Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Diabetologia ; 60(8): 1432-1441, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28547133

RESUMO

AIMS/HYPOTHESIS: Heat shock factor protein 1 (HSF1) is a transcription factor that regulates the expression of key molecular chaperones, thereby orchestrating the cellular response to stress. This system was recently implicated in the control of insulin sensitivity and is therefore being scrutinised as a novel therapeutic avenue for type 2 diabetes. However, the regulation and biological actions of HSF1 in beta cells remain elusive. Herein, we sought to investigate the regulation of HSF1 in pancreatic beta cells and to study its potential role in cell survival. METHODS: We exposed human islets and beta cell lines to glucolipotoxicity and thapsigargin. HSF1 activity was evaluated by gel shift assay. HSF1 acetylation and interaction with the protein acetylase cAMP response element binding protein (CBP) were investigated by western blot. We measured the expression of HSF1 and its canonical targets in islets from Goto-Kakizaki (GK) rat models of diabetes and delineated the effects of HSF1 acetylation using mutants mimicking constitutive acetylation and deacetylation of the protein. RESULTS: Glucolipotoxicity promoted HSF1 acetylation and interaction with CBP. Glucolipotoxicity-induced HSF1 acetylation inhibited HSF1 DNA binding activity and decreased the expression of its target genes. Restoration of HSF1 activity in beta cells prevented glucolipotoxicity-induced endoplasmic reticulum stress and apoptosis. However, overexpression of a mutant protein (K80Q) mimicking constitutive acetylation of HSF1 failed to confer protection against glucolipotoxicity. Finally, we showed that expression of HSF1 and its target genes were altered in islets from diabetic GK rats, suggesting that this pathway could participate in the pathophysiology of diabetes and constitutes a potential site for therapeutic intervention. CONCLUSIONS/INTERPRETATION: Our results unravel a new mechanism by which HSF1 inhibition is required for glucolipotoxicity-induced beta cell apoptosis. Restoring HSF1 activity may represent a novel strategy for the maintenance of a functional beta cell mass. Our study supports the therapeutic potential of HSF1/heat shock protein-targeting agents in diabetes treatment.


Assuntos
Glucose/farmacologia , Fatores de Transcrição de Choque Térmico/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ácido Palmítico/farmacologia , Fatores de Transcrição/metabolismo , Acetilação/efeitos dos fármacos , Animais , Apoptose/genética , Apoptose/fisiologia , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Fatores de Transcrição de Choque Térmico/genética , Humanos , Masculino , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Ratos
2.
Diabetes ; 63(12): 4197-205, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25008178

RESUMO

γ-Aminobutyric acid (GABA) exerts protective and regenerative effects on mouse islet ß-cells. However, in humans it is unknown whether it can increase ß-cell mass and improve glucose homeostasis. To address this question, we transplanted a suboptimal mass of human islets into immunodeficient NOD-scid-γ mice with streptozotocin-induced diabetes. GABA treatment increased grafted ß-cell proliferation, while decreasing apoptosis, leading to enhanced ß-cell mass. This was associated with increased circulating human insulin and reduced glucagon levels. Importantly, GABA administration lowered blood glucose levels and improved glucose excursion rates. We investigated GABA receptor expression and signaling mechanisms. In human islets, GABA activated a calcium-dependent signaling pathway through both GABA A receptor and GABA B receptor. This activated the phosphatidylinositol 3-kinase-Akt and CREB-IRS-2 signaling pathways that convey GABA signals responsible for ß-cell proliferation and survival. Our findings suggest that GABA regulates human ß-cell mass and may be beneficial for the treatment of diabetes or improvement of islet transplantation.


Assuntos
Glicemia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental , GABAérgicos/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Transplante das Ilhotas Pancreáticas , Ácido gama-Aminobutírico/farmacologia , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Glucagon/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
3.
Gynecol Endocrinol ; 29(6): 583-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23656386

RESUMO

Kisspeptins are known to be the principle regulators of the hypothalamic-pituitary gonadal (HPG) axis. In addition, the role of pituitary adenylate cyclase-activating polypeptide (PACAP) in the regulation of pituitary gonadotropins has been elucidated. We measured plasma concentrations of kisspeptin and PACAP and determined whether the levels of these peptides varied in proportion to circulating gonadotropin levels. Plasma luteinizing hormone (LH) levels were higher in postmenopausal women and in patients with premature ovarian failure (POF) and lower in patients with idiopathic hypogonadotropic hypogonadism (IHH) compared with the LH level in normally menstruating women. Similarly, serum follicle-stimulating hormone levels were higher in postmenopausal women and in patients with POF but lower in pregnant women and patients with IHH compared with normally menstruating women. Plasma levels of kisspeptins were significantly higher in pregnant women compared with normally menstruating women. However, no significant differences were observed in postmenopausal women, patients with POF, and patients with IHH. On the other hand, plasma levels of PACAP were significantly lower in pregnant women, patients with POF, and in IHH patients when compared with normally menstruating women. No significant differences were observed in PACAP concentration between postmenopausal women and in normally menstruating women. Our observations suggest that the serum levels of kisspeptins and PACAP did not correlate with variations in serum gonadotropin levels.


Assuntos
Gonadotropinas/sangue , Kisspeptinas/sangue , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/sangue , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Análise Química do Sangue , Feminino , Humanos , Infertilidade Feminina/sangue , Ciclo Menstrual/sangue , Pessoa de Meia-Idade , Pós-Menopausa/sangue , Gravidez/sangue , Adulto Jovem
5.
J Obstet Gynaecol Res ; 39(1): 406-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22672538

RESUMO

We describe a case of non-classical congenital adrenal hyperplasia (NCCAH) due to 21-hydroxylase deficiency in a 30-year-old Japanese woman who achieved pregnancy after treatment of primary amenorrhea. Hirsutism and clitoromegaly were present. Ultrasound examination showed polycystic appearance of the ovary. Luteinizing hormone-releasing hormone (LH-RH) test resulted in exaggerated LH response, showing a polycystic ovary syndrome (PCOS) pattern. The diagnosis was based on the elevated intial levels of 17-hydroxyprogesterone (55 ng/mL) and dihydroepiandosterone (7780 ng/mL). The first withdrawal bleeding occurred within 6 weeks after treatment with hydrocortisone (20 mg/day) combined with conjugated estrogens (1.25 mg/day) and medroxyprogesterone acetate (10 mg/day), which were continued for five courses. The bleeding remained cyclic every 28 days with maintenance doses of hydrocortisone. Subsequently, ovulation was induced using clomiphene citrate (100 mg/day). Pregnancy was achieved at the second attempt of ovulation induction and was within 10 months after initial presentation. Continuing hydrocortisone, the patient delivered a healthy baby at term.


Assuntos
Hiperplasia Suprarrenal Congênita/complicações , Amenorreia/tratamento farmacológico , Estrogênios Conjugados (USP)/uso terapêutico , Hidrocortisona/uso terapêutico , Acetato de Medroxiprogesterona/uso terapêutico , Hiperplasia Suprarrenal Congênita/tratamento farmacológico , Adulto , Amenorreia/complicações , Quimioterapia Combinada , Feminino , Hirsutismo/complicações , Hirsutismo/tratamento farmacológico , Humanos , Recém-Nascido , Indução da Ovulação , Gravidez , Resultado da Gravidez , Resultado do Tratamento
6.
Mol Cell Endocrinol ; 365(2): 139-45, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23103668

RESUMO

We used somatolactotroph GH3 cells to examine changes in response to stimulation with thyrotropin-releasing hormone (TRH) and pituitary adenylate cyclase-activating polypeptide (PACAP) after sustained treatment with these peptides. TRH and PACAP increased prolactin promoter activity in mock- and PACAP type 1 receptor (PAC1R)-transfected cells. When the cells were pretreated with TRH for 48 h, the response of the prolactin promoter to both TRH and PACAP was diminished. Similarly, in PAC1R-transfected GH3 cells pretreated with PACAP, the effects of TRH and PACAP on the prolactin promoter were eliminated. The stimulation of prolactin mRNA expression by TRH and PACAP was eliminated by prolonged pretreatment with these peptides in PAC1R-transfected cells. Both the serum response element (SRE) promoters and cAMP response element (CRE) promoters were activated by TRH and PACAP in either mock- or PAC1R-transfected cells. Pretreatment for 48 h with TRH also eliminated the effects of TRH and PACAP on the SRE and CRE promoters, and pretreatment of PAC1R-transfected cells with PACAP for 48 h reduced the responses of the SRE and CRE promoters to TRH and PACAP. These observations demonstrated that sustained stimulation with TRH and PACAP desensitizes their own and each other's receptors.


Assuntos
Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Prolactina/biossíntese , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores do Hormônio Liberador da Tireotropina/genética , Hormônio Liberador de Tireotropina/fisiologia , Animais , Células Cultivadas , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Genes Reporter , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Adeno-Hipófise/citologia , Cultura Primária de Células , Prolactina/genética , Regiões Promotoras Genéticas , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Elemento de Resposta Sérica
7.
Biol Reprod ; 88(2): 35, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23197164

RESUMO

The pituitary gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are mainly under the control of hypothalamic gonadotropin-releasing hormone (GnRH), which regulates male and female gonadal function. GnRH is released in a pulsatile manner from the hypothalamus, and the frequency of GnRH pulses determines the dominance of output of LH and FSH from pituitary gonadotrophs. That is, more rapid pulses of GnRH preferentially increase synthesis and secretion of LH, whereas FSH is preferentially stimulated by slower GnRH pulses. The detailed mechanisms underlying this phenomenon remain unknown. Pituitary adenylate cyclase-activating polypeptide (PACAP) was originally identified as a hypothalamic activator of cAMP production in pituitary cells. PACAP is produced within the pituitary gonadotroph as well as in the central nervous system. PACAP stimulates gonadotropin alpha-, LHbeta-, and FSHbeta-subunits as well as receptors for GnRH in the pituitary gonadotropin-secreting cells. In addition, its own receptor, PACAP type I receptor (PAC1R), is also regulated by PACAP in gonadotrophs. GnRH stimulates expression of PACAP as well as PAC1R, and lower frequencies of GnRH pulses preferentially increase PACAP and PAC1R expression in gonadotrophs. Increasing concentrations of PACAP further increase the levels of gonadotropin subunit and that increasing amounts of PAC1R in gonadotrophs potentiates the effects of PACAP or GnRH on gonadotropin subunit expression. In addition, we have observed that GnRH-increased FSHbeta-subunit expression was prevented in the presence of PAC1R antagonist. These observations suggest the involvement of locally produced PACAP and its PAC1R in the differential regulation of specific gonadotropin subunit expression by pulsatile GnRH stimulation. Here, we review the possible involvement of PACAP and its PAC1R in gonadotropin control on the basis of our observations with gonadotroph cell lines.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante Subunidade beta/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Hipófise/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Animais , Linhagem Celular , Subunidade beta do Hormônio Folículoestimulante/genética , Folistatina/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Gonadotrofos/citologia , Gonadotrofos/efeitos dos fármacos , Gonadotrofos/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Camundongos , Modelos Animais , Hipófise/citologia , Hipófise/efeitos dos fármacos
8.
Neuro Endocrinol Lett ; 33(6): 608-13, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23160221

RESUMO

OBJECTIVES: Sex steroid hormones play roles in the regulation of pituitary hormone synthesis and secretion. Here we investigated the role of estradiol (E2) and progesterone (P4) on pituitary gonadotropin luteinizing hormone (LH)ß- and follicle stimulating hormone (FSH)ß-transcriptional activity in a single colony of gonadotroph LßT2 cells. METHODS: Pituitary gonadotroph cell line, LßT2 cells were used in this study. Cells were transfected with LHß- or FSHß-subunit promoter region-linked luciferase vector, and stimulated with gonadotropin-releasing hormone (GnRH) in the presence or absence of sex steroids. Transcriptional activity for LHß- and FSHß-subunit were determined by luciferase assay. Effects of sex steroids on cell proliferation was also determined by measurement of 5-bromoe-2'-deoxyuridine (BrdU) incorporation. RESULTS: The basal promoter activity of the LHß subunit was not modulated by 10 nM E2, but gonadotropin releasing hormone (GnRH)-induced LHß promoter activity was significantly increased by the same concentration of E2. Similarly, although the basal FSHß promoter was not modulated by 10 nM E2, GnRH-induced FSHß promoters were significantly potentiated in the presence of E2. One micromole E2 modulated neither basal nor GnRH-induced LHß and FSHß promoters. On the other hand, basal LHß promoter activity was enhanced by 1 µM P4, but the stimulatory response of GnRH on LHß promoters was significantly inhibited in the presence of 1 µM P4. Similar to LHß promoters, the basal activity of the FSHß promoter was increased by 1 µM P4; however, the response to GnRH was not modulated in the presence of P4. Ten micromoles P4 modified neither basal nor GnRH-induced promoter activity for LHß and FSHß. E2 had no antagonistic effect on P4-induced basal promoter activities of LHß or FSHß. A cell proliferation assay showed that neither E2 nor P4 modulated the growth of LßT2 cells, even in the presence or absence of GnRH. CONCLUSION: These observations suggest that both E2 and P4 uniquely modulate basal and GnRH-stimulated gonadotropin promoters without affecting cell growth.


Assuntos
Estradiol/metabolismo , Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Luteinizante Subunidade beta/genética , Hipófise/fisiologia , Progesterona/metabolismo , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Estradiol/farmacologia , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Luciferases/genética , Hormônio Luteinizante Subunidade beta/metabolismo , Hipófise/citologia , Hipófise/efeitos dos fármacos , Progesterona/farmacologia , Regiões Promotoras Genéticas/fisiologia , Transfecção
9.
Endocr J ; 59(10): 867-79, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22785235

RESUMO

We examined the effects of sex steroids on prolactin promoter activity in rat somatolactotrophic GH3 cells. Both estradiol (E2) and progesterone (P4) were found to inhibit basal prolactin promoter activity, but to potentiate Thyrotropin-releasing hormone (TRH)-induced prolactin promoter activity. P4 had a greater inhibitory effect on basal prolactin promoter activity than E2, and P4 also potentiated TRH-induced prolactin promoter more potently than E2. Combined treatment with E2 and P4 further increased TRH-induced prolactin promoter activity. E2 and P4 also both reduced basal serum response element (SRE) promoter activity, and increased TRH-induced SRE promoter activity. Combination treatment with E2 and P4 reduced basal activity of SRE promoter and increased TRH-induced SRE activity more potently than E2 or P4 alone. In contrast, basal cAMP response element (CRE) promoter activity was not influenced by either E2 or P4, although TRH-induced CRE promoter was potentiated by each of these steroids, and was further increased by E2 and P4 combination treatment. Both E2 and P4 increased TRH-induced extracellular signal-regulated kinase (ERK) phosphorylation; however, intracellular cAMP levels was not influenced by E2 or P4. TRH-induced CRE promoter was inhibited by mitogen-activated protein kinase/ERK kinase (MEK) inhibitor and was increased by overexpression of MEK kinase (MEKK). This study showed that ERK and SRE transcriptional pathways, but not the cAMP/CRE pathway, may be involved in the suppression of basal prolactin promoter activity, whereas both the ERK/SRE and MAP kinase-mediated CRE pathways appear to be involved in the increased transcriptional efficiency of the prolactin promoter induced by TRH stimulation.


Assuntos
Estradiol/farmacologia , Lactotrofos/metabolismo , Progesterona/farmacologia , Prolactina/biossíntese , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Elemento de Resposta Sérica/genética , Elemento de Resposta Sérica/fisiologia , Hormônio Liberador de Tireotropina/farmacologia
10.
Endocr J ; 59(7): 631-40, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22641014

RESUMO

The role of Neurokinin B (NKB) and Dynorphin A (Dyn) in the regulation of the hypothalamic pituitary axis is an important area of recent investigation. These peptides are critical for the rhythmic release of GnRH, which subsequently stimulates the secretion of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The present study utilized the gonadotroph cell line LßT2 and the somatolactotroph GH3 cell line to examine the possible role of these peptides in pituitary hormone secretion. The NKB receptor (NK3R) and the Dyn receptor (the κ-opiate receptor (KOR)) were both detected in LßT2 cells and GH3 cells. NKB, by itself, failed to increase gonadotropin LHß and FSHß promoter activities and did not modulate the effects of GnRH on gonadotropin promoter activity. In GH3 cells, NKB significantly increased TRH-induced PRL promoter activity although NKB alone did not have an effect on basal PRL promoter activity. Dyn had no effect on gonadotropin promoters alone or in combination with GnRH stimulation. PRL promoters stimulated by TRH were not significantly changed by Dyn. TRH-induced PRL promoter activity was further increased in the presence of higher concentrations of NKB, whereas Dyn did not have a significant effect on the PRL promoter even at a high concentration. In addition, TRH-induced ERK (Extracelluar signal-regulated kinase) activation was enhanced in the presence of NKB. Our current study demonstrated that NKB had a stimulatory effect on PRL expression in a PRL-producing cell, but had no effect on gonadotropin secretion from a gonadotroph cell line.


Assuntos
Dinorfinas/fisiologia , Gonadotrofos/metabolismo , Lactotrofos/metabolismo , Neurocinina B/fisiologia , Somatotrofos/metabolismo , Animais , Linhagem Celular , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Gonadotrofos/efeitos dos fármacos , Gonadotropinas/genética , Gonadotropinas/metabolismo , Lactotrofos/efeitos dos fármacos , Neurocinina B/genética , Neurocinina B/metabolismo , Neurocinina B/farmacologia , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Prolactina/genética , Prolactina/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores de Taquicininas/genética , Receptores de Taquicininas/metabolismo , Somatotrofos/efeitos dos fármacos , Transfecção , Receptor de Nociceptina
11.
J Signal Transduct ; 2012: 198527, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22235371

RESUMO

The frequency of gonadotropin-releasing hormone (GnRH) pulse secreted from the hypothalamus differently regulates the expressions of gonadotropin subunit genes, luteinizing hormone ß (LHß) and follicle-stimulating hormone ß (FSHß), in the pituitary gonadotrophs. FSHß is preferentially stimulated at slower GnRH pulse frequencies, whereas LHß is preferentially stimulated at more rapid pulse frequencies. Several signaling pathways are activated, including mitogen-activated protein kinase (MAPK), protein kinase C, calcium influx, and calcium-calmodulin kinases, and these may be preferentially regulated under certain conditions. Previous studies demonstrated that MAPK pathways, especially the extracellular signal-regulated kinase (ERK), play an essential role for induction of gonadotropin subunit gene expression by GnRH, whereas, MAPK phosphatases (MKPs) inactivate MAPKs through dephosphorylation of threonine and/or tyrosine residues. MKPs are also induced by GnRH, and potential feedback regulation between MAPK signaling and MKPs within the GnRH signaling pathway is evident in gonadotrophs. In this paper, we reviewed and mainly focused on our observations of the pattern of ERK activation and the induction of MKP by different frequencies of GnRH stimulation.

12.
J Med Case Rep ; 5: 567, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22152284

RESUMO

INTRODUCTION: While thyrotropin-releasing hormone is known to be a prolactin-release stimulating factor, thyrotropin-releasing hormone-tartrate and its derivative, taltirelin hydrate, are used for the treatment of spinocerebellar degeneration, a degenerative disease characterized mainly by motor ataxia. We report the case of a patient being treated with a thyrotropin-releasing hormone preparation for spinocerebellar degeneration who developed amenorrhea after a dose increase. Her hormonal background was analyzed and the effect of prolonged exposure to thyrotropin-releasing hormone on pituitary prolactin-producing cells was examined in vitro. CASE PRESENTATION: Our patient was a 36-year-old Japanese woman who experienced worsening of gait disturbance at around 23 years of age, and was subsequently diagnosed as having spinocerebellar degeneration. She had been treated with thyrotropin-releasing hormone-tartrate for four years. Taltirelin hydrate was added to the treatment seven months prior to her presentation, followed by an improvement in gait disturbance. Around the same period, she started lactating and subsequently developed amenorrhea three months later. Taltirelin hydrate was discontinued and she was referred to our hospital. She was found to have normal sex hormone levels. A thyrotropin-releasing hormone provocation test showed a normal response of thyroid-stimulating hormone level and an over-response of prolactin at 30 minutes (142.7 ng/mL). Resumption of menstruation was noted three months after dose reduction of thyrotropin-releasing hormone. In our in vitro study, following long-term exposure to thyrotropin-releasing hormone, cells from the rat pituitary prolactin-producing cell line GH3 exhibited an increased basal prolactin promoter activity but showed a marked decrease in responsiveness to thyrotropin-releasing hormone. CONCLUSIONS: Physicians should be aware of hyperprolactinemia-associated side effects in patients receiving thyrotropin-releasing hormone treatment. Long-term treatment with a thyrotropin-releasing hormone preparation might cause a large amount of prolactin to accumulate in prolactin-producing cells and be released in response to exogenous thyrotropin-releasing hormone stimulation.

13.
Biochem Biophys Res Commun ; 410(2): 295-300, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21658369

RESUMO

We examined the effect of the pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1R) on gonadotropin-releasing hormone (GnRH)-induced gonadotropin subunit promoter activities using the LßT2 gonadotroph cell line. In mock transfected cells, GnRH-increased LHß and FSHß promoters up to 2.74 ± 0.15-fold and 1.6 ± 0.05-fold respectively. When cells were transfected with PAC1R, both LHß and FSHß promoter activities were further increased up to 6.1 ± 0.87-fold and 2.22 ± 0.43-fold following GnRH stimulation. ERK phosphorylation, serum response element (SRE) promoters, and cAMP response element (CRE) promoters stimulated by GnRH were also potentiated in the presence of increasing amounts of PAC1R. The EC50 values for LHß and FSHß gene transcription by GnRH were significantly decreased by overexpression of PAC1R. PACAP 6-38, a PACAP receptor antagonist, failed to reduce the effect of GnRH on gonadotropin promoter activities in PAC1R overexpressing cells, suggesting that the potentiation of the effects of GnRH by PAC1R expression was not related to an autocrine mechanism of PACAP produced in the gonadotrophs. Our current results show that the action of GnRH in the regulation of gonadotropin subunit expression is enhanced by the presence of PAC1Rs.


Assuntos
Regulação da Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Linhagem Celular , Expressão Gênica , Camundongos , Regiões Promotoras Genéticas
14.
Mol Cell Endocrinol ; 339(1-2): 172-9, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21539889

RESUMO

In this present study, we investigated the role of pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor, PACAP type I receptor (PAC1R) on prolactin synthesis in pituitary somatolactotroph GH3 cells. PACAP increased prolactin promoter activity up to 1.3 ± 0.1-fold. This increase, while significant, was less than the increase resulting from thyrotropin-releasing hormone (TRH) stimulation. By transfection of a PAC1R expression vector to the cells, the response to PACAP on prolactin promoter activity was dramatically potentiated to a degree proportional to the amount of PAC1R transfected. In the PAC1R expressing GH3 cells, TRH and PACAP alone increased prolactin promoter up to 3.3 ± 0.3-fold and 4.9 ± 0.2-fold, respectively, and combined treatment with TRH and PACAP further increased prolactin promoters up to 6.8 ± 0.6-fold. PACAP binds both Gs- and Gq-coupled receptors and stimulates adenylate cyclase/cAMP and protein kinase C/extracellular signal-regulated kinase (ERK) signaling pathways. PACAP increased ERK phosphorylation in PAC1R expressing cells to the same degree as TRH. Combined treatment with TRH and PACAP had a synergistic effect on ERK activation. GH3 cells produce both prolactin and growth hormone. Stimulation of GH3 cells with TRH significantly increased the mRNA level of prolactin and attenuated growth hormone mRNA expression. PACAP increased both prolactin and growth hormone mRNA levels, particularly in PAC1R expressing cells. In addition, increasing amount of PAC1R in GH3 cells potentiated the action of TRH on prolactin promoter activity, as well as on ERK phosphorylation. PAC1R was induced by PACAP itself, but not by TRH. Our current study demonstrates that PACAP and its PAC1R, functions as a stimulator of prolactin alone or with TRH in prolactin producing cells.


Assuntos
Lactotrofos/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Hipófise/citologia , Prolactina/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Somatotrofos/metabolismo , Animais , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes Reporter , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Lactotrofos/efeitos dos fármacos , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Fosforilação , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Prolactina/genética , Regiões Promotoras Genéticas , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Somatotrofos/efeitos dos fármacos , Hormônio Liberador de Tireotropina/farmacologia , Transcrição Gênica
15.
Regul Pept ; 167(2-3): 227-32, 2011 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21329727

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor, PACAP type 1 receptor (PAC1-R) play an important role in the induction of pituitary gonadotropins. In this present study, we examined whether the PAC1-R was involved in the action of gonadotropin-releasing hormone (GnRH) on gonadotropin FSHß subunit expression. In a static culture, GnRH stimulation significantly increased PAC1-R expression as well as PACAP gene expression in the gonadotroph cell line, LßT2. Stimulation with low frequency GnRH pulses, which preferentially increase FSHß, increased the expression of both the PAC1-R and the PACAP genes to a greater extent than did high frequency pulses. In the determination of transcriptional activity, the GnRH antagonist, cetrotide inhibited GnRH-induced FSHß promoter activity completely, but PACAP6-38, a PACAP antagonist, had no effect on GnRH-induced FSHß promoter activity. As expected, PACAP-induced FSHß promoter activity was significantly prevented by PACAP6-38, but was not affected by cetrotide. PACAP6-38, however, significantly prevented GnRH-increased FSHß mRNA expression. These observations suggest that GnRH-induced FSHß gene expression is stimulated partially through PAC1-R by gonadotrophs producing PACAP or PAC1-R.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Liberador de Gonadotropina/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Linhagem Celular , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo
16.
Biol Reprod ; 84(5): 996-1004, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21228211

RESUMO

In pituitary gonadotrophs, GnRH induces expression of the mitogen-activated protein kinases (MAPK3/1) dephosphorylating enzyme, dual-specificity phosphatase 1 (DUSP1). Here we examined DUSP1 expression levels following pulsatile GnRH stimulation of the LbetaT2 gonadotroph cells. DUSP1 expression was increased more prominently following high-frequency (every 30 min) GnRH pulse stimulation (7.02- ± 1.47-fold) than low-frequency (every 120 min) GnRH pulses (2.68- ± 0.09-fold). With high-frequency GnRH pulses, DUSP1 expression increased by 2.89- ± 0.32-fold 2 h after GnRH pulse initiation (four 5-min pulses). DUSP1 expression was not induced following lower frequency GnRH pulses, even when the GnRH concentration was increased. Under high-frequency conditions, MAPK3/1 phosphorylation was observed 10 min after the GnRH pulse and decreased to basal levels after 25 min. However, MAPK3/1 dephosphorylation did not occur concurrently with DUSP1 expression. Overexpression of MAP3K1, a kinase upstream of MAPK3/1, increased both the Lhb and the Fshb subunit promoter activities, which could be completely inhibited by cotransfection with DUSP1-expressing vectors. Serum response factor (Srf) promoter activities induced by MAP3K1 were also prevented by DUSP1 overexpression, confirming that MAPK3/1 has an important role in gonadotropin subunit gene expression. Both high- and low-frequency GnRH pulse stimulation failed to increase the Lhb and Fshb subunit gonadotropin gene expression levels upon DUSP1 overexpression. Our study demonstrates that DUSP1 is specifically expressed following high-frequency GnRH pulses and that this effect may participate in the differential regulation of gonadotropin subunit expression in association with MAPK3/1 phosphorylation.


Assuntos
Fosfatase 1 de Especificidade Dupla/biossíntese , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante Subunidade beta/metabolismo , Periodicidade , Hipófise/metabolismo , Animais , Linhagem Celular , Indução Enzimática , Subunidade beta do Hormônio Folículoestimulante/genética , Genes Reporter , Hormônio Luteinizante Subunidade beta/genética , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Concentração Osmolar , Fosforilação , Regiões Promotoras Genéticas , Ratos , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo
17.
Peptides ; 31(9): 1748-55, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20553777

RESUMO

We examined the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and the PACAP type 1 receptor (PAC1-R) mRNA following gonadotropin-releasing hormone (GnRH) stimulation using the gonadotroph cell line LbetaT2. GnRH stimulation increased PACAP and PAC1-R mRNA expression in a static culture. Increase in the cell surface density of the PAC1-R following transfection with PAC1-R expression vectors significantly increased gonadotropin LHbeta and FSHbeta subunit promoter activities following 100 nM PACAP stimulation. In addition, increasing concentrations of PACAP stimulation augmented the promoter activities for both LHbeta and FSHbeta in PAC-1R overexpressing cells. In the cells with PAC1-R, the effect of GnRH was further potentiated in the presence of PACAP from 5.31+/-0.93 to 9.89+/-0.38-fold for LHbeta and for FSHbeta subunit, respectively; from 2.58+/-0.31-fold by GnRH alone to 10.90+/-2.79-fold with PACAP. The combination treatment with GnRH and PACAP did not augment the ERK phosphorylation induced by GnRH alone. PACAP expectedly increased cAMP accumulation and this effect was significantly attenuated in the presence of GnRH. PACAP gene expression was more prominent following lower frequency GnRH pulses (every 120 min) in a perifused culture. Our results suggest that PACAP and PAC1-R are produced locally within the gonadotrophs following GnRH stimulation. They subsequently affect the gonadotrophs in an autocrine manner and modulate the GnRH pulse-dependent specific regulation of gonadotropin subunits.


Assuntos
Regulação da Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Subunidades Proteicas/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade beta do Hormônio Folículoestimulante , Genes Reporter , Hormônio Luteinizante Subunidade beta , Camundongos , Fosforilação , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Propriedades de Superfície , Fatores de Tempo
18.
Pituitary ; 13(3): 236-41, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20180027

RESUMO

Changes in LH and FSH levels were evaluated before and after metformin administration. In all 25 patients, plasma LH levels were significantly reduced after 3 months of metformin administration (500-1,500 mg/day). When patients were classified into a PCOS group (n = 12) or a non-PCOS group (n = 13), the reduction in LH levels only remained significant in the PCOS group. Plasma FSH levels were unchanged following metformin treatment when all patients were considered collectively and when patients were classified based on PCOS. LH/FSH ratio was significantly reduced only in the PCOS group. To examine the direct effect of metformin on gonadotropin-secreting cells, gonadotroph cell line, LbetaT2 was used for in vitro studies. Treatment of LbetaT2 cells with metformin modified neither the LHbeta nor the FSHbeta subunit promoter activity. The GnRH-induced LHbeta promoter activity was not modulated in the presence of metformin. In contrast, GnRH-induced FSHbeta promoter activity was significantly potentiated in the presence of metformin. Our results suggest that metformin does indeed modulate the basal level of LH and the LH/FSH ratio, albeit indirectly, particularly in the patients with PCOS. Additionally our results suggest that metformin does directly regulate FSH gene expression.


Assuntos
Gonadotrofos/efeitos dos fármacos , Gonadotropinas/sangue , Infertilidade Feminina/sangue , Infertilidade Feminina/tratamento farmacológico , Metformina/uso terapêutico , Adulto , Linhagem Celular , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/genética , Hormônio Liberador de Gonadotropina/sangue , Humanos , Infertilidade Feminina/genética , Hormônio Luteinizante/sangue , Hormônio Luteinizante/genética , Metformina/administração & dosagem , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética
19.
Biol Reprod ; 82(2): 352-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19846601

RESUMO

We examined the expression of dual specificity phosphatase 1 (DUSP1) by gonadotropin-releasing hormone (GnRH) stimulation and investigated the role of DUSP1 on gonadotropin gene expression using LbetaT2 gonadotroph cell line. DUSP1 expression was markedly increased 60 min after GnRH stimulation, and mitogen-activated protein kinase 3/1 (MAPK3/1) activation was gradually decreased after 60 min. GnRH-induced MAPK3/1 activation was completely inhibited by U0126, a MEK inhibitor, whereas GnRH-induced DUSP1 expression was partially inhibited by U0126. GnRH-induced DUSP1 induction was inhibited by triptolide, a diterpenoid triepoxide. In contrast, this compound potentiated MAPK3/1 activation. U0126 prevented GnRH-stimulated gonadotropin subunit promoter activation dose dependently, and 10 muM of U0126 reduced the effects of GnRH on the Lhb and Fshb promoters to 79.15% and 55.66%, respectively. GnRH-stimulated activation of Lhb and Fshb promoters as well as serum response factor (Srf) promoters were almost completely inhibited by triptolide, suggesting that this component had a nonspecific effect to the cells. Dusp1 siRNA reduced the expression of DUSP1 and augmented MAPK3/1 phosphorylation, but it did not increase of gonadotropin promoters. By overexpression of DUSP1, both GnRH-stimulated Lhb and Fshb promoters were significantly reduced. We have previously shown that insulin-like growth factor 1 (IGF1) increases MAPK3/1 but does not activate gonadotropin subunit promoters. IGF1 failed to induce DUSP1 expression. In addition, under pulsatile GnRH stimulation, DUSP1 expression was observed following high-frequency GnRH pulses but not following low-frequency pulses. Our study demonstrated that DUSP1, induced by GnRH, functions not only as an MAPK3/1-inactivating phosphatase but also as an important mediator in gonadotropin subunit gene expression regulation.


Assuntos
Fosfatase 1 de Especificidade Dupla/biossíntese , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/genética , Hipófise/enzimologia , Animais , Butadienos/farmacologia , Linhagem Celular , Diterpenos/farmacologia , Fosfatase 1 de Especificidade Dupla/genética , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Compostos de Epóxi/farmacologia , Subunidade beta do Hormônio Folículoestimulante/genética , Cinética , Hormônio Luteinizante Subunidade beta/genética , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Fenantrenos/farmacologia , Regiões Promotoras Genéticas/genética
20.
Mol Cell Endocrinol ; 307(1-2): 125-32, 2009 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-19533841

RESUMO

The purpose of the present study was to examine the signal transduction pathways involved in follistatin gene expression induced by GnRH in the LbetaT2 cell line. The LHbeta-subunit was predominantly increased by high frequency GnRH pulses (30 min interval); whereas low frequency pulses (120 min) increased FSHbeta. In a static culture, follistatin expression was significantly increased at 12 h (2.35 +/- 0.80-fold) after the addition of GnRH. Following pulsatile stimulation, follistatin mRNA was increased by high frequency GnRH pulses, but not by low frequency pulses. In a static culture, GnRH maximally activated extracellular signal-regulated kinase (ERK) 10 min (3.2 +/- 0.55-fold) after treatment. In addition, intracellular cAMP accumulated up to 2.1 +/- 0.76-fold. Follistatin promoter activity was significantly increased following transfection with either a constitutively active cAMP dependent protein kinase (PKA) or a constitutively active MEK kinase (MEKK). The induction of follistatin gene expression by GnRH was completely inhibited by H89, a protein kinase A inhibitor, and U0126, a MEK inhibitor. Follistatin gene expression was also activated by both PACAP and CPT-cAMP under static culture conditions. Maximal ERK activation levels were nearly identical regardless of GnRH pulse frequency; however, high frequency GnRH pulses elevated both the intracellular cAMP level as well as cAMP-response element (Cre) promoter activity. These results suggest that both the PKA and ERK pathways are necessary for the induction of the follistatin promoter. Furthermore, the intracellular cAMP level, but not ERK activity, determined whether follistatin was induced following high frequency GnRH pulses.


Assuntos
AMP Cíclico/metabolismo , Folistatina/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Gonadotrofos/citologia , Gonadotrofos/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Linhagem Celular , Células Clonais , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Folistatina/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Hormônio Luteinizante Subunidade beta/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Tionucleotídeos/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...