Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 428
Filtrar
1.
Reprod Toxicol ; 106: 25-31, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34597818

RESUMO

Phthalates are a family of chemicals that can be found in plastic and personal care products used by consumers every day and they are known endocrine disrupting chemicals that can disrupt female reproduction. In previous studies, an environmentally relevant phthalate mixture was shown to affect female reproduction in a transgenerational manner. However, limited information was available on the effect of phthalate mixtures on ovarian steroidogenesis and folliculogenesis. Ovarian steroidogenesis is important for producing hormones needed for reproduction and ovarian regulation, and folliculogenesis is essential for the development of ovarian follicles and successful fertility. Thus, this study tested the hypothesis that prenatal exposure to an environmentally relevant phthalate mixture adversely affects ovarian steroidogenesis and folliculogenesis in the F1 generation of adult female mice. Pregnant dams (F0 generation) were orally dosed with vehicle control or a phthalate mixture (20 µg/kg/day-500 mg/kg/day) daily from gestational day 10 to birth, and the adult F1 females were the offspring of the dosed dams. The ovaries of the F1 generation were collected at postnatal day 60. One ovary was used for histological examination of the numbers and percent of different follicle types. The other ovary was used to measure expression of theca and granulosa cell enzymes. Additionally, sera were collected for measuring hormone levels. The results indicate that prenatal exposure to the phthalate mixture decreases hormone levels and gene expression, alters the transitioning of follicle types, and leads to a higher incidence of atresia in the F1 generation offspring.


Assuntos
Poluentes Ambientais/toxicidade , Hormônios Esteroides Gonadais/biossíntese , Folículo Ovariano/efeitos dos fármacos , Ácidos Ftálicos/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Hormônio Foliculoestimulante/biossíntese , Masculino , Camundongos , Folículo Ovariano/fisiologia , Gravidez
2.
Toxicol Ind Health ; 36(12): 1010-1018, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33190593

RESUMO

Exposure to an electromagnetic field (EMF) can have adverse effects on many organs and tissues, including the reproductive system. This study aimed to investigate the effects of EMF exposure during prenatal and postnatal periods on ovarian development in rat offspring. In this study, rat pups born from eight pregnant rats were used. EMF exposure was initiated on the first day of pregnancy and continued until the 42nd postnatal day. The blood and ovarian tissue samples of female offspring in sham and EMF groups were collected when they reached the age of 42 days. Follicle-stimulating hormone levels were significantly higher in the EMF group than in the sham group. Estradiol levels were significantly lower in the EMF group than in the sham group. Tissue-inducible nitric oxide synthase (iNOS) levels and expression were significantly greater in the EMF group than in the sham group. In the EMF group, congestion, bleeding areas, and degeneration of follicle structures were observed in ovarian tissue. The findings suggest that exposure to 50-Hz, 3-mT EMF used in this study during prenatal and postnatal periods may lead to impaired ovarian structure and function in female offspring. EMF may affect ovarian physiology by increasing iNOS levels and may lead to fertility disorders.


Assuntos
Campos Eletromagnéticos/efeitos adversos , Ovário/efeitos da radiação , Efeitos Tardios da Exposição Pré-Natal/veterinária , Animais , Animais Recém-Nascidos , Estradiol/biossíntese , Feminino , Hormônio Foliculoestimulante/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Gravidez , Ratos , Ratos Sprague-Dawley
3.
J Mol Endocrinol ; 65(4): 135-148, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33048061

RESUMO

The pituitary gland functions as a prominent regulator of diverse physiologic processes by secreting multiple hormones. Circular RNAs (circRNAs) are an emerging novel type of endogenous noncoding RNA that have recently been recognized as powerful regulators participating in various biological processes. However, the physiological roles and molecular mechanisms of circRNAs in pituitary remain largely unclear. Herein, we concentrated on expounding the biological function and molecular mechanism of circRNA in rat pituitary. In this study, we identified a novel circRNA in pituitary tissue, circAkap17b, which was pituitary- and stage-specific. Then, we designed circAkap17b siRNA and constructed an overexpression plasmid to evaluate the effect of loss- and gain-of-circAkap17b function on FSH secretion. Interestingly, silencing circAkakp17b significantly inhibited FSH expression and secretion, while overexpression of circAkap17b enhanced FSH expression and secretion. Furthermore, dual luciferase reporter and RNA immunoprecipitation (RIP) assays confirmed that circAkap17b could serve as miR-7 sponge to regulate target genes. Additionally, miR-7b suppressed FSH expression and secretion by directly targeting Fshb through the dual luciferase reporter and RT-qPCR analysis. Additionally, rescue experiments showed that circAkap17b could regulate FSH secretion in pituitary cells through a circAkap17b-miR-7-Fshb axis. Collectively, we demonstrated that circAkap17b could act as a molecular sponge of miR-7 to upregulate expression of the target gene Fshb and facilitate FSH secretion. These findings provide evidence for a novel regulatory role of circRNAs in pituitary.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Hipófise/metabolismo , RNA Circular/genética , Animais , Células Cultivadas , Hipófise/citologia , Ratos , Reprodução/genética
4.
Mol Cell Endocrinol ; 517: 110963, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32745576

RESUMO

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.


Assuntos
Hormônio Antimülleriano/fisiologia , Gametogênese/fisiologia , Homeostase/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Ativinas/fisiologia , Animais , Hormônio Antimülleriano/deficiência , Hormônio Antimülleriano/genética , Sequência de Bases , Sistemas CRISPR-Cas , Retroalimentação Fisiológica , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Técnicas de Inativação de Genes , Fator 9 de Diferenciação de Crescimento/genética , Hipertrofia , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Inibinas/fisiologia , Masculino , Ovário/metabolismo , Ovário/patologia , Comunicação Parácrina , Adeno-Hipófise/metabolismo , Maturidade Sexual/genética , Testículo/metabolismo , Testículo/patologia , Peixe-Zebra
5.
Sci Rep ; 10(1): 12840, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32732973

RESUMO

Currently, the generation of cell lines for the production of recombinant proteins has the limitation of unstable gene expression due to the repeat-induced gene silencing or the loss of transgene copies resulting from recombination events. In this work, we developed a new strategy based on the sequential insertion of transgenes for generating stable clones producing high levels of a chimeric human follicle-stimulating hormone (hscFSH). Gene insertion was done by transducing HEK-293 cells with a lentiviral vector containing a bicistronic transcriptional unit for expressing hscFSH and GFP genes. Clone selection was performed by flow cytometry coupled to cell sorting, and the GFP gene was further removed by CRE-mediated site-specific recombination. High-producing clones of hscFSH were obtained after three rounds of lentiviral transduction. Expression levels increased in a step-wise manner from 7 to 23 pg/cell/day, with a relatively constant rate of 7 pg/cell/day in each round of transduction. The GFP gene was successfully removed from the cell genome without disturbing the hscFSH gene expression. Clones generated using this approach showed stable expression levels for more than two years. This is the first report describing the sequential insertion of transgenes as an alternative for increasing the expression levels of transformed cell lines. The methodology described here could notably impact on biotechnological industry by improving the capacity of mammalian cells to produce biopharmaceuticals.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Mutagênese Insercional/métodos , Transgenes/genética , Biotecnologia/métodos , Células Clonais , Citometria de Fluxo/métodos , Hormônio Foliculoestimulante/genética , Expressão Gênica , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Lentivirus/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transdução Genética
6.
Genes (Basel) ; 11(5)2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32365901

RESUMO

H3K27me3 is an epigenetic modification that results in the repression of gene transcription. The transcription factor RUNX1 (the runt-related transcription factor 1) influences granulosa cells' growth and ovulation. This research uses ELISA, flow cytometry, EDU, ChIP-PCR, WB and qPCR to investigate steroidogenesis, cell apoptosis, and the proliferation effect of RUNX1 in porcine granulosa cells (pGCs) as regulated by H3K27me3. Decreased H3K27me3 stimulates the expression of steroidogenesis-related genes, including CYP11A1, PTGS2, and STAR, as well as prostaglandin. H3K27me3 transcriptionally represses RUNX1 here, whereas RUNX1 acts as an activator of FSHR, CYP11A1, and CYP19A1, promoting the production of androgen, estrogen, and prostaglandin, as well as increasing anti-apoptotic and cell proliferation activity, but decreasing progesterone. Both the complementary recovery of the H3K27me3 antagonist with the siRUNX1 signal, and the H3K27me3 agonist with the RUNX1 signal to maintain RUNX1 lead to the activation of CYP19A1, ER1, HSD17ß4, and STAR here. Androgen and prostaglandin are significantly repressed but progesterone is markedly increased with the antagonist and siRUNX1. Prostaglandin is significantly promoted with the agonist and RUNX1. Furthermore, H3K27me3-RUNX1 affects the anti-apoptotic activity and stimulation of proliferation in pGCs. The present work verifies the transcriptional suppression of RUNX1 by H3K27me3 during antral follicular development and maturation, which determines the levels of hormone synthesis and cell apoptosis and proliferation in the pGC microenvironment.


Assuntos
Proliferação de Células/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Histona Desmetilases com o Domínio Jumonji/genética , Esteroides/biossíntese , Apoptose/genética , Estrogênios/biossíntese , Estrogênios/genética , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células da Granulosa/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Ovulação/genética , Progesterona/biossíntese , Progesterona/genética , RNA Mensageiro/genética , Esteroides/metabolismo
7.
Endocrinology ; 161(7)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32270195

RESUMO

Activins are selective regulators of FSH production by pituitary gonadotrope cells. In a gonadotrope-like cell line, LßT2, activins stimulate FSH via the activin type IIA receptor (ACVR2A) and/or bone morphogenetic protein type II receptor (BMPR2). Consistent with these observations, FSH is greatly reduced, though still present, in global Acvr2a knockout mice. In contrast, FSH production is unaltered in gonadotrope-specific Bmpr2 knockout mice. In light of these results, we questioned whether an additional type II receptor might mediate the actions of activins or related TGF-ß ligands in gonadotropes. We focused on the activin type IIB receptor (ACVR2B), even though it does not mediate activin actions in LßT2 cells. Using a Cre-lox strategy, we ablated Acvr2a and/or Acvr2b in murine gonadotropes. The resulting conditional knockout (cKO) animals were compared with littermate controls. Acvr2a cKO (cKO-A) females were subfertile (~70% reduced litter size), cKO-A males were hypogonadal, and both sexes showed marked decreases in serum FSH levels compared with controls. Acvr2b cKO (cKO-B) females were subfertile (~20% reduced litter size), cKO-B males had a moderate decrease in testicular weight, but only males showed a significant decrease in serum FSH levels relative to controls. Simultaneous deletion of both Acvr2a and Acvr2b in gonadotropes led to profound hypogonadism and FSH deficiency in both sexes; females were acyclic and sterile. Collectively, these data demonstrate that ACVR2A and ACVR2B are the critical type II receptors through which activins or related TGF-ß ligands induce FSH production in mice in vivo.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Hormônio Foliculoestimulante/biossíntese , Receptores de Activinas Tipo II/genética , Animais , Feminino , Hipogonadismo/genética , Masculino , Camundongos , Camundongos Knockout , Caracteres Sexuais
8.
J Endocrinol ; 244(3): 459-471, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31905166

RESUMO

MicroRNA-7 (miR-7) is an important modulator of a plenty of gene expressions and the interrelated biological processes, highly expressed in porcine pituitary. Norepinephrine (NE), acting as an important neurotransmitter or/and a hormone secreted excessively under stress, affects the synthesis and secretion of various hormones, including pituitary follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which are the key hormones which regulate sexual maturation and reproductive functions. However, the relationship among NE, miR-7 and gonadotropin needs to be elucidated. The aim of this study was to identify whether miR-7 involved in the NE-adrenoceptor signaling pathway affects the synthesis and secretion of FSH and LH in porcine pituitary. Our results showed that the NE intracerebroventricular injection increased pituitary miR-7 level and the synthesis and secretion of FSH and LH in porcine, whereas the inhibition of either endogenous miR-7 or ß-adrenergic receptors hindered the rise of FSH and LH synthesis induced by NE in cultured primary porcine anterior pituitary cells. Further, we identified the molecular type of ß-adrenergic receptors and the signaling pathway in porcine pituitary, and we found that NE played its roles relying on adrenoceptor beta 2 (ß2AR) and the RAF/MEK/ERK1/2 signaling pathway. The phosphorylation of ERK1/2 upregulated miR-7 level which subsequently enhanced FSH and LH synthesis by targeting to Golgi glycoprotein 1 (GLG1). These suggest that miR-7 mediates NE's effect on promoting FSH and LH synthesis in porcine pituitary.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Hormônio Luteinizante/biossíntese , Norepinefrina/metabolismo , Hipófise/metabolismo , Suínos/metabolismo , Animais , Hormônio Foliculoestimulante/genética , Hormônio Luteinizante/genética , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Suínos/genética
9.
Life Sci ; 239: 116999, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31654746

RESUMO

AIMS: The present study aimed to investigate the effects of cyclophosphamide (Cytoxan, CTX) on premature ovarian failure (POF) in mice and its regulatory mechanisms by transcriptome analysis. MAIN METHODS: Female C57BL/6 mice were treated with a single intraperitoneal injection of 70 mg/kg CTX. Serum levels of estradiol (E2) and follicle stimulating hormone (FSH) were measured by enzyme-linked immunosorbent assay (ELISA), and follicular structure differences were observed by hematoxylin and eosin (H&E) staining. The main mechanism of POF was investigated by RNA-seq data, protein-protein interaction (PPI) networks and qPCR analysis. KEY FINDINGS: The serum levels of E2 were significantly decreased and those of FSH were significantly increased compared to the control group. The ovarian weights of the mice in the CTX group were reduced, and abnormal follicular structures were also observed in the CTX group. The RNA-seq data show that the downregulated genes were related to the cholesterol biosynthesis pathway. The PPI network and qPCR analyses further confirm that the PPAR signaling pathway and the ovarian infertility genes were also involved in blocking the cholesterol biosynthesis pathway. The differences were statistically significant. SIGNIFICANCE: Our results indicate that CTX may exert its anti-tumor effects by inactivating the cholesterol biosynthesis pathway, and simultaneously reducing the supply of estrogen precursor materials, ultimately leading to the occurrence of POF. Our data provided a preliminary theoretical basis for resolving the clinical toxicity and side effects of CTX.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Colesterol/biossíntese , Ciclofosfamida/toxicidade , Perfilação da Expressão Gênica , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/biossíntese , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Receptores Ativados por Proliferador de Peroxissomo/genética , Insuficiência Ovariana Primária/genética , Mapas de Interação de Proteínas
10.
Mar Biotechnol (NY) ; 21(5): 697-706, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31372794

RESUMO

The availability of sexually mature fish often dictates the success of its captive breeding. In this study, we induced reproductive development in juvenile protogynous tiger grouper through oral administration of a plasmid (p) containing an engineered follicle-stimulating hormone (FSH). An expression construct (pcDNA3.1) was designed to express a single-chain FSH consisting of giant grouper FSH ß-subunit and glycoprotein subunit-α (CGα), linked by the carboxy-terminal peptide (CTP) sequence from the human chorionic gonadotropin (hCG). Single oral delivery of pFSH encapsulated in liposome and chitosan to tiger grouper yielded a significant increase in plasma FSH protein level after 4 days. Weekly pFSH feeding of juvenile tiger groupers for 8 weeks stimulated ovarian development as indicated by a significant increase in oocyte diameter and progression of oocytes to cortical alveolar stage. As the pFSH treatment progressed from 20 to 38 weeks, female to male sex change was initiated, characterized by oocyte regression, proliferation of spermatogonial cells, and occurrence of spermatogenic cysts. It was also associated with significantly lower mRNA expression of steroidogenic genes (cyp11b, cyp19a1a, and foxl2) and basal plasma levels of sex steroid hormones 17ß-estradiol (E2), testosterone (T), and 11-ketotestosterone (11KT). Results suggest that pFSH stimulates ovarian development up to cortical alveolar stage and then initiates sex change in tiger grouper. These findings significantly contribute to our knowledge on the role of FSH in the development of protogynous hermaphroditic fish. This study is the first to demonstrate induction of reproductive development in fish through oral delivery of plasmid gonadotropin.


Assuntos
Gonadotropina Coriônica/genética , Hormônio Foliculoestimulante/genética , Gônadas/efeitos dos fármacos , Organismos Hermafroditas/efeitos dos fármacos , Perciformes/genética , Processos de Determinação Sexual/efeitos dos fármacos , Diferenciação Sexual/efeitos dos fármacos , Administração Oral , Animais , Quitosana/química , Gonadotropina Coriônica/administração & dosagem , Gonadotropina Coriônica/biossíntese , Composição de Medicamentos , Feminino , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/biossíntese , Hormônios Esteroides Gonadais/biossíntese , Hormônios Esteroides Gonadais/genética , Gônadas/crescimento & desenvolvimento , Gônadas/metabolismo , Organismos Hermafroditas/genética , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Masculino , Oogênese/efeitos dos fármacos , Oogênese/genética , Perciformes/crescimento & desenvolvimento , Perciformes/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Pré-Seleção do Sexo/métodos , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética
11.
J Neuroendocrinol ; 31(10): e12769, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31283846

RESUMO

Diethylstilbestrol (DES) is a synthetic oestrogen known to disrupt the endocrine system and to cause reproductive toxicity mediated via the hypothalamic-pituitary-adrenal axis; however, its molecular mechanism of action is poorly understood. In the present study, we found that, after only 1 week of exposure to DES, blood testosterone dramatically decreased and that this decrease was associated with a strong induction of prolactin (PRL). Even with the increase in PRL, the luteinising hormone and follicle-stimulating hormone mRNAs slightly decreased. Our results show that, after 48 hours of a single dose of DES, there was a six-fold increase in PRL expression. After exploring the upstream mechanisms, we determined that dopamine, which inhibits PRL secretion in male rats, did not decrease in the pituitary gland of DES-treated rats, whereas vasoactive intestinal peptide (VIP), which mediates the acute release of PRL, was elevated. Serotonin (5-HT) increased in the brain of male rats 24 hours after a single DES treatment; however, PRL, VIP or 5-HT was not induced by DES in female rats. Our results indicate that DES induces the expression of pituitary PRL in male rats by stimulating VIP in the hypothalamus and 5-HT in the central nervous system.


Assuntos
Dietilestilbestrol/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Prolactina/metabolismo , Animais , Encéfalo/metabolismo , Dopamina/metabolismo , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Luteinizante/biossíntese , Masculino , Hipófise/metabolismo , Prolactina/sangue , Ratos , Serotonina/metabolismo , Caracteres Sexuais , Testosterona/sangue , Peptídeo Intestinal Vasoativo/metabolismo
12.
Endocrinology ; 160(3): 675-683, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715256

RESUMO

Bone morphogenetic proteins (BMPs) are pleiotropic ligands in the TGF-ß superfamily. In the early to mid-2000s, several BMPs, including BMP2, were shown to regulate FSH synthesis alone and in synergy with activins in immortalized gonadotrope-like cell lines and primary pituitary cultures. Activins are also TGF-ß family members, which were identified and named based on their abilities to stimulate FSH production selectively. Mechanistic analyses suggested that BMP2 promoted expression of the FSHß subunit gene (Fshb) via at least two nonmutually exclusive mechanisms. First, BMP2 stimulated the production of the inhibitor of DNA-binding proteins 1, 2, and 3 (Id1, Id2, and Id3), which potentiated the stimulatory actions of homolog of Drosophila mothers against decapentaplegic 3 (SMAD3) on the Fshb promoter. SMAD3 is an intracellular signaling protein that canonically mediates the actions of activins and is an essential regulator of Fshb production in vitro and in vivo. Second, BMP2 was shown to activate SMAD3-dependent signaling via its canonical type IA receptor, BMPR1A (also known as ALK3). This was a surprising result, as ALK3 conventionally activates distinct SMAD proteins. Although these initial results were compelling, they were challenged by contemporaneous and subsequent observations. For example, inhibitors of BMP signaling did not specifically impair FSH production in cultured pituitary cells. Of perhaps greater significance, mice lacking ALK3 in gonadotrope cells produced FSH normally. Therefore, the physiological role of BMPs in FSH synthesis in vivo is presently uncertain.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Hormônio Foliculoestimulante/biossíntese , Gonadotrofos/fisiologia , Ativinas/metabolismo , Animais , Inibinas/metabolismo , Reprodução , Proteínas Smad/metabolismo
13.
Microsc Res Tech ; 82(6): 635-642, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30582244

RESUMO

Many studies have reported that human endometrial mesenchymal stem cells (HuMenSCs) are capable of repairing damaged tissues. The aim of the present study was to investigate the effects of HuMenSCs transplantation as a treatment modality in premature ovarian failure (POF) associated with chemotherapy-induced ovarian damage. HuMenSCs were isolated from menstrual blood samples of five women. After the in vitro culture of HuMenSCs, purity of the cells was assessed by cytometry using CD44, CD90, CD34, and CD45 FITC conjugate antibody. Twenty-four female Wistar rats were randomly divided into four groups: negative control, positive control, sham, and treatment groups. The rat models of POF used in our study were established by injecting busulfan intraperitoneally into the rats during the first estrus cycle. HuMenSCs were transplanted by injection via the tail vein into the POF-induced rats. Four weeks after POF induction, ovaries were collected and the levels of Amh, Fst, and Fshr expression in the granulosa cell (GC) layer, as well as plasma estradiol (E2) and progesterone (P4) levels were evaluated. Moreover, migration and localization of DiI-labeled HuMenSCs were detected, and the labeled cells were found to be localized in GCs layer of immature follicles. In addition to DiI-labelled HuMenSCs tracking, increased levels of expression of Amh and Fshr and Fst, and the high plasma levels of E2 and P4 confirmed that HuMenSC transplantation had a significant effect on follicle formation and ovulation in the treatment group compared with the negative control (POF) group.


Assuntos
Transplante de Células/métodos , Células da Granulosa/fisiologia , Células-Tronco Mesenquimais/fisiologia , Insuficiência Ovariana Primária/terapia , Animais , Hormônio Antimülleriano/biossíntese , Bussulfano/administração & dosagem , Modelos Animais de Doenças , Feminino , Hormônio Foliculoestimulante/biossíntese , Perfilação da Expressão Gênica , Histocitoquímica , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Folículo Ovariano/patologia , Ovário/patologia , Ovário/fisiologia , Ovulação , Insuficiência Ovariana Primária/induzido quimicamente , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores do FSH/biossíntese , Resultado do Tratamento
14.
Endocrinology ; 159(12): 4077-4091, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30364975

RESUMO

Inhibins are gonadal hormones that act on pituitary gonadotrope cells to suppress FSH synthesis and secretion. Inhibin A and B are heterodimers of the inhibin ⍺-subunit disulfide-linked to one of two inhibin ß-subunits. Homodimers or heterodimers of the inhibin ß-subunits form the activins, which stimulate FSH production. Activins signal through complexes of type I and II receptor serine/threonine kinases to increase transcription of the FSHß subunit gene. According to in vitro observations, inhibins impair FSH synthesis by competitively binding to activin type II receptors, particularly in the presence of the TGFß type III receptor (TGFBR3, or betaglycan). The role of TGFBR3 in inhibin action in vivo has not been determined. Here, we ablated Tgfbr3 specifically in murine gonadotropes. Conditional knockout females were supra-fertile, exhibiting enhanced folliculogenesis, numbers of ovulated eggs per cycle, and litter sizes relative to control mice. Despite these phenotypes, FSH levels appeared to be unaltered in knockout mice, and the mechanisms underlying their enhanced fertility remain unexplained. Inhibin B is the predominant form of the hormone in males and in females during most stages of the estrous cycle. Remarkably, inhibin A, but not inhibin B, suppression of FSH synthesis was impaired in cultured pituitaries of knockout mice, which may explain the absence of discernible changes in FSH levels in vivo. Collectively, these data challenge current dogma by demonstrating that TGFBR3 (betaglycan) functions as an inhibin A, but not an inhibin B, coreceptor in gonadotrope cells in vivo. Mechanisms of inhibin B action merit further investigation.


Assuntos
Gonadotrofos/metabolismo , Inibinas/metabolismo , Proteoglicanas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Feminino , Fertilidade/genética , Hormônio Foliculoestimulante/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Ligação Proteica , Multimerização Proteica , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
15.
J Mol Endocrinol ; 60(3): R131-R155, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437880

RESUMO

Follicle-stimulating hormone (FSH) plays fundamental roles in male and female fertility. FSH is a heterodimeric glycoprotein expressed by gonadotrophs in the anterior pituitary. The hormone-specific FSHß-subunit is non-covalently associated with the common α-subunit that is also present in the luteinizing hormone (LH), another gonadotrophic hormone secreted by gonadotrophs and thyroid-stimulating hormone (TSH) secreted by thyrotrophs. Several decades of research led to the purification, structural characterization and physiological regulation of FSH in a variety of species including humans. With the advent of molecular tools, availability of immortalized gonadotroph cell lines and genetically modified mouse models, our knowledge on molecular mechanisms of FSH regulation has tremendously expanded. Several key players that regulate FSH synthesis, sorting, secretion and action in gonads and extragonadal tissues have been identified in a physiological setting. Novel post-transcriptional and post-translational regulatory mechanisms have also been identified that provide additional layers of regulation mediating FSH homeostasis. Recombinant human FSH analogs hold promise for a variety of clinical applications, whereas blocking antibodies against FSH may prove efficacious for preventing age-dependent bone loss and adiposity. It is anticipated that several exciting new discoveries uncovering all aspects of FSH biology will soon be forthcoming.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/farmacologia , Animais , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esteroides/farmacologia
16.
Mol Cell Endocrinol ; 461: 43-54, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-28830788

RESUMO

Zearalenone (ZEA) is one of the most popular endocrine-disrupting chemicals and is mainly produced by fungi of the genus Fusarium. The excessive intake of ZEA severely disrupts human and animal fertility by affecting the reproductive axis. However, most studies on the effects of ZEA and its metabolite α-zearalenol (α-ZOL) on reproductive systems have focused on gonads. Few studies have investigated the endocrine-disrupting effects of ZEA and α-ZOL on pituitary gonadotropins, including follicle-stimulating hormone (FSH) and luteinizing hormone (LH). The present study was designed to investigate the effects of ZEA and α-ZOL on the synthesis and secretion of FSH and LH and related mechanisms in female pig pituitary. Our in vivo and in vitro results demonstrated that ZEA significantly inhibited the synthesis and secretion of FSH in the pig pituitary gland, but ZEA and α-ZOL had no effects on LH. Our study also showed that ZEA and α-ZOL decreased FSH synthesis and secretion through non-classical estrogen membrane receptor GPR30, which subsequently induced protein kinase cascades and the phosphorylation of PKC, ERK and p38MAPK signaling pathways in pig pituitary cells. Furthermore, our study showed that the LIM homeodomain transcription factor LHX3 was involved in the mechanisms of ZEA and α-ZOL actions on gonadotropes in the female pig pituitary. These findings elucidate the mechanisms behind the physiological alterations resulting from endocrine-disrupting chemicals and further show that the proposed key molecules of the α-ZOL signaling pathway could be potential pharmacological targets.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Zearalenona/farmacologia , Zeranol/análogos & derivados , Animais , Feminino , Proteínas com Homeodomínio LIM/metabolismo , Hormônio Luteinizante/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Proteína Quinase C/metabolismo , Receptores de Estrogênio/metabolismo , Suínos , Fatores de Transcrição/metabolismo , Zeranol/farmacologia
17.
J Reprod Dev ; 63(6): 605-609, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29033405

RESUMO

The gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are important hormones in vertebrate reproduction. The isolation of gonadotropins from the pituitary gland is sub-optimal, as the cross-contamination of one hormone with another is common and often results in the variation in the measured activity of LH and FSH. The production of recombinant hormones is, therefore, a viable approach to solve this problem. This study aimed to express recombinant rat, mouse, and mastomys FSH and LH in Chinese hamster ovary (CHO) cells. Their common α-subunits along with their hormone-specific ß-subunits were encoded in a single mammalian expression vector. FSH from all three species was expressed, whereas expression was achieved only for the mouse LH. Immunohistochemistry for rat alpha subunit of glycoprotein hormone (αGSU) and LHß and FSHß subunits confirmed the production of the dimeric hormone in CHO cells. The recombinant rodent gonadotropins were confirmed to be biologically active; estradiol production was increased by recombinant FSH in granulosa cells, while recombinant LH increased testosterone production in Leydig cells.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Vetores Genéticos , Hormônio Luteinizante/biossíntese , Animais , Células CHO , Cricetulus , Hormônio Foliculoestimulante/genética , Hormônio Luteinizante/genética , Masculino , Camundongos Endogâmicos C57BL , Murinae , Ratos Wistar , Proteínas Recombinantes/biossíntese
18.
J Biol Chem ; 292(6): 2301-2314, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-27994055

RESUMO

Pituitary follicle-stimulating hormone (FSH) is an essential regulator of fertility in females and of quantitatively normal spermatogenesis in males. Pituitary-derived activins are thought to act as major stimulators of FSH synthesis by gonadotrope cells. In vitro, activins signal via SMAD3, SMAD4, and forkhead box L2 (FOXL2) to regulate transcription of the FSHß subunit gene (Fshb). Consistent with this model, gonadotrope-specific Smad4 or Foxl2 knock-out mice have greatly reduced FSH and are subfertile. The role of SMAD3 in vivo is unresolved; however, residual FSH production in Smad4 conditional knock-out mice may derive from partial compensation by SMAD3 and its ability to bind DNA in the absence of SMAD4. To test this hypothesis and determine the role of SMAD3 in FSH biosynthesis, we generated mice lacking both the SMAD3 DNA binding domain and SMAD4 specifically in gonadotropes. Conditional knock-out females were hypogonadal, acyclic, and sterile and had thread-like uteri; their ovaries lacked antral follicles and corpora lutea. Knock-out males were fertile but had reduced testis weights and epididymal sperm counts. These phenotypes were consistent with those of Fshb knock-out mice. Indeed, pituitary Fshb mRNA levels were nearly undetectable in both male and female knock-outs. In contrast, gonadotropin-releasing hormone receptor mRNA levels were significantly elevated in knock-outs in both sexes. Interestingly, luteinizing hormone production was altered in a sex-specific fashion. Overall, our analyses demonstrate that SMAD3 is required for FSH synthesis in vivo.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Gonadotrofos/metabolismo , Proteína Smad3/fisiologia , Animais , Éxons , Feminino , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout , Proteína Smad3/genética , Proteína Smad4/genética , Proteína Smad4/fisiologia , Espermatogênese/genética
19.
J Biol Chem ; 292(7): 2646-2659, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28007961

RESUMO

Follicle-stimulating hormone (FSH) regulates follicular growth and stimulates estrogen synthesis in the ovaries. FSH is a heterodimer consisting of an α subunit, also present in luteinizing hormone, and a unique ß subunit, which is transcriptionally regulated by gonadotropin-releasing hormone 1 (GNRH). Because most FSH is constitutively secreted, tight transcriptional regulation is critical for maintaining FSH levels within a narrow physiological range. Previously, we reported that GNRH induces FSHß (Fshb) transcription via induction of the AP-1 transcription factor, a heterodimer of c-FOS and c-JUN. Herein, we identify c-JUN-dimerization protein 2 (JDP2) as a novel repressor of GNRH-mediated Fshb induction. JDP2 exhibited high basal expression and bound the Fshb promoter at an AP-1-binding site in a complex with c-JUN. GNRH treatment induced c-FOS to replace JDP2 as a c-JUN binding partner, forming transcriptionally active AP-1. Subsequently, rapid c-FOS degradation enabled reformation of the JDP2 complex. In vivo studies revealed that JDP2 null male mice have normal reproductive function, as expected from a negative regulator of the FSH hormone. Female JDP2 null mice, however, exhibited early puberty, observed as early vaginal opening, larger litters, and early reproductive senescence. JDP2 null females had increased levels of circulating FSH and higher expression of the Fshb subunit in the pituitary, resulting in elevated serum estrogen and higher numbers of large ovarian follicles. Disruption of JDP2 function therefore appears to cause early cessation of reproductive function, a condition that has been associated with elevated FSH in women.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Insuficiência Ovariana Primária/prevenção & controle , Proteínas Repressoras/fisiologia , Animais , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Hormônio Liberador de Gonadotropina/fisiologia , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Repressoras/genética
20.
J Assist Reprod Genet ; 33(12): 1665-1675, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27638727

RESUMO

PURPOSE: The main goals of this study were to investigate the expression of anti-Müllerian hormone (AMH) and its receptor (AMHR2) during follicular development in primates, and to evaluate the potential of AMH as a biomarker for follicle growth and oocyte maturation in vitro. METHODS: The mRNA and protein expression of AMH and AMHR2 were determined using isolated follicles and ovarian sections from rhesus macaques (n = 4) by real-time PCR and immunohistochemistry, respectively. Isolated secondary follicles were cultured individually. Follicle growth and media AMH concentrations were assessed by ELISA. The mRNA expression profiles, obtained from RNA sequencing, of in vitro- and in vivo-developed antral follicles were compared. Secondary follicles from additional animals (n = 35) were cultured. Follicle growth, oocyte maturation, and media AMH concentrations were evaluated for forecasting follicular development in vitro by AMH levels. RESULTS: AMH immunostaining was heterogeneous in the population of preantral follicles that were also stained for AMHR2. The mRNA expression profiles were comparable between in vivo- and in vitro-developed follicles. AMH levels produced by growing follicles were higher than those of nongrowing follicles in culture. With a cutoff value of 1.40 ng/ml, 85 % of nongrowing follicles could be identified while eliminating only 5 % of growing follicles. Growing follicles that generated metaphase II-stage oocytes secreted greater amounts of AMH than did those yielding immature germinal vesicle-stage oocytes. CONCLUSIONS: AMH, co-expressed with AMHR2, was produced heterogeneously by preantral follicles in macaques with levels correlated positively with follicle growth and oocyte maturation. AMH may serve as a biomarker for primate follicular development in vitro.


Assuntos
Hormônio Antimülleriano/biossíntese , Técnicas de Maturação in Vitro de Oócitos , Folículo Ovariano/metabolismo , Receptores de Peptídeos/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Animais , Hormônio Antimülleriano/genética , Biomarcadores/metabolismo , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Humanos , Macaca mulatta , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Oogênese/genética , Folículo Ovariano/crescimento & desenvolvimento , Progesterona/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...