capacity to inhibit activin signaling and suppress pituitary FSH production. Inhibin is one of the major reproductive hormones involved in the regulation of folliculogenesis and steroidogenesis. Although the physiological role of inhibin as an activin antagonist in other organ systems is not as well defined as it is in the pituitary-gonadal axis, inhibin also modulates biological processes in other organs through paracrine, autocrine, and/or endocrine mechanisms. Inhibin and components of its signaling pathway are expressed in many organs. Diagnostically, inhibin is used for prenatal screening of Down syndrome as part of the quadruple test and as a biochemical marker in the assessment of ovarian reserve. In this review, we provide a comprehensive summary of our current understanding of the biological role of inhibin, its relationship with activin, its signaling mechanisms, and its potential value as a diagnostic marker for reproductive function and pregnancy-associated conditions.


Asunto(s)
Inhibinas/fisiología , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Inhibinas/historia
11.
Anim Reprod Sci ; 148(3-4): 97-108, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25001504

RESUMEN

Cystic ovarian disease (COD) is an important cause of infertility in dairy cattle. Although many researchers have focused their work on the endocrine changes related to this disease, evidence indicates that intraovarian components play an important role in follicular persistence. Activin, inhibin, and follistatin participate as intraovarian regulatory molecules involved in follicular cell proliferation, differentiation, steroidogenesis, oocyte maturation, and corpus luteum function. Given the importance of these factors in folliculogenesis, we examined the expression and immunolocalization of activin/inhibin ßA-subunit, inhibin α-subunit, and follistatin in the ovaries of healthy estrus-synchronized cows and in those of cows with spontaneous or adrenocorticotropic hormone (ACTH)-induced COD. We also studied inhibin B (α ßB) levels in serum and follicular fluid. We found an increased expression of the ßA-subunit of activin A/inhibin A, the α-subunit of inhibin, and follistatin in granulosa cells of spontaneous follicular cysts by immunohistochemistry, and decreased concentrations of inhibin B (α ßB) in the follicular fluid of spontaneous follicular cysts. These results, together with those previously obtained, indicate that the expression of the components of the activin-inhibin-follistatin system is altered. This could lead to multiple alterations in important functions in the ovary like the balance between pro- and anti-apoptotic factors, follicular proliferation/apoptosis, and steroidogenesis, which may contribute to the follicular persistence and endocrine changes found in cattle with COD.


Asunto(s)
Enfermedades de los Bovinos/etiología , Folistatina/fisiología , Subunidades beta de Inhibinas/fisiología , Inhibinas/fisiología , Quistes Ováricos/etiología , Hormona Adrenocorticotrópica , Animales , Sangre/metabolismo , Estudios de Casos y Controles , Bovinos , Enfermedades de los Bovinos/diagnóstico por imagen , Enfermedades de los Bovinos/metabolismo , Femenino , Líquido Folicular/metabolismo , Folistatina/metabolismo , Subunidades beta de Inhibinas/metabolismo , Inhibinas/metabolismo , Quistes Ováricos/diagnóstico por imagen , Quistes Ováricos/metabolismo , Quistes Ováricos/veterinaria , Subunidades de Proteína , Ultrasonografía
12.
Cytokine Growth Factor Rev ; 24(3): 285-95, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23541927

RESUMEN

The activins, as members of the transforming growth factor-ß superfamily, are pleiotrophic regulators of cell development and function, including cells of the myeloid and lymphoid lineages. Clinical and animal studies have shown that activin levels increase in both acute and chronic inflammation, and are frequently indicators of disease severity. Moreover, inhibition of activin action can reduce inflammation, damage, fibrosis and morbidity/mortality in various disease models. Consequently, activin A and, more recently, activin B are emerging as important diagnostic tools and therapeutic targets in inflammatory and fibrotic diseases. Activin antagonists such as follistatin, an endogenous activin-binding protein, offer considerable promise as therapies in conditions as diverse as sepsis, liver fibrosis, acute lung injury, asthma, wound healing and ischaemia-reperfusion injury.


Asunto(s)
Activinas/fisiología , Folistatina/metabolismo , Activinas/biosíntesis , Animales , Asma/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Fibrosis , Folistatina/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inhibinas/fisiología , Masculino , Cicatrización de Heridas/efectos de los fármacos
13.
Kidney Int ; 83(3): 351-3, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23446253

RESUMEN

Cultures of stem or progenitor cells have made critical contributions to the comprehension of tissue regeneration. In the kidney, primary cultures of human tubular progenitors became available only recently and allow dissection of the functional properties of tubular progenitors vs. differentiated tubular epithelia. Toll-like receptor-mediated activation now appears as a previously unknown mechanism of progenitor-mediated tubular regeneration, implying that proinflammatory factors activate regenerative processes in the kidney.


Asunto(s)
Lesión Renal Aguda/terapia , Decorina/fisiología , Inhibinas/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Riñón/citología , Trasplante de Células Madre , Receptor Toll-Like 2/fisiología , Humanos
14.
Kidney Int ; 83(3): 392-403, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23325086

RESUMEN

Acute kidney injury (AKI) is emerging as a worldwide public health problem. Recent studies have focused on the possibility of using human adult renal stem/progenitor cells (ARPCs) to improve the repair of AKI. Here we studied the influence of ARPCs on the healing of cisplatin-injured renal proximal tubular epithelial cells. Tubular, but not glomerular, ARPCs provided a protective effect promoting proliferation of surviving tubular cells and inhibiting cisplatin-induced apoptosis. The recovery effect was specific to tubular ARPCs, occurred only after damage sensing, and was completely cancelled by TLR2 blockade on tubular ARPCs. Moreover, tubular, but not glomerular, ARPCs were resistant to the apoptotic effect of cisplatin. Tubular ARPCs operate mainly through the engagement of TLR2, the secretion of inhibin-A protein, and microvesicle-shuttled decorin, inhibin-A, and cyclin D1 mRNAs. These factors worked synergistically and were essential to the repair process. The involvement of tubular ARPC-secreted inhibin-A and decorin mRNA in the pathophysiology of AKI was also confirmed in transplant patients affected by delayed graft function. Hence, identification of this TLR2-driven recovery mechanism may shed light on new therapeutic strategies to promote the recovery capacity of the kidney in acute tubular damage. Use of these components, derived from ARPCs, avoids injecting stem cells.


Asunto(s)
Lesión Renal Aguda/terapia , Decorina/fisiología , Inhibinas/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Riñón/citología , Trasplante de Células Madre , Receptor Toll-Like 2/fisiología , Adulto , Apoptosis , Proliferación Celular , Separación Celular , Cisplatino/toxicidad , Ciclina D1/fisiología , Decorina/genética , Humanos , Inhibinas/análisis , Túbulos Renales Proximales/patología , Regeneración , Receptor Toll-Like 2/antagonistas & inhibidores
15.
Theor Biol Med Model ; 9: 35, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22870942

RESUMEN

When hormones during the ovulatory cycle are shown in phase plane graphs, reported FSH and estrogen values form a specific pattern that resembles the leaning "&" symbol, while LH and progesterone (Pg) values form a "boomerang" shape. Graphs in this paper were made using data reported by Stricker et al. [Clin Chem Lab Med 2006;44:883-887]. These patterns were used to construct a simplistic model of the ovulatory cycle without the conventional "positive feedback" phenomenon. The model is based on few well-established relations:hypothalamic GnRH secretion is increased under estrogen exposure during two weeks that start before the ovulatory surge and lasts till lutheolysis.the pituitary GnRH receptors are so prone to downregulation through ligand binding that this must be important for their function.in several estrogen target tissue progesterone receptor (PgR) expression depends on previous estrogen binding to functional estrogen receptors (ER), while Pg binding to the expressed PgRs reduces both ER and PgR expression.Some key features of the presented model are here listed:High GnRH secretion induced by the recovered estrogen exposure starts in the late follicular phase and lasts till lutheolysis. The LH and FSH surges start due to combination of accumulated pituitary GnRH receptors and increased GnRH secretion. The surges quickly end due to partial downregulation of the pituitary GnRH receptors (64% reduction of the follicular phase pituitary GnRH receptors is needed to explain the reported LH drop after the surge). A strong increase in the lutheal Pg blood level, despite modest decline in LH levels, is explained as delayed expression of pituitary PgRs. Postponed pituitary PgRs expression enforces a negative feedback loop between Pg levels and LH secretions not before the mid lutheal phase.Lutheolysis is explained as a consequence of Pg binding to hypothalamic and pituitary PgRs that reduces local ER expression. When hypothalamic sensitivity to estrogen is diminished due to lack of local ERs, hypothalamus switches back to the low GnRH secretion rate, leading to low secretion of gonadotropins and to lutheolysis. During low GnRH secretion rates, previously downregulated pituitary GnRH receptors recover to normal levels and thus allow the next cycle.Possible implications of the presented model on several topics related to reproductive physiology are shortly discussed with some evolutionary aspects including the emergence of menopause.


Asunto(s)
Ciclo Menstrual/fisiología , Modelos Biológicos , Ovulación/fisiología , Activinas/fisiología , Evolución Biológica , Estrógenos/fisiología , Retroalimentación Fisiológica , Femenino , Hormona Folículo Estimulante/fisiología , Humanos , Hipotálamo/fisiología , Inhibinas/fisiología , Leptina/fisiología , Hormona Luteinizante/fisiología , Luteólisis/fisiología , Masculino , Hipófisis/fisiología , Progesterona/fisiología , Pubertad/fisiología , Receptores de Estrógenos/fisiología , Receptores LHRH/fisiología , Receptores de Progesterona/fisiología , Caracteres Sexuales
16.
Reprod Biol Endocrinol ; 10: 39, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22559970

RESUMEN

BACKGROUND: We characterized the spectrum and etiology of hypogonadism in a cohort of Prader-Willi syndrome (PWS) adolescents and adults. METHODS: Reproductive hormonal profiles and physical examination were performed on 19 males and 16 females ages 16-34 years with PWS. Gonadotropins, sex-steroids, inhibin B (INB) and anti-Mullerian hormone (AMH) were measured. We defined 4 groups according to the relative contribution of central and gonadal dysfunction based on FSH and INB levels: Group A: primary hypogonadism (FSH >15 IU/l and undetectable INB (<10 pg/ml); Group B: central hypogonadism (FSH <0.5 IU/l, INB <10 pg/ml); Group C: partial gonadal & central dysfunction (FSH 1.5-15 IU/l, INB >20 pg/ml); Group D: mild central and severe gonadal dysfunction (FSH 1.5-15 IU/l, INB < 10 pg/ml. RESULTS: There were 10, 8, 9 and 8 individuals in Groups A-D respectively; significantly more males in group A (9, 4, 4 and 2; P = 0.04). Significant differences between the groups were found in mean testosterone (P = 0.04), AMH (P = 0.003) and pubic hair (P = 0.04) in males and mean LH (P = 0.003) and breast development (P = 0.04) in females. Mean age, height, weight, BMI and the distribution of genetic subtypes were similar within the groups. CONCLUSIONS: Analysis of FSH and inhibin B revealed four distinct phenotypes ranging from primary gonadal to central hypogonadism. Primary gonadal dysfunction was common, while severe gonadotropin deficiency was rare. Longitudinal studies are needed to verify whether the individual phenotypes are consistent.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Trastornos Gonadales/etiología , Inhibinas/metabolismo , Síndrome de Prader-Willi/complicaciones , Adolescente , Adulto , Hormona Antimülleriana/sangre , Hormona Antimülleriana/metabolismo , Estudios de Cohortes , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/fisiología , Trastornos Gonadales/epidemiología , Trastornos Gonadales/fisiopatología , Gónadas/fisiopatología , Humanos , Hipogonadismo/sangre , Hipogonadismo/epidemiología , Hipogonadismo/etiología , Hipogonadismo/fisiopatología , Individualidad , Inhibinas/sangre , Inhibinas/fisiología , Masculino , Fenotipo , Síndrome de Prader-Willi/epidemiología , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología , Pubertad/sangre , Pubertad/metabolismo , Pubertad/fisiología , Transducción de Señal/fisiología , Adulto Joven
17.
Mol Cell Endocrinol ; 359(1-2): 66-77, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22406273

RESUMEN

The discovery of activin and inhibins as modulators of the hypothalamic-pituitary-gonadal axis has set the foundation for understanding their central importance to many facets of development and disease. This review contains an overview of the processes and cell types that are central to testis development and spermatogenesis and then provides an update focussed on information gathered over the past five years to address new concepts about how these proteins function to control testis development in fetal and juvenile life. Current knowledge about the interactive nature of the transforming growth factor-ß (TGFß) superfamily signalling network is applied to recent findings about activins and inhibins in the testis. Information about the regulated synthesis of signalling components and signalling regulators in the testis is integrated with new concepts that demonstrate their functional significance. The importance of activin bioactivity levels or dosage in controlling balanced growth of spermatogonial cells and their niche at different stages of testis development is highlighted.


Asunto(s)
Activinas/metabolismo , Inhibinas/metabolismo , Testículo/embriología , Testículo/crecimiento & desarrollo , Activinas/fisiología , Animales , Humanos , Inhibinas/fisiología , Masculino , Organogénesis , Transducción de Señal , Espermatogénesis , Proteínas de la Superfamilia TGF-beta/metabolismo , Proteínas de la Superfamilia TGF-beta/fisiología , Testículo/fisiología
18.
Mol Cell Endocrinol ; 359(1-2): 43-52, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22330643

RESUMEN

Activins are multifunctional proteins and members of the TGF-ß superfamily. Activins are expressed locally in most tissues and, analogous to the actions of other members of this large family of pleiotropic factors, play prominent roles in the regulation of diverse biological processes in both differentiated and embryonic stem cells. They have an essential role in maintaining tissue homeostasis in the adult and are known to contribute to the developmental programs in the embryo. Activins are further implicated in the growth and metastasis of tumor cells. Through distinct modes of action, inhibins and follistatins function as antagonists of activin and several other TGF-ß family members, including a subset of BMPs/GDFs, and modulate cellular responses and the signaling cascades downstream of these ligands. In the pituitary, the activin pathway is known to regulate key aspects of gonadotrope functions and also exert effects on other pituitary cell types. As in other tissues, activin is produced locally by pituitary cells and acts locally by exerting cell-type specific actions on gonadotropes. These local actions of activin on gonadotropes are modulated by the autocrine/paracrine actions of locally secreted follistatin and by the feedback actions of gonadal inhibin. Knowledge about the mechanism of activin, inhibin and follistatin actions is providing information about their importance for pituitary function as well as their contribution to the pathophysiology of pituitary adenomas. The aim of this review is to highlight recent findings and summarize the evidence that supports the important functions of activin, inhibin and follistatin in the pituitary.


Asunto(s)
Activinas/fisiología , Folistatina/fisiología , Gonadotrofos/metabolismo , Inhibinas/fisiología , Activinas/metabolismo , Animales , Folistatina/metabolismo , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/fisiología , Humanos , Inhibinas/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , Transducción de Señal , Proteínas de la Superfamilia TGF-beta/metabolismo , Proteínas de la Superfamilia TGF-beta/fisiología
19.
Mol Cell Endocrinol ; 359(1-2): 78-84, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21664246

RESUMEN

Inhibins A and B are gonadal factors which are important in fertility. Their use as predictors of female reproductive health has centred on their application to ovarian cancer, Anorexia Nervosa, Down Syndrome and preeclampsia. Inhibin B also provides an index of the endocrine feedback relationship between the ovary and pituitary particularly when the ovarian follicle reserve is low. These applications are relevant in monitoring the onset of the menopause transition, ovarian recovery following chemotherapy and disturbances in pubertal development. Currently, these applications have only found widespread use in Down Syndrome and ovarian cancer. Activins, on the other hand, appear to have a limited application.


Asunto(s)
Activinas/sangre , Inhibinas/sangre , Reproducción , Activinas/fisiología , Animales , Biomarcadores/sangre , Femenino , Salud , Humanos , Infertilidad Femenina/sangre , Inhibinas/fisiología , Menopausia/sangre , Folículo Ovárico/metabolismo , Folículo Ovárico/fisiopatología , Neoplasias Ováricas/sangre , Embarazo , Complicaciones del Embarazo/sangre
20.
Mol Cell Endocrinol ; 359(1-2): 53-65, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21664422

RESUMEN

Granulosa cells are the main ovarian source of inhibins, activins and activin-binding protein (follistatin) while germ (oogonia, oocytes) and somatic (theca, granulosa, luteal) cells express activin receptors, signaling components and inhibin co-receptor (betaglycan). Activins are implicated in various intra-ovarian roles including germ cell survival and primordial follicle assembly; follicle growth from preantral to mid-antral stages; suppression of thecal androgen production; promotion of granulosa cell proliferation, FSHR and CYP19A1 expression; enhancement of oocyte developmental competence; retardation of follicle luteinization and/or atresia and involvement in luteolysis. Inhibins (primarily inhibin A) are produced in greatest amounts by preovulatory follicles (and corpus luteum in primates) and suppress FSH secretion through endocrine negative feedback. Together with follistatin, inhibins act locally to oppose auto-/paracrine activin (and BMP) signaling thus modulating many of the above processes. The balance between activin-inhibin shifts during follicle development with activin signalling prevailing at earlier stages but declining as inhibin and betaglycan expression rise.


Asunto(s)
Activinas/fisiología , Inhibinas/fisiología , Folículo Ovárico/metabolismo , Receptores de Activinas/metabolismo , Activinas/metabolismo , Animales , Ciclo Estral/metabolismo , Femenino , Humanos , Inhibinas/metabolismo , Ciclo Menstrual/metabolismo , Folículo Ovárico/fisiología , Ovario/citología , Ovario/metabolismo , Ovario/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...