Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Toxicol Pathol ; 35(4): 321-331, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36406167

RESUMO

In subcutaneous tumor models, changes in the tumor microenvironment can lead to differences in therapeutic treatment responses between the subcutaneous and parent tumors. Accordingly, we generated a lung carcinogenesis model that combines genetically modified mice (Tg-rasH2 mice) with two-stage chemical carcinogenesis as an alternative to the subcutaneous tumor model. In this model, Tg-rasH2 mice were treated with 1-ethyl-1-nitrosourea, followed by butylhydroxytoluene. Mice developed lung adenomas five weeks after treatment initiation. Subsequently, anti-mouse PD-1 antibody (α-mPD-1) or isotype control was administered intraperitoneally twice a week for 4 weeks. Tumor growth was examined by measuring the relative tumor area in serially sliced lung histopathological specimens. No statistically significant differences were observed in the relative lung tumor areas between treated and control groups. A second experiment then examined the antitumor efficacy of α-mPD-1 combined with gemcitabine in a mouse model. Mice were treated identically as in Experiment 1, except that the treated group received once-weekly intraperitoneal injections of 10 mg/kg gemcitabine. In contrast to Experiment 1, the combined treatment significantly reduced the relative tumor areas in the lungs. This result also resembles that of a phase III clinical trial (ORIENT-12), showing that patients with non-small-cell lung carcinoma benefited from combination treatment with gemcitabine and the anti-human PD-1 antibody sintilimab. Thus, this mouse model could be a feasible means to preclinically evaluate the antitumor efficacy of different immunotherapy and chemotherapy drug combinations.

2.
Obstet Gynecol Int ; 2020: 7367403, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32328104

RESUMO

METHODS: We identified that 308 women who had undergone surgical repair of POP were followed up for at least 6 months. Recurrence rates of POP after tension-free vaginal mesh (TVM) surgery (n = 243), native tissue repair (NTR) (vaginal hysterectomy with colpopexy, anterior and posterior colpoplasty, or circumferential suturing of the levator ani muscles and apical repair by transvaginal sacrospinous ligament fixation (SSLF)) (NTR; n = 31), and laparoscopic sacrocolpopexy after subtotal hysterectomy (LSC; n = 34) were compared. Presence of mesh erosion was also recorded. RESULTS: Patients who underwent LSC were significantly younger (65.32 ± 3.23 years) than those who underwent TVM surgery (69.61 ± 8.31 years). After TVM surgery, the rate of recurrence (over POP-Q stage II) was 6.17% (15/243) and was highest in patients with advanced POP. The recurrence rate in patients who underwent NTR procedure was 3.23% (1/34) and that in patients who underwent LSC was 11.76% (4/11). There was no statistically significant difference in the recurrence rate between the three types of surgery. There were 13 cases (5.35%) of mesh erosion after TVM surgery and none after LSC surgery. The risk of mesh erosion was correlated with having had total TVM surgery but not with patient age or POP stage. Repeat procedures were performed in 5 women (2.14%) who underwent TVM surgery and 1 (2.94%) who underwent LSC. No patient underwent repeat surgery after NTR. There was no statistically significant difference in the reoperation rate between the three types of surgery. CONCLUSION: Our study suggested that TVM surgery, NTR, and LSC have comparable outcomes as for the postoperative recurrence rate and mesh erosion. However, the outcomes of each technique need to be carefully evaluated over a long period of time.

3.
Reprod Sci ; 27(3): 806-814, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32006244

RESUMO

Clomiphene citrate (CC) and letrozole stimulate the hypothalamic-pituitary-ovarian axis and are used widely as oral fertility drugs to induce folliculogenesis. We examined whether these drugs increase Kiss-1 expression in hypothalamic cell models. We utilized two hypothalamic cell models, mHypoA-50 and mHypoA-55, which originated from Kiss-1 neurons in the anteroventral periventricular (AVPV) nucleus and arcuate (ARC) nucleus of the mouse hypothalamus, respectively. The cells were stimulated with CC or letrozole, after which Kiss-1 mRNA expression was determined. CC stimulated Kiss-1 gene expression in mHypoA-50 and mHypoA-55 cells. The basal expression of Kiss-1 was significantly increased in the presence of estradiol (E2) in mHypoA-50 cells, and the CC-induced increase in Kiss-1 expression was not observed in the presence of E2 in these cells. In contrast, E2 did not modify the basal expression of Kiss-1 in mHypoA-55 cells, and CC-induced Kiss-1 expression was still observed in the presence of E2. The significant increase in Kiss-1 gene expression in mHypoA-50 and mHypoA-55 cells was blunted in the presence of estrogen receptor antagonists. Aromatase was expressed in mHypoA-50 and mHypoA-55 cells. Letrozole, an aromatase inhibitor, increased Kiss-1 expression in mHypoA-55 ARC cells but not in mHypoA-50 AVPV cells. Although the basal expression of Kiss-1 was increased by E2, letrozole did not modulate Kiss-1 expression in mHypoA-50 cells. Letrozole-induced Kiss-1 gene expression in mHypoA-55 cells was not modulated in the presence of E2. The fertility drugs CC and letrozole modulated Kiss-1 expression in hypothalamic cell models.


Assuntos
Clomifeno/administração & dosagem , Fármacos para a Fertilidade Feminina/administração & dosagem , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Letrozol/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Linhagem Celular , Estradiol/administração & dosagem , Antagonistas do Receptor de Estrogênio/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Camundongos , Receptores de Estrogênio/metabolismo
4.
Vet Pathol ; 57(1): 192-199, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31221040

RESUMO

After initiation with 7,12-dimethylbenz[a]anthracene (DMBA), the promoting potential of 12-O-tetradecanoylphorbol-13-acetate (TPA) on skin tumor development can be detected by an ultra-short-term skin carcinogenicity bioassay using Tg-rasH2 mice. In the present study, 10 chemicals were assessed using this ultra-short-term bioassay as a first step to validate this practical and easy-to-use skin carcinogenicity bioassay. These chemicals belonged to 4 categories: dermal vehicles (acetone, 99.5% ethanol, anhydrous ethanol, and Vaseline), skin noncarcinogens (oleic acid diethanolamine condensate, benzethonium chloride, and diisopropylcarbodiimide), skin tumor promoters (TPA and benzoyl peroxide), and a skin carcinogen (4-vinyl-1-cyclohexene diepoxide). In a first study, DMBA was used as the initiator at a dose of 50 µg according to previous data, but skin tumors were observed in the no-treatment and vehicle groups. Therefore, the dose of DMBA for skin tumor initiation was reevaluated using 12.5 or 25 µg, with 12.5 µg found to be sufficient for initiation activity. In the ultra-short-term assay, the vehicles and skin noncarcinogens were negative while the skin tumor promoters and the skin carcinogen were positive. The detection of skin tumor promotion and carcinogenicity was feasible in only 8 weeks. In conclusion, this carcinogenicity bioassay may represent a useful tool for the assessment of the carcinogenicity potential of topically applied chemicals.


Assuntos
Testes de Carcinogenicidade/métodos , Carcinógenos/administração & dosagem , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/administração & dosagem , Animais , Feminino , Genes ras/genética , Humanos , Camundongos , Camundongos Transgênicos , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente
5.
Urol J ; 16(6): 581-585, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31630387

RESUMO

PURPOSE: We retrospectively reviewed the postoperative outcomes of patients who underwent tension-free vaginal mesh (TVM) surgery in our institution. METHODS: In total, 195 TVM surgeries were performed at the Shimane University School of Medicine from January 2010 to May 2016 in patients with Pelvic Organ Prolapse-Quantification (POP-Q) stage II or higher. Perioperative complications and problems arising following surgery were assessed from medical charts. RESULTS: Among the 195 patients, only 1 patient required blood transfusion due to massive intraoperative blood loss. None of the patients experienced intraoperative complications, such as injury to the bladder or rectum during surgery. Mesh exposure was observed in 10 patients (5.1%). Overall, 6 of these 10 patients were asymptomatic, and surgical treatment was required in only 1 patient. Mesh exposure occurred at significantly higher frequencies in patients aged less than 60 years. Postoperative recurrence of POP, which was defined as recurrence over POP-Q stage 2, was noted in 13 of the 195 patients (6.6%). Re-operation was performed in 1 patient in whom recurrence was observed within 3 months postoperatively. Recurrence of POP was likely to occur in patients with higher POP-Q stages. Overall, 31 of the 195 patients (15.9%) required medication for postoperative stress urinary incontinence (SUI) after surgery. Among these, 2 patients underwent surgical treatment for SUI. CONCLUSION: Outcomes following the TVM procedure were satisfactory. However, caution should be exercisedagainst mesh exposure in younger patients and recurrence of POP in patients with advanced POP-Q stage.


Assuntos
Procedimentos Cirúrgicos em Ginecologia/métodos , Prolapso de Órgão Pélvico/cirurgia , Complicações Pós-Operatórias/prevenção & controle , Slings Suburetrais , Incontinência Urinária por Estresse/prevenção & controle , Micção/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Humanos , Incidência , Japão/epidemiologia , Pessoa de Meia-Idade , Prolapso de Órgão Pélvico/complicações , Complicações Pós-Operatórias/epidemiologia , Recidiva , Estudos Retrospectivos , Resultado do Tratamento , Incontinência Urinária por Estresse/epidemiologia , Incontinência Urinária por Estresse/fisiopatologia
6.
Biol Reprod ; 101(2): 405-415, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31167231

RESUMO

Kisspeptin (encoded by the Kiss-1 gene) in the arcuate nucleus (ARC) of the hypothalamus governs the hypothalamic-pituitary-gonadal (HPG) axis by regulating pulsatile release of gonadotropin-releasing hormone (GnRH). Meanwhile, kisspeptin in the anteroventral periventricular nucleus (AVPV) region has been implicated in estradiol (E2)-induced GnRH surges. Kiss-1-expressing cell model mHypoA-55 exhibits characteristics of Kiss-1 neurons in the ARC region. On the other hand, Kiss-1 expressing mHypoA-50 cells originate from the AVPV region. In the mHypoA-55 ARC cells, activin significantly increased Kiss-1 gene expression. Follistatin alone reduced Kiss-1 expression within these cells. Interestingly, activin-induced Kiss-1 gene expression was completely abolished by follistatin. Inhibin A, but not inhibin B reduced Kiss-1 expression. Activin-increased Kiss-1 expression was also abolished by inhibin A. Pretreatment of the cells with follistatin or inhibin A significantly inhibited kisspeptin- or GnRH-induced Kiss-1 gene expression in mHypoA-55 cells. In contrast, in the mHypoA-50 AVPV cell model, activin, follistatin, and inhibin A did not modulate Kiss-1 gene expression. The subunits that compose activin and inhibin, as well as follistatin were expressed in both mHypoA-55 and mHypoA-50 cells. Expression of inhibin ßA and ßB subunits and follistatin was much higher in mHypoA-55 ARC cells. Furthermore, we found that expression of the inhibin α subunit and follistatin genes was modulated in the presence of E2 in mHypoA-55 ARC cells. The results of this study suggest that activin, follistatin, and inhibin A within the ARC region participate in the regulation of the HPG axis under the influence of E2.


Assuntos
Ativinas/farmacologia , Folistatina/farmacologia , Inibinas/farmacologia , Kisspeptinas/metabolismo , Animais , Linhagem Celular , Folistatina/genética , Folistatina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas/genética , Camundongos , Subunidades Proteicas
7.
Gen Comp Endocrinol ; 270: 60-66, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30316762

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor are broadly distributed in the brain, and PACAP is known to work as a multifunctional peptide. However, it is still largely unknown how PACAP affects the hypothalamic-pituitary-gonadal (HPG) axis. In this study, we examined the effect of PACAP on hypothalamic kisspeptin expression, a known regulator of gonadotropin-releasing hormone. We used two hypothalamic cell models, mHypoA-50 and mHypoA-55, which were originated from kisspeptin-expressing neuron in anterioventral periventricular nucleus and arcuate nucleus regions in the hypothalamus, respectively. Expression of Kiss-1 gene, which encodes kisspeptin, was significantly increased by PACAP stimulation in both mHypoA-50 and mHypoA-55 cells, by up to 2.69 ±â€¯0.93-fold and 4.89 ±â€¯1.13-fold, respectively. PACAP6-38, a PACAP receptor antagonist did not antagonize the action of PACAP on Kiss-1 gene expression but increased Kiss-1 gene by itself in these cells. PACAP-induced Kiss-1 gene expression in both mHypoA-50 and mHypoA-55 cells was almost completely prevented in the presence of H89, a protein kinase A inhibitor. PACAP was expressed in both these hypothalamic cell models and its expression was up-regulated by estradiol in mHypoA-50 cells but not in mHypoA-55 cells. Stimulation of mHypoA-50 and mHypoA-55 cells with PACAP increased the expression levels of corticotropin-releasing hormone and neurotensin, both of which could modulate HPG axis. Our present observations suggest that hypothalamic PACAP might modulate the HPG axis by directly or indirectly modulating Kiss-1 gene expression.


Assuntos
Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Expressão Gênica
8.
Endocrine ; 63(1): 101-111, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30255291

RESUMO

Hypothalamic kisspeptin is a known principal activator of gonadotropin-releasing hormone neurons and governs the hypothalamic-pituitary-gonadal axis. Previous reports have shown that kisspeptin is also released into the hypophyseal portal circulation and directly affects the anterior pituitary. In this study, we examined the direct effect of kisspeptin on pituitary prolactin-producing cells. The rat pituitary somatolactotroph cell line GH3 expresses the kisspeptin receptor (Kiss1R); however, in these cells, kisspeptin failed to stimulate prolactin-promoter activity. When GH3 cells overexpressed Kiss1R, kisspeptin clearly increased prolactin-promoter activity, with a concomitant increase in extracellular signal-regulated kinase (ERK) and cAMP/protein kinase A (PKA) signaling pathways. In the experiments using GH3 cells overexpressing Kiss1R, kisspeptin did not potentiate thyrotropin-releasing hormone (TRH)-induced prolactin-promoter activity, but it potentiated the pituitary adenylate cyclase-activating polypeptide-induced prolactin-promoter activity, with a concomitant enhancement of ERK and PKA signaling pathways. Although the basal and TRH-induced prolactin-promoter activities were not modulated by increasing amounts of Kiss1R expression in GH3 cells, kisspeptin-stimulated prolactin-promoter activity was increased by the amount of Kiss1R overexpression. Endogenous Kiss1r mRNA expression in GH3 cells was significantly increased by treatment with estradiol (E2) but not by TRH. In addition, kisspeptin's ability to stimulate prolactin-promoter activity was restored after E2 treatment in non-transfected GH3 cells. Our current observations suggest that kisspeptin might have a direct effect on prolactin expression in the anterior pituitary prolactin-producing cells under the influence of E2, which may regulate Kiss1R expression and function.


Assuntos
Regulação da Expressão Gênica/genética , Kisspeptinas/genética , Prolactina/biossíntese , Prolactina/genética , Receptores de Kisspeptina-1/genética , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Hipófise/metabolismo , Regiões Promotoras Genéticas/genética , Ratos , Transdução de Sinais/genética , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo
9.
Reprod Sci ; 26(9): 1249-1255, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30458681

RESUMO

Kisspeptin, encoded by the Kiss-1 gene, plays a crucial role in reproductive function by governing the hypothalamic-pituitary-gonadal axis. The recently established Kiss-1-expressing cell model mHypoA-50 displays characteristics of neuronal cells of the anteroventral periventricular (AVPV) region of the mouse hypothalamus. Because Kiss-1 gene expression in these cells is upregulated by estradiol (E2), mHypoA-50 cells are regarded as a valuable model for the study of Kiss-1-expressing neurons in the AVPV region. These cells also express RFamide-related peptide-3 (RFRP-3), a mammalian homolog of gonadotropin inhibitory hormone. The RFRP-3 expression in mHypoA-50 cells was increased by melatonin stimulation. In addition, E2 stimulation increased RFRP-3 expression in these cells. Treatment of the mHypoA-50 cells with exogenous RFRP-3 resulted in the increase of Kiss-1 messenger RNA expression within the cells; however, RFRP-3 did not modify gonadotropin-releasing hormone or kisspeptin-induced Kiss-1 gene expression in these cells. In addition, we found that RFRP-3 stimulation increased the expression of corticotropin-releasing hormone, which may be involved in E2-induced positive feedback in mHypoA-50 cells. Our observations suggest that RFRP-3 might be involved in positive feedback regulation by directly or indirectly increasing Kiss-1 gene expression.


Assuntos
Regulação da Expressão Gênica , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Linhagem Celular , Hormônio Liberador da Corticotropina/metabolismo , Estradiol/farmacologia , Hipotálamo/efeitos dos fármacos , Kisspeptinas/genética , Melatonina/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo
10.
Biol Reprod ; 99(6): 1216-1226, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29961889

RESUMO

The recently established immortalized hypothalamic cell model mHypoA-55 possesses characteristics similar to those of Kiss-1 neurons in the arcuate nucleus (ARC) region of the hypothalamus. Here, we show that Kiss-1 gene expression in these cells was downregulated by 17ß-estradiol (E2) under certain conditions. Both neurotensin (NT) and corticotropin-releasing hormone (CRH) were expressed in these cells and upregulated by E2. Stimulation of mHypoA-55 cells with NT and CRH significantly decreased Kiss-1 mRNA expression. A mammalian gonadotropin-inhibitory hormone homolog, RFamide-related peptide-3 (RFRP-3), was also found to be expressed in mHypoA-55 cells, and RFRP-3 expression in these cells was increased by exogenous melatonin stimulation. E2 stimulation also upregulated RFRP-3 expression in these cells. Stimulation of mHypoA-55 cells with RFRP-3 significantly increased the expression of NT and CRH. Furthermore, melatonin stimulation resulted in the increase of both NT and CRH mRNA expression in mHypoA-55 cells. On the other hand, in experiments using mHypoA-50 cells, which were originally derived from hypothalamic neurons in the anteroventral periventricular nucleus, Kiss-1 gene expression was upregulated by both NT and CRH, although E2 increased both NT and CRH expression, similarly to the mHypoA-55 cells. Our observations using the hypothalamic ARC cell model mHypoA-55 suggest that NT and CRH have inhibitory effects on Kiss-1 gene expression under the influence of E2 in association with RFRP-3 expression. Thus, these neuropeptides might be involved in E2-induced negative feedback mechanisms.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Hormônio Liberador da Corticotropina/farmacologia , Estradiol/farmacologia , Neuropeptídeos/farmacologia , Neurotensina/farmacologia , Animais , Linhagem Celular , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas/genética , Kisspeptinas/metabolismo , Melatonina/farmacologia , Camundongos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Neurotensina/genética , Neurotensina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Obstet Gynecol Int ; 2018: 7918071, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29805449

RESUMO

AIM: We compared the short-term effectiveness of tension-free vaginal mesh (TVM) surgery using the Capio SLIM suture capturing device and conventional TVM surgery for treatment of pelvic organ prolapse. METHODS: We retrospectively compared postoperative pain, urinary function, and length of hospital stay between 7 patients who underwent TVM surgery using the Capio device and 9 patients who underwent conventional TVM surgery. RESULTS: There was no significant between-group difference in mean age between the Capio TVM group and the conventional TVM group (76.0 ± 5.6 years and 72.5 ± 11.5 years) or in mean operating time (86.56 ± 23.33 min and 95.28 ± 23.88 min). Four of the 7 patients in the Capio TVM group could not sense the urge to urinate after removal of the urethral catheter, but all patients in the conventional TVM group did so. The volume of the first voluntary urination was significantly smaller in the Capio TVM group than that in the conventional TVM group (102.14 ± 80.57 mL versus 472.22 ± 459.43 mL). The mean residual urine volume after the first voluntary urination was greater in the Capio TVM group than that in the conventional TVM group (285.70 ± 233.82 mL versus 34.56 ± 73.31 mL). The number of catheter days and mean maximal volume of residual urine were significantly greater in the Capio TVM group. The mean postoperative hospital stay was 6.57 ± 1.83 days in the Capio TVM group and 3.2 ± 0.42 days in the conventional TVM group. Six patients who underwent Capio TVM surgery complained of deep-seated pain in the hip region. CONCLUSION: Urinary function may worsen postoperatively when the Capio TVM device is used in patients with pelvic organ prolapse.

12.
Reprod Med Biol ; 17(1): 71-76, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29371824

RESUMO

Purpose: To identify the factors that characterize cycles with empty zona pellucida (EZP). Methods: Thirty-six oocyte retrieval cycles from which EZP were collected and another 36 cycles from which no EZP was collected were compared. The patients were divided into three groups: those with no EZP collected during any cycle, those with EZP collected during all cycles, and those experiencing cycles both with and without EZP. Results: The mean number of oocytes collected per cycle was higher in the cycles with EZP than without EZP. The fertilization rate of the collected oocytes and the rate of good embryo formation were significantly lower in the cycles with EZP. No significant difference was observed between the three groups in terms of age, number of oocytes collected, or hormone levels before and after the oocyte retrieval. The fertilization and pregnancy rates were highest in the patients with no EZP being collected during any cycle, followed by those experiencing cycles both with and without EZP, and then by those with EZP collected during all cycles. Conclusion: The observation of lower fertilization, poor embryo formation, and a low pregnancy rate in the patients with EZP suggests the poor quality of oocytes that were collected with EZP in the same cycle.

13.
Reprod Med Biol ; 16(4): 386-391, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29259493

RESUMO

Purpose: Accumulating evidence indicates that hypothalamic kisspeptin plays a pivotal role in the regulation of the hypothalamic-pituitary-gonadal (HPG) axis. In this study, the direct action of the gamma-aminobutyric acid (GABA)A receptor agonist on kisspeptin-expressing neuronal cells was examined. Methods: A hypothalamic cell model of rat hypothalamic cell line R8 (rHypoE8) cells and primary cultures of neuronal cells from fetal rat brains were stimulated with a potent and selective GABAA receptor agonist, muscimol, to determine the expression of the KiSS-1 gene. Results: Stimulation of the rHypoE8 cells with muscimol significantly increased the level of KiSS-1 messenger (m)RNA expression. The ability of muscimol to increase the level of KiSS-1 mRNA also was observed in the primary cultures of the neuronal cells from the fetal rat brains. The muscimol-induced increase in KiSS-1 mRNA expression was completely inhibited in the presence of the GABAA receptor antagonist. Although muscimol increased the expression of KiSS-1, the natural compound, GABA, failed to induce the expression of KiSS-1 in the rHypoE8 cells. Muscimol did not modulate gonadotropin-releasing hormone expression in either the rHypoE8 cells or the primary cultures of the fetal rat brains. Conclusions: This study's observations suggest that the activation of the GABAA receptor modulates the HPG axis by increasing kisspeptin expression in the hypothalamic neurons.

14.
Biol Reprod ; 97(2): 240-248, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-29044434

RESUMO

Feeding-related metabolic factors exert regulatory influences on the hypothalamic-pituitary-gonadal axis. Glucagon-like peptide-1 (GLP-1) is an anorexigenic hormone synthesized from the ileum in response to food intake. The purpose of this study was to examine the direct effect of GLP-1 on hypothalamic kisspeptin and gonadotropin-releasing hormone (GnRH) expression using the rat clonal hypothalamic cell line rHypoE-8. GLP-1 significantly increased Kiss-1 mRNA expression in rHypoE-8 cells up to 1.94 ± 0.22-fold. This effect of GLP-1 on Kiss-1 gene expression was also observed in GT1-7 GnRH-producing neurons and in primary cultures of fetal rat brain. GLP-1 increased cAMP-mediated signaling, as determined by cAMP response element activity assays, but failed to activate extracellular signal-regulated kinase pathways. Another anorexigenic factor, leptin, similarly increased Kiss-1 mRNA levels up to 1.34 ± 0.08-fold in rHypoE-8 cells. However, combined treatment with GLP-1 and leptin failed to potentiate their individual effects on Kiss-1 mRNA expression. Gnrh mRNA expression was not significantly increased by GLP-1 stimulation in rHypoE-8, but kisspeptin significantly stimulated the expression of Gnrh mRNA in these cells. Our current observations suggest that the anorexigenic peptide GLP-1 directly regulates Kiss-1 mRNA expression in these hypothalamic cell lines and in neuronal cells of fetal rat brain and affects the expression of Gnrh mRNA.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/farmacologia , Kisspeptinas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Linhagem Celular , Feminino , Feto/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Kisspeptinas/genética , Leptina/farmacologia , Ratos
15.
Biol Reprod ; 96(5): 1043-1051, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28863434

RESUMO

We examined direct effect of kisspeptin on pituitary gonadotrophs. Kisspeptin-10 (KP10) significantly increased the promoter activities of the gonadotropin subunits, common alpha-glycoprotein (Cga), luteinizing hormone beta (Lhb), and follicle-stimulatinghormone beta (Fshb) in LbetaT2 cells overexpressing kisspeptin receptor (Kiss1r). KP10 and gonadotropin-releasing hormone (GnRH) increased gonadotropin subunit levels to similar degrees and combined treatment with GnRH and KP10 did not potentiate their individual effects. Adenylate cyclase-activating polypeptide 1 (ADCYAP1) also stimulates all three gonadotropin subunits. When cells were stimulated with both KP10 and ADCYAP1, expression of gonadotropin subunits was further increased compared to KP10 or ADCYAP1 alone. KP10 and GnRH dramatically increased serum response element (Sre) promoter levels but only slightly increased cAMP response element (Cre) promoter levels. Combined stimulation with KP10 and GnRH further increased Sre promoter levels. In contrast, ADCYAP1 slightly increased Sre promoter expression but did not modify the effect of KP10. However, ADCYAP1 increased Cre promoter to greater levels than KP10 alone, and combined treatment with KP10 and ADCYAP1 further increased Cre promoter expression. KP10 increased the expression of ADCYAP1 type I receptor (Adcyap1r) and the basal activity of the Cga promoter was increased at a higher Adcyap1r transfection level. The KP10-induced fold increase in all three gonadotropin subunit promoters was not altered by transfection with a higher amount of Adcyap1r vector. Our findings using model cells show that distinct signaling activation by ADCYAP1 potentiates the action of KP10. We also found that KP10 increases Adcyap1r expression.


Assuntos
Kisspeptinas/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Regulação da Expressão Gênica/genética , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Kisspeptinas/genética , Hormônio Luteinizante Subunidade beta/metabolismo , Camundongos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Plasmídeos , Regiões Promotoras Genéticas , Receptores de Kisspeptina-1/genética , Elemento de Resposta Sérica/genética
16.
Gynecol Endocrinol ; 33(9): 721-727, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28447478

RESUMO

Hypothalamic kisspeptin is integral to the hypothalamic-pituitary-gonadal axis by stimulating gonadotropin-releasing hormone (GnRH) release. GnRH is released from the hypothalamus in a pulsatile manner and determines the output of the gonadotropins. However, the effect of kisspeptin on GnRH-secreting cells remains unknown. In an experiment using static cultures of GT1-7 cells, kisspeptin did not significantly increase GnRH mRNA expression. However, when kisspeptin was administered to the cells in a pulsatile manner, GnRH mRNA expression was significantly increased. Primary cultures of fetal rat brain containing GnRH-expressing neurons responded to kisspeptin and increased GnRH mRNA expression by 1.65 ± 0.27-fold in the static condition. When cells were stimulated with kisspeptin in a pulsatile manner, GnRH mRNA expression was increased by up to 2.40 ± 0.21-fold. In perifused GT1-7 cells, pulsatile, but not continuous kisspeptin stimulation, effectively stimulated GnRH mRNA expression. To assess the level of stimulation of GnRH neurons by kisspeptin, the expression of c-fos was examined. In GT1-7 cells, kisspeptin stimulation in the static condition failed to increase c-fos mRNA expression. However, pulsatile kisspeptin stimulation increased c-fos mRNA by 2.31 ± 0.47-fold. Similar to the phenomenon observed in GT1-7 cells, pulsatile, but not static, kisspeptin stimulation significantly increased c-fos mRNA expression in the primary cultures of fetal rat brain. These observations suggest that pulsatile kisspeptin more effectively stimulates GnRH-producing cells to increase the production of GnRH.


Assuntos
Encéfalo/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/administração & dosagem , Neurônios/efeitos dos fármacos , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Neurônios/citologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos
17.
Gen Comp Endocrinol ; 246: 382-389, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087300

RESUMO

Hypothalamic kisspeptin plays a pivotal role in the regulation of the hypothalamic-pituitary-gonadal axis by stimulating gonadotropin-releasing hormone (GnRH) release into the portal circulation, with the subsequent release of gonadotropins. Kisspeptin and its receptor, the kisspeptin 1 receptor (Kiss1R), are also expressed in the pituitary gland. This study demonstrates the interaction between GnRH and kisspeptin within the pituitary gonadotrophs by altering their individual receptor expression. Our results show that kisspeptin and Kiss1R are expressed in the mouse pituitary gonadotroph cell line LßT2. Endogenous Kiss1R did not respond to kisspeptin and failed to stimulate gonadotropin LHß and FSHß expression in LßT2 cells; however, kisspeptin increased both LHß and FSHß promoter activity in Kiss1R-overexpressing LßT2 cells. Stimulating the cells with GnRH significantly increased Kiss1R expression, whereas kisspeptin increased the expression of the GnRH receptor (GnRHR) in these cells. Elevating the Kiss1R concentration led to an increase in the basal activities of gonadotropin LHß- and FSHß-subunit promoters. In addition, the level of kisspeptin-induced LHß promoter activity, but not that of FSHß, was significantly increased when a large number of Kiss1R expression vectors was introduced into the cells. The level of induction of GnRH-induced gonadotropin promoter activities was not significantly changed by increasing Kiss1R expression. Increasing the amount of GnRHR by overexpressing cellular GnRHR did not potentiate basal gonadotropin promoter activities; however, kisspeptin- and GnRH-stimulated increases in gonadotropin promoter activities were significantly potentiated (except GnRH-induced LHß promoters). The activities of serum response element-containing promoters were also modified in cells overexpressing Kiss1R or GnRHR. Our current observations demonstrate that GnRH and kisspeptin affect each other's function to stimulate gonadotropin subunit gene expression by reciprocally increasing the expression of their receptors.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Gonadotrofos/metabolismo , Kisspeptinas/metabolismo , Receptores LHRH/metabolismo , Animais , Camundongos
18.
Reprod Sci ; 24(2): 227-233, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27313117

RESUMO

Genetic studies in humans or in vivo studies using animals have shown that kisspeptin released from the hypothalamus controls secretion of gonadotropin-releasing hormone (GnRH) from GnRH neurons, and subsequently GnRH induces gonadotropin secretion from the anterior pituitary. Kisspeptin did not stimulate GnRH expression in the GnRH-producing cell line GT1-7. Thus, we cultured GnRH and kisspeptin neurons from whole fetal rat brain and examined the regulation of GnRH and kisspeptin. Expression of GnRH messenger RNA (mRNA) was unchanged by estradiol (E2) treatment in these primary cultures. In contrast, kisspeptin mRNA expression was increased 2.00 ± 0.23-fold by E2 treatment. When these cultures were stimulated by kisspeptin-10, GnRH mRNA was significantly increased up to 1.51 ± 0.35-fold. Expression of GnRH mRNA was also stimulated 1.84 ± 0.33-fold by GnRH itself. Interestingly, kisspeptin mRNA was significantly increased up to 2.43 ± 0.40-fold by kisspeptin alone. In addition, kisspeptin mRNA expression was significantly increased by stimulation with GnRH (1.46 ± 0.21-fold). Our observations demonstrated that kisspeptin, but not GnRH, was upregulated by E2 and that kisspeptin stimulates GnRH mRNA expression in primary cultures of whole fetal rat brain. Furthermore, GnRH and kisspeptin stimulate their own neurons to produce GnRH or kisspeptin, respectively.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Estradiol/farmacologia , Kisspeptinas/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ratos
19.
Int J Mol Sci ; 17(10)2016 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-27681724

RESUMO

Gonadotropin-releasing hormone (GnRH) and gonadotropins are indispensable hormones for maintaining female reproductive functions. In a similar manner to other endocrine hormones, GnRH and gonadotropins are controlled by their principle regulators. Although it has been previously established that GnRH regulates the synthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH)-both gonadotropins-from pituitary gonadotrophs, it has recently become clear that hypothalamic GnRH is under the control of hypothalamic kisspeptin. Prolactin, which is also known as luteotropic hormone and is released from pituitary lactotrophs, stimulates milk production in mammals. Prolactin is also regulated by hypothalamic factors, and it is thought that prolactin synthesis and release are principally under inhibitory control by dopamine through the dopamine D2 receptor. In addition, although it remains unknown whether it is a physiological regulator, thyrotropin-releasing hormone (TRH) is a strong secretagogue for prolactin. Thus, GnRH, LH and FSH, and prolactin are mainly regulated by hypothalamic kisspeptin, GnRH, and TRH, respectively. However, the synthesis and release of these hormones is also modulated by other neuropeptides in the hypothalamus. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a hypothalamic peptide that was first isolated from sheep hypothalamic extracts based on its ability to stimulate cAMP production in anterior pituitary cells. PACAP acts on GnRH neurons and pituitary gonadotrophs and lactotrophs, resulting in the modulation of their hormone producing/secreting functions. Furthermore, the presence of the PACAP type 1 receptor (PAC1R) has been demonstrated in these cells. We have examined how PACAP and PAC1R affect GnRH- and pituitary hormone-secreting cells and interact with their principle regulators. In this review, we describe our understanding of the role of PACAP and PAC1R in the regulation of GnRH neurons, gonadotrophs, and lactotrophs, which are regulated mainly by kisspeptin, GnRH, and TRH, respectively.

20.
J Obstet Gynaecol Res ; 42(10): 1379-1384, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27436485

RESUMO

We report a case of a woman who was incidentally diagnosed with acromegaly after referral for prolonged post-partum amenorrhea. A 25-year-old woman, gravida 2 para 1, had a normal transvaginal delivery and breastfeeding had been discontinued more than a year after delivery. Thereafter, spontaneous menstruation did not restart and she underwent hormonal therapy. Subsequently, she was referred to our university hospital for prolonged amenorrhea. Hormonal examination revealed severe hypogonadotropic hypogonadism (luteinizing hormone 0.5 mIU/mL, follicle-stimulating hormone < 1.0 mIU/mL, estradiol 13 pg/mL), along with a modest increase in serum prolactin levels (43.2 ng/mL). She also complained of milk ejection on nipple stimulation. Magnetic resonance imaging of the head was performed, because an organic abnormality in the central nervous system was suspected to be causing the amenorrhea, which revealed mass lesions extending from the pituitary fossa to the suprasellar area with similar signal intensity as the gray matter. In addition, bitemporal hemianopsia was observed on campimetry. After further examination, the patient was diagnosed with acromegaly.


Assuntos
Acromegalia/diagnóstico , Amenorreia/diagnóstico , Neoplasias Hipofisárias/diagnóstico por imagem , Acromegalia/complicações , Acromegalia/diagnóstico por imagem , Adulto , Amenorreia/complicações , Feminino , Humanos , Neoplasias Hipofisárias/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...