Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Open Vet J ; 14(1): 428-437, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38633156

RESUMO

Background: Obesity is one of the most prevalent and perilous health affairs. Male obesity-associated secondary hypogonadism (MOSH) is one of many of its complexities, which is mounting in parallel with the aggravation of obesity. Magnetic nanoparticles seem to be an advanced favorable trend in multiple biomedical fields. Aim: In this study, we explore the therapeutic effects of superparamagnetic iron oxide nanoparticles (SPIONs) coated with carboxymethyl cellulose (CMC) on an obese male rat model with MOSH syndrome, comparing their impacts with a well-known anti-obesity medication (Orlistat). Methods: 42 male albino rats split into 7 equal groups: 1-negative control: nonobese, untreated; 35 rats fed the high fat-high fructose (HFHF) diet for a period of 12 weeks. Obese rats splitted into 6 equal groups; 2-positive control: obese untreated; 3-obese given Orlistat (30 mg/kg); 4-obese given CMC-SPIONs (25 mgFe/kg); 5-obese given CMC-SPIONs (50 mgFe/kg); 6-obese given CMC-SPIONs(25 mgFe/kg) + Orlistat (30 mg/kg), 7-obese given CMC-SPIONs (50 mgFe/kg) + Orlistat (30 mg/kg); all treatments given orally for 4 weeks. During sacrifice, blood serum and sectioned hypothalamic, pituitary, testicular, and adipose tissues were collected for biochemical and biomolecular assessments. Results: The HFHF diet for 12 weeks resulted in a significant upsurge in body weight, body mass index, serum fasting glucose, insulin resistance, TAG, total cholesterol, and LDL-c; HDL-c was dropped. Serum FSH, LH, and testosterone values declined. A significant disorder in expression levels of genes regulating the hypothalamic-pituitary-testicular-axis pathway. Hypothalamic GnRH, Kisspeptin-1, Kisspeptin-r1, and Adipo-R1 values declined. GnIH and Leptin-R1 values raised up. Pituitary GnRH-R values declined. Testicular tissue STAR, HSD17B3, and CYP19A1 values declined. Adipose tissue adiponectin declined, while leptin raised up. CMC-SPIONs 25-50 mg could modulate the deranged biochemical parameters and correct the deranged expression levels of all previous genes. Co-treatments revealed highly synergistic effects on all parameters. Overall, CMC-SPIONs have significant efficiency whether alone or with Orlisat in limiting obesity and consequence subfertility. Conclusion: CMC-SPIONs act as an incoming promising contender for obesity and MOSH disorders management, and need more studies on their mechanisms.


Assuntos
Hipogonadismo , Obesidade , Doenças dos Roedores , Ratos , Masculino , Animais , Leptina/metabolismo , Leptina/uso terapêutico , Orlistate/metabolismo , Orlistate/farmacologia , Orlistate/uso terapêutico , Testículo/metabolismo , Obesidade/genética , Obesidade/metabolismo , Obesidade/veterinária , Hipogonadismo/metabolismo , Hipogonadismo/veterinária , Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/uso terapêutico , Nanopartículas Magnéticas de Óxido de Ferro
2.
Toxicol Lett ; 369: 1-11, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-35963426

RESUMO

Isoflavones are phytoestrogens with recognized estrogenic activity but may also affect testosterone, corticosterone and thyroid hormone levels in experimental models. However, the molecular mechanisms involved in these alterations are still unclear. Isoflavones are present in soy-based infant formula, in breast milk after the consumption of soy by the mother and are widely used for the preparation of beverages consumed by toddlers and teenagers. In this sense, we proposed to investigate the effects of soy isoflavone exposure during the prepubertal period, a recognized window of sensitivity for endocrine disruption, over the hypothalamic-pituitary-testicular (HPT) axis. For this, 42 3-week-old male Wistar rats were exposed to 0.5, 5 or 50 mg of soy isoflavones/kg from postnatal day (PND) 23 to PND60. We evaluated body growth, age at puberty, serum concentrations of LH, FSH, testosterone and estradiol, and the expression of the transcripts (mRNA) of genes encoding key genes controlling the hypothalamic-pituitary-testicular (HPT) axis. In the hypothalamus, we observed an increase in Esr1 mRNA expression (0.5 and 5 mg). In the pituitary, we observed an increase in Gnrhr mRNA expression (50 mg), a reduction in Lhb mRNA expression (0.5 mg), and a reduction in Ar mRNA expression. In the testis, we observed an increase in Lhcgr mRNA expression (50 mg) and a reduction in Star mRNA expression (0.5 and 5 mg). The serum levels of LH (5 and 50 mg) and FSH (0.5 mg) were increased, while testosterone and estradiol were reduced. Puberty was delayed in all groups. Taken together, these results suggest that prepubertal consumption of relevant levels of soy isoflavones disrupts the HPT axis, causing hypergonadotropic hypogonadism and altered expression levels of key genes regulating the axis.


Assuntos
Hipogonadismo , Isoflavonas , Animais , Corticosterona , Estradiol/metabolismo , Hormônio Foliculoestimulante , Gonadotropinas Hipofisárias/metabolismo , Humanos , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Isoflavonas/farmacologia , Masculino , Fitoestrógenos/metabolismo , Fitoestrógenos/toxicidade , Puberdade , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Testosterona
3.
J Neuroendocrinol ; 33(7): e12994, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34156126

RESUMO

Prader-Willi Syndrome (PWS) is a rare and incurable congenital neurodevelopmental disorder, resulting from the absence of expression of a group of genes on the paternally acquired chromosome 15q11-q13. Phenotypical characteristics of PWS include infantile hypotonia, short stature, incomplete pubertal development, hyperphagia and morbid obesity. Hypothalamic dysfunction in controlling body weight and food intake is a hallmark of PWS. Neuroimaging studies have demonstrated that PWS subjects have abnormal neurocircuitry engaged in the hedonic and physiological control of feeding behavior. This is translated into diminished production of hypothalamic effector peptides which are responsible for the coordination of energy homeostasis and satiety. So far, studies with animal models for PWS and with human post-mortem hypothalamic specimens demonstrated changes particularly in the infundibular and the paraventricular nuclei of the hypothalamus, both in orexigenic and anorexigenic neural populations. Moreover, many PWS patients have a severe endocrine dysfunction, e.g. central hypogonadism and/or growth hormone deficiency, which may contribute to the development of increased fat mass, especially if left untreated. Additionally, the role of non-neuronal cells, such as astrocytes and microglia in the hypothalamic dysregulation in PWS is yet to be determined. Notably, microglial activation is persistently present in non-genetic obesity. To what extent microglia, and other glial cells, are affected in PWS is poorly understood. The elucidation of the hypothalamic dysfunction in PWS could prove to be a key feature of rational therapeutic management in this syndrome. This review aims to examine the evidence for hypothalamic dysfunction, both at the neuropeptidergic and circuitry levels, and its correlation with the pathophysiology of PWS.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Rede Nervosa/fisiopatologia , Síndrome de Prader-Willi , Animais , Humanos , Hiperfagia/etiologia , Hiperfagia/metabolismo , Hiperfagia/psicologia , Hipogonadismo/etiologia , Hipogonadismo/metabolismo , Hipogonadismo/psicologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Neuropeptídeos/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/psicologia , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Síndrome de Prader-Willi/psicologia
4.
Endocrinology ; 162(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33674833

RESUMO

Testosterone (T) reduces male fat mass, but the underlying mechanisms remain elusive, limiting its clinical relevance in hypogonadism-associated obesity. Here, we subjected chemically castrated high-fat diet-induced adult obese male mice to supplementation with T or the nonaromatizable androgen dihydrotestosterone (DHT) for 20 weeks. Both hormones increased lean mass, thereby indirectly increasing oxygen consumption and energy expenditure. In addition, T but not DHT decreased fat mass and increased ambulatory activity, indicating a role for aromatization into estrogens. Investigation of the pattern of aromatase expression in various murine tissues revealed the absence of Cyp19a1 expression in adipose tissue while high levels were observed in brain and gonads. In obese hypogonadal male mice with extrahypothalamic neuronal estrogen receptor alpha deletion (N-ERαKO), T still increased lean mass but was unable to decrease fat mass. The stimulatory effect of T on ambulatory activity was also abolished in N-ERαKO males. In conclusion, our work demonstrates that the fat-burning action of T is dependent on aromatization into estrogens and is at least partially mediated by the stimulation of physical activity via extrahypothalamic ERα signaling. In contrast, the increase in lean mass upon T supplementation is mediated through the androgen receptor and indirectly leads to an increase in energy expenditure, which might also contribute to the fat-burning effects of T.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Receptor alfa de Estrogênio/fisiologia , Atividade Motora/fisiologia , Testosterona/farmacologia , Tecido Adiposo/metabolismo , Animais , Di-Hidrotestosterona/farmacologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Atividade Motora/efeitos dos fármacos , Obesidade/genética , Obesidade/metabolismo , Condicionamento Físico Animal/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Congêneres da Testosterona/farmacologia
5.
Development ; 147(21)2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-32994169

RESUMO

Börjeson-Forssman-Lehmann syndrome (BFLS) is an intellectual disability and endocrine disorder caused by plant homeodomain finger 6 (PHF6) mutations. Individuals with BFLS present with short stature. We report a mouse model of BFLS, in which deletion of Phf6 causes a proportional reduction in body size compared with control mice. Growth hormone (GH) levels were reduced in the absence of PHF6. Phf6-/Y animals displayed a reduction in the expression of the genes encoding GH-releasing hormone (GHRH) in the brain, GH in the pituitary gland and insulin-like growth factor 1 (IGF1) in the liver. Phf6 deletion specifically in the nervous system caused a proportional growth defect, indicating a neuroendocrine contribution to the phenotype. Loss of suppressor of cytokine signaling 2 (SOCS2), a negative regulator of growth hormone signaling partially rescued body size, supporting a reversible deficiency in GH signaling. These results demonstrate that PHF6 regulates the GHRH/GH/IGF1 axis.


Assuntos
Regulação para Baixo , Epilepsia/metabolismo , Face/anormalidades , Dedos/anormalidades , Transtornos do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio do Crescimento/metabolismo , Hipogonadismo/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Obesidade/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Epilepsia/sangue , Epilepsia/patologia , Face/patologia , Dedos/patologia , Transtornos do Crescimento/sangue , Transtornos do Crescimento/patologia , Hormônio do Crescimento/sangue , Hipogonadismo/sangue , Hipogonadismo/patologia , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/genética , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/sangue , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sistema Nervoso/metabolismo , Obesidade/sangue , Obesidade/patologia , Especificidade de Órgãos , Hipófise/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo
6.
Mol Cell Endocrinol ; 518: 110996, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32860862

RESUMO

The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.


Assuntos
Células Endócrinas/fisiologia , Hipogonadismo/etiologia , Animais , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Neurônios/fisiologia , Sistemas Neurossecretores/metabolismo , Sistemas Neurossecretores/fisiologia , Reprodução/fisiologia
7.
Gene ; 754: 144885, 2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32535046

RESUMO

BACKGROUND: Congenital hypogonadotropic hypogonadism (CHH) is a rare genetically heterogeneous disorder. We aimed to determine the prevalence and pathogenesis of NECL2 (Nectin-like molecule 2) variants in a cohort of female patients with CHH. METHODS: We sequenced and determined the prevalence of NECL2 variants in 68 female patients with CHH and 243 healthy controls collected from an academic medical center. Further cellular and animal studies were performed to verify the pathogenicity of the mutations. Necl2 knockout female mice were generated, and their puberty development was observed. RESULTS: A novel NECL2 variant (c.1052_1060del, p.Thr351_Thr353del) was detected in 4 of 68 (5.9%) patients with CHH. Its prevalence was significantly higher in CHH patients than in healthy controls (0%). At the cellular level, the necl2 variant leads to a decrease in gonadotropin-releasing hormone. In animal models, we found that the Necl2 protein was expressed in the hypothalamus, especially in the ventromedial hypothalamic nucleus of mice. Necl2 knockout female mice showed delayed puberty and an irregular estrous cycle, consistent with CHH patient phenotypes. CONCLUSIONS: Our findings predict that NECL2 may be a new candidate gene for CHH and that the NECL2 protein plays a critical role in the progression of puberty development.


Assuntos
Molécula 1 de Adesão Celular/genética , Molécula 1 de Adesão Celular/metabolismo , Hipogonadismo/patologia , Mutação , Puberdade , Maturidade Sexual , Adolescente , Adulto , Animais , Apoptose , Estudos de Casos e Controles , Adesão Celular , Proliferação de Células , Células Cultivadas , Estudos de Coortes , Ciclo Estral , Feminino , Hormônio Liberador de Gonadotropina , Humanos , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Prognóstico , Adulto Jovem
8.
J Reprod Dev ; 66(4): 359-367, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32307336

RESUMO

The present study aimed to evaluate whether novel conditional kisspeptin neuron-specific Kiss1 knockout (KO) mice utilizing the Cre-loxP system could recapitulate the infertility of global Kiss1 KO models, thereby providing further evidence for the fundamental role of hypothalamic kisspeptin neurons in regulating mammalian reproduction. We generated Kiss1-floxed mice and hypothalamic kisspeptin neuron-specific Cre-expressing transgenic mice and then crossed these two lines. The conditional Kiss1 KO mice showed pubertal failure along with a suppression of gonadotropin secretion and ovarian atrophy. These results indicate that newly-created hypothalamic Kiss1 KO mice obtained by the Cre-loxP system recapitulated the infertility of global Kiss1 KO models, suggesting that hypothalamic kisspeptin, but not peripheral kisspeptin, is critical for reproduction. Importantly, these Kiss1-floxed mice are now available and will be a valuable tool for detailed analyses of roles of each population of kisspeptin neurons in the brain and peripheral kisspeptin-producing cells by the spatiotemporal-specific manipulation of Cre expression.


Assuntos
Hipogonadismo/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Neurônios/metabolismo , Animais , Hipogonadismo/metabolismo , Kisspeptinas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo
9.
J Diabetes Res ; 2019: 7894950, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281852

RESUMO

OBJECTIVE: In diabetes mellitus, vitamin D3 deficiency affects sex hormone levels and male fertility; however, the mechanism leading to the disorder is unclear. This research was designed to investigate the mechanism of vitamin D3 deficiency and hypogonadism in diabetic rats. Our aim was to assess serum vitamin D3 levels and the relationship among vitamin D3, insulin-like growth factor-1 (IGF-1), and testicular function. MATERIALS AND METHODS: Rats with streptozotocin-induced diabetes were randomly divided into four groups and treated with different doses of vitamin D3: no vitamin D3, low (0.025 µg/kg/day), high (0.1 µg/kg/day), and high (0.1 µg/kg/day) with JB-1 (the insulin-like growth factor-1 receptor inhibitor group, 100 µg/kg/day). The groups were compared with wild-type rats, which function as the control group. Various parameters such as vitamin D3 and IGF-1 were compared between the experimental and wild-type groups, and their correlations were determined. RESULTS: Twelve weeks of vitamin D3 supplementation improved the testosterone levels, as shown by the increase in the level of serum IGF-1 in diabetic rats. Phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT), which was a downstream of the signaling pathway of IGF-1, was significantly increased after vitamin D3 treatment. CONCLUSIONS: The study shows that vitamin D3 may promote the expression of testosterone and improve testicular function in diabetic rats by activating PI3K/AKT via IGF-1.


Assuntos
Colecalciferol/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Testículo/fisiologia , Animais , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Hipogonadismo/metabolismo , Masculino , Tamanho do Órgão , Fosforilação , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
10.
Med Hypotheses ; 128: 54-57, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31203909

RESUMO

Hypogonadotropic hypogonadism (secondary hypogonadism), congenital or acquired, is a form of hypogonadism that is due to problems with either the hypothalamus or pituitary gland affecting gonadotropin levels. Pulsatile secretion of gonadotropin-releasing hormone (GnRH) by hypothalamus is a primer step to initiate the release of pituitary gonadotropins. Kisspeptin and gonadotropin-inhibitory hormone (GnIH) are accepted as two major players in the activation and inhibition of GnRH regarding the neuroendocrine functioning of the hypothalamic pituitary gonadal axis. Kisspeptin is known as the most potent activator of GnRH. Regarding the inhibition of GnRH, RF-amide-related peptide-3 (RFRP-3) is accepted as the mammalian orthologue of GnIH in avian species. RF9 (1-adamantane carbonyl-Arg-Phe-NH2) is an antagonist of RFRP-3/GnIH receptor (neuropeptide FF receptor 1 (NPFFR1; also termed as GPR147). In recent years, several studies have indicated that RF9 activates GnRH neurons and gonadotropins in a kisspeptin receptor (Kiss1r, formerly known as GPR54) dependent manner. These results suggest that RF9 may have a bimodal function as both an RFRP-3 antagonist and a kisspeptin agonist or it may be a kiss1r agonist rather than an RFRP-3/GnIH receptor antagonist. These interactions are possible because Kisspeptin and GnIH are members of the RF-amide family, and both possibilities are not far from explaining the potent gonadotropin stimulating effects of RF9. Therefore, we hypothesize that RF9 may be a new therapeutic option for the hypogonadotropic hypogonadism due to its potent GnRH stimulating effects. A constant or repeated administration of RF9 provides a sustained increase in plasma gonadotrophin levels. However, applications in the same way with GnRH analogues and kisspeptin may result in desensitization of the gonadotropic axis. The reasons reported above contribute to our hypothesis that RF9 may be a good option in the GnRH stimulating as a kisspeptin agonist. We suggest that further studies are needed to elucidate the potential effects of RF9 in the treatment of the hypogonadotropic hypogonadism.


Assuntos
Adamantano/análogos & derivados , Dipeptídeos/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Adamantano/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Hipogonadismo/terapia , Camundongos , Modelos Biológicos , Modelos Teóricos , Neuropeptídeos/metabolismo , Ratos , Receptores de Kisspeptina-1/metabolismo , Receptores de Neuropeptídeos/metabolismo
11.
Andrologia ; 51(8): e13323, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31134680

RESUMO

High-fat diets (HFDs) are detrimental to steroidogenesis and male fertility. This study aimed to investigate the protective effects of melatonin (MT) treatment on testicular dysfunction in mice fed with HFD. C57BL/6J male mice were randomly divided into three groups: CTRL, HFD and HFD + MT. MT treatment mitigated the increase in body weight and adipose tissue in HFD-fed mice. Serum levels of sex hormones were improved upon MT supplementation, and the expression of the testosterone synthesis proteins, StAR and P450scc was rescued as well. MT treatment significantly up-regulated the expression of SIRT1, SOD2, and GPx4 and down-regulated the expression of GRP78 and CHOP, indicating an attenuation of oxidative stress (OS) and endoplasmic reticulum (ER) stress. In TM3 cells, MT treatment protected against H2 O2 -induced steroidogenic collapse by improving mitochondrial function and attenuating OS and ER stress. These results indicate that MT treatment can improve steroidogenesis in mice fed with HFD and may have therapeutic value in the treatment of obesity-associated hypogonadism.


Assuntos
Hipogonadismo/tratamento farmacológico , Células Intersticiais do Testículo/efeitos dos fármacos , Melatonina/administração & dosagem , Obesidade/complicações , Testosterona/biossíntese , Animais , Linhagem Celular , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/toxicidade , Hipogonadismo/etiologia , Hipogonadismo/metabolismo , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Obesidade/etiologia , Obesidade/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfoproteínas/metabolismo , Resultado do Tratamento
12.
Neuropeptides ; 74: 34-43, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30503692

RESUMO

To explore the role of nesfatin-1 in regulating male reproductive function during energy balance variation, we employed an obese mouse model which was first induced by a high-fat diet (HFD) and followed by interventions of a normal diet (ND) and/or moderate exercise, and then serum reproductive hormones of male mice, hypothalamic nucleobindin 2 (NUCB2)/nesfatin-1, inflammatory factors, and gonadotropin-releasing hormone (GnRH) levels were tested. Our findings showed that both serum nesfatin-1, follicle-stimulating hormone (FSH), luteinizing hormone (LH), and testosterone (T) levels and hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels were reduced, whereas, the mRNA and protein levels of hypothalamic tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, inhibitor kappa B kinase ß (IKKß), and nuclear factor (NF)-κB were increased in obese male mice. Diet, exercise, and diet combined with exercise interventions reversed the decreases in serum nesfatin-1, FSH, LH, and T levels; increased hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels; and reduced hypothalamic TNF-α, IL-1ß, IKKß, and NF-κB levels. These changes were accompanied by reduced adiposity, and these effects were more obvious in the diet combined with exercise group. Overall, our findings suggested that the hypogonadotropic hypogonadism associated with obesity may be induced by reduced hypothalamic NUCB2/nesfatin-1 levels, which attenuated the stimulatory effect on GnRH directly or indirectly by suppressing its anti-inflammatory effect in the brain. Diet and/or exercise interventions were able to alleviate the hypogonadotropic hypogonadism associated with obesity, potentially by increasing hypothalamic NUCB2/nesfatin-1 levels.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Encefalite/metabolismo , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Obesidade/metabolismo , Condicionamento Físico Animal , Animais , Dieta Hiperlipídica , Encefalite/complicações , Hormônio Liberador de Gonadotropina/metabolismo , Hipogonadismo/complicações , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Nucleobindinas , RNA Mensageiro/metabolismo
13.
Cell Mol Biol (Noisy-le-grand) ; 64(10): 20-27, 2018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-30084791

RESUMO

Late-onset hypogonadism (LOH) is associated with advancing age and is caused by a deficiency in serum testosterone levels. The aim of this study was to examine the effect of a Dendropanax morbiferus H.Lév. leaf extract (DME) on LOH using TM3 cells and aging male rats as in vitro and in vivo models, respectively. The in vitro effects of DME on testosterone levels and 3ß-hydroxysteroid dehydrogenase (3ß-HSD) protein expression in TM3 cells were analyzed. In the in vivo experiments, DME was orally administered to rats at three doses (50, 100, and 200 mg/kg/day) for 4 weeks. DME significantly increased the testosterone levels and 3ß-HSD protein expression in TM3 cells. The DME groups showed significantly increased levels of androgenic hormones such as testosterone and dehydroepiandrosterone sulfate. The sex hormone-binding globulin production was significantly lower in the DME groups than that in the control group, while the neurohormone levels in the hypothalamic-pituitary-gonadal axis markedly increased. No significant differences were observed in the glutamic pyruvic transaminase, glutamic oxaloacetic transaminase, and prostate-specific antigen levels among the DME and control groups. The triglyceride and low-density lipoprotein cholesterol levels were significantly lower, while the high-density lipoprotein cholesterol levels were significantly higher in the DME groups than those in the control group. The latency time in the rotarod, treadmill, and swimming tests increased with the DME treatment. Furthermore, the sperm counts in the epididymis markedly increased. These results suggest that DME can be effectively used to alleviate the symptoms of LOH.


Assuntos
Araliaceae/química , Hipogonadismo/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Folhas de Planta/química , Testosterona/metabolismo , 17-Hidroxiesteroide Desidrogenases/análise , 17-Hidroxiesteroide Desidrogenases/metabolismo , Envelhecimento , Animais , Linhagem Celular , Hipogonadismo/sangue , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/patologia , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia , Masculino , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Ratos , Ratos Sprague-Dawley , Testosterona/análise , Testosterona/sangue
14.
EBioMedicine ; 35: 295-306, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30131307

RESUMO

BACKGROUND: Nerve growth factor (NGF) plays essential roles in regulating the development and maintenance of central sympathetic and sensory neurons. However, the effects of NGF on hypogonadism remain unexplored. METHODS: To assess the effects of NGF on hypogonadism, we established a convenient and noninvasive way to deliver NGF to the hypothalamus by spraying liposome-encapsulated NGF into the nasal cavity. The ten-month-old aging male senescence accelerate mouse P8 (SAMP8) mice with age-related hypogonadotrophic hypogonadism were used to study the role of NGF in hypogonadism. The age-matched accelerated senescence-resistant mouse R1 (SAMR1) served as a control. The ten-month-old SAMP8 mice were treated with NGF twice per week for 12 weeks. Sexual hormones, sexual behaviors, and fertility were analyzed after NGF treatment. And the mechanisms of NGF in sex hormones sexual function were also studied. FINDINGS: NGF could enhance the sexual function, improve the quality of the sperm, and restore the fertility of aging male SAMP8 mice with age-related hypogonadism by activating gonadotropin-releasing hormone (GnRH) neuron and regulating secretion of GnRH. And NGF regulated the GnRH release through the PKC/p-ERK1/2/p-CREB signal pathway. INTERPRETATION: These results suggest that NGF treatment could alleviate various age-related hypogonadism symptoms in male SAMP8 and may be usefulness for age-related hypogonadotrophic hypogonadism and its related subfertility. FUND: National Natural Science Foundation of China, Natural Science Foundation of Guangdong Province, the Science and Technology Plan Project of Guangzhou, Wenzhou Science & Technology Bureau, Guangdong Province Pearl River Scholar Fund, Guangdong province science and technology innovation leading Scholar Fund.


Assuntos
Envelhecimento/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Fator de Crescimento Neural/administração & dosagem , Fator de Crescimento Neural/uso terapêutico , Testosterona/metabolismo , Regulação para Cima , Administração Intranasal , Animais , Feminino , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Fator de Crescimento Neural/farmacologia , Neurônios/metabolismo , Comportamento Sexual Animal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
15.
J Clin Endocrinol Metab ; 103(12): 4419-4427, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30032193

RESUMO

Context: Testosterone (T) increases GH secretion in older men with a relative lack of T, in hypogonadal men of all ages, and in patients undergoing sex reassignment. The role of estradiol (E2) in men is less well defined. Objective: To assess the contribution of aromatization of T to spontaneous nocturnal and stimulated GH secretion. Participants: Four groups of healthy older men (N = 74, age range 57 to 77 years) were studied. The gonadotropic axis was clamped with the gonadotropin-releasing hormone antagonist degarelix. Three groups received T and one group placebo addback. Two T-replaced groups were treated with anastrozole (an aromatase inhibitor) and either placebo or E2 addback. Main Outcome Measures: Ten-minute GH concentration profiles were quantified by deconvolution analysis, after overnight (2200 to 0800 hours) sampling, and after combined IV injection of GHRH (0.3 µg/kg) and GHRH-2 (0.3 µg/kg) and withdrawal of a 2-hour somatostatin infusion (1 µg/kg/h). Results: E2 addback during aromatase inhibition increased basal (P = 0.046), pulsatile (P = 0.020), and total (P = 0.018) GH secretion by 60% to 70%. E2 did not potentiate GH secretory stimuli. Logarithmically transformed pulsatile GH secretion correlated strongly and positively with concurrent E2 concentrations overall (P = 0.028) and under anastrozole treatment (P = 0.005). Conclusion: E2 administration in older men transdermally stimulates overnight pulsatile GH secretion. The exact site of E2 action cannot be ascertained from these experiments but may include hypothalamic loci involved in GH regulation, especially because GH secretagogue effects on somatotrope pituitary cells were not affected.


Assuntos
Envelhecimento/metabolismo , Estradiol/administração & dosagem , Hormônio do Crescimento Humano/metabolismo , Hipogonadismo/tratamento farmacológico , Testosterona/administração & dosagem , Administração Cutânea , Adulto , Idoso , Envelhecimento/efeitos dos fármacos , Anastrozol/administração & dosagem , Aromatase/metabolismo , Inibidores da Aromatase/administração & dosagem , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Voluntários Saudáveis , Hormônio do Crescimento Humano/sangue , Humanos , Hipogonadismo/induzido quimicamente , Hipogonadismo/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Placebos/administração & dosagem , Testosterona/metabolismo
16.
Endocrine ; 62(1): 195-206, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29948931

RESUMO

PURPOSE: Obesity may lead to male hypogonadism, the underlying mechanism of which remains unclear. In the present study, we established a murine model of male hypogonadism caused by high-fat diet-induced obesity to verify the following hypotheses: 1) an increased leptin level may be related to decreased secretion of GnRH in obese males, and 2) repression of kisspeptin/GPR54 in the hypothalamus, which is associated with increased leptin levels, may account for the decreased secretion of GnRH and be involved in secondary hypogonadism (SH) in obese males. METHODS: Male mice were fed high-fat diet for 19 weeks and divided by body weight gain into diet-induced obesity (DIO) and diet-induced obesity resistant (DIO-R) group. The effect of obesity on the reproductive organs in male mice was observed by measuring sperm count and spermatozoid motility, relative to testis and epididymis weight, testosterone levels, and pathologic changes. Leptin, testosterone, estrogen, and LH in serum were detected by ELISA method. Leptin receptor (Ob-R), Kiss1, GPR54, and GnRH mRNA were measured by real-time PCR in the hypothalamus. Expression of kisspeptin and Ob-R protein was determined by Western blotting. Expression of GnRH and GPR54 protein was determined by immunohistochemical analysis. RESULTS: We found that diet-induced obesity decreased spermatozoid motility, testis and epididymis relative coefficients, and plasma testosterone and luteinizing hormone levels. An increased number and volume of lipid droplets in Leydig cells were observed in the DIO group compared to the control group. Significantly, higher serum leptin levels were found in the DIO and DIO-R groups. The DIO and DIO-R groups showed significant downregulation of the GnRH, Kiss1, GPR54, and Ob-R genes. We also found decreased levels of GnRH, kisspeptin, GPR54, and Ob-R protein in the DIO and DIO-R groups. CONCLUSIONS: These lines of evidence suggest that downregulation of Ob-R and kisspeptin/GPR54 in the murine hypothalamus may contribute to male hypogonadism caused by high-fat diet-induced obesity.


Assuntos
Regulação para Baixo , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Obesidade/metabolismo , Receptores de Kisspeptina-1/metabolismo , Receptores para Leptina/metabolismo , Animais , Peso Corporal , Dieta Hiperlipídica , Modelos Animais de Doenças , Hormônio Liberador de Gonadotropina/metabolismo , Hipogonadismo/etiologia , Hipogonadismo/genética , Kisspeptinas/genética , Leptina/sangue , Masculino , Camundongos , Obesidade/complicações , Obesidade/genética , Receptores de Kisspeptina-1/genética , Receptores para Leptina/genética , Motilidade dos Espermatozoides/fisiologia , Testículo/metabolismo
17.
Obes Surg ; 28(8): 2272-2280, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29508270

RESUMO

BACKGROUND: Obesity is a metabolic disease with a serious health burden in children and adults, and it induces a variety of conditions including subfecundity. Sleeve gastrectomy showed encouraging results in terms of weight loss and improve quality of life, and this study aimed to determine whether sleeve gastrectomy could reverse obesity-induced impaired fertility in male Sprague-Dawley rats. METHODS: After 16 weeks of a chow diet (CD) or a high-fat diet (HFD) challenge, rats on the HFD were given a sleeve gastrectomy or sham operation and then fed an HFD for another 8 weeks. Serum glucose, insulin, lipids, sex hormone, sperm quality, inflammatory profile of the testis, and hypothalamic Kiss1 expression in the three study groups were compared. RESULTS: Sleeve gastrectomy significantly decreased HFD-induced obesity and serum glucose and insulin levels. It also reversed the HFD-induced increase in teratozoospermia and decreases in sperm motility and progressive motility. Testicular morphological abnormalities were also improved after sleeve gastrectomy. Enzyme-linked immunosorbent assay showed that the expression of sex hormones increased after sleeve gastrectomy and that expression of inflammatory factors decreased. The HFD induced a hypothalamic inflammatory response that inhibited Kiss1 expression, which in turn mediated sex hormone expression. Sleeve gastrectomy treatment improved the hypothalamic response. CONCLUSIONS: The results consistently showed that sleeve gastrectomy reversed obesity-induced male fertility impairment by decreasing the inflammatory responses of the testis and hypothalamus.


Assuntos
Dieta Hiperlipídica , Gastrectomia , Hipotálamo , Obesidade Mórbida , Obesidade , Testículo , Animais , Modelos Animais de Doenças , Gastrectomia/métodos , Humanos , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Kisspeptinas/metabolismo , Masculino , Obesidade/cirurgia , Obesidade Mórbida/cirurgia , Qualidade de Vida , Ratos , Ratos Sprague-Dawley , Motilidade dos Espermatozoides , Testículo/metabolismo , Redução de Peso
18.
Adv Exp Med Biol ; 1034: 25-28, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29256124

RESUMO

Due in part to aggressive marketing, the prevalence of exogenous androgen use has increased to disturbing levels. Prescribing practitioners are often unaware of the severity of the anti-fertility effects. Exogenous androgens should only be prescribed if hypogonadism has been established by appropriate investigation, and preferably the patient does not intend to father a child. There are alternative medications, or combinations of medications, that can be used if hypogonadism is present and fertility is desired.It is somewhat counterintuitive that testosterone treatment will decrease or abolish fertility. Exogenous testosterone inhibits spermatogenesis by removing the feedback response to low testosterone at the hypothalamus and pituitary. This results in reduced synthesis and secretion of gonadotropins required to stimulate endogenous testosterone production and to support spermatogenesis. It is important to realize that the normal testicular levels of testosterone are approximately 100 times the concentration in circulation. These high levels are required locally to support spermatogenesis. So even with circulating androgen levels within the normal range, spermatogenesis fails due to insufficient gonadotropin and local testosterone support. Androgenic herbal supplements and illicit use of anabolic steroids have contributed to this serious challenge in the treatment of infertile men. Most men will recover normal spermatogenesis after cessation of exogenous testosterone treatment, but this requires 6 months or more in most men. In rare cases fertility is permanently impaired.


Assuntos
Androgênios/efeitos adversos , Androgênios/uso terapêutico , Hipogonadismo , Reprodução/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Gonadotropinas/metabolismo , Humanos , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Masculino , Hipófise/metabolismo , Hipófise/patologia , Testosterona/metabolismo
19.
Eur J Endocrinol ; 176(6): R283-R308, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28264815

RESUMO

Aging and its underlying pathophysiological background has always attracted the attention of the scientific society. Defined as the gradual, time-dependent, heterogeneous decline of physiological functions, aging is orchestrated by a plethora of molecular mechanisms, which vividly interact to alter body homeostasis. The ability of an organism to adjust to these alterations, in conjunction with the dynamic effect of various environmental stimuli across lifespan, promotes longevity, frailty or disease. Endocrine function undergoes major changes during aging, as well. Specifically, alterations in hormonal networks and concomitant hormonal deficits/excess, augmented by poor sensitivity of tissues to their action, take place. As hypothalamic-pituitary unit is the central regulator of crucial body functions, these alterations can be translated in significant clinical sequelae that can impair the quality of life and promote frailty and disease. Delineating the hormonal signaling alterations that occur across lifespan and exploring possible remedial interventions could possibly help us improve the quality of life of the elderly and promote longevity.


Assuntos
Envelhecimento/metabolismo , Sistema Endócrino/metabolismo , Estresse Oxidativo , Adjuvantes Imunológicos/uso terapêutico , Androgênios/uso terapêutico , Antioxidantes/uso terapêutico , Ritmo Circadiano , Desidroepiandrosterona/uso terapêutico , Diabetes Mellitus Tipo 2/metabolismo , Dietoterapia , Terapia de Reposição de Estrogênios , Retroalimentação Fisiológica , Feminino , Preservação da Fertilidade , Gonadotropinas/metabolismo , Terapia de Reposição Hormonal , Humanos , Hiperandrogenismo/metabolismo , Hipertireoidismo/metabolismo , Hipertireoidismo/terapia , Hipoglicemiantes/uso terapêutico , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Menopausa/metabolismo , Reserva Ovariana , Medicina de Precisão , Qualidade de Vida , Transplante de Células-Tronco , Células-Tronco , Testosterona/uso terapêutico , Glândula Tireoide , Equilíbrio Hidroeletrolítico
20.
J Clin Endocrinol Metab ; 102(3): 1067-1075, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28359097

RESUMO

Context: Middle-aged and older men (≥50 years), especially those who are obese and suffer from comorbidities, not uncommonly present with clinical features consistent with androgen deficiency and modestly reduced testosterone levels. Commonly, such men do not demonstrate anatomical hypothalamic-pituitary-testicular axis pathology but have functional hypogonadism that is potentially reversible. Evidence Acquisition: Literature review from 1970 to October 2016. Evidence Synthesis: Although definitive randomized controlled trials are lacking, evidence suggests that in such men, lifestyle measures to achieve weight loss and optimization of comorbidities, including discontinuation of offending medications, lead to clinical improvement and a modest increase in testosterone. Also, androgen deficiency-like symptoms and end-organ deficits respond to targeted treatments (such as phosphodiesterase-5 inhibitors for erectile dysfunction) without evidence that hypogonadal men are refractory. Unfortunately, lifestyle interventions remain difficult and may be insufficient even if successful. Testosterone therapy should be considered primarily for men who have significant clinical features of androgen deficiency and unequivocally low testosterone levels. Testosterone should be initiated either concomitantly with a trial of lifestyle measures, or after such a trial fails, after a tailored diagnostic work-up, exclusion of contraindications, and appropriate counseling. Conclusions: There is modest evidence that functional hypogonadism responds to lifestyle measures and optimization of comorbidities. If achievable, these interventions may have demonstrable health benefits beyond the potential for increasing testosterone levels. Therefore, treatment of underlying causes of functional hypogonadism and of symptoms should be used either as an initial or adjunctive approach to testosterone therapy.


Assuntos
Androgênios/uso terapêutico , Dietoterapia , Disfunção Erétil/tratamento farmacológico , Exercício Físico , Hipogonadismo/terapia , Obesidade/terapia , Inibidores da Fosfodiesterase 5/uso terapêutico , Testosterona/uso terapêutico , Idoso , Disfunção Erétil/etiologia , Humanos , Hipogonadismo/complicações , Hipogonadismo/metabolismo , Estilo de Vida , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/metabolismo , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA