Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
J Biol Chem ; 291(40): 21322-21334, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27466366

RESUMO

Reproductive function is controlled by the pulsatile release of hypothalamic gonadotropin-releasing hormone (GnRH), which regulates the expression of the gonadotropins luteinizing hormone and FSH in pituitary gonadotropes. Paradoxically, Fshb gene expression is maximally induced at lower frequency GnRH pulses, which provide a very low average concentration of GnRH stimulation. We studied the role of secreted factors in modulating gonadotropin gene expression. Inhibition of secretion specifically disrupted gonadotropin subunit gene regulation but left early gene induction intact. We characterized the gonadotrope secretoproteome and global mRNA expression at baseline and after Gαs knockdown, which has been found to increase Fshb gene expression (1). We identified 1077 secreted proteins or peptides, 19 of which showed mRNA regulation by GnRH or/and Gαs knockdown. Among several novel secreted factors implicated in Fshb gene regulation, we focused on the neurosecretory protein VGF. Vgf mRNA, whose gene has been implicated in fertility (2), exhibited high induction by GnRH and depended on Gαs In contrast with Fshb induction, Vgf induction occurred preferentially at high GnRH pulse frequency. We hypothesized that a VGF-derived peptide might regulate Fshb gene induction. siRNA knockdown or extracellular immunoneutralization of VGF augmented Fshb mRNA induction by GnRH. GnRH stimulated the secretion of the VGF-derived peptide NERP1. NERP1 caused a concentration-dependent decrease in Fshb gene induction. These findings implicate a VGF-derived peptide in selective regulation of the Fshb gene. Our results support the concept that signaling specificity from the cell membrane GnRH receptor to the nuclear Fshb gene involves integration of intracellular signaling and exosignaling regulatory motifs.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Regulação da Expressão Gênica/fisiologia , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neuropeptídeos/metabolismo , Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Gonadotrofos/citologia , Camundongos , Fatores de Crescimento Neural , RNA Mensageiro/biossíntese
12.
Stem Cell Reports ; 6(6): 858-872, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27304916

RESUMO

Human pluripotent stem cells (hPSCs) provide an unlimited cell source for regenerative medicine. Hormone-producing cells are particularly suitable for cell therapy, and hypopituitarism, a defect in pituitary gland function, represents a promising therapeutic target. Previous studies have derived pituitary lineages from mouse and human ESCs using 3D organoid cultures that mimic the complex events underlying pituitary gland development in vivo. Instead of relying on unknown cellular signals, we present a simple and efficient strategy to derive human pituitary lineages from hPSCs using monolayer culture conditions suitable for cell manufacturing. We demonstrate that purified placode cells can be directed into pituitary fates using defined signals. hPSC-derived pituitary cells show basal and stimulus-induced hormone release in vitro and engraftment and hormone release in vivo after transplantation into a murine model of hypopituitarism. This work lays the foundation for future cell therapy applications in patients with hypopituitarism.


Assuntos
Corticotrofos/metabolismo , Células-Tronco Embrionárias/metabolismo , Hipopituitarismo/terapia , Células-Tronco Pluripotentes/metabolismo , Tireotrofos/metabolismo , Hormônio Adrenocorticotrópico/biossíntese , Hormônio Adrenocorticotrópico/metabolismo , Animais , Benzamidas/farmacologia , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Terapia Baseada em Transplante de Células e Tecidos , Corticotrofos/citologia , Corticotrofos/efeitos dos fármacos , Dioxóis/farmacologia , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Expressão Gênica , Hormônio do Crescimento/biossíntese , Hormônio do Crescimento/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Hipopituitarismo/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Hipófise/metabolismo , Hipófise/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Tireotrofos/citologia , Tireotrofos/efeitos dos fármacos , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Reprod Toxicol ; 58: 174-83, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26476359

RESUMO

Endocrine modulating effects of Simvastatin (SV) and its metabolite, Simvastatin ß-hydroxy acid (SVA), were investigated in H295R cells and in female Sprague-Dawley (SPRD) rats. H295R cells were exposed to SV and SVA concentrations from 0 to 10µM for 48h. Four groups of SPRD rats received 0 (CT), 1.3 (L), 5.0 (M), and 20.0 (H)mg SV/kg bw/day for 14 days. 10 Steroids were investigated in H295R growth media, and in tissues and plasma from rats using GC-MS/MS. Plasma LH and FSH were quantified by ELISA. In the H295R assay, SV and SVA particularly decreased progestagens with IC50-values from 0.10-0.13µM for SV and from 0.019-0.055µM for SVA. In rats, SV decreased progestagens in ovaries, brain and plasma, and plasma FSH in the M (72.4% decrease) and H group (76.6% decrease). Because progestagens and gonadotropins are major players in fertility, administration of SV might exert negative effects on female reproduction.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Hormônio Foliculoestimulante/biossíntese , Hormônios Esteroides Gonadais/biossíntese , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Sinvastatina/toxicidade , Córtex Suprarrenal/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Fertilidade/efeitos dos fármacos , Hormônio Foliculoestimulante/sangue , Cromatografia Gasosa-Espectrometria de Massas , Hormônios Esteroides Gonadais/sangue , Hormônio Luteinizante/biossíntese , Hormônio Luteinizante/sangue , Ratos Sprague-Dawley , Reprodução/efeitos dos fármacos , Medição de Risco , Espectrometria de Massas em Tandem , Fatores de Tempo
14.
Biol Reprod ; 93(5): 114, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26400402

RESUMO

Kisspeptins are key players in the neuroendocrine control of puberty and other reproductive processes in mammals. Several studies have demonstrated that the KISS/GPR54 system is expressed by gonadotrophs, but in vitro studies assessing the direct stimulatory effects of kisspeptin on gonadotropin secretion in the pituitary have provided conflicting results. In this study, we investigated whether kisspeptin directly influences the reproductive function of sea bass pituitary. First, the highly active peptides Kiss1-15 and Kiss2-12 were used to stimulate dispersed sea bass pituitary cells obtained from mature males. Our results show that, first, Kiss2-12 induced luteinizing hormone (Lh) and follicle-stimulating hormone (Fsh) release, whereas Kiss1-15 had no effect on gonadotropin secretion at full spermiation stage. Second, the distribution and nature of Kiss2 and its potential interactions with the gonadotropin-releasing hormone 1 (Gnrh1) system in the pituitary were analyzed using dual fluorescence immunohistochemistry. Kiss2 cells were found in the proximal pars distalis and colocalized with gonadotropin-immunoreactive cells. In summary, our results provide, for the first time in a teleost species, functional and neuroanatomical evidence that Kiss2 may act through different routes to directly modulate the activity of gonadotrophs, either as a hypophysiotropic neuropeptide or as an autocrine/paracrine factor.


Assuntos
Bass/metabolismo , Hormônio Foliculoestimulante/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Comunicação Autócrina , Células Cultivadas , Hormônio Foliculoestimulante/biossíntese , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Luteinizante/biossíntese , Masculino , Comunicação Parácrina
15.
Endocrinology ; 156(10): 3818-27, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26252060

RESUMO

Due to increased numbers of young cancer patients and improved survival, the impact of anticancer treatments on fertility has become a major health concern. Despite mounting research on ovarian toxicity, there is paucity of data regarding reliable biomarkers of testicular toxicity. Our aim was to evaluate anti-Müllerian hormone (AMH) as a marker for chemotherapy-induced testicular toxicity. Serum AMH and a panel of gonadal hormones were measured in male cancer patients at baseline and after chemotherapy. In the preclinical setting, mice were injected with diverse chemotherapies and were killed 1 week or 1, 3, or 6 months later. We evaluated spermatogenesis by AMH as well as qualitative and quantitative sperm parameters. Nineteen patients were enrolled, the median age was 38 years (21-44 y). Serum AMH was correlated with increased FSH and T and decreased inhibin-B in gonadotoxic protocols (cisplatin or busulfan) and remained unchanged in nongonadotoxic protocols (capecitabine). AMH expression had the same pattern in mice serum and testes; it was negatively correlated with testicular/epididymal weight and sperm motility. The increase in testicular AMH expression was also correlated with elevated apoptosis (terminal transferase-mediated deoxyuridine 5-triphosphate nick-end labeling) and reduced proliferation (Ki67, proliferating cell nuclear antigen; all seminiferous tubules cells were analyzed). Severely damaged mice testes demonstrated a marked costaining of AMH and GATA-4, a Sertoli cell marker; staining that resembled the pattern of the Sertoli cell-only condition. Our study indicates that the pattern of serum AMH expression, in combination with other hormones, can delineate testicular damage, as determined in both experimental settings. Future large-scale clinical studies are warranted to further define the role of AMH as a biomarker for testicular toxicity.


Assuntos
Hormônio Antimülleriano/sangue , Antineoplásicos/efeitos adversos , Biomarcadores Tumorais/sangue , Testículo/efeitos dos fármacos , Adulto , Animais , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Western Blotting , Hormônio Foliculoestimulante/biossíntese , Expressão Gênica , Humanos , Subunidades beta de Inibinas/sangue , Masculino , Camundongos Endogâmicos ICR , Microscopia Confocal , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Tamanho do Órgão/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia , Testosterona/sangue , Adulto Jovem
16.
Appl Biochem Biotechnol ; 176(2): 399-411, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25805018

RESUMO

Callithrix jacchus (common marmoset) is a New World primate monkey, used as an animal model in biomedical research. Marmoset-specific follicle-stimulating hormone (FSH) preparation is required to improve superovulation protocols and to develop homologous FSH monitoring assays in these monkeys. In this study, we document the large-scale expression of recombinant marmoset FSH in methylotropic yeast, Pichia pastoris. The recombinant preparation was found to be immunologically active in Western blotting and radioimmunoassay. The preparation displayed receptor binding ability in radioreceptor assay. Based on the receptor binding ability, the yield of fermentation was estimated to be 7.2 mg/L. FSH-induced cAMP assay and estradiol assay revealed that the recombinant hormone is able to induce signal transduction. Both immunological and in vitro biological activity of marmoset FSH was found to be comparable to purified human pituitary FSH, which served as reference hormone for these assays. Thus, the study suggests that a Pichia expression system can be used for large-scale expression of bioactive recombinant marmoset FSH.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica , Pichia/genética , Animais , Callithrix , Feminino , Hormônio Foliculoestimulante/genética , Células HEK293 , Humanos , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
17.
Biochim Biophys Acta ; 1849(3): 328-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25591470

RESUMO

The genes encoding luteinizing hormone and follicle stimulating hormone are activated by gonadotropin-releasing hormone (GnRH), and we hypothesized that this involves GnRH-induction of various histone modifications. At basal conditions in an immature gonadotrope-derived cell line, the hormone-specific ß-subunit gene promoters are densely packed with histones, and contain low levels of H3K4 trimethylation (H3K4me3). GnRH both induces this modification and causes histone loss, creating a more active chromatin state. The H3K4me3 appears to be mediated by menin and possibly catalyzed by the menin-mixed-lineage leukemia (MLL) 1/2 methyl transferase complex, as inhibition of MLL recruitment or menin knockdown reduced gene expression and the levels of H3K4me3 on all three promoters. Menin recruitment to the ß-subunit gene promoters is increased by GnRH, possibly involving transcription factors such as estrogen receptor α and/or steroidogenic factor 1, with which menin interacts. Menin also interacts with ring finger protein 20, which ubiquitylates H2BK120 (H2BK120ub), which was reported to be a pre-requisite for H3K4me3 at various gene promoters. Although levels of H2BK120ub are increased by GnRH in the coding regions of these genes, levels at the promoters do not correlate with those of H3K4me3, nor with gene expression, suggesting that H3K4me3 is not coupled to H2BK120ub in transcriptional activation of these genes.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Hormônio Liberador de Gonadotropina/biossíntese , Gonadotropinas/genética , Hormônio Luteinizante/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/genética , Gonadotropinas/biossíntese , Histona Desmetilases/genética , Histonas/genética , Hormônio Luteinizante/genética , Camundongos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Ativação Transcricional
18.
Am J Physiol Endocrinol Metab ; 307(11): E1038-46, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25315693

RESUMO

Female reproductive success is closely associated with nutritional status and energy balance. In this context, adiponectin appears to be a key hormone connecting reproductive system function and metabolism regulation. It is hypothesized that adiponectin expression in the pituitary depends on the phase of the estrous cycle. The effect of adiponectin on luteinizing hormone (LH) and follicle-stimulating hormone (FSH) secretion is also postulated. Changes in the adiponectin gene and protein expression in the porcine anterior (AP) and posterior (NP) pituitaries as well as the effect of in vitro administration of adiponectin on basal and gonadotropin-releasing hormone (GnRH)- and/or insulin-stimulated LH and FSH secretion were investigated on days 2-3, 10-12, 14-16, and 17-19 of the estrous cycle. Adiponectin gene was more pronounced on days 2-3 in AP but on days 10-12 in NP. Protein concentration in AP was the highest on days 10-12 and in NP on days 10-12 and 17-19 of the cycle. In vitro, adiponectin did not affect basal LH secretion but increased FSH release by AP cells. Adiponectin administration affected GnRH- and/or insulin-induced LH and FSH output in a manner dependent on the phase of the estrous cycle. In this study we indicated for the first time adiponectin expression in the porcine AP and NP that was dependent on the phase of the estrous cycle. In vitro studies indicated that adiponectin may affect gonadotropin secretion. The above suggests that the studied adipokine may influence female reproductive functions via its effect on LH and FSH secretion by gonadotrophs, but the cellular mechanism of its action remains unknown.


Assuntos
Adiponectina/biossíntese , Ciclo Estral/fisiologia , Hormônio Foliculoestimulante/biossíntese , Hormônio Luteinizante/biossíntese , Hipófise/metabolismo , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Insulina/farmacologia , Adeno-Hipófise/metabolismo , Neuro-Hipófise/metabolismo , Suínos
19.
Ultrastruct Pathol ; 38(6): 430-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25080040

RESUMO

Abstract Metastases to the pituitary occur more frequently in patients with widespread cancer and mainly involve the posterior lobe. A few cases of metastatic carcinoma to a pituitary adenoma have been described so far. Here, the authors present an additional case of a clear cell renal cell carcinoma (CCRCC) metastatic to a FSH/LH/α-subunit pituitary adenoma and systematically review the literature. Immunohistochemistry and electron microscopy were performed to characterize both neoplastic components at the morphological level. Moreover, it was hypothesized that expression of VEGF and of the corresponding receptor VEGFR1 could be implicated in the development of the carcinomatous metastasis within the adenoma.


Assuntos
Adenoma/patologia , Carcinoma de Células Renais/secundário , Neoplasias Renais/patologia , Neoplasias Primárias Múltiplas/ultraestrutura , Neoplasias Hipofisárias/patologia , Idoso , Carcinoma de Células Renais/ultraestrutura , Feminino , Hormônio Foliculoestimulante/biossíntese , Humanos , Imuno-Histoquímica , Hormônio Luteinizante/biossíntese , Microscopia Eletrônica de Transmissão
20.
Carcinogenesis ; 35(11): 2447-51, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24997853

RESUMO

Estetrol (E4) is a fetal estrogen with estrogenic effects on reproductive organs and bone in preclinical models and in postmenopausal women. However, E4 exerts antiestrogenic effects on breast cancer (BC) cell growth in vitro and in vivo. We have investigated the effect of 14 days preoperative treatment with 20mg E4 per day on tumor proliferation markers, sex steroid receptor expression and endocrine parameters in a prospective, randomized, placebo-controlled, preoperative window trial in 30 pre- and post-menopausal women with estrogen-receptor positive early BC. E4 had a significant pro-apoptotic effect on tumor tissue, whereas Ki67 expression remained unchanged in both pre- and post-menopausal women. E4 increased sex-hormone-binding globulin significantly thereby reducing the concentrations of bioavailable estradiol. Follicle-stimulating hormone levels decreased in postmenopausal women only and luteinizing hormone levels remained unchanged. Systemic insulin growth factor-1 levels decreased significantly. Intratumoral epithelial ERα expression decreased significantly and a trend was found towards an increased expression of ERß. This clinical data support the preclinical findings that E4 has antiestrogenic effects on BC cells, whereas earlier studies have shown that E4 has estrogenic effects on reproductive tissues and bone. Further clinical studies seem acceptable and are needed to confirm the safety and efficacy of E4 for the breast in hormone replacement therapy, including hormone replacement therapy in women who have or have had BC, especially in those BC patients treated with aromatase inhibitors and suffering from serious complaints due to estrogen deficiency.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Estetrol/administração & dosagem , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Adulto , Idoso , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Receptor alfa de Estrogênio/genética , Feminino , Hormônio Foliculoestimulante/biossíntese , Terapia de Reposição Hormonal , Humanos , Fator de Crescimento Insulin-Like I/biossíntese , Antígeno Ki-67/biossíntese , Pessoa de Meia-Idade , Período Pré-Operatório
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA