Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 287
Filtrar
2.
Exp Anim ; 70(1): 63-72, 2021 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-32981898

RESUMEN

Aromatase, an estrogen synthase, exists in the gastric parietal cells of Wistar rats. The stomach synthesizes large amounts of estrogens and secretes them into the portal vein. We have been particularly studying gastric estrogen synthesis using Wistar rats. However, estrogen synthesis in the stomach of Sprague-Dawley (SD) rats, which are used as frequently as those of the Wistar strain, has not been clarified. We examined steroid synthesis in the stomach of SD rats using immunohistochemistry, in situ hybridization, Western blotting, real-time PCR, and LC-MS/MS. Aromatase also exists in the stomach of SD rats. Its distribution was not found to be different from that of Wistar rats. Results show that H+/K+-ATPase ß-subunit and aromatase colocalized in double immunofluorescence staining. Each steroid synthase downstream from progesterone was present in the gastric mucosa. These results suggest that steroid hormones are synthesized in the parietal cells in the same pathway as Wistar rats. Although mRNA expression of steroid synthases were higher in SD, no significant difference was found in the amount of protein and each steroid hormone level in the portal vein. Although differences between strains might exist in steroid hormone synthesis, results show that SD rats are as useful as Wistar rats for gastric estrogen synthesis experimentation.


Asunto(s)
Estrógenos/biosíntesis , Células Parietales Gástricas/metabolismo , Ratas Sprague-Dawley , Animales , Aromatasa/genética , Aromatasa/metabolismo , Aromatasa/fisiología , Estrógenos/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/enzimología , Mucosa Gástrica/metabolismo , Expresión Génica , Masculino , Células Parietales Gástricas/enzimología , Vena Porta/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar
3.
Domest Anim Endocrinol ; 74: 106509, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32653739

RESUMEN

MicroRNAs (miRNAs) are 21- to 24-nucleotide long small noncoding RNAs, which play an important role in follicular atresia and granulosa cell (GC) apoptosis in the mammalian ovary. Here, we report that miR-146b, a conserved and ovary-enriched miRNA, modulates estradiol (E2) secretion, GC apoptosis, and follicular atresia in pigs. Genome-wide analysis and quantitative real-time PCR revealed that miR-146b was significantly upregulated during follicular atresia, and fluorescence-activated cell sorting showed that miR-146b functioned as a proapoptotic factor to induce GC apoptosis. MicroRNA-mRNA network analysis and luciferase reporter assays showed that CYP19A1, the pivotal enzyme for E2 synthesis signaling, was directly targeted by miR-146b. Furthermore, miR-146b interacted with the 3'untranslated region of CYP19A1 to prevent translation, thereby regulating CYP19A1-mediated E2 secretion and GC apoptosis. However, miR-146b was not regulated by the transcription factor SMAD4 or oxidative stress, both of which are critical regulators of CYP19A1. We, thus, conclude that miR-146b is a novel epigenetic factor regulating GC functions, follicular development, and female reproduction.


Asunto(s)
Apoptosis/genética , Aromatasa/genética , Aromatasa/fisiología , Células de la Granulosa/fisiología , MicroARNs/fisiología , Sus scrofa/fisiología , Animales , Inhibidores de la Aromatasa , Secuencia de Bases , Células Cultivadas , Epigénesis Genética , Femenino , Expresión Génica/genética , MicroARNs/genética , Sus scrofa/genética , Transfección , Regulación hacia Arriba
4.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153202

RESUMEN

Endometriosis is defined as the presence of endometrial foci, localized beyond their primary site, i.e., the uterine cavity. The etiology of this disease is rather complex. Its development is supported by hormonal, immunological, and environmental factors. During recent years, particular attention has been focused on the genetic mechanisms that may be of particular significance for the increased incidence rates of endometriosis. According to most recent studies, ESR2 and CYP19A1 genes may account for the potential risk factors of infertility associated with endometriosis. The paper presents a thorough review of the latest reports and data concerning the genetic background of the risk for endometriosis development.


Asunto(s)
Aromatasa/fisiología , Endometriosis/genética , Receptor beta de Estrógeno/fisiología , Antecedentes Genéticos , Endometriosis/epidemiología , Endometriosis/etiología , Endometrio/metabolismo , Endometrio/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Infertilidad/genética , Factores de Riesgo
5.
Mol Cell Endocrinol ; 516: 110947, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32702473

RESUMEN

Estrogens are important in regulating mood, especially for females. However, whether tissue-specific estrogen, such as brain estrogen, contributes to the effects of antidepressant treatment has not been determined. The present study used middle-aged aromatase gene knockout (Ar-/-) mice or overexpression (Thy1-Ar; hGFAP-Ar) mice as brain estrogen models to investigate whether brain estrogen synthesis alters the anti-depressive behaviors of sertraline treatment. Our results showed that depletion of brain estrogen increased depressive-like behavior in females, and elevated brain estrogen reduced depression-like behavior, regardless of sex. These genotype-related behaviors correlated with alterations of monoamine metabolism in the hippocampus (HPC) and the prefrontal cortex (PFC). We also demonstrated that male and female Ar-/- mice exhibited an attenuation of sertraline-induced anti-depressive behaviors compared to wild-type (WT) mice. The present data suggest that brain estrogen alters depressive-like behaviors and changes the effectiveness of antidepressants in middle-aged mice, regardless of sex.


Asunto(s)
Aromatasa/fisiología , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Estrógenos/farmacología , Sertralina/farmacología , Animales , Antidepresivos/farmacología , Depresión/metabolismo , Depresión/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurotransmisores/metabolismo
6.
Bull Math Biol ; 82(7): 89, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32638157

RESUMEN

In many oviparous species, the incubation temperature of the egg determines the sex of the offspring. This is known as temperature-dependent sex determination (TSD). The probability of the hatched offspring being male or female varies across the incubation temperature range. This leads to the appearance of different TSD patterns in species such as FM pattern where females are predominately born at lower temperature and males at higher temperature, FMF pattern where the probability of female being born is higher at extreme temperatures and of the male being born is high at intermediate temperatures. We analyze an enzymatic reaction system proposed in the literature involving sex hormones with positive feedback effect to understand the emergence of different TSD patterns. The nonlinearity in the model is accounted through temperature sensitivity of the reaction rates affecting the catalytic mechanism in the reaction system. We employ a dynamical systems approach of singularity theory and bifurcation analysis to divide the parameter plane of temperature sensitivities into different regions where different TSD patterns are observed. Bifurcation analysis in association with the delineation of the parameter space for different TSD pattern has led to the identification of a subspace where all the TSD patterns observed in nature can be realized. We also show how modulation of the sex hormone in the species can be used to change the probability of occurrence of a specific sex, thereby preventing the extinction of endangered species.


Asunto(s)
Modelos Biológicos , Oviparidad/fisiología , Procesos de Determinación del Sexo/fisiología , Animales , Aromatasa/fisiología , Estrógenos/fisiología , Femenino , Masculino , Conceptos Matemáticos , Dinámicas no Lineales , Razón de Masculinidad , Análisis de Sistemas , Temperatura , Testosterona/fisiología
7.
Aging (Albany NY) ; 12(15): 15414-15435, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32651991

RESUMEN

Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in reproductive women and is characterized by polycystic ovaries, hyperandrogenism and chronic anovulation. Abnormal folliculogenesis is considered as a common characteristic of PCOS. Our aim is to identify the altered circRNA expression profile in exosomes isolated from follicular fluid (FF) of PCOS patients to investigate the molecular function of exosomal circRNA, as a vital mediator in follicular microenvironment, in the aetiology and pathobiology of PCOS. In this study, the circRNA expression profile of FF exosomes were compared between PCOS and control patients by RNA sequencing (N=5 vs 5). Sixteen circRNAs showed significantly different expression. GO and KEGG pathway analyses indicated that their parental genes were enriched in PCOS-related pathways, including ovarian steroidogenesis, aldosterone synthesis and secretion, and Jak-STAT signaling. Among sixteen differentially expressed circRNAs, hsa_circ_0006877 (circLDLR) was processed from its parental LDLR (low density lipoprotein receptor) transcript, which participated in ovarian steroidogenesis. Its depletion in PCOS FF exosomes was further verified in an additional cohort (N=25 vs 25) by qRT-PCR. And a circLDLR-miR-1294-CYP19A1 competing endogenous RNA (ceRNA) network was predicted by cytoscape software, and confirmed by luciferase assay and correlative expression in the cumulus cells of PCOS patients. Mechanistically, the intercellular transfer of functional circLDLR assay and its withdrawal experiments in KGN cells showed that depleting circLDLR in exosomes increased miR-1294 expression and inhibited CYP19A1 expression in recipient cells, as well as reduced their estrogen (E2) secretion. Our findings revealed a ceRNA network of circLDLR and provided new information on abnormal follicle development in PCOS.


Asunto(s)
Aromatasa/fisiología , Estradiol/biosíntesis , Exosomas , Líquido Folicular , MicroARNs/fisiología , Síndrome del Ovario Poliquístico/etiología , Síndrome del Ovario Poliquístico/genética , ARN Circular/genética , Receptores de LDL/genética , Adulto , Femenino , Humanos
8.
Development ; 147(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32001440

RESUMEN

Sex determination and differentiation are complex processes controlled by many different factors; however, the relationships among these factors are poorly understood. Zebrafish gonadal differentiation exhibits high plasticity involving multiple factors and pathways, which provides an excellent model for investigating the interactions between them. Ovarian aromatase (cyp19a1a) and dmrt1 are key factors in directing vertebrate ovary and testis differentiation, respectively. Knockout of zebrafish cyp19a1a leads to all-male offspring, whereas the loss of dmrt1 results in a female-biased sex ratio. In the present study, we established dmrt1-/- ;cyp19a1a-/- double mutant zebrafish and discovered that the introduction of the dmrt1 mutation into the cyp19a1a mutant could rescue the all-male phenotype of the latter. Interestingly, despite the lack of aromatase/estrogens, the follicles in the ovary of the rescued cyp19a1a mutant could develop normally up to the previtellogenic stage. Further evidence suggested the ovarian aromatase directed ovarian differentiation by suppressing dmrt1 expression via nuclear estrogen receptors (nERs). Our results provide solid evidence for an interaction between cyp19a1a and dmrt1 in zebrafish gonadal differentiation, and for the dispensability of estrogens in controlling early folliculogenesis.


Asunto(s)
Aromatasa/genética , Aromatasa/fisiología , Folículo Ovárico/embriología , Testículo/embriología , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Alelos , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Estrógenos/fisiología , Femenino , Técnicas de Inactivación de Genes , Genotipo , Heterocigoto , Masculino , Mutación , Fenotipo , Receptores de Estrógenos/fisiología , Procesos de Determinación del Sexo , Diferenciación Sexual , Pez Cebra
9.
Eur J Histochem ; 64(1)2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-31988532

RESUMEN

The goal of this study was to evaluate P450 aromatase localization in the epididymis of two different vertebrates: the lizard Podarcis sicula, a seasonal breeder, and Rattus rattus, a continuous breeder. P450 aromatase is a key enzyme involved in the local control of spermatogenesis and steroidogenesis and we proved for the first time that this enzyme is represented in the epididymis of both P. sicula and R. rattus. In details, P450 aromatase was well represented in epithelial and myoid cells and in the connective tissue of P. sicula epididymis during the reproductive period; instead, during autumnal resumption this enzyme was absent in the connective tissue. During the non-reproductive period, P450 aromatase was localized only in myoid cells of P. sicula epididymis, whereas in R. rattus it was localized both in myoid cells and connective tissue. Our findings, the first on the epididymis aromatase localization in the vertebrates, suggest a possible role of P450 aromatase in the control of male genital tract function, particularly in sperm maturation.


Asunto(s)
Aromatasa/fisiología , Epidídimo/enzimología , Animales , Tejido Conectivo/enzimología , Inmunohistoquímica , Lagartos , Masculino , Ratas , Reproducción/fisiología
10.
Differentiation ; 109: 42-52, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31520742

RESUMEN

Hypospadias is the abnormal opening of the urethra on the underside of the penis and occurs in approximately 1/125 live male births worldwide. The incidence rate of hypospadias has dramatically increased over the past few decades. This is now attributed, at least in part, to our exposure to endocrine-disrupting chemicals (EDCs) which alter the hormonal signals required for development of the penis. In humans androgens are the main drivers of fusion of the urethral folds to form the urethra within the shaft of the penis, a process required for termination of the urethra in its normal location at the tip of the penis. However, recent research has suggested that estrogen also plays a role in this process. To better understand how EDCs impact urethral development it is essential that we understand the normal function of hormones during development of the penis. To define the role of estrogen in urethral development we examined development of the penis in the aromatase (Cyp19a1) Knockout (ArKO) mouse strain in which endogenous estrogen production is completely ablated. We found that the ArKO penis had a mild hypospadias phenotype. The developing ArKO postnatal penis displayed an early disruption in preputial development, which likely causes the mild hypospadias observed in adults. Using qPCR, we found altered expression of keratin genes and key urethral patterning genes in response to the disrupted estrogen signaling. The hypospadias phenotype was almost identical to that reported for the estrogen receptor α (ERα) knockout confirming that ERα is the predominant receptor for mediating estrogen action during development of the mouse penis. Our results show that estrogen is required for normal prepucial development and placement of the mature urethral opening at the distal aspect of the penis. We also identified several genes which are potential downstream targets of estrogen during normal urethral closure. With this knowledge, we can now better understand how anti-estrogenic as well as estrogenic EDCs disrupt urethral closure to cause mild hypospadias in both mice and humans.


Asunto(s)
Aromatasa/fisiología , Estrógenos/metabolismo , Hipospadias/etiología , Organogénesis , Pene/anomalías , Receptores de Estrógenos/metabolismo , Animales , Hipospadias/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pene/enzimología , Transducción de Señal
11.
Psychoneuroendocrinology ; 98: 161-167, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30165284

RESUMEN

BACKGROUND: Polymorphisms in sex steroid receptors have been associated with transsexualism. However, published replication studies have yielded inconsistent findings, possibly because of a limited sample size and/or the heterogeneity of the transsexual population with respect to the onset of dysphoria and sexual orientation. We assessed the role of androgen receptor (AR), estrogen receptors alpha (ERα) and beta (ERß), and aromatase (CYP19A1) in two large and homogeneous transsexual male-to-female (MtF) and female-to-male (FtM) populations. METHODS: The association of each polymorphism with transsexualism was studied with a twofold subject-control analysis: in a homogeneous population of 549 early onset androphilic MtF transsexuals versus 728 male controls, and 425 gynephilic FtMs versus 599 female controls. Associations and interactions were investigated using binary logistic regression. RESULTS: Our data show that specific allele and genotype combinations of ERß, ERα and AR are implicated in the genetic basis of transsexualism, and that MtF gender development requires AR, which must be accompanied by ERß. An inverse allele interaction between ERß and AR is characteristic of the MtF population: when either of these polymorphisms is short, the other is long. ERß and ERα are also associated with transsexualism in the FtM population although there was no interaction between the polymorphisms. Our data show that ERß plays a key role in the typical brain differentiation of humans. CONCLUSION: ERß plays a key role in human gender differentiation in males and females.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Disforia de Género/genética , Disforia de Género/metabolismo , Adulto , Alelos , Andrógenos/metabolismo , Aromatasa/metabolismo , Aromatasa/fisiología , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Estrógenos/metabolismo , Femenino , Identidad de Género , Frecuencia de los Genes/genética , Genotipo , Humanos , Masculino , Oportunidad Relativa , Polimorfismo Genético/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Conducta Sexual , Desarrollo Sexual/fisiología , Transexualidad/genética
12.
Conscious Cogn ; 55: 205-213, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28888137

RESUMEN

Testosterone and estrogen are involved in self-related behavioral dispositions and experiences of subjective well-being. In this study, we investigated to what extent the aromatase (CYP19A1) gene, which encodes an enzyme in converting testosterone into estrogen, contributes to subjective well-being and in another self-related disposition: independent and interdependent self-construal. In study 1, a meta-analysis showed that the GG genotype of CYP19A1 (a G/A substitution at Val80, rs700518) was associated with higher testosterone and lower estradiol. In study 2, an empirical study of individuals with the GG (n=115), AG (n=286) and AA (n=193) genotypes indicated that individuals with the GG genotype exhibited higher independent self-construal and higher subjective well-being. The association between the GG genotype of CYP19A1 Val80 and subjective well-being was mediated by the independent self-construal. Our findings reinforce the idea that personality traits such as independent self-construal explain the link between genetic variant and subjective well-being.


Asunto(s)
Aromatasa/fisiología , Estradiol/sangre , Satisfacción Personal , Personalidad/fisiología , Autoimagen , Testosterona/sangre , Adulto , Aromatasa/genética , Femenino , Genotipo , Humanos , Masculino , Adulto Joven
13.
Endocrinology ; 158(9): 3030-3041, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575219

RESUMEN

Aromatase (encoded by the cyp19a1a and cyp19a1b genes) plays a central role in sex differentiation in fish, but its precise roles during sex differentiation are still largely unknown. Here, we systematically generated cyp19a1a and cyp19a1b mutant lines as well as a cyp19a1a;cyp19a1b double mutant line in zebrafish using transcription activatorlike effector nucleases. Our results showed that cyp19a1a mutants and cyp19a1a;cyp19a1b double mutants, but not cyp19a1b mutants, had impaired sex differentiation, and all cyp19a1a mutants and cyp19a1a;cyp19a1b double mutants were males. During sex differentiation, the ovary-like gonads were not observed and the male sex differentiation program was delayed in the cyp19a1a-null fish, and these phenotypes could be partially rescued by 17ß-estradiol treatment. Gene expression analysis indicated that male and female sex differentiation-related genes were significantly decreased in the cyp19a1a mutant. Collectively, our results revealed dual functions of the cyp19a1a gene during sex differentiation: cyp19a1a is not only indispensable for female sex differentiation but also required for male sex differentiation.


Asunto(s)
Aromatasa/fisiología , Diferenciación Sexual/genética , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Aromatasa/genética , Embrión no Mamífero , Estradiol/farmacología , Femenino , Fertilidad/efectos de los fármacos , Fertilidad/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Gónadas/efectos de los fármacos , Gónadas/embriología , Gónadas/metabolismo , Masculino , Mutación , Diferenciación Sexual/efectos de los fármacos , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
14.
J Steroid Biochem Mol Biol ; 168: 19-25, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28109841

RESUMEN

Human health is beset with a legion of ailments, which is exacerbated by lifestyle errors. Out of the numerous enzymes in human body, aromatase, a cytochrome P450 enzyme is particularly very critical. Occurring at the crossroads of multiple signalling pathways, its homeostasis is vital for optimal health. Unfortunately, medications, hormone therapy, chemical additives in food, and endocrine-disrupting personal care products are oscillating the aromatase concentration beyond the permissible level. As this enzyme converts androgens (C19) into estrogens (C18), its agitation has different outcomes in different genders and age groups. Some common pathologies associated with aromatase disruption include breast cancer, prostate cancer, polycystic ovary syndrome (PCOS), endometriosis, osteoporosis, ovarian cancer, gastric cancer, pituitary cancer, Alzheimer's disease, schizophrenia, male hypogonadism, and transgender issues. Several drugs, cosmetics and pesticides act as the activators and suppressors of this enzyme. This carefully-compiled critical review is expected to increase public awareness regarding the threats resultant of the perturbations of this enzyme and to motivate researchers for further investigation of this field.


Asunto(s)
Aromatasa/efectos de los fármacos , Aromatasa/fisiología , Homeostasis , Andrógenos/química , Aromatasa/genética , Inhibidores de la Aromatasa/química , Neoplasias de la Mama/sangre , Sistema Endocrino , Endometriosis/sangre , Estrógenos/química , Femenino , Humanos , Masculino , Obesidad/sangre , Osteoporosis/sangre , Neoplasias Ováricas/sangre , Plaguicidas/química , Síndrome del Ovario Poliquístico/sangre , Embarazo , Neoplasias de la Próstata/sangre , Transducción de Señal , Personas Transgénero
15.
J Neurosci ; 37(6): 1532-1545, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28028198

RESUMEN

The basolateral amygdala (BLA) integrates sensory input from cortical and subcortical regions, a function that requires marked synaptic plasticity. Here we provide evidence that cytochrome P450 aromatase (AROM), the enzyme converting testosterone to 17ß-estradiol (E2), contributes to the regulation of this plasticity in a sex-specific manner. We show that AROM is expressed in the BLA, particularly in the basolateral nucleus (BL), in male and female rodents. Systemic administration of the AROM inhibitor letrozole reduced spine synapse density in the BL of adult female mice but not in the BL of male mice. Similarly, in organotypic corticoamygdalar slice cultures from immature rats, treatment with letrozole significantly reduced spine synapses in the BL only in cultures derived from females. In addition, letrozole sex-specifically altered synaptic properties in the BL: in acute slices from juvenile (prepubertal) female rats, wash-in of letrozole virtually abolished long-term potentiation (LTP), whereas it did not prevent the generation of LTP in the slices from males. Together, these data indicate that neuron-derived E2 modulates synaptic plasticity in rodent BLA sex-dependently. As protein expression levels of AROM, estrogen and androgen receptors did not differ between males and females and were not sex-specifically altered by letrozole, the findings suggest sex-specific mechanisms of E2 signaling.SIGNIFICANCE STATEMENT The basolateral amygdala (BLA) is a key structure of the fear circuit. This research reveals a sexually dimorphic regulation of synaptic plasticity in the BLA involving neuronal aromatase, which produces the neurosteroid 17ß-estradiol (E2). As male and female neurons in rodent BLA responded differently to aromatase inhibition both in vivo and in vitro, our findings suggest that E2 signaling in BLA neurons is regulated sex-dependently, presumably via mechanisms that have been established during sexual determination. These findings could be relevant for the understanding of sex differences in mood disorders and of the side effects of cytochrome P450 aromatase inhibitors, which are frequently used for breast cancer therapy.


Asunto(s)
Inhibidores de la Aromatasa/farmacología , Aromatasa/fisiología , Complejo Nuclear Basolateral/fisiología , Plasticidad Neuronal/fisiología , Caracteres Sexuales , Animales , Complejo Nuclear Basolateral/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Letrozol , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Nitrilos/farmacología , Técnicas de Cultivo de Órganos , Ratas , Triazoles/farmacología
16.
Gen Comp Endocrinol ; 241: 69-79, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-26873632

RESUMEN

Radial glial cells (RGCs) in teleost brain are progenitor cells that express aromatase B and produce estrogens. Controversial data suggest that estrogens are critical for brain repair and neurogenesis in teleosts. Using a goldfish model for neurotoxin-induced Parkinson-like syndrome, we investigated the possible roles of RGCs, especially estrogen synthetic function, in the processes underlying dopamine neuron regeneration. The data indicate that dopamine neuron degeneration and aromatase activity inhibition could be respectively achieved in vivo with treatments with the neurotoxin 1-methyl-1,2,3,6-tetrahydropyridine (MPTP) and fadrozole in female goldfish. The expression of genes in the telencephalon and hypothalamus related to RGC functions including gfap, gdnf and bdnf as well as genes related to mature dopamine neuron functions including th, slc6a3 and pitx3 are under modulation of estrogens. Together these results revealed that the activation of radial glial cells and dopamine neuron recovery after MPTP insult is aromatase-dependent. Findings in this study provide support for the hypothesis that endogenous estrogens are neuroprotective in goldfish. Future studies focus on the molecular pathways for enhancing protective functions of estrogens and understanding global effects of estrogens in the central nervous system.


Asunto(s)
Aromatasa/fisiología , Neuronas Dopaminérgicas/fisiología , Células Ependimogliales/fisiología , Degeneración Nerviosa , Regeneración , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Fadrozol/farmacología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Carpa Dorada , Degeneración Nerviosa/inducido químicamente , Neurogénesis/efectos de los fármacos , Neurotoxinas/farmacología , Regeneración/efectos de los fármacos
17.
Physiology (Bethesda) ; 31(4): 258-69, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27252161

RESUMEN

Aromatase (estrogen synthetase; EC 1.14.14.1) catalyzes the demethylation of androgens' carbon 19, producing phenolic 18-carbon estrogens. Aromatase is most widely known for its roles in reproduction and reproductive system diseases, and as a target for inhibitor therapy in estrogen-sensitive diseases including cancer, endometriosis, and leiomyoma (141, 143). However, all tissues contain estrogen receptor-expressing cells, the majority of genes have a complete or partial estrogen response element that regulates their expression (61), and there are plentiful nonreceptor effects of estrogens (79); therefore, the effect of aromatase through the provision of estrogen is almost universal in terms of health and disease. This review will provide a brief but comprehensive overview of the enzyme, its role in steroidogenesis, the problems that arise with its functional mutations and mishaps, the roles in human physiology of aromatase and its product estrogens, its current clinical roles, and the effects of aromatase inhibitors. While much of the story is that of the consequences of the formation of its product estrogens, we also will address alternative enzymatic roles of aromatase as a demethylase or nonenzymatic actions of this versatile molecule. Although this short review is meant to be thorough, it is by no means exhaustive; rather, it is meant to reflect the cutting-edge, exciting properties and possibilities of this ancient enzyme and its products.


Asunto(s)
Aromatasa/fisiología , Estrógenos/fisiología , Animales , Aromatasa/genética , Aromatasa/inmunología , Inhibidores de la Aromatasa/uso terapéutico , Encéfalo/enzimología , Enfermedad , Femenino , Homeostasis , Desarrollo Humano , Humanos , Masculino
18.
Horm Behav ; 83: 45-59, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27189762

RESUMEN

Although aromatase is expressed in both male and female brains, its functional significance in females remains poorly understood. In female quail, sexual receptivity is activated by estrogens. However it is not known whether sexual motivation is similarly estrogen-dependent and whether estrogens locally produced in the brain contribute to these behavioral responses. Four main experiments were designed to address these questions. In Experiment 1 chronic treatment of females with the anti-estrogen tamoxifen decreased their receptivity, confirming that this response is under the control of estrogens. In Experiment 2 chronic treatment with tamoxifen significantly decreased sexual motivation as treated females no longer approached a sexual partner. In Experiment 3 (a) ovariectomy (OVX) induced a significant decrease of time spent near the male and a significantly decreased receptivity compared to gonadally intact females, (b) treatment with testosterone (OVX+T) partially restored these responses and (c) this effect of T was prevented when estradiol synthesis was inhibited by the potent aromatase inhibitor Vorozole (OVX+T+VOR). Serum estradiol concentration was significantly higher in OVX+T than in OVX or OVX+T+VOR females. Together these data demonstrate that treatment of OVX females with T increases sexual motivation and that these effects are mediated at least in part by non-gonadal aromatization of the androgen. Finally, assays of aromatase activity on brain and peripheral tissues (Experiment 4) strongly suggest that brain aromatization contributes to behavioral effects observed here following T treatment but alternative sources of estrogens (e.g. liver) should also be considered.


Asunto(s)
Aromatasa/metabolismo , Coturnix , Motivación/fisiología , Conducta Sexual Animal/fisiología , Testosterona/farmacología , Animales , Aromatasa/fisiología , Inhibidores de la Aromatasa/farmacología , Coturnix/sangre , Coturnix/fisiología , Estradiol/sangre , Femenino , Masculino , Motivación/efectos de los fármacos , Ovariectomía/veterinaria , Ovario/efectos de los fármacos , Ovario/metabolismo , Codorniz/sangre , Codorniz/fisiología , Conducta Sexual Animal/efectos de los fármacos , Triazoles/farmacología
19.
Asian J Androl ; 18(3): 435-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26908066

RESUMEN

Traditionally, testosterone and estrogen have been considered to be male and female sex hormones, respectively. However, estradiol, the predominant form of estrogen, also plays a critical role in male sexual function. Estradiol in men is essential for modulating libido, erectile function, and spermatogenesis. Estrogen receptors, as well as aromatase, the enzyme that converts testosterone to estrogen, are abundant in brain, penis, and testis, organs important for sexual function. In the brain, estradiol synthesis is increased in areas related to sexual arousal. In addition, in the penis, estrogen receptors are found throughout the corpus cavernosum with high concentration around neurovascular bundles. Low testosterone and elevated estrogen increase the incidence of erectile dysfunction independently of one another. In the testes, spermatogenesis is modulated at every level by estrogen, starting with the hypothalamus-pituitary-gonadal axis, followed by the Leydig, Sertoli, and germ cells, and finishing with the ductal epithelium, epididymis, and mature sperm. Regulation of testicular cells by estradiol shows both an inhibitory and a stimulatory influence, indicating an intricate symphony of dose-dependent and temporally sensitive modulation. Our goal in this review is to elucidate the overall contribution of estradiol to male sexual function by looking at the hormone's effects on erectile function, spermatogenesis, and libido.


Asunto(s)
Estradiol/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Espermatogénesis/fisiología , Testículo/fisiología , Testosterona/fisiología , Aromatasa/fisiología , Células Germinativas/fisiología , Humanos , Células Intersticiales del Testículo/fisiología , Libido/fisiología , Masculino , Erección Peniana/fisiología , Células de Sertoli/fisiología
20.
Fish Physiol Biochem ; 42(2): 795-805, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26643906

RESUMEN

Female summer flounder grow considerably faster and larger than males, and a tremendous increase in performance can therefore be realized through production of monosex female populations. Rearing temperature has been shown to affect sex differentiation in other teleost species by influencing expression of genes encoding transcription factors or enzymes involved in endocrine function. Cyp19a1a is a well-studied gene that had been shown to play a role in ovarian development, and exhibits sexually dimorphic expression in other species. In the present study, summer flounder (37 days post-hatch; DPH) were raised at 13, 16 or 19 °C. Fish from all three treatments were sampled throughout development and analyzed in qPCR to determine cyp19a1a gene expression levels. Sex ratios of additional fish grown to ≥150 mm at each temperature treatment were determined. Low female production was achieved overall (26.9, 17.6 and 0% at 13, 16 and 19 °C, respectively). Cyp19a1a expression was significantly lower at 52 DPH (~15 mm total length) at the male-producing temperature (19 °C) and increased to similar levels as other treatments at 66 DPH. Expression levels later in juvenile development (66-191 DPH) largely decreased with fish size. The period of sex differentiation in summer flounder remains unknown, but cyp19a1a expression patterns suggest that it may occur earlier in development than that of congenerics. Further research is necessary to understand the sex-determining mechanisms in this species before sexually dimorphic growth can be used to achieve economic advantages in commercial production.


Asunto(s)
Aromatasa/fisiología , Lenguado/fisiología , Diferenciación Sexual/fisiología , Animales , Femenino , Masculino , Razón de Masculinidad , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...