Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 267
Filtrar
1.
Ann Med ; 56(1): 2329259, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38738380

RESUMEN

Opioids are the most prescribed drugs for the alleviation of pain. Both clinical and preclinical studies have reported strong evidence for sex-related divergence regarding opioid analgesia. There is an increasing amount of evidence indicating that gonadal hormones regulate the analgesic efficacy of opioids. This review presents an overview of the importance of gonadal steroids in modulating opioid analgesic responsiveness and focuses on elaborating what is currently known regarding the underlyingmechanism. We sought to identify the link between gonadal hormones and the effect of oipiod antinociception.


Gonadal hormones contribute to the sexual dimorphism of opioid antinociception.Generally, oestradiol is a negative modulator of opioid analgesia via both non-genomic and genomic effects.Testosterone facilitates opioid analgesia mainly through the transcriptional activities of androgen receptors.Under normal physiological conditions, progestin and oestrogen exist in parallel and have a combined effect. However, progestin alone could promote opioid analgesia by increasing the expression of opioid receptors.


Asunto(s)
Analgésicos Opioides , Hormonas Gonadales , Dolor , Analgésicos Opioides/farmacología , Humanos , Animales , Hormonas Gonadales/metabolismo , Masculino , Dolor/tratamiento farmacológico , Dolor/metabolismo , Femenino
2.
Physiol Genomics ; 56(2): 179-193, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38047312

RESUMEN

Epigenetic alterations such as dysregulation of miRNAs have been reported to play important roles in interactions between genetic and environmental factors. In this study, we tested the hypothesis that induction of lung inflammation by inhaled allergens triggers a sex-specific miRNA regulation that is dependent on chromosome complement and hormonal milieu. We challenged the four core genotypes (FCGs) model through intranasal sensitization with a house dust mite (HDM) solution (or PBS as a control) for 5 wk. The FCG model allows four combinations of gonads and sex chromosomes: 1) XX mice with ovaries (XXF), 2) XY mice with testes (XYM), 3) XX mice with testes (XXM), and 4) XY mice with ovaries (XYF). Following the challenge (n = 5-7/group), we assessed the expression of 84 inflammatory miRNAs in lung tissue using a PCR array and cytokine levels in bronchoalveolar lavage fluid (BAL) by a multiplex protein assay (n = 4-7 animals/group). Our results showed higher levels of the chemokine KC (an Il-8 homolog) and IL-7 in BAL from XYF mice challenged with HDM. In addition, IL-17A was significantly higher in BAL from both XXF and XYF mice. A three-way interaction among treatment, gonads, and sex chromosome revealed 60 of 64 miRNAs that differed in expression depending on genotype; XXF, XXM, XYF, and XYM mice had 45, 32, 4, and 52 differentially expressed miRNAs, respectively. Regulatory networks of miRNAs identified in this study were implicated in pathways associated with asthma. Female gonadal hormonal effects may alter miRNA expression and contribute to the higher susceptibility of females to asthma.NEW & NOTEWORTHY miRNAs play important roles in regulating gene and environmental interactions. However, their role in mediating sex differences in allergic responses and lung diseases has not been elucidated. Our study used a targeted omics approach to characterize the contributions of gonadal hormones and chromosomal components to lung responses to an allergen challenge. Our results point to the influence of sex hormones in miRNA expression and proinflammatory markers in allergic airway inflammation.


Asunto(s)
Asma , MicroARNs , Femenino , Animales , Ratones , Masculino , Citocinas/genética , MicroARNs/genética , MicroARNs/metabolismo , Pulmón/metabolismo , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Asma/genética , Asma/metabolismo , Inflamación/genética , Inflamación/metabolismo , Líquido del Lavado Bronquioalveolar , Hormonas Gonadales/genética , Hormonas Gonadales/metabolismo , Modelos Animales de Enfermedad
3.
Horm Behav ; 158: 105463, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37995608

RESUMEN

The socially monogamous prairie vole (Microtus ochrogaster) and promiscuous meadow vole (Microtus pennsylvanicus) are closely related, but only prairie voles display long-lasting pair bonds, biparental care, and selective aggression towards unfamiliar individuals after pair bonding. These social behaviors in mammals are largely mediated by steroid hormone signaling in the social behavior network (SBN) of the brain. Hormone receptors are reproducible markers of sex differences that can provide more information than anatomy alone and can even be at odds with anatomical dimorphisms. We reasoned that behaviors associated with social monogamy in prairie voles may emerge in part from unique expression patterns of steroid hormone receptors in this species, and that these expression patterns would be more similar across males and females in prairie than in meadow voles or the laboratory mouse. To obtain insight into steroid hormone signaling in the developing prairie vole brain, we assessed expression of estrogen receptor alpha (Esr1), estrogen receptor beta (Esr2), and androgen receptor (Ar) within the SBN, using in situ hybridization at postnatal day 14 in mice, meadow, and prairie voles. We found species-specific patterns of hormone receptor expression in the hippocampus and ventromedial hypothalamus, as well as species differences in the sex bias of these markers in the principal nucleus of the bed nucleus of the stria terminalis. These findings suggest the observed differences in gonadal hormone receptor expression may underlie species differences in the display of social behaviors.


Asunto(s)
Encéfalo , Pradera , Femenino , Animales , Masculino , Ratones , Encéfalo/metabolismo , Conducta Social , Arvicolinae/metabolismo , Hormonas/metabolismo , Hormonas Gonadales/metabolismo , Esteroides/metabolismo
4.
Brain Behav Immun ; 115: 680-695, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37972878

RESUMEN

There is a strong male bias in the prevalence of many neurodevelopmental disorders such as autism spectrum disorder. However, the mechanisms underlying this sex bias remain elusive. Infection during the perinatal period is associated with an increased risk of neurodevelopmental disorder development. Here, we used a mouse model of early-life immune activation that reliably induces deficits in social behaviors only in males. We demonstrate that male-biased alterations in social behavior are dependent upon microglial immune signaling and are coupled to alterations in mitochondrial morphology, gene expression, and function specifically within microglia, the innate immune cells of the brain. Additionally, we show that this behavioral and microglial mitochondrial vulnerability to early-life immune activation is programmed by the male-typical perinatal gonadal hormone surge. These findings demonstrate that social behavior in males over the lifespan are regulated by microglia-specific mechanisms that are shaped by events that occur in early development.


Asunto(s)
Trastorno del Espectro Autista , Microglía , Animales , Ratones , Embarazo , Femenino , Masculino , Microglía/metabolismo , Encéfalo/metabolismo , Hormonas Gonadales/metabolismo , Mitocondrias/metabolismo
5.
Mar Biotechnol (NY) ; 25(6): 983-996, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37831333

RESUMEN

The insulin-like androgenic gland hormone gene (IAG), primarily expressed in the androgenic gland (AG), plays a crucial role in controlling male sex differentiation and maintaining male secondary sexual characteristics in decapods. In this study, we investigated the mRNA and microRNA expression profiles of male Procambarus clarkii to understand the transcriptomic regulatory mechanism of IAG after the injection of an efficient siRNA (GsiRNA) designed based on IAG. The results revealed that several differentially expressed genes were enriched in reproduction-related pathways, such as the wnt signaling pathway, MAPK signaling pathway, and GnRH signaling pathway. In the testis (Te), the injection of GsiRNA led to the up-regulation of many ovary-related genes and down-regulation of testis-related genes. Moreover, the brain (Br) and abdominal nerve cord (AN) appeared to be involved in the regulation of IAG, with numerous differentially expressed genes found in Br and AN. Notably, the expression of five neuropeptide genes, Crustacean hyperglycemic hormone, pigment-dispersing hormone, red pigment concentrating hormone precursor, corazonin, and gonadotropin-releasing hormone II receptor isoform X1 in Br/AN, was significantly changed. Additionally, three ovary-related miRNAs (miR-263a, miR-263b, miR-133) highly expressed in Te/AG showed significant up-regulation after GsiRNA injection. Furthermore, the long-term interference of GsiRNA was found to inhibit the development of male external sexual characteristics during the juvenile stage and delay it during the adult stage. This research provides valuable insights into the molecular regulatory mechanism and function of IAG in P. clarkii.


Asunto(s)
MicroARNs , Tejido Nervioso , Animales , Femenino , Masculino , Hormonas Gonadales/genética , Hormonas Gonadales/metabolismo , Astacoidea/genética , Astacoidea/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Andrógenos/metabolismo , Tejido Nervioso/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
6.
Int J Mol Sci ; 24(11)2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37298172

RESUMEN

Statins have been shown to cause diverse male reproductive function impairment, and in some cases, orchialgia. Therefore, the current study investigated the possible mechanisms through which statins may alter male reproductive parameters. Thirty adult male Wistar rats (200-250 g) were divided into three groups. The animals were orally administered rosuvastatin (50 mg/kg), simvastatin (50 mg/kg), or 0.5% carboxy methyl cellulose (control), for a 30-day period. Spermatozoa were retrieved from the caudal epididymis for sperm analysis. The testis was used for all biochemical assays and immunofluorescent localization of biomarkers of interest. Rosuvastatin-treated animals presented with a significant decrease in sperm concentration when compared to both the control and simvastatin groups (p < 0.005). While no significant difference was observed between the simvastatin and the control group. The Sertoli cells, Leydig cells and whole testicular tissue homogenate expressed transcripts of solute carrier organic anion transporters (SLCO1B1 and SLCO1B3). There was a significant decrease in the testicular protein expression of the luteinizing hormone receptor, follicle stimulating hormone receptor, and transient receptor potential vanilloid 1 in the rosuvastatin and simvastatin-treated animals compared to the control. The expression of SLCO1B1, SLCO1B2, and SLCO1B3 in the different spermatogenic cells portray that un-bio transformed statin can be transported into the testicular microenvironment, which can subsequently alter the regulation of the gonadal hormone receptors, dysregulate pain-inflammatory biomarkers, and consequently impair sperm concentration.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Ratas , Animales , Masculino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Rosuvastatina Cálcica/farmacología , Ratas Wistar , Semen , Testículo/metabolismo , Espermatozoides/metabolismo , Hormona Folículo Estimulante/metabolismo , Simvastatina/farmacología , Simvastatina/metabolismo , Hormonas Gonadales/metabolismo , Testosterona/metabolismo
7.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293143

RESUMEN

For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.


Asunto(s)
Diferenciación Sexual , Cromosoma Y , Femenino , Masculino , Animales , Diferenciación Sexual/genética , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Caracteres Sexuales , Hormonas Gonadales/metabolismo , Encéfalo/metabolismo , Epigénesis Genética , Cromosoma X
8.
Nat Rev Endocrinol ; 18(9): 574-583, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35705742

RESUMEN

Understanding sex differences in physiology and disease requires the identification of the molecular agents that cause phenotypic sex differences. Two groups of such agents are genes located on the sex chromosomes, and gonadal hormones. The former have coherent linkage to chromosomes that form differently in the two sexes under the influence of genomic forces that are not related to reproductive function, whereas the latter have a direct or indirect relationship to reproduction. Evidence published in the past 5 years supports the identification of several agents of sexual differentiation encoded by the X chromosome in mice, including Kdm5c, Kdm6a, Ogt and Xist. These X chromosome agents have wide pleiotropic effects, potentially influencing sex differences in many different tissues, a characteristic shared with the gonadal hormones. The identification of X chromosome agents of sexual differentiation will facilitate understanding of complex intersecting gene pathways underlying sex differences in disease.


Asunto(s)
Diferenciación Sexual , Cromosoma X , Animales , Femenino , Hormonas Gonadales/metabolismo , Humanos , Masculino , Ratones , Caracteres Sexuales , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Diferenciación Sexual/genética , Cromosoma X/metabolismo
9.
Circ Res ; 130(12): 1747-1762, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35679362

RESUMEN

Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.


Asunto(s)
Enfermedades Cardiovasculares , Caracteres Sexuales , Adiposidad , Animales , Enfermedades Cardiovasculares/genética , Femenino , Hormonas Gonadales/metabolismo , Humanos , Masculino , Ratones , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo
10.
J Psychiatr Res ; 152: 128-138, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35724494

RESUMEN

Gonadal hormone deficiency is associated with the development of depression, but what mediates this association is unclear. To test the possibility that it reflects neuroimmune and neuroinflammatory processes, we analyzed how gonadal hormone deficiency and replacement affect microglial activation and inflammatory response during the development of depressive symptomatology in gonadectomized male mice. Testosterone level and the ratio of testosterone to estradiol in the serum and brain tissue of mice exposed to 3-35 days of chronic unpredictable stress were much lower than in control animals. Gonadal hormone sustained deficiency in gonadectomized mice and subsequent led to acute inflammation at day 7 following castration. Activating microglia in mice exposed to 7 days of castration subsequently suppressed the proliferation of microglia, such that their numbers in hippocampus and cortex were lower than the numbers in sham-operated mice after 30 days of castration. Here, we showed that gonadal hormone deficiency induces Traf6-mediated microglia activation, a type of inflammatory mediator. Microglia treated in this way for long time showed down-regulation of activation markers, abnormal morphology and depressive-like behaviors. Restoration and maintenance of a fixed ratio of testosterone to estradiol significantly suppressed microglial activation, neuronal necroptosis, dramatically inducing hippocampal neurogenesis and reducing depressive behaviors via the suppression of Traf6/TAK1 pathway. These findings suggest that activated or immunoreactive microglia contribute to gonadal hormone deficiency-induced depression, as well as testosterone and estradiol exert synergistic anti-depressant effects via suppressing microglial activaton in gonadectomized male mice, possibly through Traf6 signaling.


Asunto(s)
Microglía , Factor 6 Asociado a Receptor de TNF , Animales , Depresión/etiología , Depresión/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Hormonas Gonadales/metabolismo , Hormonas Gonadales/farmacología , Hipocampo , Masculino , Ratones , Microglía/metabolismo , Receptores de Esteroides , Receptores de Hormona Tiroidea , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/farmacología , Testosterona
11.
Trop Anim Health Prod ; 54(3): 161, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35428914

RESUMEN

Previous investigations have revealed that lipopolysaccharide (LPS) endotoxin from certain Gram-negative bacteria could adversely affect the reproductive system of female animals. However, it is unknown whether LPS endotoxin of Mannheimia haemolytica serotype A2, the principal causative bacteria that cause pneumonic mannheimiosis in small ruminants, may also induce similar insidious effects. Therefore, this study aimed to investigate the effects of M. haemolytica serotype A2 and its LPS endotoxin on the responses of female gonadal hormones (progesterone and oestrogen), pro-inflammatory cytokines (interleukin-1ß, interleukin-6), acute-phase proteins (haptoglobin and serum amyloid A) and cellular changes via histopathology study of female reproductive organs of the treatment does. Twelve clinically healthy, non-pregnant, crossbred does were randomly allocated into three equal groups. Group 1 was administered intranasally with 2 ml of sterile phosphate-buffered saline (PBS) and served as a negative control group. Group 2 was challenged intranasally with 2 ml of bacterial inoculum containing 109 colony-forming units (CFU)/ml of M. haemolytica serotype A2, while Group 3 was challenged intravenously with 2 ml of LPS endotoxin extracted from 109 CFU/ml of M. haemolytica serotype A2. Following that, blood samples were collected serially at pre-determined intervals for serological analyses. All does were euthanised 60 days post-challenges, and tissue samples from the ovaries, oviducts, uterine horns, uterine body, cervix and vagina were collected for histopathological study. The serological result revealed a significant increase (p < 0.05) in the mean concentrations of progesterone, oestrogen, interleukin-1ß, interleukin-6, haptoglobin and serum amyloid A for both challenged groups. Histopathologically, all reproductive organs (except the cervix and vagina) from both challenged groups displayed significant cellular alterations (p < 0.05) characterised by haemorrhage and congestion, necrosis and degeneration, inflammatory cell infiltration and oedema. This study provides new information that elucidates the potential role of pneumonic mannheimiosis in the pathogenesis of female infertility amongst small ruminants.


Asunto(s)
Mannheimia haemolytica , Proteínas de Fase Aguda/metabolismo , Animales , Citocinas/metabolismo , Perros , Endotoxinas/metabolismo , Endotoxinas/toxicidad , Estrógenos , Femenino , Genitales , Hormonas Gonadales/metabolismo , Haptoglobinas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Mannheimia haemolytica/fisiología , Progesterona/metabolismo , Serogrupo , Proteína Amiloide A Sérica/metabolismo
12.
Genome Res ; 32(5): 807-824, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35396276

RESUMEN

Sex differences in physiology and disease in mammals result from the effects of three classes of factors that are inherently unequal in males and females: reversible (activational) effects of gonadal hormones, permanent (organizational) effects of gonadal hormones, and cell-autonomous effects of sex chromosomes, as well as genes driven by these classes of factors. Often, these factors act together to cause sex differences in specific phenotypes, but the relative contribution of each and the interactions among them remain unclear. Here, we used the four core genotypes (FCG) mouse model with or without hormone replacement to distinguish the effects of each class of sex-biasing factors on transcriptome regulation in liver and adipose tissues. We found that the activational hormone levels have the strongest influence on gene expression, followed by the organizational gonadal sex effect, and last, sex chromosomal effect, along with interactions among the three factors. Tissue specificity was prominent, with a major impact of estradiol on adipose tissue gene regulation and of testosterone on the liver transcriptome. The networks affected by the three sex-biasing factors include development, immunity and metabolism, and tissue-specific regulators were identified for these networks. Furthermore, the genes affected by individual sex-biasing factors and interactions among factors are associated with human disease traits such as coronary artery disease, diabetes, and inflammatory bowel disease. Our study offers a tissue-specific account of the individual and interactive contributions of major sex-biasing factors to gene regulation that have broad impact on systemic metabolic, endocrine, and immune functions.


Asunto(s)
Caracteres Sexuales , Cromosomas Sexuales , Animales , Femenino , Hormonas Gonadales/metabolismo , Hormonas Gonadales/farmacología , Hormonas Esteroides Gonadales/metabolismo , Gónadas/metabolismo , Masculino , Mamíferos/genética , Ratones , Cromosomas Sexuales/genética
13.
J Integr Neurosci ; 20(3): 651-657, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34645098

RESUMEN

Hypobaric hypoxia is a stressful condition known to decrease fertility both in humans and animals. However, the mechanism by which the hypothalamus-pituitary-gonad axis is altered remains unknown. The aim of the present study was to analyze the effects of chronic intermittent and continuous exposure to hypoxia on hypothalamic-pituitary-gonadal axis regulation in male rats. Thirty adult male Wistar rats were assigned to one of the following three groups: control group; chronic intermittent hypoxia: subjected to 600 mbar for 18 h/d five days a week; and chronic continuous hypoxia: subjected to 600 mbar for 23.5 hours/day seven days a week, for 30 days. Plasma luteinizing hormone and testosterone concentration, hypothalamic GnRh, Kiss1 and Rfrp3 mRNA levels and PGE2 content were determined. Levels of Rfrp3, a negative regulator of GnRH and LH release, were higher in intermittently exposed animals than in controls. Levels of Kiss1, a neuropeptide that stimulates the release of GnRH only increased in animals exposed to continuous hypoxia. Plasma luteinizing hormone and testosterone concentrations and body weight were lower in rats subjected to intermittent hypoxia as compared to the remaining groups. GnRh mRNA levels as well as PGE2 content remained unchanged in all groups. Taken together, results suggest that besides the well documented direct effects of hypoxia on the testes, infertility observed in male rats exposed to hypoxia may also be due to overexpression of negative regulators of GnRH and luteinizing hormone release. Intermittent, rather than continuous, to hypoxia exposure would seem to be more detrimental to fertility.


Asunto(s)
Fertilidad/fisiología , Hormonas Gonadales/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipoxia/metabolismo , Neuropéptidos/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Wistar
14.
J Clin Pharm Ther ; 46(6): 1764-1775, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34490645

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Metformin was found to reduce elevated gonadotropin levels. The aim of the present study was to determine whether metformin modulates the impact of discontinuation of testosterone therapy on hypothalamic-pituitary-gonadal axis activity and sexual function in men with low testosterone levels. METHODS: The study included 28 men with late-onset hypogonadism (defined according to the criteria of the European Male Aging Study group) receiving testosterone undecanoate (120 mg in three equal doses), 12 of whom had been treated with oral metformin (1.7-3 g daily). Both testosterone and metformin had been administered for at least six months before enrolment. In all patients, testosterone replacement required to be discontinued. The control group included 16 testosterone- and metformin-treated men with late-onset hypogonadism who during the entire study period continued their treatment. Glucose homeostasis markers, as well as plasma levels of insulin, gonadotropins, testosterone, calculated bioavailable testosterone, dehydroepiandrosterone-sulphate, oestradiol, thyrotropin, free thyroxine, prolactin, insulin-growth factor-1 and cortisol were measured at the beginning of the study and four months later. Moreover, at the beginning and the end of the study, all enrolled patients completed a questionnaire assessing their sexual functioning (IIEF-15). RESULTS AND DISCUSSION: Discontinuation of testosterone therapy resulted in a decrease in total testosterone and bioavailable testosterone (by 42% and 45% in metformin-treated patients, and by 52% and 54% in metformin-naïve patients), as well as impaired all aspects of male sexual function. Changes in bioavailable testosterone, as well as in erectile function, orgasmic function and sexual desire were less pronounced if subjects received metformin. Only in metformin-naïve men, follow-up FSH and LH levels were higher than at baseline (by 75% and 62%). Moreover, discontinuation of testosterone therapy in metformin-naïve men increased glycated haemoglobin, as well as worsened insulin sensitivity. There were no differences between baseline and follow-up levels of the remaining hormones. In metformin-naïve subjects, the increase in gonadotropin levels correlated with the changes in testosterone levels and insulin sensitivity. No effect on glucose homeostasis markers, hormone levels and sexual functioning was observed in the control group. WHAT IS NEW AND CONCLUSION: The obtained results suggest that metformin treatment mitigates the unfavourable effect of discontinuation of testosterone treatment on hypothalamic-pituitary-testicular axis activity and sexual function in men with late-onset hypogonadism.


Asunto(s)
Hormonas Gonadales/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Metformina/farmacología , Disfunciones Sexuales Fisiológicas/fisiopatología , Adulto , Anciano , Glucemia/efectos de los fármacos , Gonadotropinas/sangre , Terapia de Reemplazo de Hormonas/métodos , Humanos , Hipogonadismo/tratamiento farmacológico , Insulina/sangre , Masculino , Persona de Mediana Edad , Proyectos Piloto , Testosterona/uso terapéutico
15.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34061778

RESUMEN

The main mechanisms underlying sexually dimorphic outcomes in neonatal lung injury are unknown. We tested the hypothesis that hormone- or sex chromosome-mediated mechanisms interact with hyperoxia exposure to impact injury and repair in the neonatal lung. To distinguish sex differences caused by gonadal hormones versus sex chromosome complement (XX versus XY), we used the Four Core Genotypes (FCG) mice and exposed them to hyperoxia (95% FiO2, P1-P4: saccular stage) or room air. This model generates XX and XY mice that each have either testes (with Sry, XXM, or XYM) or ovaries (without Sry, XXF, or XYF). Lung alveolarization and vascular development were more severely impacted in XYM and XYF compared with XXF and XXM mice. Cell cycle-related pathways were enriched in the gonadal or chromosomal females, while muscle-related pathways were enriched in the gonadal males, and immune-response-related pathways were enriched in chromosomal males. Female gene signatures showed a negative correlation with human patients who developed bronchopulmonary dysplasia (BPD) or needed oxygen therapy at 28 days. These results demonstrate that chromosomal sex - and not gonadal sex - impacted the response to neonatal hyperoxia exposure. The female sex chromosomal complement was protective and could mediate sex-specific differences in the neonatal lung injury.


Asunto(s)
Displasia Broncopulmonar , Hormonas Gonadales/metabolismo , Hiperoxia , Lesión Pulmonar , Terapia por Inhalación de Oxígeno , Cromosomas Sexuales , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/terapia , Femenino , Humanos , Hiperoxia/etiología , Hiperoxia/genética , Hiperoxia/metabolismo , Recién Nacido , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Masculino , Ratones , Ovario/metabolismo , Terapia por Inhalación de Oxígeno/efectos adversos , Terapia por Inhalación de Oxígeno/métodos , Factores Protectores , Factores de Riesgo , Caracteres Sexuales , Testículo/metabolismo
16.
Int J Mol Sci ; 22(6)2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33808655

RESUMEN

Chronic stress is encountered in our everyday life and is thought to contribute to a number of diseases. Many of these stress-related disorders display a sex bias. Because glucocorticoid hormones are the main biological mediator of chronic stress, researchers have been interested in understanding the sexual dimorphism in glucocorticoid stress response to better explain the sex bias in stress-related diseases. Although not yet demonstrated for glucocorticoid regulation, sex chromosomes do influence sex-specific biology as soon as conception. Then a transient rise in testosterone start to shape the male brain during the prenatal period differently to the female brain. These organizational effects are completed just before puberty. The cerebral regions implicated in glucocorticoid regulation at rest and after stress are thereby impacted in a sex-specific manner. After puberty, the high levels of all gonadal hormones will interact with glucocorticoid hormones in specific crosstalk through their respective nuclear receptors. In addition, stress occurring early in life, in particular during the prenatal period and in adolescence will prime in the long-term glucocorticoid stress response through epigenetic mechanisms, again in a sex-specific manner. Altogether, various molecular mechanisms explain sex-specific glucocorticoid stress responses that do not exclude important gender effects in humans.


Asunto(s)
Glucocorticoides/metabolismo , Caracteres Sexuales , Estrés Fisiológico , Estrés Psicológico , Adolescente , Animales , Niño , Desarrollo Infantil , Desarrollo Embrionario/genética , Estudios de Asociación Genética , Hormonas Gonadales/metabolismo , Humanos , Hidrocortisona/metabolismo , Pubertad/genética , Pubertad/metabolismo , Factores Sexuales , Esteroides/metabolismo , Estrés Fisiológico/genética , Estrés Psicológico/genética
17.
Mech Ageing Dev ; 196: 111473, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33766745

RESUMEN

Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.


Asunto(s)
Envejecimiento/fisiología , Encéfalo , Neuroglía , Neuroprotección/fisiología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Encéfalo/fisiopatología , Senescencia Celular/fisiología , Hormonas Gonadales/metabolismo , Humanos , Neuroglía/patología , Neuroglía/fisiología , Caracteres Sexuales
18.
PLoS One ; 16(2): e0247311, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606835

RESUMEN

The serotonin transporter (5-HTT) is a key molecule of serotoninergic neurotransmission and target of many anxiolytics and antidepressants. In humans, 5-HTT gene variants resulting in lower expression levels are associated with behavioral traits of anxiety. Furthermore, functional magnetic resonance imaging (fMRI) studies reported increased cerebral blood flow (CBF) during resting state (RS) and amygdala hyperreactivity. 5-HTT deficient mice as an established animal model for anxiety disorders seem to be well suited for investigating amygdala (re-)activity in an fMRI study. We investigated wildtype (5-HTT+/+), heterozygous (5-HTT+/-), and homozygous 5-HTT-knockout mice (5-HTT-/-) of both sexes in an ultra-high-field 17.6 Tesla magnetic resonance scanner. CBF was measured with continuous arterial spin labeling during RS, stimulation state (SS; with odor of rats as aversive stimulus), and post-stimulation state (PS). Subsequently, post mortem c-Fos immunohistochemistry elucidated neural activation on cellular level. The results showed that in reaction to the aversive odor CBF in total brain and amygdala of all mice significantly increased. In male 5-HTT+/+ mice amygdala RS CBF levels were found to be significantly lower than in 5-HTT+/- mice. From RS to SS 5-HTT+/+ amygdala perfusion significantly increased compared to both 5-HTT+/- and 5-HTT-/- mice. Perfusion level changes of male mice correlated with the density of c-Fos-immunoreactive cells in the amygdaloid nuclei. In female mice the perfusion was not modulated by the 5-Htt-genotype, but by estrous cycle stages. We conclude that amygdala reactivity is modulated by the 5-Htt genotype in males. In females, gonadal hormones have an impact which might have obscured genotype effects. Furthermore, our results demonstrate experimental support for the tonic model of 5-HTTLPR function.


Asunto(s)
Amígdala del Cerebelo/irrigación sanguínea , Ansiedad/diagnóstico por imagen , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Femenino , Hormonas Gonadales/metabolismo , Homocigoto , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Caracteres Sexuales
19.
J Environ Sci Health B ; 56(1): 30-40, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33052060

RESUMEN

Clarias gariepinus juveniles were exposed to environmentally relevant concentrations of 0 (control), 2.5, 25, 250 and 500 µg L-1 atrazine in a quality-controlled 28-day laboratory procedure. Findings revealed a significant decrease in the levels of follicle-stimulating hormone, luteinizing hormone and prolactin relative to control (p < 0.05). Atrazine reduced the levels of testosterone while increasing the concentration of progesterone. Histologically, the control and treatments presented three stages of oocyte maturation: the chromatin nucleolar oocyte stage, early perinucleolar oocyte stage and the vitellogenic oocyte stage. However, in the ovaries of the treatment group with the lowest treatment concentration (2.5 µg L-1), atretic oocytes with broken membranes invaded many of the dead ova and empty spaces. In other treatments (25, 250 and 500 µg L-1), interfollicular spaces, vacuolation in oocyte formation, and dissolution of oocyte walls were observed. Disruption of the yolk vesicle and clumping of the cytoplasm in maturing oocytes was observed only at the highest atrazine concentration (500 µg L-1). Gross alterations in ovarian histoarchitecture and reproductive hormone levels observed in this study showed interference with oogenesis which may result in reduced egg viability and fecundity in fish with ecological implications in water bodies exposed to atrazine even at reduced concentrations.


Asunto(s)
Atrazina/toxicidad , Bagres/fisiología , Herbicidas/toxicidad , Oogénesis/efectos de los fármacos , Ovario/efectos de los fármacos , Animales , Femenino , Hormonas Gonadales/metabolismo , Hormona Luteinizante/metabolismo , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Ovario/patología
20.
Horm Behav ; 127: 104874, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33191199

RESUMEN

Current food supply is a major driver of timing of breeding in income-breeding animals, likely because increased net energy balance directly increases reproductive hormones and advances breeding. In capital breeders, increased net energy balance increases energy reserves, which eventually leads to improved reproductive readiness and earlier breeding. To test the hypothesis that phenology of income-breeding birds is independent of energy reserves, we conducted an experiment on food-supplemented ("fed") and control female black-legged kittiwakes (Rissa tridactyla). We temporarily increased energy costs (via weight handicap) in a 2 × 2 design (fed/unfed; handicapped/unhandicapped) during the pre-laying period and observed movement via GPS-accelerometry. We measured body mass, baseline hormones (corticosterone; luteinising hormone) before and after handicap manipulation, and conducted a gonadotropin-releasing hormone challenge. Females from all treatment groups foraged in similar areas, implying that individuals could adjust time spent foraging, but had low flexibility to adjust foraging distance. Consistent with the idea that income breeders do not accumulate reserves in response to increased food supply, fed birds remained within an energy ceiling by reducing time foraging instead of increasing energy reserves. Moreover, body mass remained constant until the onset of follicle development 20 days prior to laying regardless of feeding or handicap, implying that females were using a 'lean and fit' approach to body mass rather than accumulating lipid reserves for breeding. Increased food supply advanced endocrine and laying phenology and altered interactions between the hypothalamic-pituitary-adrenal axis and the hypothalamic-pituitary-gonadal axis, but higher energy costs (handicap) had little effect. Consistent with our hypothesis, increased food supply (but not net energy balance) advanced endocrine and laying phenology in income-breeding birds without any impact on energy reserves.


Asunto(s)
Charadriiformes/fisiología , Abastecimiento de Alimentos , Hormonas Gonadales/metabolismo , Conducta Sexual Animal/fisiología , Animales , Aves/fisiología , Composición Corporal , Corticosterona/metabolismo , Metabolismo Energético/fisiología , Conducta Exploratoria/fisiología , Conducta Alimentaria/fisiología , Femenino , Alimentos , Hormona Liberadora de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , Reproducción/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...