Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.434
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 13: 995491, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36714560

RESUMO

Introduction: DEHP is an endocrine disruptor widely used in the production of malleable plastics. DEHP exposure was associated with altered hypothalamic-pituitary-thyroid (HPT) axis function. Although previous studies reported deleterious effects of DEHP exposure during the intrauterine period, few studies have evaluated the direct effects triggered by this endocrine disruptor on the offspring animals' thyroid function. This study aimed to investigate the impact of intrauterine exposure to DEHP on the HPT axis function programming of the offspring animals during adulthood. Methods: Pregnant Wistar rats were orally treated with corn oil or corn oil supplemented with DEHP (0.48 or 4.8 mg/kg/day) throughout the gestational period. The offspring rats were euthanized on the 90th postnatal day. Hypothalamus, pituitary, thyroid, and liver were collected to analyze gene expression and protein content through qPCR and Western Blot. Blood was collected to determine TSH and thyroid hormone levels through fluorometric or chemiluminescence immunoassays. Results: In the adult F1 female rats, the highest dose of DEHP decreased TSH serum levels. In the thyroid, DEHP reduced the gene expression and/or protein content of NIS, TSHR, TG, TPO, MCT8, NKX2.1, PAX8, and FOXE1. These data are consistent with the reduction in T4 serum levels of the F1 DEHP-exposed female rats. In the liver, DEHP exposure increased the mRNA expression of Dio1 and Ttr, while the highest dose of DEHP reduced the mRNA expression of Ugt1a1 and Ugt1a6. Conversely, in the F1 male adult rats, TSHB expression and TSH serum levels were increased in DEHP-exposed animals. In the thyroid, except for the reduced protein content of TSHR, none of the evaluated genes/proteins were altered by DEHP. TH serum levels were not changed in the DEHP-exposed F1 male rats compared to the control group. Additionally, there were no significant alterations in the expression of hepatic enzymes in these animals. Discussion/Conclusions: Our results demonstrated, for the first time, that intrauterine exposure to DEHP disrupts the HPT axis function in male and female offspring rats and strongly suggest that DEHP exposure increases the susceptibility of the offspring animals to develop thyroid dysfunctions during adulthood.


Assuntos
Dietilexilftalato , Disruptores Endócrinos , Hipotálamo , Hipófise , Efeitos Tardios da Exposição Pré-Natal , Glândula Tireoide , Animais , Feminino , Masculino , Gravidez , Ratos , Óleo de Milho , Dietilexilftalato/toxicidade , Disruptores Endócrinos/toxicidade , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Ratos Wistar , RNA Mensageiro/metabolismo , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Tireotropina , Hipófise/efeitos dos fármacos , Hipófise/metabolismo
2.
Reprod Biol Endocrinol ; 19(1): 120, 2021 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-34344365

RESUMO

BACKGROUND: This study aimed to detect the effect of angiotensin receptor 1 (AT1) knock out (KO) on spermatogenesis and hypothalamic-pituitary-gonadal (HPG) axis hormone expression. METHODS: Normal C57BL/6 male mice were used as control group or treated with angiotensin receptor blocker, in addition heterozygous ± AT1KO mice were generated. After caged at a ratio of 2 to 1 with females, pregnancy rates of female mice were determined by detection of vaginal plugs. Deformity rate of spermatozoa was evaluated by eosin staining and morphology evaluation. The AT1 mRNA expression in the testes of male ± AT1KO mice was detected by quantitative real-time polymerase chain reaction (QRT-PCR). Serum GnRH level was determined by ELISA. RESULTS: Compared to control, ± AT1KO mice showed reduced expression of AT1 in testes, pituitary and hypothalamus. In addition, decreased level of GnRH, but not follicle stimulating hormone (FSH) or luteinizing hormone (LH), in ± AT1KO mice was detected. Treatment with angiotensin receptor blocker (ARB) did not have significant effects on HPG hormones. ± AT1KO mice exhibited male infertility and significant abnormality of sperm morphology. CONCLUSION: Reduced AT1 knockout resulted in male infertility, potentially by inducing abnormal spermatogenesis. Both testis and HPG axis signaling may be involved.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Infertilidade Masculina/genética , Receptor Tipo 1 de Angiotensina/genética , Espermatogênese/genética , Testículo/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Infertilidade Masculina/metabolismo , Losartan/farmacologia , Masculino , Camundongos , Camundongos Knockout , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos
3.
Reproduction ; 162(2): 171-179, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34128825

RESUMO

To elucidate the mechanism by which nerve growth factor (NGF) influences the LH secretory pathway in camelids, a series of experiments were done to determine the involvement of the hypothalamus (Experiment 1), the role of GnRH neurons (Experiment 2), and the effect of progesterone (Experiment 3) on the NGF-induced LH surge and ovulation in llamas. In Experiment 1, the declining phase of the NGF-induced LH surge was used to determine if the decline is a result of pituitary depletion or hypothalamic unresponsiveness. Female llamas were treated with NGF and, 7 h later, assigned to three groups and given a second dose of NGF (n = 5), a dose of GnRH (n = 5), or saline (n = 6). The LH response was attenuated after the second dose of NGF vs GnRH. In Experiment 2, Fos expression (marker of neuronal activation) in GnRH neurons was examined in the hypothalamus of llamas after NGF or saline treatment (n = 3 per group). Despite an LH surge in the NGF group but not in the saline group, no differences were detected between groups in Fos/GnRH co-expression. In Experiment 3, llamas in low-, medium-, and high-plasma progesterone groups (n = 4 per group) were treated with NGF. The NGF-induced LH surge did not differ among treatment groups. Results from the present study show that the induction of a preovulatory LH surge by NGF may be controlled by a novel pathway involving GnRH neuro-terminals downstream of the hypothalamus and is independent of progesterone influence.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Fator de Crescimento Neural/farmacologia , Hipófise/metabolismo , Progesterona/metabolismo , Animais , Camelídeos Americanos , Feminino , Hipotálamo/efeitos dos fármacos , Hipófise/efeitos dos fármacos
4.
Pharmacol Rep ; 73(5): 1465-1472, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34086261

RESUMO

BACKGROUND: Intense statin therapy was found to impair testosterone production in men. Metformin administered to subjects with hypergonadotropic hypogonadism decreased gonadotropin production. The current study was aimed at investigating whether metformin treatment modulates the impact of high-dose rosuvastatin therapy on hypothalamic-pituitary-testicular axis activity in men. METHODS: The study included 43 very high cardiovascular risk men with late-onset hypogonadism, 20 of whom had been treated with metformin (1.7-3 g daily) for at least 6 months. In all subjects, unsuccessful initial statin treatment was replaced with rosuvastatin (20-40 mg daily). Plasma lipid levels, glucose homeostasis markers, as well as circulating levels of gonadotropins, testosterone, bioavailable testosterone, dehydroepiandrosterone-sulfate, prolactin, estradiol and creatinine were measured at the beginning of the study and 4 months later in 28 individuals in whom rosuvastatin reduced LDL cholesterol levels to below 70 mg/dL. RESULTS: There were no differences between treatment-induced changes in plasma lipids. In both study groups, rosuvastatin reduced total and bioavailable testosterone levels. However, only in metformin-naïve men, rosuvastatin increased LH and FSH levels and slightly impaired insulin sensitivity. The impact on gonadotropin concentrations correlated with treatment-induced decrease in testosterone levels. There were no significant differences between baseline and posttreatment values of dehydroepiandrosterone-sulfate, prolactin, estradiol and the glomerular filtration rate. CONCLUSION: The obtained results suggest that metformin prevents the compensatory increase in gonadotrope function induced by intense statin therapy.


Assuntos
Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Metformina/farmacologia , Hipófise/efeitos dos fármacos , Rosuvastatina Cálcica/efeitos adversos , Rosuvastatina Cálcica/uso terapêutico , Testículo/efeitos dos fármacos , Adulto , Idoso , Anticolesterolemiantes/efeitos adversos , Anticolesterolemiantes/uso terapêutico , Humanos , Hipoglicemiantes/farmacologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Testículo/metabolismo , Testosterona/sangue , Testosterona/metabolismo
5.
Cells ; 10(3)2021 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800837

RESUMO

Central actions of leptin and insulin on hepatic lipid metabolism can be opposing and the mechanism underlying this phenomenon remains unclear. Both hormones can modulate the central somatostatinergic system that has an inhibitory effect on growth hormone (GH) expression, which plays an important role in hepatic metabolism. Using a model of chronic central leptin infusion, we evaluated whether an increase in central leptin bioavailability modifies the serum lipid pattern through changes in hepatic lipid metabolism in male rats in response to an increase in central insulin and the possible involvement of the GH axis in these effects. We found a rise in serum GH in leptin plus insulin-treated rats, due to an increase in pituitary GH mRNA levels associated with lower hypothalamic somatostatin and pituitary somatostatin receptor-2 mRNA levels. An augment in hepatic lipolysis and a reduction in serum levels of non-esterified fatty acids (NEFA) and triglycerides were found in leptin-treated rats. These rats experienced a rise in lipogenic-related factors and normalization of serum levels of NEFA and triglycerides after insulin treatment. These results suggest that an increase in insulin in leptin-treated rats can act on the hepatic lipid metabolism through activation of the GH axis.


Assuntos
Hipotálamo/efeitos dos fármacos , Insulina/farmacologia , Leptina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Animais , Ácidos Graxos não Esterificados/sangue , Regulação da Expressão Gênica , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Injeções Intravenosas , Injeções Intraventriculares , Insulina/metabolismo , Leptina/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Hipófise/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Triglicerídeos/sangue
6.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799501

RESUMO

The growth hormone (GH)/insulin-like growth factor I (IGF-I) axis is involved in metabolic control. Malnutrition reduces IGF-I and modifies the thermogenic capacity of brown adipose tissue (BAT). Leptin has effects on the GH/IGF-I axis and the function of BAT, but its interaction with IGF-I and the mechanisms involved in the regulation of thermogenesis remains unknown. We studied the GH/IGF-I axis and activation of IGF-I-related signaling and metabolism related to BAT thermogenesis in chronic central leptin infused (L), pair-fed (PF), and control rats. Hypothalamic somatostatin mRNA levels were increased in PF and decreased in L, while pituitary GH mRNA was reduced in PF. Serum GH and IGF-I concentrations were decreased only in PF. In BAT, the association between suppressor of cytokine signaling 3 and the IGF-I receptor was reduced, and phosphorylation of the IGF-I receptor increased in the L group. Phosphorylation of Akt and cyclic AMP response element binding protein and glucose transporter 4 mRNA levels were increased in L and mRNA levels of uncoupling protein-1 (UCP-1) and enzymes involved in lipid anabolism reduced in PF. These results suggest that modifications in UCP-1 in BAT and changes in the GH/IGF-I axis induced by negative energy balance are dependent upon leptin levels.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Hormônio do Crescimento/genética , Fator de Crescimento Insulin-Like I/genética , Leptina/farmacologia , Termogênese/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Metabolismo Energético/genética , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Hormônio do Crescimento/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraventriculares , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Fosforilação/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Termogênese/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
7.
PLoS One ; 16(4): e0249482, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33882080

RESUMO

The effects of hormonal contraceptives on structural features of the hypothalamus and pituitary are incompletely understood. One prior study reported microstructural changes in the hypothalamus with oral contraceptive pill (OCP) use. However, effects on hypothalamic volume have not been reported. One prior study reported volumetric changes in the pituitary. However, this study was limited by including participants evaluated for neurological symptoms. We sought to determine if OCP use is associated with alteration of hypothalamic or pituitary volume. High-resolution 3T MRI was performed for a prospective cohort of 50 healthy women from 2016 to 2018, which comprised 21 OCP users (age, 19-29) and 29 naturally cycling women (age, 18-36). Participants were excluded if they were pregnant or had significant medical conditions including neurological, psychiatric, and endocrine disorders. After confirming reliability of the image analysis techniques, 5 raters independently performed manual segmentation of the hypothalamus and semi-automated intensity threshold-based segmentation of the pituitary using ITK-SNAP. Total intracranial volume was estimated using FreeSurfer. A general linear model tested the association of OCP use with hypothalamic and pituitary volumes. Hypothalamic (B = -81.2 ± 24.9, p = 0.002) and pituitary (B = -81.2 ± 38.7, p = 0.04) volumes in OCP users were smaller than in naturally cycling women. These findings may be related to interference with known trophic effects of sex hormones and suggest a structural correlate of central OCP effects.


Assuntos
Anticoncepcionais Orais Combinados/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/crescimento & desenvolvimento , Hipófise/efeitos dos fármacos , Hipófise/crescimento & desenvolvimento , Adolescente , Adulto , Feminino , Humanos , Hipotálamo/diagnóstico por imagem , Imageamento por Ressonância Magnética , Tamanho do Órgão/efeitos dos fármacos , Hipófise/diagnóstico por imagem , Gravidez , Adulto Jovem
8.
J Ethnopharmacol ; 273: 113947, 2021 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-33617969

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Eucommia ulmoides Oliv. leaves are the dry leaves of Eucommia ulmoides Oliv. Modern studies have shown that Eucommia ulmoides Oliv. leaves and its extracts have many pharmacological effects, such as regulating hypothalamus pituitary ovary (HPO) axis function, estrogen like effects, correcting insulin resistance (IR), regulating lipids, and reducing weight, which are consistent with the clinical manifestations in polycystic ovary syndrome (PCOS) patients. PCOS patients often have HPO axis disorder, low estrogen, high androgen, high IR complication rate, and obesity. Previous preclinical studies have shown that total flavonoids from Eucommia ulmoides Oliv. leaves (TFEL) can improve the imbalance in sex hormone secretion in perimenopausal animal models by regulating the function of the HPO axis. Thus, it is important to understand if flavonoids are the active parts of Eucommia ulmoides Oliv. leaves that interfere with polycystic ovary syndrome with insulin resistance (PCOS-IR), and determine the regulatory role they play in sex hormones and IR? AIM OF THE STUDY: Investigate the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway in the ovary and kisspeptin/insulin like growth factor/leptin receptor1/androgen receptor (Kiss1/IGF-1/LEPR/AR) in the HPO axis to determine the mechanism of TFEL intervention in a rat model of PCOS-IR model rats. MATERIALS AND METHODS: A rat model of PCOS-IR was established using a high-fat diet (49 d) combined with letrozole (1 mg/kg·d, for 28 d). Then, metformin (300 mg/kg·d) and TFEL (220 mg/kg·d, 110 mg/kg·d, and 55 mg/kg·d) were administered continuously for 21 days. At the end of the experiment, samples were taken and the related indexes were measured. RESULTS: TFEL reduced the body weight, Lee's index, ovarian index, ovarian area and ovarian volume, increased serum E2, SHBG levels and ISI, decreased serum levels of T, LEP, INS, and FBG (whole blood), and reduced the HOMA-IR in rats with PCOS-IR. TFEL downregulate Kiss1, IGF-1, and AR in the arcuate nucleus of hypothalamus, and upregulate Kiss1, downregulate IGF-1 and AR in the pituitary gland, and upregulate Kiss1, downregulate IGF-1, LEPR, and AR in the ovary of rats with PCOS-IR. TFEL could downregulate p-IRS-1Ser307, upregulate IRS-1, p-IRS-1Tyr895, PI3Kp85α, p-PI3Kp85α, AKT, p-AKT, and GLUT4 in the ovary, and ameliorated histopathological changes in the ovary and pancreas of rats with PCOS-IR. CONCLUSION: TFEL can inhibit ovarian hyperplasia, regulate disorders of glucose and lipid metabolism and improve the secretion of sex hormones, by regulating the expression of PI3K/AKT signaling pathway-related proteins in the ovary and Kiss1/IGF-1/LEPR/AR in the HPO axis.


Assuntos
Dieta Hiperlipídica , Eucommiaceae/química , Flavonoides/farmacologia , Resistência à Insulina , Extratos Vegetais/farmacologia , Síndrome do Ovário Policístico/tratamento farmacológico , Animais , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Flavonoides/química , Flavonoides/uso terapêutico , Hormônios Esteroides Gonadais/sangue , Hipotálamo/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Kisspeptinas/metabolismo , Letrozol/toxicidade , Metformina/farmacologia , Metformina/uso terapêutico , Ovário/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Hipófise/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Folhas de Planta/química , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Receptores Androgênicos/metabolismo , Receptores para Leptina/metabolismo
9.
Int J Mol Sci ; 21(23)2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33276521

RESUMO

Anthropogenic endocrine-disrupting chemicals (EDCs) can contaminate air, soil, and water. Human exposures to EDCs occur through inhalation, absorption, and ingestion. EDCs act by disrupting various pathways in the endocrine system. When the hypothalamic-pituitary-gonadal (HPG) axis is disrupted by EDCs, there can be effects on fertility in both men and women. Not only can fertility be indirectly affected by EDC disruptions of the HPG axis, but EDCs can also directly affect the menstrual cycle and sperm morphology. In this review, we will discuss the current findings on EDCs that can be inhaled. This review examines effects of exposure to prominent EDCs: brominated and organophosphate flame retardants, diesel exhaust, polycyclic aromatic hydrocarbons, cadmium and lead, TCDD, and polychlorinated biphenyls on fertility through alterations that disrupt the HPG axis and fertility through inhalation. Although the studies included herein include multiple exposure routes, all the studies indicate receptor interactions that can occur from inhalation and the associated effects of all compounds on the HPG axis and subsequent fertility.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Gônadas/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Poluentes Atmosféricos/classificação , Animais , Disruptores Endócrinos/classificação , Fertilidade/efeitos dos fármacos , Gônadas/metabolismo , Humanos , Hipotálamo/metabolismo , Metais Pesados/química , Hipófise/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/efeitos adversos , Fatores Sexuais , Emissões de Veículos/toxicidade
10.
Endocrinology ; 161(12)2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075809

RESUMO

Androgens can affect the reproductive axis of both sexes. In healthy women, as in men, elevated exogenous androgens decrease gonad function and lower gonadotropin levels; such circumstances occur with anabolic steroid abuse or in transgender men (genetic XX individuals) taking androgen supplements. The neuroendocrine mechanisms by which endogenous or exogenous androgens regulate gonadotropin release, including aspects of pulsatile luteinizing hormone (LH) secretion, remain unknown. Because animal models are valuable for interrogating neural and pituitary mechanisms, we studied effects of androgens in the normal male physiological range on in vivo LH secretion parameters in female mice and in vitro LH secretion patterns from isolated female pituitaries. We also assessed androgen effects on hypothalamic and gonadotrope gene expression in female mice, which may contribute to altered LH secretion profiles. We used a nonaromatizable androgen, dihydrotestosterone (DHT), to isolate effects occurring specifically via androgen receptor (AR) signaling. Compared with control females, DHT-treated females exhibited markedly reduced in vivo LH pulsatility, with decreases in pulse frequency, amplitude, peak, and basal LH levels. Correlating with reduced LH pulsatility, DHT-treated females also exhibited suppressed arcuate nucleus Kiss1 and Tac2 expression. Separate from these neural effects, we determined in vitro that the female pituitary is directly inhibited by AR signaling, resulting in lower basal LH levels and reduced LH secretory responses to gonadotropin-releasing hormone pulses, along with lower gonadotropin gene expression. Thus, in normal adult females, male levels of androgen acting via AR can strongly inhibit the reproductive axis at both the neural and pituitary levels.


Assuntos
Androgênios/farmacologia , Di-Hidrotestosterona/farmacologia , Hipotálamo/efeitos dos fármacos , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Neurônios/efeitos dos fármacos , Precursores de Proteínas/metabolismo , Taquicininas/metabolismo , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Masculino , Camundongos , Neurônios/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Precursores de Proteínas/genética , Transdução de Sinais/efeitos dos fármacos , Taquicininas/genética
11.
Chem Res Toxicol ; 33(10): 2605-2622, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32972137

RESUMO

Silver nanoparticles (AgNPs) have potent antimicrobial activity and, for this reason, are incorporated into a variety of products, raising concern about their potential risks and impacts on human health and the environment. The developmental period is highly dependent on thyroid hormones (THs), and puberty is a sensitive period, where changes in the hormonal environment may have permanent effects. We evaluated the hypothalamic-pituitary (HP)-thyroid axis after exposure to low doses of AgNPs using a validated protocol to assess pubertal development and thyroid function in immature male rats. For stimulatory events of the HP-thyroid axis, we observed an increase in the expression of Trh mRNA and serum triiodothyronine. Negative feedback reduced the hypothalamic expression of Dio2 mRNA and increased the expression of Thra1, Thra2, and Thrb2 mRNAs. In the pituitary, there was a reduced expression of Mct-8 mRNA and Dio2 mRNA. For peripheral T3-target tissues, a reduced expression of Mct-8 mRNA was observed in the heart and liver. An increased expression of Dio3 mRNA was observed in the heart and liver, and an increased expression of Thrb2 mRNA was observed in the liver. The quantitative proteomic profile of the thyroid gland indicated a reduction in cytoskeletal proteins (Cap1, Cav1, Lasp1, Marcks, and Tpm4; 1.875 µg AgNP/kg) and a reduction in the profile of chaperones (Hsp90aa1, Hsp90ab1, Hspa8, Hspa9, P4hb) and proteins that participate in the N-glycosylation process (Ddost, Rpn1 and Rpn2) (15 µg AgNP/kg). Exposure to low doses of AgNPs during the window of puberty development affects the regulation of the HP-thyroid axis with further consequences in thyroid gland physiology.


Assuntos
Hipotálamo/efeitos dos fármacos , Nanopartículas Metálicas/química , Hipófise/efeitos dos fármacos , Proteômica , Prata/farmacologia , Glândula Tireoide/efeitos dos fármacos , Animais , Expressão Gênica , Masculino , Ratos , Ratos Wistar , Prata/química , Tireotropina/sangue , Tri-Iodotironina/sangue
12.
Neurosci Lett ; 738: 135335, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32891671

RESUMO

Proopiomelanocortin (POMC) is a neuropeptide precursor produced in the anterior and intermediate pituitary lobes, the hypothalamic arcuate nucleus (ARC), and solitary tract nucleus. Alpha-melanocyte-stimulating hormone (α-MSH) is a cell type specific POMC derivative that is essential for regulating feeding, and energy homeostasis. However, the molecular mechanism underlying POMC/α-MSH secretion remains unclear. Ca2+-dependent activator protein for secretion 2 (CAPS2) is a regulatory protein involved in the exocytosis of dense-core vesicles containing neuropeptides. We previously reported CAPS2 localization in the intermediate pituitary lobe and reduced body weights in Caps2-knockout (Caps2-KO) mice, compared to control mice. Here, we aimed to investigate CAPS2 expression in POMC-expressing neurons and the effects of CAPS2 deficiency on the secretion of POMC-related peptides and feeding behavior phenotype. CAPS2 was localized in the POMC-expressing neurons of the intermediate pituitary lobe, hypothalamic ARC, and the paraventricular nucleus, which is innervated by hypothalamic neurons. POMC protein levels in the intermediate pituitary lobe of Caps2-KO mice were significantly higher than that in the control mice, suggesting a possible accumulation of POMC-derived peptides in the intermediate pituitary lobe of Caps2-KO mice. Moreover, administration of low-dose melanotan-2, an α-MSH receptor (MC4R) agonist, decreased food intake per body weight in Caps2-KO mice; no such effect was observed in the wildtype mice. Collectively, these results suggest that CAPS2 is involved in regulating the secretion of POMC-derived peptides, including α-MSH, is partially associated with feeding, and affects energy metabolism.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Ingestão de Alimentos/genética , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/genética , Hipófise/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Proteínas de Ligação ao Cálcio/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Peptídeos Cíclicos/farmacologia , Hipófise/efeitos dos fármacos , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
13.
Aquat Toxicol ; 225: 105547, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32623180

RESUMO

Some chemicals in the environment disrupt thyroid hormone (TH) systems leading to alterations in organism development, but their effect mechanisms are poorly understood. In fish, this has been limited by a lack of fundamental knowledge on thyroid gene ontogeny and tissue expression in early life stages. Here we established detailed expression profiles for a suite of genes in the hypothalamic-pituitary-thyroid (HPT) axis of zebrafish (Danio rerio) between 24-120 h post fertilisation (hpf) and quantified their responses following exposure to 3,3',5-triiodo-L-thyronine (T3) using whole mount in situ hybridisation (WISH) and qRT-PCR (using whole-body extracts). All of the selected genes in the HPT axis demonstrated dynamic transcript expression profiles across the developmental stages examined. The expression of thyroid receptor alpha (thraa) was observed in the brain, gastrointestinal tract, craniofacial tissues and pectoral fins, while thyroid receptor beta (thrb) expression occurred in the brain, otic vesicles, liver and lower jaw. The TH deiodinases (dio1, dio2 and dio3b) were expressed in the liver, pronephric ducts and brain and the patterns differed depending on life stage. Both dio1 and dio2 were also expressed in the intestinal bulb (96-120 hpf), and dio2 expression occurred also in the pituitary (48-120 hpf). Exposure of zebrafish embryo-larvae to T3 (30 and 100 µg L-1) for periods of 48, 96 or 120 hpf resulted in the up-regulation of thraa, thrb, dio3b, thyroid follicle synthesis proteins (pax8) and corticotropin-releasing hormone (crhb) and down-regulation of dio1, dio2, glucuronidation enzymes (ugt1ab) and thyroid stimulating hormone (tshb) (assessed via qRT-PCR) and responses differed across life stage and tissues. T3 induced thraa expression in the pineal gland, pectoral fins, brain, somites, gastrointestinal tract, craniofacial tissues, liver and pronephric ducts. T3 enhanced thrb expression in the brain, jaw cartilage and intestine, while thrb expression was suppressed in the liver. T3 exposure suppressed the transcript levels of dio1 and dio2 in the liver, brain, gastrointestinal tract and craniofacial tissues, while dio2 signalling was also suppressed in the pituitary gland. Dio3b expression was induced by T3 exposure in the jaw cartilage, pectoral fins and brain. The involvement of THs in the development of numerous body tissues and the responsiveness of these tissues to T3 in zebrafish highlights their potential vulnerability to exposure to environmental thyroid-disrupting chemicals.


Assuntos
Tri-Iodotironina/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Hormônio Liberador da Corticotropina , Hipotálamo/efeitos dos fármacos , Larva/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Glândula Tireoide/efeitos dos fármacos , Hormônios Tireóideos/metabolismo , Tironinas/metabolismo , Tironinas/farmacologia , Tireotropina , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
14.
World Neurosurg ; 142: 142-146, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32615288

RESUMO

BACKGROUND: Adult-onset Langerhans cell histiocytosis (LCH) with simultaneous involvement of the high cervical spine and the hypothalamus is rare. CASE DESCRIPTION: We have reported a case of adult-onset LCH in the second cervical vertebra with bony destruction and subsequent diabetes insipidus due to simultaneous involvement of the hypothalamus and pituitary stalk. Magnetic resonance imaging of the hypothalamus and pituitary lesion and immunohistochemistry of the cervical lesion revealed LCH. Posterior fusion of the cervical spine (first, third, and fourth cervical vertebrae) was performed, followed by systemic chemotherapy. The cervical fusion was well maintained, and the patient achieved clinical remission. No new LCH lesion was found during the follow-up of >2 years. CONCLUSIONS: Patients with known LCH of the spine showing new symptoms of diabetes insipidus should be examined for infiltrating lesions of the pituitary stalk or hypothalamus. In cases of severe instability of the spine, surgical treatment should be performed. If multiple and systemic LCH lesions are found, systemic chemotherapy should be administered.


Assuntos
Vértebras Cervicais/diagnóstico por imagem , Histiocitose de Células de Langerhans/diagnóstico por imagem , Hipotálamo/diagnóstico por imagem , Doenças da Hipófise/diagnóstico por imagem , Hipófise/diagnóstico por imagem , Adulto , Antimetabólitos Antineoplásicos/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Diabetes Insípido/diagnóstico por imagem , Diabetes Insípido/tratamento farmacológico , Diabetes Insípido/etiologia , Histiocitose de Células de Langerhans/complicações , Histiocitose de Células de Langerhans/tratamento farmacológico , Humanos , Hipotálamo/efeitos dos fármacos , Masculino , Doenças da Hipófise/tratamento farmacológico , Doenças da Hipófise/etiologia , Hipófise/efeitos dos fármacos
15.
Aging (Albany NY) ; 12(14): 14019-14036, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32697764

RESUMO

BACKGROUND: Gestational diabetes mellitus (GDM) is a metabolic disease that can have long-term adverse effects on the cognitive function of mothers. In our study, we explored the changes in metabolic health and cognitive function in mice of middle- and old- age after exposure to GDM, and whether metformin therapy during pregnancy provided long-term benefits. RESULTS: Mice with GDM demonstrated significant cognitive impairment in old age, which was associated with insulin resistance. Gestational metformin therapy was shown to increase insulin sensitivity and improve cognition. The ovarian aging rate was also accelerated in mice exposed to GDM during pregnancy, which may be related to fatty acid metabolism in the ovaries. CONCLUSION: Treatment with metformin during pregnancy was shown to improve fatty acid metabolism in ovarian tissues. METHOD: During pregnancy, mice were fed with a high-fat diet (GDM group) or a low-fat diet (Control group), and a third group received metformin while receiving a high-fat diet (Treatment group). At 12 months old, the mice completed an oral glucose tolerance test, insulin tolerance test, Morris water maze test, female sex hormones were measured, and metabolite profiles of tissue from the ovaries, hypothalamus, and pituitary glands were analysed using gas chromatography-mass spectrometry.


Assuntos
Disfunção Cognitiva/prevenção & controle , Diabetes Gestacional/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Animais , Disfunção Cognitiva/etiologia , Diabetes Gestacional/metabolismo , Diabetes Gestacional/psicologia , Dieta com Restrição de Gorduras , Dieta Hiperlipídica , Feminino , Teste de Tolerância a Glucose , Hormônios Esteroides Gonadais/sangue , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Ovário/efeitos dos fármacos , Ovário/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Gravidez
16.
PLoS Comput Biol ; 16(6): e1007848, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32598357

RESUMO

Contraceptive drugs intended for family planning are used by the majority of married or in-union women in almost all regions of the world. The two most prevalent types of hormones associated with contraception are synthetic estrogens and progestins. Hormonal based contraceptives contain a dose of a synthetic progesterone (progestin) or a combination of a progestin and a synthetic estrogen. In this study we use mathematical modeling to understand better how these contraceptive paradigms prevent ovulation, special focus is on understanding how changes in dose impact hormonal cycling. To explain this phenomenon, we added two autocrine mechanisms essential to achieve contraception within our previous menstrual cycle models. This new model predicts mean daily blood concentrations of key hormones during a contraceptive state achieved by administering progestins, synthetic estrogens, or a combined treatment. Model outputs are compared with data from two clinical trials: one for a progestin only treatment and one for a combined hormonal treatment. Results show that contraception can be achieved with synthetic estrogen, with progestin, and by combining the two hormones. An advantage of the combined treatment is that a contraceptive state can be obtained at a lower dose of each hormone. The model studied here is qualitative in nature, but can be coupled with a pharmacokinetic/pharamacodynamic (PKPD) model providing the ability to fit exogenous inputs to specific bioavailability and affinity. A model of this type may allow insight into a specific drug's effects, which has potential to be useful in the pre-clinical trial stage identifying the lowest dose required to achieve contraception.


Assuntos
Anticoncepcionais/uso terapêutico , Contracepção Hormonal , Ciclo Menstrual/efeitos dos fármacos , Progestinas/uso terapêutico , Adulto , Estrogênios/uso terapêutico , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Modelos Biológicos , Ovário/efeitos dos fármacos , Hipófise/efeitos dos fármacos
17.
Domest Anim Endocrinol ; 72: 106473, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32361423

RESUMO

The action of glucocorticoids on target tissues is regulated by the glucocorticoid and mineralocorticoid receptors (codified by the NR3C1 and NR3C2 gene, respectively). Moreover, the prereceptor system, represented by the hydroxysteroid 11-beta dehydrogenases (HSD11Bs), catalyzes the interconversion from active glucocorticoids into inactive compounds. This study aimed to determine whether the expression of the prereceptor system, the corticosteroid receptors, and the molecules regulating their intracellular trafficking (FKBP prolyl isomerase 4 and FKBP prolyl isomerase 5) could be regulated in the hypothalamic-pituitary-adrenal axis and in different type of adipose tissue of calves by the administration of dexamethasone in combination with estradiol or prednisolone. Research about the glucocorticoid effects on bovine target tissues may allow development of new diagnostic methods that use potential molecular biomarkers of glucocorticoid treatment. The administration of dexamethasone in combination with estradiol increased the gene expression of HSD11B1 (P < 0.01), HSD11B2 (P < 0.05), NR3C1 (P < 0.01), and NR3C2 (P < 0.01) in the adrenal glands; NR3C2 in the intramuscular adipose tissue (P < 0.01), and HSD11B1 in the subcutaneous adipose tissue (P < 0.01). Prednisolone administration increased the gene expression of HSD11B1 (P < 0.01), NR3C1 (P < 0.05), and NR3C2 (P < 0.05) in the adrenal glands and HSD11B1 (P < 0.01) in the subcutaneous adipose tissue. Interestingly, most of the examined tissues/organs showed a significant variation of FKBP5 gene expression after the administration of dexamethasone in combination with estradiol. So, these changes suggest that the FKBP5 gene expression could be a possible biomarker of the illegal dexamethasone administration in calves.


Assuntos
Tecido Adiposo/metabolismo , Bovinos/fisiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Chaperonas Moleculares/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Esteroides/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Dexametasona/administração & dosagem , Dexametasona/farmacologia , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Hipófise/efeitos dos fármacos , Hipófise/metabolismo
18.
Curr Protein Pept Sci ; 21(8): 744-750, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32392109

RESUMO

In-depth studies have identified many hormones important for controlling mammary growth and maintaining lactation. One of these is melatonin, which is synthesized and secreted by the pineal gland to regulate circadian rhythms, improve antioxidant capacity, and enhance immunity. Prolactin is secreted by the pituitary gland and is associated with the growth and development of mammary glands as well as initiation and maintenance of lactation. The hypothalamus-pituitary system, the most important endocrine system in the body, regulates prolactin secretion mainly through dopamine released from tuberoinfundibular dopaminergic neurons. This review provides a reference for further study and describes the regulation of lactation and prolactin secretion by melatonin, primarily via the protection and stimulation of tuberoinfundibular dopaminergic neurons.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Lactação/efeitos dos fármacos , Melatonina/metabolismo , Hipófise/efeitos dos fármacos , Prolactina/biossíntese , Animais , Ritmo Circadiano/fisiologia , Neurônios Dopaminérgicos/metabolismo , Feminino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/fisiologia , Lactação/fisiologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/fisiologia , Melatonina/farmacologia , Glândula Pineal/metabolismo , Hipófise/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-32344129

RESUMO

In recent years, the adverse effects of cadmium (Cd2+) on aquatic systems have attracted much attention because Cd2+ can induce endocrine disorders and toxicity in aquatic organisms at low levels. However, its effects on the thyroid system in native fish in Lhasa are still unclear. In the present study, Schizopygopsis younghusbandi larvae were exposed to Cd2+ (0.25, 2.5, 25 or 250 µg/L) for 7 or 14 days to determine its toxic effects on thyroid function. The results showed that whole-body total T4 and T3 levels were significantly decreased, which was accompanied by the significant upregulation of the expression of the dio1 and dio2 genes after exposure to Cd2+ for 7 or 14 days. Genes related to thyroid hormone synthesis (crh and tshß) were upregulated after both 7 and 14 days of Cd2+ exposure, possibly due to the negative feedback regulation of the hypothalamic-pituitary-thyroid (HPT) axis caused by a decrease in thyroid hormone. In addition, survival rates and body lengths were reduced after treatment with Cd2+. This suggests that Cd2+ caused developmental toxicity in Schizopygopsis younghusbandi larvae. An integrated assessment of biomarker response (IBR) showed that there were dose-dependent and time-dependent effects of Cd2+ exposure on Schizopygopsis younghusbandi larvae. Schizopygopsis younghusbandi larvae were sensitive to Cd2+, which caused adverse effects at a concentration as low as 2.5 µg/L. In summary, the results indicated that Cd2+ causes thyroid disruption and developmental toxicity in Schizopygopsis younghusbandi larvae and that wild Schizopygopsis younghusbandi larvae living in the Lhasa River are at potential ecological risk.


Assuntos
Cádmio/toxicidade , Cyprinidae/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipotálamo/patologia , Larva/efeitos dos fármacos , Hipófise/patologia , Glândula Tireoide/patologia , Animais , Hipotálamo/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Glândula Tireoide/efeitos dos fármacos
20.
Life Sci ; 253: 117704, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32339542

RESUMO

Adverse effects of drugs on male reproductive system can be categorized as pre-testicular, testicular, and post-testicular. Pre-testicular adverse effects disrupt the hypothalamic-pituitary-gonadal (HPG) axis, generally by interfering with endocrine function. It is known that the HPG axis has roles in the maintenance of spermatogenesis and sexual function. The hypothalamus secretes gonadotropin-releasing hormone (GnRH) which enters the hypophyseal portal system to stimulate the anterior pituitary. The anterior pituitary secretes gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH) which are vital for spermatogenesis, into the blood. The FSH stimulates the Sertoli cells for the production of regulatory molecules and nutrients needed for the maintenance of spermatogenesis, while the LH stimulates the Leydig cells to produce and secrete testosterone. Many neurotransmitters influence the hypothalamic-pituitary regulation, consequently the HPG axis, and can consequently affect spermatogenesis and sexual function. Psychotropic drugs including antipsychotics, antidepressants, and mood stabilizers that all commonly modulate dopamine, serotonin, and GABA, can affect male spermatogenesis and sexual function by impairment of the hypothalamic-pituitary regulation, act like endocrine-disrupting chemicals. Otherwise, studies have shown the relationship between decreased sperm quality and psychotropic drugs treatment. Therefore, it is important to investigate the adverse reproductive effects of psychotropic drugs which are frequently used during reproductive ages in males and to determine the role of the hypothalamic-pituitary regulation axis on possible pathologies.


Assuntos
Disruptores Endócrinos/efeitos adversos , Psicotrópicos/efeitos adversos , Reprodução/efeitos dos fármacos , Animais , Disruptores Endócrinos/farmacologia , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/efeitos dos fármacos , Masculino , Hipófise/efeitos dos fármacos , Psicotrópicos/farmacologia , Espermatogênese/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA