Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(44): 13699-704, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26483456

RESUMEN

Epigenetic signatures in germ cells, capable of both responding to the parental environment and shaping offspring neurodevelopment, are uniquely positioned to mediate transgenerational outcomes. However, molecular mechanisms by which these marks may communicate experience-dependent information across generations are currently unknown. In our model of chronic paternal stress, we previously identified nine microRNAs (miRs) that were increased in the sperm of stressed sires and associated with reduced hypothalamic-pituitary-adrenal (HPA) stress axis reactivity in offspring. In the current study, we rigorously examine the hypothesis that these sperm miRs function postfertilization to alter offspring stress responsivity and, using zygote microinjection of the nine specific miRs, demonstrated a remarkable recapitulation of the offspring stress dysregulation phenotype. Further, we associated long-term reprogramming of the hypothalamic transcriptome with HPA axis dysfunction, noting a marked decreased in the expression of extracellular matrix and collagen gene sets that may reflect an underlying change in blood-brain barrier permeability. We conclude by investigating the developmental impact of sperm miRs in early zygotes with single-cell amplification technology, identifying the targeted degradation of stored maternal mRNA transcripts including sirtuin 1 and ubiquitin protein ligase E3a, two genes with established function in chromatin remodeling, and this potent regulatory function of miRs postfertilization likely initiates a cascade of molecular events that eventually alters stress reactivity. Overall, these findings demonstrate a clear mechanistic role for sperm miRs in the transgenerational transmission of paternal lifetime experiences.


Asunto(s)
Epigénesis Genética , MicroARNs/genética , Exposición Paterna , Espermatozoides/metabolismo , Estrés Fisiológico , Animales , Sistema Hipotálamo-Hipofisario , Masculino , Ratones , Ratones Endogámicos C57BL , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 110(13): 5169-74, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23487789

RESUMEN

Maternal stress is a key risk factor for neurodevelopmental disorders, including schizophrenia and autism, which often exhibit a sex bias in rates of presentation, age of onset, and symptom severity. The placenta is an endocrine tissue that functions as an important mediator in responding to perturbations in the intrauterine environment and is accessible for diagnostic purposes, potentially providing biomarkers predictive of disease. Therefore, we have used a genome-wide array approach to screen placental expression across pregnancy for gene candidates that are sex-biased and stress-responsive in mice and translate to human tissue. We identifed O-linked-N-acetylglucosamine (O-GlcNAc) transferase (OGT), an X-linked gene important in regulating proteins involved in chromatin remodeling, as fitting these criteria. Levels of both OGT and its biochemical mark, O-GlcNAcylation, were significantly lower in males and further reduced by prenatal stress. Examination of human placental tissue found similar patterns related to X chromosome dosage. As a demonstration of the importance of placental OGT in neurodevelopment, we found that hypothalamic gene expression and the broad epigenetic microRNA environment in the neonatal brain of placental-specific hemizygous OGT mice was substantially altered. These studies identified OGT as a promising placental biomarker of maternal stress exposure that may relate to sex-biased outcomes in neurodevelopment.


Asunto(s)
N-Acetilglucosaminiltransferasas/biosíntesis , Placenta/enzimología , Complicaciones del Embarazo/enzimología , Proteínas Gestacionales/biosíntesis , Estrés Fisiológico , Animales , Biomarcadores/metabolismo , Cromosomas Humanos X/genética , Cromosomas Humanos X/metabolismo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Epigénesis Genética/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Hipotálamo/embriología , Hipotálamo/enzimología , Hipotálamo/patología , Masculino , Ratones , Ratones Transgénicos , Neurogénesis/genética , Placenta/patología , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/patología , Proteínas Gestacionales/genética , Caracteres Sexuales
3.
J Neurosci ; 33(21): 9003-12, 2013 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23699511

RESUMEN

Neuropsychiatric disease frequently presents with an underlying hyporeactivity or hyperreactivity of the HPA stress axis, suggesting an exceptional vulnerability of this circuitry to external perturbations. Parental lifetime exposures to environmental challenges are associated with increased offspring neuropsychiatric disease risk, and likely contribute to stress dysregulation. While maternal influences have been extensively examined, much less is known regarding the specific role of paternal factors. To investigate the potential mechanisms by which paternal stress may contribute to offspring hypothalamic-pituitary-adrenal (HPA) axis dysregulation, we exposed mice to 6 weeks of chronic stress before breeding. As epidemiological studies support variation in paternal germ cell susceptibility to reprogramming across the lifespan, male stress exposure occurred either throughout puberty or in adulthood. Remarkably, offspring of sires from both paternal stress groups displayed significantly reduced HPA stress axis responsivity. Gene set enrichment analyses in offspring stress regulating brain regions, the paraventricular nucleus (PVN) and the bed nucleus of stria terminalis, revealed global pattern changes in transcription suggestive of epigenetic reprogramming and consistent with altered offspring stress responsivity, including increased expression of glucocorticoid-responsive genes in the PVN. In examining potential epigenetic mechanisms of germ cell transmission, we found robust changes in sperm microRNA (miR) content, where nine specific miRs were significantly increased in both paternal stress groups. Overall, these results demonstrate that paternal experience across the lifespan can induce germ cell epigenetic reprogramming and impact offspring HPA stress axis regulation, and may therefore offer novel insight into factors influencing neuropsychiatric disease risk.


Asunto(s)
Sistema Hipotálamo-Hipofisario/fisiopatología , MicroARNs/metabolismo , Sistema Hipófiso-Suprarrenal/fisiología , Efectos Tardíos de la Exposición Prenatal/patología , Espermatozoides/metabolismo , Estrés Psicológico/patología , Estimulación Acústica , Adaptación Ocular/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Citalopram/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/fisiología , Suspensión Trasera , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Reflejo de Sobresalto , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Factores Sexuales , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/fisiopatología
4.
Nat Commun ; 15(1): 7900, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261485

RESUMEN

Semen quality and fertility has declined over the last 50 years, corresponding to ever-increasing environmental stressors. However, the cellular mechanisms involved and their impact on sperm functions remain unknown. In a repeated sampling human cohort study, we identify a significant effect of prior perceived stress to increase sperm motility 2-3 months following stress, timing that expands upon our previous studies revealing significant stress-associated changes in sperm RNA important for fertility. We mechanistically examine this post-stress timing in mice using an in vitro stress model in the epididymal epithelial cells responsible for sperm maturation and find 7282 differentially H3K27me3 bound DNA regions involving genes critical for mitochondrial and metabolic pathways. Further, prior stress exposure significantly changes the composition and size of epithelial cell-secreted extracellular vesicles that when incubated with mouse sperm, increase mitochondrial respiration and sperm motility, adding to our prior work showing impacts on embryo development. Together, these studies identify a time-dependent, translational signaling pathway that communicates stress experience to sperm, ultimately affecting reproductive functions.


Asunto(s)
Mitocondrias , Motilidad Espermática , Espermatozoides , Animales , Masculino , Motilidad Espermática/fisiología , Espermatozoides/metabolismo , Espermatozoides/fisiología , Humanos , Ratones , Mitocondrias/metabolismo , Respiración de la Célula , Vesículas Extracelulares/metabolismo , Epidídimo/metabolismo , Ratones Endogámicos C57BL , Estrés Fisiológico , Adulto , Células Epiteliales/metabolismo , Análisis de Semen , Estudios de Cohortes
5.
Sci Rep ; 13(1): 707, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639735

RESUMEN

Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of specificity in transmitting signals and cargo to coordinate local and distant cellular functions. A key example of this is the essential role that EVs secreted by epithelial cells lining the lumen of the male reproductive tract play in post-spermatogenic sperm maturation. We recently showed in a preclinical mouse model that this fundamental process had a causal role in somatic-to-germline transmission of biological information regarding prior stress experience capable of altering the rate of fetal development. However, critical mechanistic questions remain unanswered as to the processes by which signaling occurs between EVs and sperm, and whether EVs or their cargo are delivered at conception and are detectable in the early embryo. Unfortunately, notable methodological limitations shared across EV biology, particularly in the isolation and labeling of EVs, complicate efforts to answer these important questions as well as questions on EV targeting specificity and mechanisms. In our current studies, we developed a novel approach to track EVs using a conditional transgenic construct designed to label EVs via conditional Cre-induced hemagglutinin (HA) tagging of the EV endogenous tetraspanin, CD63. In our exhaustive validation steps, this internal small molecular weight tag did not affect EV secretion or functionality, a common problem found in the previous design of EV tags using larger molecular weight proteins, including fluorescent proteins. Utilizing a stably transfected immortalized epididymal epithelial cell line, we first validated key parameters of the conditional HA-tagged protein packaged into secreted EVs. Importantly, we systematically confirmed that expression of the CD63-HA had no impact on the production, size distribution, or surface charge of secreted EVs, nor did it alter the tetraspanin or miRNA composition of these EVs. We also utilized the CD63-HA EVs to verify physical interactions with sperm. Finally, using in vitro fertilization we produced some of the first images confirming sperm delivered EV cargo at conception and still detectable in the early-stage embryo. As such, this construct serves as a methodological advance and as a valuable tool, with applications in the study of EV function across biomedical research areas.


Asunto(s)
Vesículas Extracelulares , Hemaglutininas , Masculino , Animales , Ratones , Hemaglutininas/metabolismo , Semen , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Animales Modificados Genéticamente , Tetraspaninas/metabolismo , Espermatozoides
6.
J Neurosci ; 31(33): 11748-55, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21849535

RESUMEN

Studies have linked sex-biased neurodevelopmental disorders, including autism and schizophrenia, with fetal antecedents such as prenatal stress. Further, these outcomes can persist into subsequent generations, raising the possibility that aspects of heritability in these diseases involve epigenetic mechanisms. Utilizing a mouse model in which we previously identified a period in early gestation when stress results in dysmasculinized and stress-sensitive male offspring, we have examined programming effects in second-generation offspring of prenatally stressed (F2-S) or control (F2-C) sires. Examination of gene expression patterns during the perinatal sensitive period, when organizational gonadal hormones establish the sexually dimorphic brain, confirmed dysmasculinization in F2-S males, where genes important in neurodevelopment showed a female-like pattern. Analyses of the epigenomic miRNA environment detected significant reductions in miR-322, miR-574, and miR-873 in the F2-S male brain, levels that were again more similar to those of control females. Increased expression of a common gene target for these three miRNAs, ß-glycan, was confirmed in these males. These developmental effects were associated with the transmission of a stress-sensitive phenotype and shortened anogenital distance in adult F2-S males. As confirmation that the miRNA environment is responsive to organizational testosterone, neonatal males administered the aromatase inhibitor formestane exhibited dramatic changes in brain miRNA patterns, suggesting that miRNAs may serve a previously unappreciated role in organizing the sexually dimorphic brain. Overall, these data support the existence of a sensitive period of early gestation when epigenetic programming of the male germline can occur, permitting transmission of specific phenotypes into subsequent generations.


Asunto(s)
Epigenómica/métodos , MicroARNs/genética , Exposición Paterna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/psicología , Caracteres Sexuales , Estrés Psicológico/genética , Estrés Psicológico/psicología , Factores de Edad , Animales , Femenino , Zorros , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Embarazo , Distribución Aleatoria , Estrés Psicológico/complicaciones
7.
Horm Behav ; 59(3): 290-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20483359

RESUMEN

Prenatal programming of the epigenome is a critical determinant in offspring outcome and stands at the interface between environment and genetics. Maternal experiences such as stress and obesity are associated with a host of neurodevelopmental and metabolic diseases, some of which have been characterized into the second and third generations. The mechanism through which determinants such as maternal diet or stress contribute to disease development likely involves a complex interaction between the maternal environment, placental changes, and epigenetic programming of the embryo. While we have begun to more fully appreciate and explore the epigenome in determination of disease risk, we know little as to the contribution embryo sex makes in epigenetic regulation. This review discusses the importance of sex differences in the transmission and inheritance of traits that are generated in the prenatal environment using models of maternal stress and diet.


Asunto(s)
Epigénesis Genética/fisiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Caracteres Sexuales , Estrés Fisiológico/fisiología , Animales , Femenino , Masculino , Embarazo , Factores Sexuales
8.
Sci Rep ; 10(1): 17498, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060642

RESUMEN

Epidemiological studies from the last century have drawn strong associations between paternal life experiences and offspring health and disease outcomes. Recent studies have demonstrated sperm small non-coding RNA (sncRNA) populations vary in response to diverse paternal insults. However, for studies in retrospective or prospective human cohorts to identify changes in paternal germ cell epigenetics in association with offspring disease risk, a framework must first be built with insight into the expected biological variation inherent in human populations. In other words, how will we know what to look for if we don't first know what is stable and what is dynamic, and what is consistent within and between men over time? From sperm samples from a 'normative' cohort of healthy human subjects collected repeatedly from each subject over 6 months, 17 healthy male participants met inclusion criteria and completed donations and psychological evaluations of perceived stress monthly. sncRNAs (including miRNA, piRNA, and tRNA) isolated from mature sperm from these samples were subjected to Illumina small RNA sequencing, aligned to subtype-specific reference transcriptomes, and quantified. The repeated measures design allowed us to define both within- and between-subject variation in the expression of 254 miRNA, 194 tRNA, and 937 piRNA in sperm over time. We developed screening criteria to identify a subset of potential environmentally responsive 'dynamic' sperm sncRNA. Implementing complex modeling of the relationships between individual dynamic sncRNA and perceived stress states in these data, we identified 5 miRNA (including let-7f-5p and miR-181a-5p) and 4 tRNA that are responsive to the dynamics of prior stress experience and fit our established mouse model. In the current study, we aligned repeated sampling of human sperm sncRNA expression data with concurrent measures of perceived stress as a novel framework that can now be applied across a range of studies focused on diverse environmental factors able to influence germ cell programming and potentially impact offspring development.


Asunto(s)
ARN Pequeño no Traducido/genética , Espermatozoides/metabolismo , Transcriptoma , Adulto , Estudios de Cohortes , Epigénesis Genética , Humanos , Masculino , MicroARNs/metabolismo , ARN Interferente Pequeño/metabolismo , ARN de Transferencia/metabolismo , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN , Investigación Biomédica Traslacional , Adulto Joven
9.
Nat Commun ; 11(1): 1499, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32198406

RESUMEN

Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of incredible specificity in transmitting signals involved in cellular function, including germ cell maturation. Spermatogenesis occurs in the testes, behind a protective barrier to ensure safeguarding of germline DNA from environmental insults. Following DNA compaction, further sperm maturation occurs in the epididymis. Here, we report reproductive tract EVs transmit information regarding stress in the paternal environment to sperm, potentially altering fetal development. Using intracytoplasmic sperm injection, we found that sperm incubated with EVs collected from stress-treated epididymal epithelial cells produced offspring with altered neurodevelopment and adult stress reactivity. Proteomic and transcriptomic assessment of these EVs showed dramatic changes in protein and miRNA content long after stress treatment had ended, supporting a lasting programmatic change in response to chronic stress. Thus, EVs as a normal process in sperm maturation, can also perform roles in intergenerational transmission of paternal environmental experience.


Asunto(s)
Vesículas Extracelulares/metabolismo , Sistema Nervioso/crecimiento & desarrollo , Proteómica , Reproducción/fisiología , Adolescente , Animales , Técnicas de Cultivo de Célula , Epidídimo/metabolismo , Epigénesis Genética , Epigenómica , Femenino , Células Germinativas , Histonas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Nanopartículas , Maduración del Esperma/genética , Maduración del Esperma/fisiología , Espermatogénesis/genética , Espermatogénesis/fisiología , Espermatozoides/metabolismo , Estrés Fisiológico , Testículo
10.
Mol Genet Metab ; 97(4): 284-91, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19447653

RESUMEN

GM2 gangliosidosis (GM2g) is an inherited neurodegenerative disorder caused by deficiency of lysosomal beta-hexosaminidase A, resulting in accumulation of GM2 ganglioside, principally in the brain. Substrate reduction therapy is currently under investigation as a treatment. The study investigated the pharmacokinetics and safety of miglustat given as single and multiple doses in infantile and juvenile GM2g patients for 6- and 24-months, respectively. Eleven patients with infantile (n = 6) and juvenile (n = 5) GM2g received oral miglustat at 30-200 mg t.i.d. adjusted to the body surface area. Patients underwent pharmacokinetic assessments on day 1 and at month 3. The pharmacokinetics of miglustat were described by a 2-compartmental model with a lag time, median time to maximum concentration of 2.5 h, and terminal half-life of about 10 h. The pharmacokinetics were time-independent, and did not differ between infantile and juvenile cohorts. The accumulation index was 1.7. Among infantile GM2g patients, the major drug-related adverse events (DRAEs) were abdominal discomfort and flatulence. In the juvenile group, however, the major DRAEs observed were diarrhea and weight loss. One juvenile patient developed peripheral neuropathy, and others showed progression of already established neuropathy, which was judged to be part of the natural progression of the disease. Some mild laboratory abnormalities observed were either transient or attributable to concomitant medications. Miglustat showed similar pharmacokinetic parameters in all patients, with no specific difference between infantile and juvenile forms. Miglustat was shown to be a safe drug, with mild to moderate diarrhea, as an age-dependent DRAE, which was controlled by dietary modification.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Gangliosidosis GM2/tratamiento farmacológico , 1-Desoxinojirimicina/efectos adversos , 1-Desoxinojirimicina/farmacocinética , Administración Oral , Adolescente , Adulto , Niño , Preescolar , Diarrea/inducido químicamente , Diarrea/tratamiento farmacológico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Glucosiltransferasas/antagonistas & inhibidores , Humanos , Lactante
11.
Biol Psychiatry ; 85(2): 164-171, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30580777

RESUMEN

Epidemiological studies provide strong evidence for the impact of diverse paternal life experiences on offspring neurodevelopmental disease risk. While these associations are well established, the molecular mechanisms underlying these intergenerational transmissions remain elusive, though recent studies focusing on the influence of paternal experience before conception have implicated germ cell epigenetic programming. Any model accounting for the germline transfer of nongenetic information from sire to offspring must include certain components, such as 1) a vector to carry any signal from the paternal compartment to the maternal reproductive tract and future embryo; 2) a molecular signal, encoded by a paternal experience, to carry this memory and enact downstream responses; and 3) a target cell or tissue to receive the signal and convert it into an effect on embryonic development. We explore the current understanding of the potential processes and candidate factors that may serve as these components. We specifically discuss the growing appreciation for the importance of extracellular vesicles in these processes, beginning with their known role in delivering potential signals, including small RNAs, to sperm, the prototypical vector, during their posttesticular maturation. Finally, we explore the possibility that paternal extracellular vesicles could themselves serve as vectors, delivering signals not only to gametes or the zygote but also to tissues of the maternal reproductive tract to influence fetal development.


Asunto(s)
Desarrollo Embrionario/fisiología , Vesículas Extracelulares/fisiología , Acontecimientos que Cambian la Vida , ARN/fisiología , Espermatozoides/fisiología , Animales , Humanos , Masculino
12.
Biol Sex Differ ; 8(1): 27, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810930

RESUMEN

BACKGROUND: Sexual differentiation of the male brain, and specifically the stress circuitry in the hypothalamus, is primarily driven by estrogen exposure during the perinatal period. Surprisingly, this single hormone promotes diverse programs of sex-specific development that vary widely between different cell types and across the developing male brain. The complexity of this phenomenon suggests that additional layers of gene regulation, including microRNAs (miRNAs), must act downstream of estrogen to mediate this specificity. METHODS: To identify noncanonical mediators of estrogen-dependent sex-specific neural development, we assayed the miRNA complement of the mouse PN2 hypothalamus by microarray following an injection of vehicle or the aromatase inhibitor, formestane. Initially, multivariate analyses were used to test the influence of sex and experimental group on the miRNA environment as a whole. Then, we utilized traditional hypothesis testing to identify individual miRNA with significantly sex-biased expression. Finally, we performed a transcriptome-wide mapping of Argonaute footprints by high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (Ago HITS-CLIP) to empirically characterize targeting relationship between estrogen-responsive miRNAs and their messenger RNA (mRNA) targets. RESULTS: In this study, we demonstrated that the neonatal hypothalamic miRNA environment has robust sex differences and is dynamically responsive to estrogen. Analyses identified 162 individual miRNAs with sex-biased expression, 92 of which were estrogen-responsive. Examining the genomic distribution of these miRNAs, we found three miRNA clusters encoded within a 175-kb region of chromosome 12 that appears to be co-regulated by estrogen, likely acting broadly to alter the epigenetic programming of this locus. Ago HITS-CLIP analysis uncovered novel miRNA-target interactions within prototypical mediators of estrogen-driven sexual differentiation of the brain, including Esr1 and Cyp19a1. Finally, using Gene Ontology annotations and empirically identified miRNA-mRNA connections, we identified a gene network regulated by estrogen-responsive miRNAs that converge on biological processes relevant to sexual differentiation of the brain. CONCLUSIONS: Sexual differentiation of the perinatal brain, and that of stress circuitry in the hypothalamus specifically, seems to be particularly susceptible to environmental programming effects. Integrating miRNA into our conceptualization of factors, directing differentiation of this circuitry could be an informative next step in efforts to understand the complexities behind these processes.


Asunto(s)
Epigénesis Genética , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Caracteres Sexuales , Androstenodiona/análogos & derivados , Androstenodiona/farmacología , Animales , Animales Recién Nacidos , Inhibidores de la Aromatasa/farmacología , Estrógenos/metabolismo , Redes Reguladoras de Genes , Hipotálamo/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Análisis por Micromatrices , Análisis Multivariante , Transcriptoma/fisiología
13.
Biol Sex Differ ; 3(1): 22, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23009289

RESUMEN

Males and females differ widely in morphology, physiology, and behavior leading to disparities in many health outcomes, including sex biases in the prevalence of many neurodevelopmental disorders. However, with the exception of a relatively small number of genes on the Y chromosome, males and females share a common genome. Therefore, sexual differentiation must in large part be a product of the sex biased expression of this shared genetic substrate. microRNAs (miRs) are small non-coding RNAs involved in the post-transcriptional regulation of up to 70% of protein-coding genes. The ability of miRs to regulate such a vast amount of the genome with a high degree of specificity makes them perfectly poised to play a critical role in programming of the sexually dimorphic brain. This review describes those characteristics of miRs that make them particularly amenable to this task, and examines the influences of both the sex chromosome complement as well as gonadal hormones on their regulation. Exploring miRs in the context of sex differences in disease, particularly in sex-biased neurodevelopmental disorders, may provide novel insight into the pathophysiology and potential therapeutic targets in disease treatment and prevention.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA