Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.441
Filtrar
1.
Cell Genom ; 4(5): 100542, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38663407

RESUMEN

Cis-regulatory elements control transcription levels, temporal dynamics, and cell-cell variation or transcriptional noise. However, the combination of regulatory features that control these different attributes is not fully understood. Here, we used single-cell RNA-seq during an estrogen treatment time course and machine learning to identify predictors of expression timing and noise. We found that genes with multiple active enhancers exhibit faster temporal responses. We verified this finding by showing that manipulation of enhancer activity changes the temporal response of estrogen target genes. Analysis of transcriptional noise uncovered a relationship between promoter and enhancer activity, with active promoters associated with low noise and active enhancers linked to high noise. Finally, we observed that co-expression across single cells is an emergent property associated with chromatin looping, timing, and noise. Overall, our results indicate a fundamental tradeoff between a gene's ability to quickly respond to incoming signals and maintain low variation across cells.


Asunto(s)
Elementos de Facilitación Genéticos , Estrógenos , Regiones Promotoras Genéticas , Transcripción Genética , Humanos , Cromatina/genética , Estrógenos/fisiología , Regulación de la Expresión Génica , Aprendizaje Automático , Análisis de la Célula Individual
2.
Immunity ; 56(11): 2472-2491, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37967530

RESUMEN

Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.


Asunto(s)
Hormonas Esteroides Gonadales , Neoplasias , Embarazo , Femenino , Masculino , Humanos , Estrógenos/farmacología , Estrógenos/fisiología , Progestinas , Andrógenos/farmacología , Esteroides , Inmunidad , Caracteres Sexuales
3.
Science ; 382(6666): 33-34, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37797022

RESUMEN

Progesterone and estrogen exert separate effects on the brains of pregnant mice.


Asunto(s)
Encéfalo , Estrógenos , Conducta Materna , Responsabilidad Parental , Progesterona , Animales , Femenino , Ratones , Embarazo , Estrógenos/fisiología , Progesterona/fisiología
4.
PLoS One ; 18(7): e0278080, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37471329

RESUMEN

The development of human brain is shaped by both genetic and environmental factors. Sex differences in cognitive function have been found in humans as a result of sexual dimorphism in neural information transmission. Numerous studies have reported the positive effects of education on cognitive functions. However, little work has investigated the effect of education on attenuating cognitive sex differences and the neural mechanisms behind it based on healthy population. In this study, the Wisconsin Card Sorting Test (WCST) was employed to examine sex differences in cognitive function in 135 Thai healthy subjects, and label-free quantitative proteomic method and bioinformatic analysis were used to study sex-specific neurotransmission-related protein expression profiles. The results showed sex differences in two WCST sub-scores: percentage of Total corrects and Total errors in the primary education group (Bayes factor>100) with males performed better, while such differences eliminated in secondary and tertiary education levels. Moreover, 11 differentially expressed proteins (DEPs) between men and women (FDR<0.1) were presented in both education groups, with majority of them upregulated in females. Half of those DEPs interacted directly with nAChR3, whereas the other DEPs were indirectly connected to the cholinergic pathways through interaction with estrogen. These findings provided a preliminary indication that a cholinergic-estrogen interaction relates to, and might underpin, the effect of education on attenuating cognitive sex differences in a Thai healthy population.


Asunto(s)
Encéfalo , Neuronas Colinérgicas , Cognición , Escolaridad , Estrógenos , Caracteres Sexuales , Femenino , Humanos , Masculino , Teorema de Bayes , Cognición/fisiología , Pruebas Neuropsicológicas , Proteómica , Pueblos del Sudeste Asiático , Factores Sexuales , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Voluntarios Sanos , Estrógenos/fisiología , Neuronas Colinérgicas/fisiología
5.
Med Oncol ; 40(6): 171, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37162589

RESUMEN

Hormones and its regulation plays vital role in causing breast, prostate, ovarian and endometrial cancers collectively known as hormone-sensitive cancers. This review discusses the various functions of the sex hormones and the biological pathways involved in causing hormone-associated cancer under differential regulation. We have also attempted to explore the biomarkers associated with the cancers and the current therapeutic availability to treat such cancers. Among various sex hormones such as estrogen, progesterone and androgen, estrogen the female sex hormone and its receptor had a major contribution in causing cancer and hence are considered a predominant target in treating the associated cancers. Other hormones and receptors such a androgen, progesterone, and their respective receptors were also reported to have a significant correlation in causing cancers. Apart from these receptors certain enzymes that act as precursors or as promoters are also targeted for treatment strategies. The drugs commonly used belong to the selective drug classes such as selective estrogen receptor modulators and selective progesterone receptor modulators. In the case of androgen regulation androgen deprivation therapies are practiced. It is also suggested that the use of natural substances to treat cancer could prevent resistance and reduce side effects. Identification of significant targets and the discovery of many efficient drugs shall be possible in the future with better understanding of hormone regulation and its influence on cancer causative mechanisms.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Próstata , Masculino , Humanos , Femenino , Progesterona/uso terapéutico , Andrógenos , Antagonistas de Andrógenos/uso terapéutico , Biomarcadores de Tumor , Neoplasias de la Próstata/tratamiento farmacológico , Hormonas Esteroides Gonadales/uso terapéutico , Estrógenos/fisiología , Estrógenos/uso terapéutico , Receptores de Progesterona , Receptores Androgénicos/fisiología , Neoplasias de la Mama/tratamiento farmacológico
6.
Bone ; 172: 116781, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37100360

RESUMEN

External mechanostimulation applied by whole-body low-magnitude high-frequency vibration (LMHFV) was demonstrated to cause no or negative effects on fracture healing in estrogen-competent rodents, while in ovariectomized (OVX), estrogen-deficient rodents bone formation after fracture was improved. Using mice with an osteoblast-specific deletion of the estrogen receptor α (ERα), we demonstrated that ERα signaling in osteoblasts is required for both the anabolic and catabolic effects of LMHFV during bone fracture healing in OVX and non-OVX mice, respectively. Because the vibration effects mediated by ERα were strictly dependent on the estrogen status, we hypothesized different roles of ligand-dependent and -independent ERα signaling. To investigate this assumption in the present study, we used mice with a deletion of the C-terminal activation function (AF) domain-2 of the ERα receptor, which mediated ligand-dependent ERα signaling (ERαAF-20). OVX and non-OVX ERαAF-20 animals were subjected to femur osteotomy followed by vibration treatment. We revealed that estrogen-competent mice lacking the AF-2 domain were protected from LMHFV-induced impaired bone regeneration, while the anabolic effects of vibration in OVX mice were not affected by the AF-2 knockout. RNA sequencing further showed that genes involved in Hippo/Yap1-Taz and Wnt signaling were significantly downregulated upon LMHFV in the presence of estrogen in vitro. In conclusion, we demonstrated that the AF-2 domain is crucial for the negative effects of vibration during bone fracture healing in estrogen-competent mice suggesting that the osteoanabolic effects of vibration are rather mediated by ligand-independent ERα signaling.


Asunto(s)
Receptor alfa de Estrógeno , Curación de Fractura , Ratones , Animales , Curación de Fractura/fisiología , Receptor alfa de Estrógeno/genética , Furilfuramida , Receptores de Estrógenos , Mecanotransducción Celular , Ligandos , Estrógenos/fisiología
7.
J Recept Signal Transduct Res ; 43(1): 19-26, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36883690

RESUMEN

Endometrial cancer (EC) is one of the most common gynecological carcinomas in both developed and developing countries. Majority of the gynecological malignancies are hormonally driven where estrogen signaling acts as an oncogenic signal. Estrogen's effects are mediated via classical nuclear estrogen receptors; estrogen receptor alpha and beta (ERα and ERß) and a trans-membrane G protein-coupled estrogen receptor (GPR30 and GPER). ERs and GPER through ligand binding triggers multiple downstream signaling pathways causing cell cycle regulation, cell differentiation, migration, and apoptosis in various tissues including endometrium. Although the molecular aspect of estrogen function in ER-mediated signaling is now partly understood, the same is not true for GPER-mediated signaling in endometrial malignancies. Understanding the physiological roles of ERα and GPER in EC biology therefore leads to the identification of some novel therapeutic targets. Here we review the effect of estrogen signaling through ERα-and GPER in EC, major types, and some affordable treatment approaches for endometrial tumor patients which has interesting implications in understanding uterine cancer progression.


Asunto(s)
Neoplasias Endometriales , Receptores de Estrógenos , Femenino , Humanos , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Estrógenos/fisiología , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
8.
Reprod Sci ; 30(7): 2092-2106, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36696041

RESUMEN

Ultimately, the Mongolian gerbils (Meriones unguiculatus) have acquired a relevant role in biological and biomedical experiments alongside other rodents. The use of gerbils in research has been mainly oriented to physiological and pharmacological studies, with special attention to nervous, digestive, and auditory systems as well as microbiology and parasitology. Ultimately, gerbils have also been applied for studying carcinogenesis in different organs and systems, since these animals show a natural propensity to develop spontaneous proliferative lesions, especially in steroid-responsive organs. This characteristic shed light on the reproductive aspects of this rodent model regarding morphological features in male and female individuals. This review of literature summarizes the significance of this model as an alternative to the use of inbred mice and rats in reproductive experimental research, highlighting recent findings. Gerbils have contributed to the expansion of knowledge in prostate biology in male and female individuals, providing studies related to prostatic morphogenesis and neoplasia. In the testes, spermiogenesis occurs in 15 steps, differently from other experimental models. Also, the complete maturation of the testis-epididymal complex occurs between the second and third months. Mammary gland alterations related to the estrous cycle and pregnancy were described, as well as its modulation under endogenous and exogenous estrogenic compounds. The ovaries frequently present ovarian cysts. Furthermore, this organ shows predominantly interstitial steroidogenic glands in the stroma, especially at aging. Adrenal gland shows a large size compared to other animals, presenting three distinct zones with a remarkable role in steroidogenesis.


Asunto(s)
Estrógenos , Reproducción , Embarazo , Masculino , Femenino , Ratas , Ratones , Animales , Gerbillinae/fisiología , Estrógenos/fisiología , Próstata/fisiología , Biología
9.
J Endocrinol ; 257(2)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36524723

RESUMEN

The fact that growth hormone (GH) plays an important role in health after the cessation of growth requiring replacement therapy in adult life has only been recognised in the last three decades. This has only been made possible by recombinant technology providing GH supplies required to undertake investigations in the physiology of GH action and the benefits of replacement therapy in patients identified by rigorously validated diagnostic tests for GH deficiency (GHD). Human studies have revealed important regulatory roles in substrate metabolism, sodium homeostasis, body composition, and physical function. GH-induced anabolism is achieved by stimulating amino acid incorporation into protein while reducing oxidative loss simultaneously enhancing lipid utilisation by stimulating fatty acid oxidation and reducing lipid storage. Sodium and fluid retention are enhanced by activating the renin-angiotensin system and distal renal tubular reabsorption. GH stimulates the aerobic and anaerobic energy systems that underpin muscle and cardiovascular function. These pleiotropic actions explain the clinical picture of increased adiposity, reduced lean mass, and impaired physical and psychological function in the GHD adult, all of which are reversed when GH is replaced. Women require a greater replacement dose of GH than men. This is because androgens enhance while oestrogens attenuate GH action. The oestrogen effect is route-dependent, occurring with oral delivery blunting the liver-mediated actions of GH by directly inhibiting GH receptor signalling, global experience spanning over 30 years has attested to the safety, efficacy, and benefits of replacement therapy for adults with GHD.


Asunto(s)
Hormona del Crecimiento , Hormona de Crecimiento Humana , Adulto , Femenino , Humanos , Masculino , Estrógenos/fisiología , Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/fisiología , Terapia de Reemplazo de Hormonas/efectos adversos , Hormona de Crecimiento Humana/uso terapéutico , Hormona de Crecimiento Humana/farmacología , Lípidos , Sodio
10.
Adv Exp Med Biol ; 1390: 21-39, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36107311

RESUMEN

The female reproductive system which consists of the ovaries, uterus (myometrium, endometrium), Fallopian tubes, cervix and vagina is exquisitely sensitive to the actions of steroid hormones. The ovaries play a key role in the synthesis of bioactive steroids (oestrogens, androgens, progestins) that act both within the tissue (intracrine/paracrine) as well as on other reproductive organs following release into the blood stream (endocrine action). Sex steroid receptors encoded by the oestrogen (ESR1, ESR2), progesterone (PR) and androgen (AR) receptor genes, which are members of the superfamily of ligand activated transcription factors are widely expressed within these tissues. These receptors play critical role(s) in regulation of cell proliferation, ovulation, endometrial receptivity, myometrial cell function and inflammatory cell infiltration. Our understanding of their importance has been informed by studies on human tissues and cells, which have employed immunohistochemistry as well as a wide range of molecular and genetic methods to identify which processes are dependent steroid ligand activation. The development of mice with targeted deletions of each of these receptors has provided complementary data that has extended our appreciation of cell-cell interactions in the fine tuning of reproductive tissue function. This large body of work has formed the basis of new and improved therapeutics to treat conditions such as infertility.


Asunto(s)
Andrógenos , Receptores de Esteroides , Animales , Estrógenos/fisiología , Femenino , Genitales Femeninos/metabolismo , Humanos , Ligandos , Ratones , Progesterona/metabolismo , Progestinas , Receptores de Esteroides/fisiología
11.
Front Endocrinol (Lausanne) ; 13: 967711, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36072933

RESUMEN

Bone age (BA) is a clinical marker of bone maturation which indicates the developmental stage of endochondral ossification at the epiphysis and the growth plate. Hormones that promote the endochondral ossification process include growth hormone, insulin-like growth factor-1, thyroid hormone, estrogens, and androgens. In particular, estrogens are essential for growth plate fusion and closure in both sexes. Bone maturation in female children is more advanced than in male children of all ages. The promotion of bone maturation seen in females before the onset of puberty is thought to be an effect of estrogen because estrogen levels are higher in females than in males before puberty. Sex hormones are essential for bone maturation during puberty. Since females have their pubertal onset about two years earlier than males, bone maturation in females is more advanced than in males during puberty. In the present study, we aimed to review the factors affecting prepubertal and pubertal BA progression, BA progression in children with hypogonadism, and bone maturation and deformities in children with Turner syndrome.


Asunto(s)
Desarrollo Óseo , Hormona de Crecimiento Humana , Pubertad , Niño , Estrógenos/fisiología , Femenino , Hormona de Crecimiento Humana/fisiología , Humanos , Masculino
12.
Hypertens Res ; 45(11): 1743-1753, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35999282

RESUMEN

Previous studies have shown that female offspring are resistant to fetal high-fat diet (HFD)-induced programming of heightened vascular contraction; however, the underlying mechanisms remain unclear. The present study tested the hypothesis that estrogen plays a key role in protecting females from fetal programming of increased vascular contraction induced by maternal HFD exposure. Pregnant rats were fed a normal diet (ND) or HFD (60% kcal from fat). Ovariectomy (OVX) and 17ß-estradiol (E2) replacement were performed on 8-week-old female offspring. Aortas were isolated from adult female offspring. Maternal HFD exposure increased angiotensin II (Ang II)-induced contractions of the aorta in adult OVX offspring, which was abrogated by E2 replacement. The AT1 receptor (AT1R) antagonist losartan (10 µM), but not the AT2 receptor (AT2R) antagonist PD123319 (10 µM), completely blocked Ang II-induced contractions in both ND and HFD offspring. In addition, HFD exposure caused a decrease in endothelium-dependent relaxations induced by acetylcholine (ACh) in adult OVX but not OVX-E2 offspring. However, it had no effect on sodium nitroprusside (SNP)-induced endothelium-independent aorta relaxation in any of the six groups. Maternal HFD feeding increased AT1R, but not AT2R, leading to an increased AT1R/AT2R ratio in HFD-exposed OVX offspring, associated with selective decreases in DNA methylation at the AT1aR promoter, which was ameliorated by E2 replacement. Our results indicated that estrogen play a key role in sex differences of maternal HFD-induced vascular dysfunction and development of hypertensive phenotype in adulthood by differently regulating vascular AT1R and AT2R gene expression through a DNA methylation mechanism.


Asunto(s)
Dieta Alta en Grasa , Estrógenos , Hipertensión , Animales , Femenino , Embarazo , Ratas , Angiotensina II/farmacología , Dieta Alta en Grasa/efectos adversos , Estrógenos/fisiología , Losartán , Fenómenos Fisiologicos Nutricionales Maternos
13.
Drug Metab Pharmacokinet ; 46: 100460, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35820204

RESUMEN

Gender is a crucial factor determining susceptibility to drug-induced liver injury (DILI) in humans and experimental animals. However, no general concept of sex differences in DILI has been established, as metabolic events specific to one DILI model are difficult to apply to other DILI models. Herein, we examined sex differences in carbon tetrachloride (CCl4)-induced hepatotoxicity, a widely employed DILI model. Male and female CD-1 mice were intraperitoneally administered CCl4. Additionally, some male mice were administered genistein or another isoflavone to evaluate the effects of exogenous estrogens. Dose-dependent alanine aminotransferase leakage was observed at a CCl4 range of 0.5-10 mmol/kg, with male-dominant sex differences mainly observed at lower doses. No sex differences in hepatic glutathione levels or thiobarbituric acid-reactive substance formation were detected. CCl4 induced hepatic inflammatory genes, interleukin (IL)-6 and tumor necrosis factor (TNF)-α, predominantly in female mice, which might be involved in DILI resistance, observed in female mice. Treatment of male mice with phytoestrogens, especially genistein, attenuated CCl4-induced hepatotoxicity. Moreover, genistein inhibited IL-6 and TNF-α expression, suggesting possible hepatoprotection via immunosuppression. In conclusion, female mice are resistant to CCl4-induced hepatotoxicity, and male mice were afforded protection by genistein, probably via mechanisms based on anti-estrogenic, antioxidant and/or anti-inflammatory effects.


Asunto(s)
Antiinflamatorios , Antioxidantes , Enfermedad Hepática Inducida por Sustancias y Drogas , Estrógenos , Isoflavonas , Animales , Femenino , Masculino , Ratones , Alanina Transaminasa/metabolismo , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Antioxidantes/fisiología , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Estrógenos/farmacología , Estrógenos/fisiología , Genisteína/farmacología , Glutatión/metabolismo , Interleucina-6 , Fitoestrógenos , Factor de Necrosis Tumoral alfa/metabolismo
14.
Front Endocrinol (Lausanne) ; 13: 906381, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35712256

RESUMEN

Anti-Müllerian hormone (AMH) is a distinctive biomarker of the immature Sertoli cell. AMH expression, triggered by specific transcription factors upon fetal Sertoli cells differentiation independently of gonadotropins or sex steroids, drives Müllerian duct regression in the male, preventing the development of the uterus and Fallopian tubes. AMH continues to be highly expressed by Sertoli until the onset of puberty, when it is downregulated to low adult levels. FSH increases testicular AMH output by promoting immature Sertoli cell proliferation and individual cell expression. AMH secretion also showcases a differential regulation exerted by intratesticular levels of androgens and estrogens. In the fetus and the newborn, Sertoli cells do not express the androgen receptor, and the high androgen concentrations do not affect AMH expression. Conversely, estrogens can stimulate AMH production because estrogen receptors are present in Sertoli cells and aromatase is stimulated by FSH. During childhood, sex steroids levels are very low and do not play a physiological role on AMH production. However, hyperestrogenic states upregulate AMH expression. During puberty, testosterone inhibition of AMH expression overrides stimulation by estrogens and FSH. The direct effects of sex steroids on AMH transcription are mediated by androgen receptor and estrogen receptor α action on AMH promoter sequences. A modest estrogen action is also mediated by the membrane G-coupled estrogen receptor GPER. The understanding of these complex regulatory mechanisms helps in the interpretation of serum AMH levels found in physiological or pathological conditions, which underscores the importance of serum AMH as a biomarker of intratesticular steroid concentrations.


Asunto(s)
Hormona Antimülleriana , Testículo , Andrógenos/fisiología , Hormona Antimülleriana/fisiología , Biomarcadores , Estrógenos/fisiología , Hormona Folículo Estimulante/fisiología , Humanos , Masculino , Receptores Androgénicos/fisiología , Testículo/crecimiento & desarrollo , Testosterona/fisiología
15.
J Geriatr Psychiatry Neurol ; 35(6): 832-839, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35410535

RESUMEN

OBJECTIVES: Two-thirds of individuals living with Alzheimer's disease are women. Declining estrogen levels influence mood and cognition. Cumulative lifetime estrogen exposure (CLEE) correlates with cognition later in life. We examined the relationship of CLEE to depression and cognition in older women with major depression compared to non-depressed women. DESIGN: Older women (age ≥60 years) with depression were compared to non-depressed women using a lifetime estrogen exposure questionnaire. CLEE was defined as combined durations of reproductive span (age of menopause minus age of menarche) and any post-menopausal hormone replacement therapy use. Higher vs lower CLEE groups were based on a median of 474 months of estrogen exposure. SETTING: University hospital outpatient research program. PARTICIPANTS: 135 women ≥60 years; 64 depressed and 71 non-depressed. MEASURMENTS: Participants completed a comprehensive cognitive test battery. General linear models were used to examine the association between cognitive domain scores and CLEE in depressed and non-depressed women, controlling for age, education, and ethnicity. RESULTS: Depressed and non-depressed groups had significantly different levels of CLEE, measured in months: mean 495.7 (SD 108.6) vs 456.4 (SD 66.0) months, F(1,130) = 5.01, p = .03. Within the non-depressed participants, higher CLEE was associated with improved delayed recall (F(1,59) = 5.94, p = .02, effect size = .61), while no such relationship was observed in the depressed group. CONCLUSION: Higher CLEE was associated with improvement in delayed recall among non-depressed, but not among depressed participants. This suggests a protective role of estrogen on memory in non-depressed older postmenopausal women. Further research should examine the role of the CLEE in antidepressant response and cognitive decline.


Asunto(s)
Estrógenos , Posmenopausia , Femenino , Humanos , Anciano , Masculino , Posmenopausia/psicología , Estrógenos/uso terapéutico , Estrógenos/farmacología , Estrógenos/fisiología , Terapia de Reemplazo de Estrógeno , Cognición/fisiología , Pruebas Neuropsicológicas
16.
Endocrinology ; 163(5)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35325097

RESUMEN

We have shown that normal weight offspring born to estrogen-deprived baboons exhibited insulin resistance, although liver and adipose function and insulin receptor and glucose transporter expression were unaltered. The blood microvessels have an important role in insulin action by delivering insulin and glucose to target cells. Although little is known about the regulation of microvessel development during fetal life, estrogen promotes capillary proliferation and vascular function in the adult. Therefore, we tested the hypothesis that estrogen promotes fetal microvessel development and thus vascular function and insulin sensitivity in offspring. Capillary/myofiber ratio was decreased 75% (P < 0.05) in skeletal muscle, a major insulin target tissue, of fetal baboons in which estradiol levels were depleted by administration of aromatase inhibitor letrozole. This was sustained after birth, resulting in a 50% reduction (P < 0.01) in microvessel expansion; 65% decrease (P < 0.01) in arterial flow-mediated dilation, indicative of vascular endothelial dysfunction; and 35% increase (P < 0.01) in blood pressure in offspring from estrogen-deprived baboons, changes prevented by letrozole and estradiol administration. Along with vascular dysfunction, peak insulin and glucose levels during a glucose tolerance test were greater (P < 0.05 to P < 0.01) and the homeostasis model of insulin resistance 2-fold higher (P < 0.01) in offspring of letrozole-treated than untreated animals, indicative of insulin resistance. This study makes the novel discovery that estrogen promotes microvascularization in the fetus and thus normal vascular development and function required for eliciting insulin sensitivity in offspring and that placental hormonal secretions, independent from improper fetal growth, are an important determinant of risk of developing insulin resistance.


Asunto(s)
Resistencia a la Insulina , Animales , Estradiol/farmacología , Estrógenos/farmacología , Estrógenos/fisiología , Femenino , Feto , Glucosa , Insulina , Letrozol/farmacología , Nitrilos/farmacología , Papio , Placenta , Embarazo , Triazoles/farmacología
17.
Best Pract Res Clin Endocrinol Metab ; 36(1): 101580, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34538723

RESUMEN

Estrogens regulate pubertal development and reproductive function in women, spermatogenesis in men, and bone turnover and metabolic conditions in individuals of both sexes. Estradiol, the major estrogen in humans, is synthesized from testosterone by the action of aromatase and exerts its effects though binding to estrogen receptors. Germline loss- and gain-of-function variants in CYP19A1, the gene encoding aromatase, lead to aromatase deficiency and aromatase excess syndrome, respectively. Germline loss-of-function variants in ESR1, the gene encoding estrogen receptor α, are known to cause of estrogen insensitivity/resistance. In addition, rare variants in ESR1 and ESR2 have been implicated in various disease phenotypes. Clinical studies on these rare endocrine disorders provided clues to understand the biological functions of estrogens in the human body. This review introduces the genetic basis, phenotypes, and current management procedures of congenital disorders in estrogen biosynthesis and action.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX , Ginecomastia , Errores Innatos del Metabolismo , Trastornos del Desarrollo Sexual 46, XX/genética , Trastornos del Desarrollo Sexual 46, XX/metabolismo , Aromatasa/genética , Aromatasa/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/fisiología , Femenino , Ginecomastia/genética , Ginecomastia/metabolismo , Humanos , Masculino , Errores Innatos del Metabolismo/genética
18.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34927202

RESUMEN

Two highly prevalent pulmonary diseases, lung cancer and chronic obstructive lung disease (COPD), show both sex and gender differences in their presentations and outcomes. Sex differences are defined as biological differences associated with the male vs female genotype, and gender differences are defined as behavioral or social differences that primarily arise because of gender identity. The incidence of both lung cancer and COPD has increased dramatically in women over the past 50 years, and both are associated with chronic pulmonary inflammation. Development of COPD is also a risk factor for lung cancer. In this review, the main differences in lung cancer and COPD biology observed between men and women will be summarized. Potential causative factors will be discussed, including the role of estrogen in promoting pro-growth and inflammatory phenotypes which may contribute to development of both lung cancer and COPD. Response of the innate and adaptive immune system to estrogen is a likely factor in the biology of both lung cancer and COPD. Estrogen available from synthesis by reproductive organs as well as local pulmonary estrogen synthesis may be involved in activating estrogen receptors expressed by multiple cell types in the lung. Estrogenic actions, although more pronounced in women, may also have importance in the biology of lung cancer and COPD in men. Effects of estrogen are also timing and context dependent; the multiple cell types that mediate estrogen action in the lungs may confer both positive and negative effects on disease processes.


Asunto(s)
Neoplasias Pulmonares/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Caracteres Sexuales , Factores Sexuales , Factores de Edad , Animales , Terapia de Reemplazo de Estrógeno , Estrógenos/fisiología , Femenino , Humanos , Sistema Inmunológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Fumar/epidemiología
19.
Endocrinology ; 163(3)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34918063

RESUMEN

Successful lactation and the risk for developing breast cancer depend on growth and differentiation of the mammary gland (MG) epithelium that is regulated by ovarian steroids (17ß-estradiol [E] and progesterone [P]) and pituitary-derived prolactin (PRL). Given that the MG of pigs share histomorphogenic features present in the normal human breast, we sought to define the transcriptional responses within the MG of pigs following exposure to all combinations of these hormones. Hormone-ablated female pigs were administered combinations of E, medroxyprogesterone 17-acetate (source of P), and either haloperidol (to induce PRL) or 2-bromo-α-ergocryptine. We subsequently monitored phenotypic changes in the MG including mitosis, receptors for E and P (ESR1 and PGR), level of phosphorylated STAT5 (pSTAT5), and the frequency of terminal ductal lobular unit (TDLU) subtypes; these changes were then associated with all transcriptomic changes. Estrogen altered the expression of approximately 20% of all genes that were mostly associated with mitosis, whereas PRL stimulated elements of fatty acid metabolism and an inflammatory response. Several outcomes, including increased pSTAT5, highlighted the ability of E to enhance PRL action. Regression of transcriptomic changes against several MG phenotypes revealed 1669 genes correlated with proliferation, among which 29 were E inducible. Additional gene expression signatures were associated with TDLU formation and the frequency of ESR1 or PGR. These data provide a link between the hormone-regulated genome and phenome of the MG in a species having a complex histoarchitecture like that in the human breast, and highlight an underexplored synergy between the actions of E and PRL during MG development.


Asunto(s)
Estrógenos/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Progesterona/fisiología , Prolactina/fisiología , Porcinos Enanos/fisiología , Transcriptoma/fisiología , Animales , Bromocriptina/administración & dosificación , Sinergismo Farmacológico , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/genética , Estrógenos/deficiencia , Femenino , Haloperidol/administración & dosificación , Glándulas Mamarias Animales/química , Glándulas Mamarias Animales/efectos de los fármacos , Acetato de Medroxiprogesterona/administración & dosificación , Modelos Animales , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Ovariectomía , Progesterona/deficiencia , Prolactina/deficiencia , Receptores de Progesterona/análisis , Receptores de Progesterona/genética , Porcinos , Transcriptoma/efectos de los fármacos
20.
Neuropharmacology ; 204: 108879, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34785164

RESUMEN

Gonadal steroid hormone influences behavioral choice of adult animals toward pups, parental or aggressive. We previously reported that long-term administration of 17ß-estradiol (E2) to male mice during sexual maturation induces aggressive behavior toward conspecific pups, which is called "infanticide," and significantly enhanced excitatory synaptic transmission in the rhomboid nucleus of bed nucleus of the stria terminalis (BSTrh), which is an important brain region for infanticide. However, it is unclear how estrogen receptor-dependent signaling after sexual maturity regulates neural circuits including the BSTrh. Here we revealed that E2 administration to gonadectomized mice in adulthood elicited infanticidal behavior and enhanced excitatory synaptic transmission in the BSTrh by increasing the probability of glutamate release from the presynaptic terminalis. Next, we performed whole-brain mapping of E2-sensitive brain regions projecting to the BSTrh and found that amygdalohippocampal area (AHi) neurons that project to the BSTrh densely express estrogen receptor 1 (Esr1). Moreover, E2 treatment enhanced synaptic connectivity in the AHi-BSTrh pathway. Together, these results suggest that reinforcement of excitatory inputs from AHi neurons into the BSTrh by estrogen receptor-dependent signaling may contribute to the expression of infanticide.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Animales Recién Nacidos , Conducta Animal/fisiología , Conducta de Elección/fisiología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Estrógenos/fisiología , Hipocampo/metabolismo , Núcleos Talámicos de la Línea Media/metabolismo , Núcleos Septales/metabolismo , Transducción de Señal/fisiología , Agresión/efectos de los fármacos , Agresión/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Estradiol/administración & dosificación , Estradiol/farmacología , Glutamatos/metabolismo , Masculino , Ratones , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...