Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 38(5): e23506, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38411466

RESUMO

The reserve pool of primordial follicles (PMFs) is finely regulated by molecules implicated in follicular growth or PMF survival. Anti-Müllerian hormone (AMH), produced by granulosa cells of growing follicles, is known for its inhibitory role in the initiation of PMF growth. We observed in a recent in vivo study that injection of AMH into mice seemed to induce an activation of autophagy. Furthermore, injection of AMH into mice activates the transcription factor FOXO3A which is also known for its implication in autophagy regulation. Many studies highlighted the key role of autophagy in the ovary at different stages of folliculogenesis, particularly in PMF survival. Through an in vitro approach with organotypic cultures of prepubertal mouse ovaries, treated or not with AMH, we aimed to understand the link among AMH, autophagy, and FOXO3A transcription factor. Autophagy and FOXO3A phosphorylation were analyzed by western blot. The expression of genes involved in autophagy was quantified by RT-qPCR. In our in vitro model, we confirmed the decrease in FOXO3A phosphorylation and the induction of autophagy in ovaries incubated with AMH. AMH also induces the expression of genes involved in autophagy. Interestingly, most of these genes are known to be FOXO3A target genes. In conclusion, we have identified a new role for AMH, namely the induction of autophagy, probably through FOXO3A activation. Thus, AMH protects the ovarian reserve not only by inhibiting the growth of PMFs but also by enabling their survival through activation of autophagy.


Assuntos
Hormônio Antimülleriano , Hormônios Peptídicos , Feminino , Animais , Camundongos , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/farmacologia , Folículo Ovariano , Ovário , Fator de Crescimento Transformador beta , Autofagia , Fatores de Transcrição
2.
FASEB J ; 33(1): 1278-1287, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30113879

RESUMO

The follicular ovarian reserve, constituted by primordial follicles (PMFs), is established early in life, then keeps declining regularly along reproductive life. The maintenance of a normal female reproductive function implies the presence of a vast amount of dormant PMFs. This process involves a continuous repression of PMF activation into early growing follicle through the balance between factors activating the initiation of follicular growth, mainly actors of the PI3K signaling pathway, and inhibiting factors such as anti-Müllerian hormone (AMH). Any disruption of this balance may induce follicle depletion and subsequent infertility. It has been recently proposed that cyclophosphamide (Cy), an alkylating agent commonly used for treating breast cancer, triggers PMF activation, further leading to premature ovarian insufficiency. Preventing chemotherapy-induced ovarian dysfunction might represent an interesting option for preserving optimal chances of natural or medically assisted conceptions after healing. The aim of the present study was to evaluate, in a model of Cy-treated pubertal mice, whether AMH administration might restrain PMF depletion. The counting of the total PMF number within mouse ovaries showed that recombinant AMH prevented Cy-induced PMF loss. Western blot analysis revealed activation of PI3K signaling pathway after Cy administration. After AMH injection, FOXO3A phosphorylation, a main actor of PMF activation, was significantly decreased. Taken together, these results support a protective role of AMH against Cy-induced follicular loss. We also provide evidence for a possible role of autophagy in the preservation of follicular pool reserve. Therefore, concomitant recombinant AMH administration during chemotherapy might offer a new option for preserving young patients' fertility.-Sonigo, C., Beau, I., Grynberg, M., Binart, N. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice.


Assuntos
Hormônio Antimülleriano/fisiologia , Antineoplásicos Alquilantes/farmacologia , Ciclofosfamida/farmacologia , Fertilidade/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Reserva Ovariana , Animais , Hormônio Antimülleriano/farmacologia , Autofagia , Estro/efeitos dos fármacos , Feminino , Camundongos , Folículo Ovariano/metabolismo , Fosforilação
3.
Hum Mutat ; 40(1): 25-30, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30304577

RESUMO

Primary ovarian insufficiency (POI) is a frequently occurring disease affecting women under 40 years old. Recently, we have analyzed unrelated POI women via whole exome sequencing (WES) and identified NOTCH2 mutations underlying possible functional effects. The present study involved reanalyzing of WES assays. We used in the KGN granulosa-like cell model, a synthetic gene reporter construct driving luciferase gene expression to assess the functional effects of five NOTCH2 mutations identified in POI patients. We found that NOTCH2-p.Ser1804Leu, p.Ala2316Val, and p.Pro2359Ala mutations had a functional impact on the protein's transcriptional activity. The results have demonstrated for the first time that NOTCH2 mutations contribute to POI etiology. We therefore recommend sequencing NOTCH2's open reading frame in large panels of POI patients to establish an accurate genotype-phenotype correlation. We cannot rule out the fact that patients affected by Alagille syndrome carrying NOTCH2 mutations may suffer ovarian dysfunction.


Assuntos
Predisposição Genética para Doença , Mutação de Sentido Incorreto/genética , Insuficiência Ovariana Primária/genética , Receptor Notch2/genética , Sequência de Aminoácidos , Feminino , Humanos , Receptor Notch2/química , Transcrição Gênica
4.
Genet Med ; 21(4): 930-938, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30224786

RESUMO

PURPOSE: Primary ovarian insufficiency (POI) is a frequent disorder that affects ~1% of women under 40 years of age. POI, which is characterized by the premature depletion of ovarian follicles and elevated plasma levels of follicle-stimulating hormone (FSH), leads to infertility. Although various etiological factors have been described, including chromosomal abnormalities and gene variants, most cases remain idiopathic. The aim of the present study was to identify and validate functionally new sequence variants in ATG (autophagy-related genes) leading to POI. METHODS: We have reanalyzed, in silico, the exome sequencing data from a previously reported work performed in 69 unrelated POI women. Functional experiments using a classical hallmark of autophagy, the microtubule-associated protein 1 light chain 3ß (LC3), were then used to link these genes to this lysosomal degradation pathway. RESULTS: We venture a functional link between ATG7 and ATG9A variants and POI. We demonstrated that variant ATG7 and ATG9A led to a decrease in autophagosome biosynthesis and consequently to an impairment of autophagy, a key biological process implicated in the preservation of the primordial follicles forming the ovarian reserve. CONCLUSION: Our results unveil that impaired autophagy is a novel pathophysiological mechanism involved in human POI.


Assuntos
Proteína 7 Relacionada à Autofagia/genética , Proteínas Relacionadas à Autofagia/genética , Autofagia/genética , Proteínas de Membrana/genética , Insuficiência Ovariana Primária/genética , Proteínas de Transporte Vesicular/genética , Adulto , Feminino , Hormônio Foliculoestimulante/genética , Predisposição Genética para Doença , Humanos , Mutação com Perda de Função/genética , Menopausa Precoce/genética , Insuficiência Ovariana Primária/patologia , Sequenciamento do Exoma
5.
FASEB J ; 32(9): 4791-4797, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29596024

RESUMO

Prolactin (PRL), whose principal role is regulation of lactation, is mainly synthesized and secreted by lactotroph anterior pituitary cells. Its signaling is exerted via a transmembrane PRL receptor (PRLR) expressed in a wide variety of tissues, including the anterior pituitary. Dopamine, which is secreted by tuberoinfundibular hypothalamic neurons, is the major inhibitory regulator of prolactin secretion. Although PRL is well established to stimulate hypothalamic dopamine secretion, thereby exerting a negative feedback regulation on its own release, autocrine or paracrine actions of PRL on lactotroph cells have also been suggested. Within the pituitary, PRL may inhibit both lactotroph proliferation and secretion, but in vivo evaluation of these putative functions is limited. To determine whether the autocrine actions of prolactin have a significant role in the physiologic function of lactotrophs in vivo, we examined the consequences of conditional deletion of Prlr in lactotroph cells using a novel mouse line with loxP sites flanking the Prlr gene ( Prlrlox/lox) and Cre-recombinase (Cre) expressed under the control of the pituitary-specific Prl promoter. Prlrlox/lox/Prl-Cre mice have normal PRL levels and did not develop any pituitary lactotroph adenoma, even at 20 mo of age. Nevertheless, Prlrlox/lox/Prl-Cre mice displayed an increased dopaminergic inhibitory tone compared with control Prlrlox/lox mice. These results elegantly confirm an autocrine/paracrine feedback of PRL on lactotroph cells in vivo, which can be fully compensated by an intact hypothalamic feedback system.-Bernard, V., Lamothe, S., Beau, I., Guillou, A., Martin, A., Le Tissier, P., Grattan, D., Young, J., Binart, N. Autocrine actions of prolactin contribute to the regulation of lactotroph function in vivo.


Assuntos
Comunicação Autócrina/fisiologia , Lactotrofos/metabolismo , Prolactina/metabolismo , Receptores da Prolactina/metabolismo , Animais , Hipotálamo/metabolismo , Integrases/metabolismo , Lactação/metabolismo , Camundongos Transgênicos , Hipófise/metabolismo , Adeno-Hipófise/metabolismo , Receptores da Prolactina/genética , Transdução de Sinais/fisiologia
6.
Int J Mol Sci ; 20(21)2019 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-31717833

RESUMO

Cancer treatment, such as chemotherapy, induces early ovarian follicular depletion and subsequent infertility. In order to protect gametes from the gonadotoxic effects of chemotherapy, several fertility preservation techniques-such as oocyte or embryo cryopreservation with or without ovarian stimulation, or cryopreservation of the ovarian cortex-should be considered. However, these methods may be difficult to perform, and the future use of cryopreserved germ cells remains uncertain. Therefore, improving the methods currently available and developing new strategies to preserve fertility represent major challenges in the area of oncofertility. Animal and ovarian culture models have been used to decipher the effects of different cytotoxic agents on ovarian function and several theories regarding chemotherapy gonadotoxicity have been raised. For example, cytotoxic agents might (i) have a direct detrimental effect on the DNA of primordial follicles constituting the ovarian reserve and induce apoptosis; (ii) induce a massive growth of dormant follicles, which are then destroyed; or (ii) induce vascular ovarian damage. Thanks to improvements in the understanding of the mechanisms involved, a large number of studies have been carried out to develop molecules limiting the negative impact of chemotherapy on the ovaries.


Assuntos
Antineoplásicos/efeitos adversos , Preservação da Fertilidade/métodos , Ovário/citologia , Insuficiência Ovariana Primária/induzido quimicamente , Animais , Criopreservação , Feminino , Humanos , Modelos Animais , Ovário/efeitos dos fármacos
7.
Hum Mol Genet ; 25(23): 5223-5233, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798098

RESUMO

Premature ovarian insufficiency (POI) is a clinical syndrome defined by a loss of ovarian activity before the age of 40. Its pathogenesis is still largely unknown, but increasing evidences support a genetic basis in most cases. Among these, heterozygous mutations in NOBOX, a homeobox gene encoding a transcription factor expressed specifically by oocyte and granulosa cells within the ovary, have been reported in ∼6% of women with sporadic POI. The pivotal role of NOBOX in early folliculogenesis is supported by findings in knock-out mice. Here, we report the genetic screening of 107 European women with idiopathic POI, recruited in various settings, and the molecular and functional characterization of the identified variants to evaluate their involvement in POI onset. Specifically, we report the identification of two novel and two recurrent heterozygous NOBOX variants in 7 out of 107 patients, with a prevalence of 6.5% (upper 95% confidence limit of 11.17%). Furthermore, immunolocalization, Western Blot and transcriptional assays conducted in either HEK293T or CHO cells revealed that all the studied variants (p.R44L, p.G91W, p.G111R, p.G152R, p.K273*, p.R449* and p.D452N) display variable degrees of functional impairment, including defects in transcriptional activity, autophagosomal degradation, nuclear localization or protein instability. Several variants conserve the ability to interact with FOXL2 in intracellular aggregates. Their inability to sustain gene expression, together with their likely aberrant effects on protein stability and degradation, make the identified NOBOX mutations a plausible cause of POI onset.


Assuntos
Núcleo Celular/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Menopausa Precoce/genética , Insuficiência Ovariana Primária/genética , Estabilidade Proteica , Fatores de Transcrição/genética , Adolescente , Adulto , Animais , Células CHO , Cricetulus , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Predisposição Genética para Doença , Células HEK293 , Heterozigoto , Humanos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Insuficiência Ovariana Primária/epidemiologia , Insuficiência Ovariana Primária/patologia , Agregados Proteicos/genética
8.
BMC Med Genet ; 18(1): 44, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446136

RESUMO

BACKGROUND: Spontaneous ovarian hyperstimulation syndrome (sOHSS) is a rare event occurring mostly during natural pregnancy. Among described etiologies, some activating mutations of FSH receptor (FSHR) have been identified. CASE PRESENTATION: We report hereby the case of a non-pregnant women with three episodes of sOHSS. Hormonal evaluation was normal and no pituitary adenoma was detected. However, genetic analysis identified a novel heterozygous FSHR mutation (c.1901 G > A). This R634H mutation is the first described in the cytoplasmic tail of the receptor. Functional analysis failed to reveal constitutive activity of the mutant but a decreased cAMP production in response to FSH. The weak activity of this mutant is correlated with a markedly reduced cell surface expression. CONCLUSION: Pathophysiology of non gestationnal sOHSS is still ill established. The molecular characterization of this new mutant indicates that it might not be at play. Therefore, further investigations are needed to improve knowledge of the molecular mechanism of this syndrome.


Assuntos
Citoplasma/metabolismo , Mutação , Síndrome de Hiperestimulação Ovariana/genética , Receptores do FSH/genética , Adulto , Sequência de Aminoácidos , Animais , Feminino , Humanos , Receptores do FSH/química , Homologia de Sequência de Aminoácidos
9.
Hum Reprod ; 32(7): 1512-1520, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28505269

RESUMO

STUDY QUESTION: Is it possible to identify new mutations potentially associated with non-syndromic primary ovarian insufficiency (POI) via whole-exome sequencing (WES)? SUMMARY ANSWER: WES is an efficient tool to study genetic causes of POI as we have identified new mutations, some of which lead to protein destablization potentially contributing to the disease etiology. WHAT IS KNOWN ALREADY: POI is a frequently occurring complex pathology leading to infertility. Mutations in only few candidate genes, mainly identified by Sanger sequencing, have been definitively related to the pathogenesis of the disease. STUDY DESIGN, SIZE, DURATION: This is a retrospective cohort study performed on 69 women affected by POI. PARTICIPANTS/MATERIALS, SETTING, METHODS: WES and an innovative bioinformatics analysis were used on non-synonymous sequence variants in a subset of 420 selected POI candidate genes. Mutations in BMPR1B and GREM1 were modeled by using fragment molecular orbital analysis. MAIN RESULTS AND THE ROLE OF CHANCE: Fifty-five coding variants in 49 genes potentially related to POI were identified in 33 out of 69 patients (48%). These genes participate in key biological processes in the ovary, such as meiosis, follicular development, granulosa cell differentiation/proliferation and ovulation. The presence of at least two mutations in distinct genes in 42% of the patients argued in favor of a polygenic nature of POI. LIMITATIONS, REASONS FOR CAUTION: It is possible that regulatory regions, not analyzed in the present study, carry further variants related to POI. WIDER IMPLICATIONS OF THE FINDINGS: WES and the in silico analyses presented here represent an efficient approach for mapping variants associated with POI etiology. Sequence variants presented here represents potential future genetic biomarkers. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Universidad del Rosario and Colciencias (Grants CS/CIGGUR-ABN062-2016 and 672-2014). Colciencias supported Liliana Catherine Patiño´s work (Fellowship: 617, 2013). The authors declare no conflict of interest.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Predisposição Genética para Doença , Peptídeos e Proteínas de Sinalização Intercelular/genética , Modelos Moleculares , Mutação , Insuficiência Ovariana Primária/genética , Adulto , Substituição de Aminoácidos , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/química , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Estudos de Coortes , Biologia Computacional , Sistemas Inteligentes , Feminino , França , Estudo de Associação Genômica Ampla , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único , Insuficiência Ovariana Primária/metabolismo , Estabilidade Proteica , Encaminhamento e Consulta , Estudos Retrospectivos , Sequenciamento do Exoma , Adulto Jovem
10.
FASEB J ; 30(2): 1002-10, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26567005

RESUMO

The anterior pituitary hormone prolactin exerts important physiologic actions in the brain. However, the mechanism by which prolactin crosses the blood-brain barrier and enters the brain is not completely understood. On the basis of high expression of the prolactin receptor in the choroid plexus, it has been hypothesized that the receptor may bind to prolactin in the blood and translocate it into the cerebrospinal fluid (CSF). This study aimed to test this hypothesis by investigating transport of (125)I-labeled prolactin ((125)I-prolactin) into the brain of female mice in the presence and absence of the prolactin receptor (PRLR(-/-)). Peripherally administered prolactin rapidly activates brain neurons, as evidenced by prolactin-induced phosphorylation of signal transducer and activator of transcription 5 (pSTAT5) in neurons within 30 min of administration. The transport of prolactin into the brain was saturable, with transport effectively blocked only by a very high dose of unlabeled ovine prolactin. Transport was regulated, as in lactating mice with chronically elevated levels of prolactin, the rate of (125)I-prolactin transport into the brain was significantly increased compared to nonlactating controls. There was no change in the rate of (125)I-prolactin transport into the brain in PRLR(-/-) mice lacking functional prolactin receptors compared to control mice, indicating transport is independent of the prolactin receptor. These data suggest that prolactin transport into the brain involves another as yet unidentified transporter molecule. Because CSF levels of (125)I-prolactin were very low, even up to 90 min after administration, the data suggest that CSF is not the major route by which blood prolactin gains access to neurons in the brain.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Prolactina/metabolismo , Receptores da Prolactina/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Prolactina/genética , Transporte Proteico/fisiologia , Receptores da Prolactina/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo
11.
Neuroendocrinology ; 103(6): 738-45, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26641246

RESUMO

BACKGROUND/AIMS: No genetic anomalies specifically predisposing humans to prolactinomas have so far been identified. The prolactin receptor (PRLR) is a good candidate, however, as Prlr knockout mice develop prolactinomas, and a case of familial hyperprolactinemia has been linked to PRLR mutation. The main objective of this study was to detect germline PRLR mutations in patients with sporadic prolactinomas unrelated to AIP or MEN1 mutation. METHODS: We sequenced all PRLR exons and intron-exon junctions on genomic DNA from 88 patients with a median age of 24 years. RESULTS: We identified 4 PRLR variations (p.Ile76Val, p.Ile146Leu, p.Glu108Lys and p.Glu554Gln) in 16 patients. One patient had the rare variant p.Glu554Gln in the heterozygous state. Another patient had the extremely rare p.Glu108Lys variant described here for the first time. The other 2 variants (p.Ile76Val and p.Ile146Leu) are relatively common in the general population. All these 4 variants have been functionally tested in vitro and have no effect on PRLR expression, localization and signaling after prolactin stimulation. CONCLUSION: Inactivating germline variations of PRLR are not associated with sporadic prolactinoma in this series. Nevertheless, somatic disruption of PRLR has not been excluded in this subset of pituitary tumors.


Assuntos
Mutação em Linhagem Germinativa/genética , Neoplasias Hipofisárias/genética , Prolactinoma/genética , Receptores da Prolactina/genética , Adolescente , Adulto , Análise de Variância , Animais , Células COS , Criança , Chlorocebus aethiops , Estudos de Coortes , Simulação por Computador , Células HEK293 , Humanos , Imunoprecipitação , Pessoa de Meia-Idade , Modelos Moleculares , Mutagênese Sítio-Dirigida/métodos , Receptores da Prolactina/metabolismo , Fator de Transcrição STAT5/metabolismo , Transfecção , Adulto Jovem
12.
J Neurosci ; 34(5): 1868-78, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24478366

RESUMO

Retinal degeneration is characterized by the progressive destruction of retinal cells, causing the deterioration and eventual loss of vision. We explored whether the hormone prolactin provides trophic support to retinal cells, thus protecting the retina from degenerative pressure. Inducing hyperprolactinemia limited photoreceptor apoptosis, gliosis, and changes in neurotrophin expression, and it preserved the photoresponse in the phototoxicity model of retinal degeneration, in which continuous exposure of rats to bright light leads to retinal cell death and retinal dysfunction. In this model, the expression levels of prolactin receptors in the retina were upregulated. Moreover, retinas from prolactin receptor-deficient mice exhibited photoresponsive dysfunction and gliosis that correlated with decreased levels of retinal bFGF, GDNF, and BDNF. Collectively, these data unveiled prolactin as a retinal trophic factor that may regulate glial-neuronal cell interactions and is a potential therapeutic molecule against retinal degeneration.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neuroglia/fisiologia , Prolactina/sangue , Degeneração Retiniana/prevenção & controle , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Modelos Animais de Doenças , Eletrorretinografia , Feminino , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Hiperprolactinemia/induzido quimicamente , Hiperprolactinemia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Luz/efeitos adversos , Masculino , Camundongos , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Degeneração Retiniana/complicações , Degeneração Retiniana/etiologia , Degeneração Retiniana/genética , Doenças Retinianas/genética
13.
Am J Physiol Renal Physiol ; 308(8): F799-808, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25587121

RESUMO

Unique situations in female physiology require volume retention. Accordingly, a dimorphic regulation of the thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) has been reported, with a higher activity in females than in males. However, little is known about the hormones and mechanisms involved. Here, we present evidence that estrogens, progesterone, and prolactin stimulate NCC expression and phosphorylation. The sex difference in NCC abundance, however, is species dependent. In rats, NCC phosphorylation is higher in females than in males, while in mice both NCC expression and phosphorylation is higher in females, and this is associated with increased expression and phosphorylation of full-length STE-20 proline-alanine-rich kinase (SPAK). Higher expression/phosphorylation of NCC was corroborated in humans by urinary exosome analysis. Ovariectomy in rats resulted in decreased expression and phosphorylation of the cotransporter and promoted the shift of SPAK isoforms toward the short inhibitory variant SPAK2. Conversely, estradiol or progesterone administration to ovariectomized rats restored NCC phosphorylation levels and shifted SPAK expression and phosphorylation towards the full-length isoform. Estradiol administration to male rats induced a significant increase in NCC phosphorylation. NCC is also modulated by prolactin. Administration of this peptide hormone to male rats induced increased phosphorylation of NCC, an effect that was observed even using the ex vivo kidney perfusion strategy. Our results indicate that estradiol, progesterone, and prolactin, the hormones that are involved in sexual cycle, pregnancy and lactation, upregulate the activity of NCC.


Assuntos
Estradiol/metabolismo , Rim/metabolismo , Ovário/metabolismo , Progesterona/metabolismo , Prolactina/metabolismo , Animais , Estradiol/administração & dosagem , Terapia de Reposição de Estrogênios , Feminino , Humanos , Isoenzimas , Rim/efeitos dos fármacos , Masculino , Camundongos Knockout , Ovariectomia , Fosforilação , Progesterona/administração & dosagem , Prolactina/administração & dosagem , Proteínas Serina-Treonina Quinases/metabolismo , Ratos Wistar , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Fatores Sexuais , Transdução de Sinais , Membro 3 da Família 12 de Carreador de Soluto/efeitos dos fármacos , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Regulação para Cima
14.
J Immunol ; 191(5): 2082-8, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23885109

RESUMO

Predominance of multiple sclerosis (MS) in women, reductions of disease flares during pregnancy, and their increase in the postpartum period have suggested a hormonal influence on MS activity. The hormone prolactin (PRL) has long been debated as a potential immune-stimulating factor in several autoimmune disorders, including MS and its animal model experimental autoimmune encephalomyelitis (EAE). However, to date, no data clearly ascribe a pathogenic role to PRL in these diseases. Using PRL receptor-deficient (Prlr(-/-)) and PRL-deficient (Prl(-/-)) mice, we show that PRL plays a redundant role in the development of chronic EAE. In Prlr(-/-) and Prl(-/-) mice, EAE developed with a delayed onset compared with littermate control mice, but with full clinical severity. In line with the clinical outcome, T cell proliferation and production of IFN-γ, IL-17A, and IL-6 induced by myelin Ag were delayed in Prlr(-/-) and Prl(-/-) mice. Ag-specific IgG Ab responses were not affected by PRLR or PRL deficiency. We also show that mouse lymph node cells and purified CD4(+) T cells express transcript for Prlr, but not for Prl. These results reveal that PRL does not play a central role in the development of chronic EAE and optimal Th1 and Th17 responses against myelin. Moreover, they also rule out a possible contribution of PRL secreted by immune cells to the modulation of autoreactive T cell response in this model.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Prolactina/imunologia , Animais , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prolactina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
15.
FASEB J ; 27(4): 1511-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23271057

RESUMO

As a result of advances in medical treatment, almost 80% of children who are diagnosed with cancer survive long-term. The adverse consequences of cancer treatments include impaired puberty and fertility. In prepubertal girls, the only therapeutic option is the cryopreservation of an ovary. To date, a dozen births have been reported after reimplantation of cryopreserved mature ovaries. To analyze ovarian function after immature grafts, we performed ovarian grafting in a ewe model. Fresh or cryopreserved ovaries from immature ewes were autografted in prepubertal or adult ewes. Cyclic hormonal activity was recovered 3 mo after grafting. Histological analysis demonstrated the presence of all follicle populations and corpora lutea not affected by cryopreservation. After 3 reproductive seasons, births had been observed in all groups, and the follicle-stimulating hormone status was under the limit, which indicated an exhausted ovary. As an indicator of potential imprinting default, the methylation status of the Igf2r gene was analyzed and did not show significant alteration compared with that of nonmanipulated animals. Taken together, these results demonstrate that immature ovarian grafting is able to restore spontaneous puberty and fertility and could guide the reimplantation of immature cortex in women.


Assuntos
Fertilidade/fisiologia , Hormônio Foliculoestimulante/metabolismo , Folículo Ovariano/transplante , Ovário/transplante , Animais , Criopreservação , Feminino , Ovário/fisiologia , Ovinos , Transplante Autólogo/métodos
16.
Am J Physiol Endocrinol Metab ; 305(10): E1309-18, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24064341

RESUMO

Prolactin (PRL) and placental lactogens stimulate ß-cell replication and insulin production in pancreatic islets and insulinoma cells through binding to the PRL receptor (PRLR). However, the contribution of PRLR signaling to ß-cell ontogeny and function in perinatal life and the effects of the lactogens on adaptive islet growth are poorly understood. We provide evidence that expansion of ß-cell mass during both embryogenesis and the postnatal period is impaired in the PRLR(-/-) mouse model. PRLR(-/-) newborns display a 30% reduction of ß-cell mass, consistent with reduced proliferation index at E18.5. PRL stimulates leucine incorporation and S6 kinase phosphorylation in INS-1 cells, supporting a role for ß-cell mTOR signaling in PRL action. Interestingly, a defect in the development of acini is also observed in absence of PRLR signaling, with a sharp decline in cellular size in both endocrine and exocrine compartments. Of note, a decrease in levels of IGF-II, a PRL target, in the Goto-Kakizaki (GK) rat, a spontaneous model of type 2 diabetes, is associated with a lack of PRL-mediated ß-cell proliferation in embryonic pancreatic buds. Reduced pancreatic IGF-II expression in both rat and mouse models suggests that this factor may constitute a molecular link between PRL signaling and cell ontogenesis. Together, these results provide evidence that PRL signaling is essential for pancreas ontogenesis during the critical perinatal window responsible for establishing functional ß-cell reserve.


Assuntos
Células Secretoras de Insulina/fisiologia , Pâncreas/embriologia , Prolactina/metabolismo , Receptores da Prolactina/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos , Feminino , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Knockout , Pâncreas/efeitos dos fármacos , Pâncreas/crescimento & desenvolvimento , Gravidez , Prolactina/farmacologia , Ratos , Ratos Wistar , Receptores da Prolactina/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Am J Physiol Endocrinol Metab ; 305(7): E845-52, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23921141

RESUMO

Transgenic female mice overexpressing the hCGß subunit (hCGß(+)) and producing elevated levels of luteinizing hormone (LH)/hCG bioactivity present as young adults with enhanced ovarian steroidogenesis, precocious puberty, and infertility. They subsequently develop pituitary prolactinomas, high circulating prolactin (PRL) levels, and marked mammary gland lobuloalveolar development followed by adenocarcinomas. None of these phenotypes appear in gonadectomized mice, indicating that the hCG-induced aberrations of ovarian function are responsible for the extragonadal phenotypes. PRL receptor-deficient (PRLR(-/-)) female mice are sterile, despite ovulating, due to a failure of embryo implantation, as a consequence of decreased ovarian LH receptor (Lhcgr) expression and inadequate corpus luteum formation and progesterone production. To study further the presumed permissive role of PRL in the maintenance of gonadal responsiveness to LH/hCG stimulation, we crossed the hCGß(+) and PRLR(-/-) mice. The double-mutant hCGß(+)/PRLR(-/-) females remained sterile with an ovarian phenotype similar to PRLR(-/-) mice, indicating that LH action, Lhcgr expression, and consequent luteinization are not possible without simultaneous PRL signaling. The high frequency of pituitary prolactinomas in PRLR(-/-) mice was not affected by transgenic hCGß expression. In contrast, none of the hCGß(+)/PRLR(-/-) females showed either mammary gland lobuloalveolar development or tumors, and the increased mammary gland Wnt-5b expression, possibly responsible for the tumorigenesis in hCGß(+) mice, was absent in double-mutant mice. Hence, high LH/hCG stimulation is unable to compensate for missing PRL signaling in the maintenance of luteal function. PRL thus appears to be a major permissive regulator of LH action in the ovary and of its secondary extragonadal effects.


Assuntos
Carcinogênese/metabolismo , Hormônio Luteinizante/metabolismo , Ovário/metabolismo , Prolactina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Carcinogênese/patologia , Gonadotropina Coriônica Humana Subunidade beta/genética , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Corpo Lúteo/metabolismo , Feminino , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Progesterona/metabolismo , Prolactina/sangue , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 305(7): R720-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23948778

RESUMO

Prolactin (PRL) is a potent liver mitogen and proangiogenic hormone. Here, we used hyperprolactinemic rats and PRL receptor-null mice (PRLR(-/-)) to study the effect of PRL on liver growth and angiogenesis before and after partial hepatectomy (PH). Liver-to-body weight ratio (LBW), hepatocyte and sinusoidal endothelial cell (SEC) proliferation, and hepatic expression of VEGF were measured before and after PH in hyperprolactinemic rats, generated by placing two anterior pituitary glands (AP) under the kidney capsule. Also, LBW and hepatic expression of IL-6, as well as suppressor of cytokine signaling-3 (SOCS-3), were evaluated in wild-type and PRLR(-/-) mice before and after PH. Hyperprolactinemia increased the LBW, the proliferation of hepatocytes and SECs, and VEGF hepatic expression. Also, liver regeneration was increased in AP-grafted rats and was accompanied by elevated hepatocyte and SEC proliferation, and VEGF expression compared with nongrafted controls. Lowering circulating PRL levels with CB-154, an inhibitor of AP PRL secretion, prevented AP-induced stimulation of liver growth. Relative to wild-type animals, PRLR(-/-) mice had smaller livers, and soon after PH, they displayed an approximately twofold increased mortality and elevated and reduced hepatic IL-6 and SOCS-3 expression, respectively. However, liver regeneration was improved in surviving PRLR(-/-) mice. PRL stimulates normal liver growth, promotes survival, and regulates liver regeneration by mechanisms that may include hepatic downregulation of IL-6 and upregulation of SOCS-3, increased hepatocyte proliferation, and angiogenesis. PRL contributes to physiological liver growth and has potential clinical utility for ensuring survival and regulating liver mass in diseases, injuries, or surgery of the liver.


Assuntos
Hiperprolactinemia/sangue , Interleucina-6/metabolismo , Regeneração Hepática , Fígado/irrigação sanguínea , Fígado/metabolismo , Neovascularização Fisiológica , Prolactina/sangue , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Hepatectomia , Hiperprolactinemia/imunologia , Hiperprolactinemia/patologia , Hiperprolactinemia/fisiopatologia , Fígado/imunologia , Fígado/patologia , Fígado/cirurgia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Adeno-Hipófise/metabolismo , Adeno-Hipófise/transplante , Ratos , Ratos Wistar , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
FASEB J ; 26(11): 4584-91, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22859371

RESUMO

Brown fat or brown adipose tissue (BAT), found in newborn mammals as small depots localized in the interscapular region, plays a prominent role in regulating thermogenesis perinatally. The physiological importance of functional BAT has been recently reasserted in human adults. Because myoblasts and adipoblasts emerge from a common mesodermal precursor, we investigated developmental determination and the reciprocal relationship between muscle and adipocyte commitment. Here we show that a mutant mouse defective for both Igf2 and Myod genes exhibits massive BAT hypertrophy compared with wild-type and single-mutant newborns. The increased adipocyte proliferation in BAT of double-mutant newborns was associated with overexpression of the brown fat-specific marker Ucp1. More strikingly, expression of the master key gene Prdm16 involved in the switch between myogenic and brown adipogenic lineages was drastically enhanced. We further demonstrate that concomitant Myod and Igf2 inactivation accelerates differentiation of a brown preadipocyte cell line and induces lipid accumulation and increased Ucp1 and Prdm16 expression. This in vitro approach brings additional support for the implication of both Myod and Igf2 in BAT development. These results provide the first in vivo evidence that a myogenic regulator together with a growth factor act simultaneously but through independent pathways to repress Prdm16, which opens potential therapeutic perspectives for human metabolic disorders.


Assuntos
Tecido Adiposo Marrom/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Proteína MyoD/metabolismo , Fatores de Transcrição/metabolismo , Adipócitos Marrons/citologia , Adipócitos Marrons/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos , Heterozigoto , Homozigoto , Fator de Crescimento Insulin-Like II/genética , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteína MyoD/genética , RNA Interferente Pequeno , Fatores de Transcrição/genética , Proteína Desacopladora 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA