Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Obstet Gynaecol Res ; 49(1): 253-264, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36307043

RESUMO

AIM: The abnormal secretion of luteinizing hormone (LH) is one of the typical features of polycystic ovary syndrome (PCOS) and adopted in the diagnostic criteria of the Japan Society of Obstetrics and Gynecology (JSOG). We investigated cut-off values for LH and the LH/follicle-stimulating hormone (FSH) ratio in resent two measurement systems for the diagnosis of PCOS. METHODS: Ninety-nine controls and 106 patients with PCOS were enrolled. Serum LH and FSH levels were measured using an electrochemiluminescence immunoassay (ARCHITECT) and chemiluminescence immunoassay (ECLusys). We examined the distribution of the measured levels, selected the conversion closest to the standard normal distribution in the control group, and calculated mean + 1 SD values for LH and the LH/FSH ratio as candidates. Cut-off values coincided with the medians of the candidates in the two assay systems using a regression equation. We calculated the endocrinological abnormality rate in PCOS according to the JSOG criteria by abnormal LH secretion and elevated T. RESULTS: Cut-off values for LH (mIU/mL) and the LH/FSH ratio were 7.1 and 1.21, respectively, in ARCHITECT, and 9.9 and 1.51, respectively, in ECLusys. The detection rates of endocrinological abnormalities in PCOS were 72.2% and 70.6% in the nonoverweight/obese PCOS group and overweight/obese PCOS group, respectively, in ARCHITECT, and 69.4% and 73.5%, respectively, in ECLusys. CONCLUSION: We obtained cut-off values of LH and the LH/FSH ratio for diagnostic criteria of JSOG criteria for PCOS, that were highly compatible between two major assay systems. These cut-off values will contribute to the diagnosis of PCOS in Japan and presumably in women of Asian ethnicities.


Assuntos
Hormônio Foliculoestimulante Humano , Hormônio Luteinizante , Síndrome do Ovário Policístico , Feminino , Humanos , Hormônio Foliculoestimulante Humano/sangue , Japão , Hormônio Luteinizante/sangue , Síndrome do Ovário Policístico/diagnóstico
2.
Reprod Med Biol ; 21(1): e12416, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34934399

RESUMO

BACKGROUND: Although animal models of PCOS have been used in many studies, none of them can reproduce both the reproductive and metabolic phenotypes of PCOS. In addition, behavioral parameters have not been evaluated in PCOS animal models. PURPOSE: We tried to produce an improved rat model of PCOS, and the reproductive, metabolic, and behavioral phenotypes of the model rats were evaluated. METHODS: Female rats were implanted with silicon tubes containing oil-dissolved dihydrotestosterone (Oil-DHT) as a new PCOS model. Their phenotypes were compared with those of conventional PCOS model rats (DHT), into which tubes containing crystalline DHT were implanted, and non-DHT-treated rats (control). RESULTS: Both the Oil-DHT and DHT rats showed greater body weight gain, food intake, and fat depot weight than the control rats. Furthermore, these groups showed fewer estrous stages and increased numbers of cystic follicles. The DHT rats exhibited lower ovarian and uterine weights than the control rats, whereas no such changes were observed in the Oil-DHT rats. The Oil-DHT and DHT rats showed less locomotor activity in the light phase than the control rats. CONCLUSIONS: Our proposed PCOS model reproduced both the reproductive and metabolic phenotypes of PCOS and may have potential for PCOS research.

3.
Horm Behav ; 133: 105005, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34116458

RESUMO

Oxytocin (OT) and its receptor (OTR) play various roles in the central and peripheral regulation of appetite and body weight. Previously, we have shown that the administration of OT markedly decreased appetite and body weight gain in ovariectomized (OVX) obese rats. In addition, recent studies have shown that the endogenous OT system is also affected by endogenous or exogenous estrogen. In this study, we showed that ovariectomy decreased rats' hypothalamic OT/OTR mRNA and serum OT levels, but did not affect their visceral fat OTR mRNA levels. The chronic administration of estradiol (E2) abrogated these ovariectomy-induced changes; i.e., it increased the rats' hypothalamic OT/OTR mRNA and serum OT levels, and may be associated with reductions in food intake and body weight gain. In addition, acute E2 administration increased the rats' hypothalamic OTR mRNA and serum OT levels, but did not affect their hypothalamic OT mRNA levels. Taken together, these results suggest that endogenous OT and/or OTR expression might be positively regulated by E2 and that the suppressive effects of E2 on appetite and body weight gain might be mediated, at least in part, by the OT system. Thus, we consider that OT might be a target hormone to pursue subsequent interventions of menopause for menopause-induced metabolic disorders.


Assuntos
Ocitocina , Receptores de Ocitocina , Animais , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Expressão Gênica , Humanos , Ovariectomia , Ocitocina/genética , Ocitocina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Ocitocina/genética
4.
Gen Comp Endocrinol ; 288: 113360, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31830472

RESUMO

PURPOSE: We examined the mechanism by which neonatal immune stress reduces the sexual behavior of female rats in adulthood. METHODS: Neonatal female rats were randomly divided into 3 groups: control (n = 11), postnatal day 10 lipopolysaccharide (PND10LPS) (n = 23), and PND25LPS (n = 11) groups, which received intraperitoneal injections of LPS (100 µg/kg) or saline on PND10 and 25. Daily inspections of the vaginal opening (VO) were performed from PND27 to PND37. Thereafter, the frequency of estrus was assessed for 15 days. Female rats (at 11-12 weeks of age) were placed in a cage with male rats, and their sexual behavior was monitored for 30 min. The hypothalamic mRNA expression levels of factors related to sexual behavior were examined via real-time PCR. RESULTS: VO occurred later and the frequency of estrus was lower in the PND10LPS group compared to the control group. The number of lordosis behaviors and the total number of mounts performed by male partners were lower in the PND10LPS and PND25LPS groups than in the control group. Acceptability: The lordosis quotient and lordosis rating were lower in the PND10LPS group than in the control group. Proceptive behavior: the number of ear wiggling events was lower in the PND10LPS group than in the other groups, and the number of hops/darts was lower in the PND10LPS group than in the control group. The hypothalamic mRNA expression level of progesterone receptors (PR)A + B was lower in the PND10LPS group than in the control group, and the hypothalamic PRB mRNA expression level was lower in the PND10LPS and PND25LPS groups than in the control group. CONCLUSION: Neonatal immune stress impeded sexual behavior and hypothalamic PR mRNA expression in female rats. Decreased progesterone activity in the hypothalamus might explain the reduction in sexual behavior seen in these rats.


Assuntos
Hipotálamo/metabolismo , Lipopolissacarídeos/administração & dosagem , Receptores de Progesterona/genética , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia , Estresse Fisiológico/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Regulação para Baixo/efeitos dos fármacos , Esquema de Medicação , Feminino , Expressão Gênica/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/fisiopatologia , Lipopolissacarídeos/farmacologia , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Fatores de Tempo
5.
Gynecol Endocrinol ; 36(1): 55-60, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31220962

RESUMO

Polycystic ovary syndrome (PCOS) is commonly associated with metabolic disorders, which are exacerbated by obesity. Recent studies have revealed that oxytocin contributes to metabolic, appetite, and body weight regulation. In the present study, we evaluated the effects of chronic administration of oxytocin on body weight, food intake, and fat mass in a dihydrotestosterone-induced rat model of PCOS. Body weight, body weight change, and relative cumulative food intake were significantly lower in the oxytocin-treated PCOS rats than in the vehicle-treated control PCOS rats. Similarly, visceral adipocyte size was significantly smaller in the oxytocin-treated PCOS rats than in the vehicle-treated control PCOS rats. On the other hand, the numbers of cystic follicles in the ovary did not differ between the two groups. The chronic administration of oxytocin did not affect the rats' serum aspartate aminotransferase, alanine aminotransferase, or lactate dehydrogenase levels, indicating that it does not have adverse effects on hepatic function. These findings suggest that oxytocin could be a candidate drug for preventing the onset of obesity-related metabolic disorders in PCOS patients.


Assuntos
Adipócitos/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ocitócicos/farmacologia , Ocitocina/farmacologia , Adipócitos/patologia , Alanina Transaminase/efeitos dos fármacos , Alanina Transaminase/metabolismo , Androgênios/toxicidade , Animais , Aspartato Aminotransferases/efeitos dos fármacos , Aspartato Aminotransferases/metabolismo , Tamanho Celular/efeitos dos fármacos , Di-Hidrotestosterona/toxicidade , Modelos Animais de Doenças , Feminino , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/patologia , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Cistos Ovarianos/patologia , Ovário/patologia , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Síndrome do Ovário Policístico/fisiopatologia , Ratos
6.
Reprod Med Biol ; 19(2): 135-141, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32273818

RESUMO

PURPOSE: We studied the influence of psychological stress during the early neonatal period on sexual maturation and sexual behavior in rats. METHODS: Neonatal male and female rats were divided into control (C) and maternal separation (MS) groups (n = 20-24 per group). The pups in the MS groups were placed in isolation cages for 240 minutes/d from postnatal days 2-11. Vaginal opening (VO) in females and preputial separation (PS) in males (indicators of sexual maturation) were monitored, as was the estrous cycle in females. Thereafter, sexual behavior was monitored twice at 13 and 15 weeks of age. RESULTS: As for sexual maturation, the onset of PS occurred significantly earlier in the MS group than in the C group, whereas the onset of VO did not differ between the groups. The length of the estrous cycle did not differ between the groups. The frequencies of sexual behaviors did not differ between the groups in either sex. CONCLUSIONS: In conclusion, early-life psychological stress induced by MS advanced sexual maturation in male rats, whereas it did not affect sexual maturation in female rats. On the other hand, early-life psychological stress might not affect sexual behavior in adulthood in either sex.

7.
Gen Comp Endocrinol ; 269: 46-52, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30099033

RESUMO

Infectious, psychological and metabolic stresses in the prenatal and early neonatal period induce long-lasting effects in physiological function and increase the risk of metabolic disorders later in life. We examined the sexual behavior of female rats that were subjected to undernutrition in the prenatal period. Eight pregnant rats were divided into two groups: a maternal normal nutrition group (mNN; n = 4) and a maternal undernutrition group (mUN; n = 4), which received 50% of the daily food intake amount of the mNN group from gestation day 13 to delivery. Nine and seven female offspring were randomly selected from the mNN and mUN groups, respectively. Vaginal opening (VO), estrous cycle length, sexual behavior and mRNA expression levels of the factors that regulate sexual behavior were observed. In the mUN group, VO day was later, the estrous cycle was longer, and the lordosis quotient and lordosis rating were lower than in the mNN group; such differences were not seen in other sexual performances, such as ear wiggles, darts, kick bouts and box. The hypothalamic mRNA expression level of progesterone receptor (PR) A + B and oxytocin (OT) were significantly lower in the mUN group than in the mNN group. These findings indicated that prenatal undernutrition disrupted puberty onset, the estrous cycle, sexual behavior and hypothalamic mRNA expression of PR and OT in female rat pups.


Assuntos
Desnutrição/complicações , Efeitos Tardios da Exposição Pré-Natal/patologia , Comportamento Sexual , Tonsila do Cerebelo/metabolismo , Animais , Peso Corporal , Ingestão de Alimentos , Ciclo Estral , Feminino , Hipotálamo/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Maturidade Sexual
8.
Endocr J ; 65(4): 485-492, 2018 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-29479004

RESUMO

Kisspeptin/neurokinin B (NKB)/dynorphin (Dyn) (KNDy) neuron in hypothalamic arcuate nucleus plays a key role in GnRH/LH pulsatile secretion. We aimed to determine whether stimulation of NKB/neurokinin 3 receptor (NK3R) signaling and inhibition of Dyn/kappa-opioid receptor (KOR) signaling recover LH secretion that is suppressed by acute fasting in male rats. Furthermore, we determined dose dependent effect of NKB/NK3R signaling on serum LH level under acute fasting condition in male mice. Mature male rats were injected saline (0.1 mL) and senktide (20 µg/kg), a NK3R agonist, or nor-BNI (800 µg/kg), a KOR antagonist intraperitoneally (ip) after 72 h fasting. And mature male mice were injected multiple doses of senktide, ip after 48 h fasting. Blood and brain sample were collected 90 min after injections for LH measurement and hypothalamic mRNA expressions. All three studies showed significantly lower LH concentration in fasted groups than non-fasted groups. Senktide did not recover LH suppressed by acute fasting in male rats, whereas nor-BNI injected male rats showed significantly higher LH than 72 h fasted male rats (p < 0.05). Mice study showed significantly higher LH concentration in higher doses senktide groups than 48 h fasted group and one of lower doses senktide group. These results suggest that stimulation of NKB/NK3R signaling and attenuation of Dyn/KOR signaling could recover suppressed LH secretion under acute fasting condition in male rodents.


Assuntos
Hormônio Luteinizante/sangue , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptores da Neurocinina-3/agonistas , Receptores Opioides/metabolismo , Substância P/análogos & derivados , Animais , Jejum/sangue , Masculino , Naltrexona/farmacologia , Neurocinina B/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Substância P/farmacologia
9.
Gynecol Endocrinol ; 34(5): 437-441, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29187003

RESUMO

The effects of androgens on gonadotropin-releasing hormone (GnRH) secretion in females have not been fully established. To clarify the direct effects of androgens on hypothalamic reproductive factors, we evaluated the effects of chronic testosterone administration on hypothalamic GnRH regulatory factors in ovariectomized (OVX) female rats. Both testosterone and estradiol reduced the serum luteinizing hormone levels of OVX female rats, indicating that, as has been found for estrogen, testosterone suppresses GnRH secretion via negative feedback. Similarly, the administration of testosterone or estradiol suppressed the hypothalamic mRNA levels of kisspeptin and neurokinin B, both of which are positive regulators of GnRH, whereas it did not affect the hypothalamic mRNA levels of the kisspeptin receptor or neurokinin-3 receptor. On the contrary, the administration of testosterone, but not estradiol, suppressed the hypothalamic mRNA expression of prodynorphin, which is a negative regulator of GnRH. The administration of testosterone did not alter the rats' serum estradiol levels, indicating that testosterone's effects on hypothalamic factors might be induced by its androgenic activity. These findings suggest that as well as estrogen, androgens have negative feedback effects on GnRH in females and that the underlying mechanisms responsible for these effects are similar, but do not completely correspond, to the mechanisms underlying the effects of estrogen on GnRH.


Assuntos
Dinorfinas/metabolismo , Hipotálamo/efeitos dos fármacos , Kisspeptinas/metabolismo , Neurocinina B/metabolismo , Testosterona/farmacologia , Animais , Dinorfinas/genética , Estradiol/farmacologia , Feminino , Hipotálamo/metabolismo , Kisspeptinas/genética , Leptina/sangue , Hormônio Luteinizante/sangue , Neurocinina B/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Receptores da Neurocinina-3/genética , Receptores da Neurocinina-3/metabolismo
10.
Reprod Med Biol ; 17(3): 315-324, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30013434

RESUMO

PURPOSE: To evaluate the optimized protocol of low dose follicle-stimulating hormone (FSH) therapy that has a starting dose of 50 IU/62.5 IU with a small increment dose (12.5 IU) for women with World Health Organization (WHO) II ovulatory disorder and unexplained infertility. METHODS: Anovulatory women with WHO group II ovulatory disorder (ovulation induction [OI] patients, n = 29), and with an unexplained infertility (ovarian stimulation [OS] patients, n = 21) were enrolled. The protocol of low dose step-up FSH therapy was optimized for the starting dose as 50 IU (body mass index [BMI] < 20 group) and 62.5 IU (BMI ≥ 20 group) with the increment dose of 12.5 IU. Study outcomes were ovulation, monofollicular development and other variables. RESULTS: In the OIpatients, the ovulation rate was 100% (BMI < 20 group) and 90.9% (BMI ≥ 20 group). Monofollicular development was 80.0% (BMI < 20) and 77.3% (BMI ≥ 20). The pregnancy rate was 60% (3/5 BMI < 20) and 18.2% (4/22 BMI ≥ 20). There was no multiple pregnancy. In the OSpatients, the ovulation rate was 100%. Monofollicular development was 85.7% (BMI < 20) and 76.6% (BMI ≥ 20). No pregnancy was achieved in the OSpatients. CONCLUSION: Optimized protocol of low dose FSH therapy setting a starting dose 50 IU/62.5 IU by BMI with an increment dose of 12.5 IU was safe and highly effective in WHO group II anovulatory patients. However, this protocol seemed uneffective for patients with unexplained infertility.

11.
Horm Behav ; 93: 53-61, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28522306

RESUMO

In females, estrogens play pivotal roles in preventing excess body weight (BW) gain. On the other hand, the roles of androgens in female BW, appetite, and energy metabolism have not been fully examined. We hypothesized that androgens' effects on food intake (FI) and BW regulation change according to the estrogens' levels. To evaluate this hypothesis, the effects of chronic testosterone administration in ovariectomized (OVX) female rats with or without estradiol supplementation were examined in this study. Chronic testosterone administration decreased BW, FI, white adipose tissue (WAT) weight, and adipocyte size in OVX rats, whereas it increased BW, WAT weight, and adipocyte size in OVX with estradiol-administered rats. In addition, chronic testosterone administration increased hypothalamic CYP19a1 mRNA levels in OVX rats, whereas it did not alter CYP19a1 mRNA levels in OVX with estradiol-administered rats, indicating that conversion of testosterone to estrogens in the hypothalamus may be activated in testosterone-administered OVX rats. Furthermore, chronic testosterone administration decreased hypothalamic TNF-α mRNA levels in OVX rats, whereas it increased hypothalamic IL-1ß mRNA levels in OVX with estradiol-administered rats. On the other hand, IL-1ß and TNF-α mRNA levels in visceral and subcutaneous WAT and liver were not changed by chronic testosterone administration in both groups. These data indicate that the effects of chronic testosterone administration on BW, FI, WAT weight, and adipocyte size were changed by estradiol treatment in female rats. Testosterone has facilitative effects on BW gain, FI, and adiposity under the estradiol-supplemented condition, whereas it has inhibitory effects in the non-supplemented condition. Differences in the responses of hypothalamic factors, such as aromatase and inflammatory cytokines, to testosterone might underlie these opposite effects.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Estrogênios/farmacologia , Testosterona/farmacologia , Tecido Adiposo/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Estradiol/farmacologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Ovariectomia , Ratos , Ratos Wistar , Testosterona/administração & dosagem , Aumento de Peso/efeitos dos fármacos
12.
Endocr J ; 64(5): 531-541, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28381699

RESUMO

Polycystic ovary syndrome (PCOS) is an ovulatory disorder that affects 6-10% of women of reproductive age. Serum AMH level may be an additional factor, or surrogate of PCOM, in the diagnostic criteria of PCOS. We evaluated the correlations between the serum AMH level and various endocrine and metabolic features in PCOS using the latest fully automated assay. Serum AMH level was compared between 114 PCOS patient (PCOS group) and 95 normal menstrual cycle women (Control group). Correlations between serum AMH level and various endocrine and metabolic factors were analysed in PCOS group. The serum AMH level was significantly higher in the PCOS group (8.35±8.19 ng/mL) than in the Control group (4.99±3.23 ng/mL). The serum AMH level was independently affected by age and the presence of PCOS on multiple regression analysis. Ovarian volume per ovary (OPVO) showed the strongest positive correlation (r=0.62) with the serum AMH level among related factors. On receiver operating characteristic (ROC) curve analysis, the cut-off value of AMH for the diagnosis of PCOS was 7.33 ng/mL, but this value did not have high efficacy (sensitivity 44.7%, specificity 76.8%). A cut-off value of 10 ng/mL had a high specificity of 92.6%, although the sensitivity was low (24.6%). The serum AMH level was elevated and reflected ovarian size in PCOS patients. The serum AMH level could be a surrogate for ultrasound findings of the ovaries in PCOS and might be useful for estimating ovarian findings without transvaginal ultrasound in the diagnosis of PCOS.


Assuntos
Hormônio Antimülleriano/sangue , Ovário/diagnóstico por imagem , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/diagnóstico , Adolescente , Adulto , Fatores Etários , Feminino , Humanos , Tamanho do Órgão/fisiologia , Sensibilidade e Especificidade , Ultrassonografia , Adulto Jovem
13.
Endocr J ; 64(1): 7-14, 2017 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-27665725

RESUMO

Hypersecretion of luteinizing hormone (LH) is a common endocrinological finding of polycystic ovary syndrome (PCOS). This derangement might have a close relationship with hypothalamic kisspeptin expression that is thought to be a key regulator of gonadotropin-releasing hormone (GnRH). We evaluated the relationship between the hypothalamic-pituitary-gonadal axis (HPG axis) and kisspeptin using a rat model of PCOS induced by letrozole. Letrozole pellets (0.4 mg/day) and control pellets were placed subcutaneously onto the backs of 3-week-old female Wistar rats. Body weight, vaginal opening and vaginal smear were checked daily. Blood and tissues of ovary, uterus and brain were collected at 12-weeks of age. An hypothalamic block was cut into anterior and posterior blocks, which included the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC), respectively, in order to estimate hypothalamic kisspeptin expression in each area. The letrozole group showed a similar phenotype to human PCOS such as heavier body weight, heavier ovary, persistent anovulatory state, multiple enlarged follicles with no corpus luteum and higher LH and testosterone (T) levels compared to the control group. Kisspeptin mRNA expression in the posterior hypothalamic block including ARC was higher in the letrozole group than in the control group although its expression in the anterior hypothalamic block was similar between groups. These results suggest that enhanced KNDy neuron activity in ARC contributes to hypersecretion of LH in PCOS and might be a therapeutic target to rescue ovulatory disorder of PCOS in the future.


Assuntos
Hipotálamo Posterior/metabolismo , Kisspeptinas/genética , Síndrome do Ovário Policístico/genética , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Hipotálamo Posterior/patologia , Kisspeptinas/metabolismo , Letrozol , Nitrilas , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Triazóis , Regulação para Cima
14.
Reprod Med Biol ; 16(2): 179-187, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-29259467

RESUMO

Aim: Polycystic ovary syndrome (PCOS) is an ovulatory disorder and insulin resistance and diabetes are involved in its pathophysiology. Metformin, an anti-diabetic agent, has been reported to be useful to induce ovulation. Methods: Metformin treatment was classified into four types: (1) clomiphene-metformin combination treatment for clomiphene-resistant patients; (2) clomiphene-metformin combination for clomiphene-sensitive patients; (3) clomiphene-metformin combination for naïve patients; and (4) metformin monotherapy. The patients underwent physical, endocrinological, and clinical examinations for their ovulation rates, pregnancy rates, and follicular development. Results: The ovulation rates, pregnancy rates, and single follicular development were not significantly different among the clomiphene-metformin combination treatment groups. In the Body Mass Index (BMI) subanalysis, the pregnancy rate was higher in the BMI≥30 kg/m2 group than in the other three groups with a BMI of ≤30 kg/m2 in both cycles and cases. The ovulation rates and pregnancy rates were significantly higher in the group with a fasting insulin of ≥15 µU/mL than in the groups with a fasting insulin of <15 µU/mL in both cycles and cases. Conclusion: Clomiphene-metformin combination treatment appears to be useful, at least for clomiphene-resistant patients, and a BMI of >30 kg/m2 and a fasting insulin of ≥15 µU/mL appear to be predictors of a good result with this treatment.

15.
Reprod Med Biol ; 16(4): 325-329, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29259485

RESUMO

Purpose: Exposure to various stressors, including psychological, metabolic, and immune, in the perinatal period induces long-lasting effects in physiological function and increase the risk of metabolic disorders in later life. In the present study, sexual maturation and sexual behavior were assessed in prenatally undernourished mature male rats. Methods: All the pregnant rats were divided into the maternal normal nutrition (mNN) group and the maternal undernutrition (mUN) group. The mUN mothers received 50% of the amount of the daily food intake of the mNN mothers. Preputial separation and sexual behavior were observed in randomly selected pups of the mNN and mUN groups. Results: The body weight of the mothers was significantly lighter in the mUN group than in the mNN group. Similarly, the pups in the mUN group showed a significantly lower body weight than those in the mNN group from postnatal day (PND) 1 to PND 15. The preputial separation day was significantly delayed in the mUN group, compared to the mNN group. Sexual behavior did not show any significant difference between the two groups. Conclusion: These findings indicated that prenatal undernutrition delayed sexual maturation, but did not suppress sexual behavior, in mature male rats.

16.
J Med Invest ; 69(1.2): 65-69, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35466148

RESUMO

It has been shown that biotin, a water-soluble vitamin (B7), plays roles in reproductive functions, such as oocyte maturation and embryo development, in experimental animals. On the other hand, little is known about the clinical effects of biotin on human reproduction. In this study, serum and follicular fluid biotin levels were measured in patients who underwent in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI), and their associations with reproductive outcomes were evaluated. As a result, biotin was detected in follicular fluid, as well as serum, and the biotin levels of follicular fluid were found to be positively correlated with those of serum. The biotin levels of serum were higher than those of follicular fluid, suggesting that biotin may be taken up into the follicular fluid from the blood. Although serum and follicular fluid biotin levels tended to be higher in pregnant patients than in non-pregnant patients, these data did not show the significant statistical difference. These findings indicate that biotin does not contribute to the maintenance of oocyte quality, and hence, it does not increase fertilization and pregnancy rates. J. Med. Invest. 69 : 65-69, February, 2022.


Assuntos
Líquido Folicular , Injeções de Esperma Intracitoplásmicas , Biotina , Feminino , Fertilização in vitro , Humanos , Gravidez , Resultado da Gravidez
17.
J Med Invest ; 68(3.4): 228-231, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759135

RESUMO

The metabolic effects of androgens and their underlying mechanisms in females have been revealed by recent studies. An excess of androgens can have adverse effects on feeding behavior and metabolic functions and induce metabolic disorders / diseases, such as obesity, insulin resistance, and diabetes, in women and experimental animals of reproductive age. Interestingly, these effects of androgens are not observed in ovariectomized animals, indicating that their effects might be dependent on the estrogen milieu. Central and peripheral mechanisms, such as alterations in the activity of hypothalamic factors, reductions in energy expenditure, skeletal muscle insulin resistance, and ß-cell dysfunction, might be related to these androgens' effects. J. Med. Invest. 68 : 228-231, August, 2021.


Assuntos
Androgênios , Resistência à Insulina , Animais , Feminino , Humanos , Músculo Esquelético , Obesidade
18.
Neuropeptides ; 75: 49-57, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30885500

RESUMO

Recent studies have shown that oxytocin reduces food intake and body weight gain and promotes lipolysis in some species, including humans. Interestingly, these effects of oxytocin are more marked in obese individuals. Although the menopausal loss of ovarian function induces increased visceral adiposity and some metabolic disorders, no safe medical interventions for these conditions have been established. In this study, we evaluated the effects of oxytocin on appetite, body weight, and fat mass in ovariectomized rats. Six-day oxytocin treatment attenuated cumulative food intake and body weight gain, and reduced visceral and subcutaneous fat weight and adipocyte cell area in ovariectomized rats. Blood examinations indicated that 6-day oxytocin treatment did not alter renal or hepatic functions. Instead, it might prevent ovariectomy-induced liver damage. In addition, acute oxytocin treatment did not affect body temperature or locomotor activity. These results indicate that oxytocin might be useful for treating or preventing menopause-induced metabolic disorders, without causing any adverse effects.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Obesidade/tratamento farmacológico , Ocitocina/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Obesidade/metabolismo , Ovariectomia , Ocitocina/uso terapêutico , Ratos
19.
J Clin Med ; 7(7)2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29976877

RESUMO

It is known that metabolic disturbances suppress reproductive functions in females. The mechanisms underlying metabolic and nutritional effects on reproductive functions have been established based on a large body of clinical and experimental data. From the 1980s to 1990s, it was revealed that disrupted gonadotropin-releasing hormone (GnRH) secretion is the main cause of reproductive impairments in metabolic and nutritional disorders. From the late 1990s to early 2000s, it was demonstrated that, in addition to their primary functions, some appetite- or metabolism-regulating factors affect GnRH secretion. Furthermore, in the early 2000s, kisspeptin, which is a potent positive regulator of GnRH secretion, was newly discovered, and it has been revealed that kisspeptin integrates the effects of metabolic status on GnRH neurons. Recent studies have shown that kisspeptin mediates at least some of the effects of appetite- and metabolism-regulating factors on GnRH neurons. Thus, kisspeptin might be a useful clinical target for treatments aimed at restoring reproductive functions in individuals with metabolic or nutritional disturbances, such as those who exercise excessively, experience marked weight loss, or suffer from eating disorders. This paper presents a review of what is currently known about the effects of metabolic status on reproductive functions and their underlying mechanisms by summarizing the available evidence.

20.
J Endocrinol ; 2018 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-30089683

RESUMO

Although polycystic ovary syndrome (PCOS) is among the most common endocrine disorders in women of reproductive age, its etiology remains poorly understood. From the perspective of developmental origins of health and disease, some studies have investigated the relationship between low birth weight and the prevalence of PCOS and/or PCOS phenotypes in humans; however, the results of these studies were inconclusive. Here, we evaluated the effects of prenatal undernutrition on the metabolic and reproductive phenotypes of dihydrotestosterone-induced PCOS model rats. The PCOS model rats showed increased body weight, food intake, fat weight, adipocyte size, and upregulation of inflammatory cytokines in adipose tissue; prenatal undernutrition exacerbated these metabolic changes. Prenatal undernutrition also increased the gene expression of hypothalamic orexigenic factor and decreased the gene expression of anorexigenic factor in the PCOS model rats. In addition, the PCOS model rats exhibited irregular cyclicity, polycystic ovaries, and disrupted gene expression of ovarian steroidogenic enzymes. Interestingly, prenatal undernutrition attenuated these reproductive changes in the PCOS model rats. Our results suggest that in dihydrotestosterone-induced PCOS model rats, prenatal undernutrition exacerbates the metabolic phenotypes, whereas it improves the reproductive phenotypes, and that such phenotypic changes may be induced by the alteration of some peripheral and central factors.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA