Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(20)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37831056

RESUMO

Compelling epidemiological and animal experimental data demonstrate that cardiometabolic and neuropsychiatric diseases originate in a suboptimal intrauterine environment. Here, we review evidence suggesting that altered placental function may, at least in part, mediate the link between the maternal environment and changes in fetal growth and development. Emerging evidence indicates that the placenta controls the development and function of several fetal tissues through nutrient sensing, modulation of trophoblast nutrient transporters and by altering the number and cargo of released extracellular vesicles. In this Review, we discuss the development and functions of the maternal-placental-fetal interface (in humans and mice) and how cross-talk between these compartments may be a mechanism for in utero programming, focusing on mechanistic target of rapamycin (mTOR), adiponectin and O-GlcNac transferase (OGT) signaling. We also discuss how maternal diet and stress influences fetal development and metabolism and how fetal growth restriction can result in susceptibility to developing chronic disease later in life. Finally, we speculate how interventions targeting placental function may offer unprecedented opportunities to prevent cardiometabolic disease in future generations.


Assuntos
Desenvolvimento Fetal , Placenta , Gravidez , Feminino , Humanos , Camundongos , Animais , Placenta/metabolismo , Trofoblastos/metabolismo , Transdução de Sinais , Retardo do Crescimento Fetal
2.
Biol Reprod ; 110(2): 339-354, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-37971364

RESUMO

Entering pregnancy with a history of adversity, including adverse childhood experiences and racial discrimination stress, is a predictor of negative maternal and fetal health outcomes. Little is known about the biological mechanisms by which preconception adverse experiences are stored and impact future offspring health outcomes. In our maternal preconception stress (MPS) model, female mice underwent chronic stress from postnatal days 28-70 and were mated 2 weeks post-stress. Maternal preconception stress dams blunted the pregnancy-induced shift in the circulating extracellular vesicle proteome and reduced glucose tolerance at mid-gestation, suggesting a shift in pregnancy adaptation. To investigate MPS effects at the maternal:fetal interface, we probed the mid-gestation placental, uterine, and fetal brain tissue transcriptome. Male and female placentas differentially regulated expression of genes involved in growth and metabolic signaling in response to gestation in an MPS dam. We also report novel offspring sex- and MPS-specific responses in the uterine tissue apposing these placentas. In the fetal compartment, MPS female offspring reduced expression of neurodevelopmental genes. Using a ribosome-tagging transgenic approach we detected a dramatic increase in genes involved in chromatin regulation in a PVN-enriched neuronal population in females at PN21. While MPS had an additive effect on high-fat-diet (HFD)-induced weight gain in male offspring, both MPS and HFD were necessary to induce significant weight gain in female offspring. These data highlight the preconception period as a determinant of maternal health in pregnancy and provides novel insights into mechanisms by which maternal stress history impacts offspring developmental programming.


Assuntos
Placenta , Aumento de Peso , Humanos , Gravidez , Camundongos , Feminino , Masculino , Animais , Placenta/metabolismo , Feto/metabolismo , Transdução de Sinais , Dieta Hiperlipídica/efeitos adversos
3.
Front Neuroendocrinol ; 55: 100797, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31574280

RESUMO

The maternal microbiota is positioned to regulate the development of offspring immunity, metabolism, as well as brain function and behavior. The mechanisms by which maternal microbial signals drive these processes are beginning to be elucidated. In this review, we provide a brief overview on the importance of the microbiome in brain function and behavior, define the maternal vaginal and gut microbiota as distinct influences on offspring development, and outline current concepts in microbial origins of offspring health outcomes. We propose that the maternal microbiota influences prenatal and early postnatal offspring development and health outcomes through two overlapping processes. First, during pregnancy maternal gut microbiota provide metabolites and substrates essential for fetal growth through metabolic provisioning, driving expansion and maturation of central and peripheral immune cells, and formation of neural circuits. Second, vertical transmission of maternal microbiota during birth and in the early postnatal window elicits a potent immunostimulatory effect in offspring that induces metabolic and developmental transcriptional programs, primes the immune system for subsequent microbial exposure, and provides substrates for brain metabolism. Finally, we explore the possibility that environmental factors, such as malnutrition, stress and infection, may exert programmatic effects by disrupting the functional contributions of the maternal microbiome during prenatal and postnatal development to influence offspring outcomes across the lifespan.


Assuntos
Encéfalo , Desenvolvimento Infantil/fisiologia , Desenvolvimento Fetal/fisiologia , Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Vagina/microbiologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/imunologia , Encéfalo/metabolismo , Feminino , Desenvolvimento Fetal/imunologia , Microbioma Gastrointestinal/imunologia , Humanos , Recém-Nascido , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/microbiologia
4.
Nat Rev Neurosci ; 16(6): 332-44, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25921815

RESUMO

Neurodevelopmental programming - the implementation of the genetic and epigenetic blueprints that guide and coordinate normal brain development - requires tight regulation of transcriptional processes. During prenatal and postnatal time periods, epigenetic processes fine-tune neurodevelopment towards an end product that determines how an organism interacts with and responds to exposures and experiences throughout life. Epigenetic processes also have the ability to reprogramme the epigenome in response to environmental challenges, such as maternal stress, making the organism more or less adaptive depending on the future challenges presented. Epigenetic marks generated within germ cells as a result of environmental influences throughout life can also shape future generations long before conception occurs.


Assuntos
Encéfalo/embriologia , Reprogramação Celular/fisiologia , Epigênese Genética/fisiologia , Neurogênese/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Humanos , Gravidez
5.
Proc Natl Acad Sci U S A ; 112(44): 13699-704, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26483456

RESUMO

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.


Assuntos
Epigênese Genética , MicroRNAs/genética , Exposição Paterna , Espermatozoides/metabolismo , Estresse Fisiológico , Animais , Sistema Hipotálamo-Hipofisário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
7.
J Physiol ; 600(20): 4417-4418, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36190177
8.
Proc Natl Acad Sci U S A ; 111(26): 9639-44, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24979775

RESUMO

Maternal stress is a key risk factor in neurodevelopmental disorders, which often have a sex bias in severity and prevalence. We previously identified O-GlcNAc transferase (OGT) as a placental biomarker in our mouse model of early prenatal stress (EPS), where OGT levels were lower in male compared with female tissue and were further decreased following maternal stress. However, the function of placental OGT in programming the developing brain has not been determined. Therefore, we generated a transgenic mouse with targeted placental disruption of Ogt (Pl-OGT) and examined offspring for recapitulation of the adult EPS phenotype. Pl-OGT hemizygous and EPS male placentas showed similar robust changes in gene expression patterns suggestive of an altered ability to respond to endocrine and inflammatory signals, supporting placental OGT as an important mediator of EPS effects. ChIP-Seq for the O-GlcNAc mark identified the 17 beta hydroxysteroid dehydrogenase-3 (Hsd17b3) locus in male EPS placentas, which correlated with a reduction in Hsd17b3 expression and concordant reduced testosterone conversion. Remarkably, Pl-OGT adult offspring had reduced body weights and elevated hypothalamic-pituitary-adrenal stress axis responsivity, recapitulating phenotypes previously reported for EPS males. Further, hypothalamic microarray gene-set enrichment analyses identified reduced mitochondrial function in both Pl-OGT and EPS males. Cytochrome c oxidase activity assays verified this finding, linking reduced placental OGT with critical brain programming. Together, these studies confirm OGT as in important placental biomarker of maternal stress and demonstrate the profound impact a single placental gene has on long-term metabolic and neurodevelopmental programming that may be related to an increased risk for neurodevelopmental disorders.


Assuntos
Doenças Fetais/fisiopatologia , N-Acetilglucosaminiltransferases/metabolismo , Placenta/metabolismo , Estresse Fisiológico/fisiologia , Análise de Variância , Animais , Biomarcadores/metabolismo , Imunoprecipitação da Cromatina , Feminino , Doenças Fetais/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Doenças Mitocondriais/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
9.
Front Neuroendocrinol ; 39: 28-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26368654

RESUMO

Fetal development could be considered a sensitive period wherein exogenous insults and changes to the maternal milieu can have long-term impacts on developmental programming. The placenta provides the fetus with protection and necessary nutrients for growth, and responds to maternal cues and changes in nutrient signaling through multiple epigenetic mechanisms. The X-linked enzyme O-linked-N-acetylglucosamine transferase (OGT) acts as a nutrient sensor that modifies numerous proteins to alter various cellular signals, including major epigenetic processes. This review describes epigenetic alterations in the placenta in response to insults during pregnancy, the potential links of OGT as a nutrient sensor to placental epigenetics, and the implications of placental epigenetics in long-term neurodevelopmental programming. We describe the role of placental OGT in the sex-specific programming of hypothalamic-pituitary-adrenal (HPA) axis programming deficits by early prenatal stress as an example of how placental signaling can have long-term effects on neurodevelopment.


Assuntos
Epigênese Genética/fisiologia , Desenvolvimento Fetal/genética , Desenvolvimento Fetal/fisiologia , Feto/metabolismo , Placenta/fisiologia , Adulto , Animais , Feminino , Humanos , Gravidez
10.
Proc Natl Acad Sci U S A ; 110(13): 5169-74, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23487789

RESUMO

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.


Assuntos
N-Acetilglucosaminiltransferases/biossíntese , Placenta/enzimologia , Complicações na Gravidez/enzimologia , Proteínas da Gravidez/biossíntese , Estresse Fisiológico , Animais , Biomarcadores/metabolismo , Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/patologia , Epigênese Genética/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/embriologia , Hipotálamo/enzimologia , Hipotálamo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neurogênese/genética , Placenta/patologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/patologia , Proteínas da Gravidez/genética , Caracteres Sexuais
11.
J Neurosci ; 34(21): 7113-23, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24849347

RESUMO

Impulsivity, risk-taking behavior, and elevated stress responsivity are prominent symptoms of mania, a behavioral state common to schizophrenia and bipolar disorder. Though inflammatory processes activated within the brain are involved in the pathophysiology of both disorders, the specific mechanisms by which neuroinflammation drives manic behavior are not well understood. Serotonin cell bodies originating within the dorsal raphe (DR) play a major role in the regulation of behavioral features characteristic of mania. Therefore, we hypothesized that the link between neuroinflammation and manic behavior may be mediated by actions on serotonergic neurocircuitry. To examine this, we induced local neuroinflammation in the DR by viral delivery of Cre recombinase into interleukin (IL)-1ß(XAT) transgenic male and female mice, resulting in overexpressing of the proinflammatory cytokine, IL-1ß. For assertion of brain-region specificity of these outcomes, the prefrontal cortex (PFC), as a downstream target of DR serotonergic projections, was also infused. Inflammation within the DR, but not the PFC, resulted in a profound display of manic-like behavior, characterized by increased stress-induced locomotion and responsivity, and reduced risk-aversion/fearfulness. Microarray analysis of the DR revealed a dramatic increase in immune-related genes, and dysregulation of genes important in GABAergic, glutamatergic, and serotonergic neurotransmission. Behavioral and physiological changes were driven by a loss of serotonergic neurons and reduced output as measured by high-performance liquid chromatography, demonstrating inflammation-induced serotonergic hypofunction. Behavioral changes were rescued by acute selective serotonin reuptake inhibitor treatment, supporting the hypothesis that serotonin dysregulation stemming from neuroinflammation in the DR underlies manic-like behaviors.


Assuntos
Encefalite/patologia , Núcleos da Rafe/citologia , Núcleos da Rafe/fisiologia , Neurônios Serotoninérgicos/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Citalopram/farmacologia , Dependovirus/genética , Modelos Animais de Doenças , Encefalite/genética , Encefalite/fisiopatologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Estresse Psicológico/sangue , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/genética
12.
Curr Psychiatry Rep ; 17(2): 5, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25617041

RESUMO

The British epidemiologist Dr. David J. Barker documented the relationship between infant birth weight and later onset of hypertension, coronary heart disease, insulin resistance, and type II diabetes. A stressful in utero environment can cause long-term consequences for offspring through prenatal programming. Prenatal programming most commonly occurs through epigenetic mechanisms and can be dependent on the type and timing of exposure as well as the sex of the fetus. In this review, we highlight the most recent evidence that prenatal programming is implicated in the development of psychiatric disorders in offspring exposed to maternal stress during pregnancy. Methodological differences between studies contribute to unavoidable heterogeneity in study findings. Current data suggest that fetal exposure to maternal hypothalamic-pituitary-adrenal axis dysregulation, excessive glucocorticoids, and inflammation with resulting epigenetic changes at both the placental and fetal levels are important areas of continued investigation.


Assuntos
Transtornos Mentais/etiologia , Complicações na Gravidez/psicologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Animais , Feminino , Humanos , Masculino , Gravidez , Complicações na Gravidez/fisiopatologia , Fatores Sexuais , Estresse Fisiológico/fisiologia , Estresse Psicológico/fisiopatologia
13.
J Neurosci ; 33(17): 7122-9, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616522

RESUMO

Hedonic overconsumption contributing to obesity involves altered activation within the mesolimbic dopamine system. Dysregulation of dopamine signaling in the nucleus accumbens shell (NAS) has been implicated in reward-seeking behaviors, such as binge eating, which contributes to treatment resistance in obesity (Wise, 2012). Direct modulation of the NAS with deep brain stimulation (DBS), a surgical procedure currently under investigation in humans for the treatment of major depression, obsessive-compulsive disorder, and addiction, may also be effective in ameliorating binge eating. Therefore, we examined the ability of DBS of the NAS to block this behavior in mice. c-Fos immunoreactivity was assessed as a marker of DBS-mediated neuronal activation. NAS DBS was found to reduce binge eating and increased c-Fos levels in this region. DBS of the dorsal striatum had no influence on this behavior, demonstrating anatomical specificity for this effect. The dopamine D2 receptor antagonist, raclopride, attenuated the action of DBS, whereas the D1 receptor antagonist, SCH-23390, was ineffective, suggesting that dopamine signaling involving D2 receptors underlies the effect of NAS DBS. To determine the potential translational relevance to the obese state, chronic NAS DBS was also examined in diet-induced obese mice and was found to acutely reduce caloric intake and induce weight loss. Together, these findings support the involvement of the mesolimbic dopamine pathways in the hedonic mechanisms contributing to obesity, and the efficacy of NAS DBS to modulate this system.


Assuntos
Bulimia/fisiopatologia , Bulimia/terapia , Estimulação Encefálica Profunda/métodos , Núcleo Accumbens/fisiologia , Receptores de Dopamina D2/fisiologia , Animais , Benzazepinas/farmacologia , Antagonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória
14.
J Neurosci ; 33(21): 9003-12, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23699511

RESUMO

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.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiopatologia , MicroRNAs/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Espermatozoides/metabolismo , Estresse Psicológico/patologia , Estimulação Acústica , Adaptação Ocular/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Citalopram/uso terapêutico , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/fisiologia , Elevação dos Membros Posteriores , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Reflexo de Sobressalto , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Fatores Sexuais , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/fisiopatologia
15.
Biol Psychiatry ; 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38821195

RESUMO

BACKGROUND: Adverse childhood experiences (ACEs) increase risk for mental illness in women and their children, and dysregulation of the hypothalamic-pituitary-adrenal axis may play a role. The impact of ACEs on the hypothalamic-pituitary-adrenal axis may be strongest when ACEs occur prepubertally and in people who are exposed to abuse ACEs. METHODS: To test this, we measured salivary cortisol in 96 mother-infant dyads while mothers were separated from their infants, who were experiencing a laboratory stressor. Mothers completed the Adverse Childhood Experiences Questionnaire; ACEs that occurred prepubertally (pACEs) were measured, and mother-infant dyads were grouped based on maternal pACE history as follows: no pACEs, ≥1 pACEs with abuse, or ≥1 pACEs but no abuse. RESULTS: Mothers with ≥1 pACEs exhibited decreases in cortisol (relative to preinfant stressor), which differed significantly from the cortisol increase experienced by mothers with no pACEs, regardless of abuse presence (p = .001) or absence (p = .002). These pACE groups did not differ from one another (p = .929). Significant sex differences in infant cortisol were observed in infants of mothers with ≥1 pACEs (regardless of abuse) but not in infants of mothers with no pACEs. When mothers had experienced ≥1 pACEs, males showed decreases in cortisol in response to a stressor whereas females demonstrated increases, and males and females differed significantly when their mothers had ≥1 pACEs with (p = .025) and without (p = .032) abuse. CONCLUSIONS: Regardless of maternal exposure to childhood abuse, in response to a stressor, pACEs were associated with lower cortisol response in mothers and sex differences in 6-month-old infants, with males showing a lower cortisol response than females.

16.
Neuropsychopharmacology ; 49(2): 443-454, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37833589

RESUMO

Trauma and chronic stress exposure are the strongest predictors of lifetime neuropsychiatric disease presentation. These disorders often have significant sex biases, with females having higher incidences of affective disorders such as major depression, anxiety, and PTSD. Understanding the mechanisms by which stress exposure heightens disease vulnerability is essential for developing novel interventions. Current rodent stress models consist of a battery of sensory, homeostatic, and psychological stressors that are ultimately integrated by corticotropin-releasing factor (CRF) neurons to trigger corticosteroid release. These stress paradigms, however, often differ between research groups in the type, timing, and duration of stressors utilized. These inconsistencies, along with the variability of individual animals' perception and response to each stressor, present challenges for reproducibility and translational relevance. Here, we hypothesized that a more direct approach using chemogenetic activation of CRF neurons would recapitulate the effects of traditional stress paradigms and provide a high-throughput method for examining stress-relevant phenotypes. Using a transgenic approach to express the Gq-coupled Designer Receptor Exclusively Activated by Designer Drugs (DREADD) receptor hM3Dq in CRF-neurons, we found that the DREADD ligand clozapine-N-oxide (CNO) produced an acute and robust activation of the hypothalamic-pituitary-adrenal (HPA) axis, as predicted. Interestingly, chronic treatment with this method of direct CRF activation uncovered a novel sex-specific dissociation of glucocorticoid levels with stress-related outcomes. Despite hM3Dq-expressing females producing greater corticosterone levels in response to CNO than males, hM3Dq-expressing males showed significant typical physiological stress sensitivity with reductions in body and thymus weights. hM3Dq-expressing females while resistant to the physiological effects of chronic CRF activation, showed significant increases in baseline and fear-conditioned freezing behaviors. These data establish a novel mouse model for interrogating stress-relevant phenotypes and highlight sex-specific stress circuitry distinct for physiological and limbic control that may underlie disease risk.


Assuntos
Hormônio Liberador da Corticotropina , Neurônios , Camundongos , Masculino , Animais , Feminino , Hormônio Liberador da Corticotropina/farmacologia , Reprodutibilidade dos Testes , Ansiedade , Medo
17.
Sci Rep ; 13(1): 707, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36639735

RESUMO

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.


Assuntos
Vesículas Extracelulares , Hemaglutininas , Masculino , Animais , Camundongos , Hemaglutininas/metabolismo , Sêmen , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Animais Geneticamente Modificados , Tetraspaninas/metabolismo , Espermatozoides
18.
Sci Rep ; 13(1): 4568, 2023 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-36941297

RESUMO

Homeostatic regulation of the maternal milieu during pregnancy is critical for maternal and fetal health. The placenta facilitates critical communication between maternal and fetal compartments, in part, through the production of extracellular vesicles (EVs). EVs enable tissue synchrony via cell-cell and long-distance communication and are at their highest circulating concentration during pregnancy. While much work has been done investigating how physiological challenges in pregnancy affect the fetus, the role of placental communication in maternal health has not been well examined. We previously identified placental O-glycosyl transferase (OGT), a glucose-sensing enzyme, as a target of maternal stress where OGT levels and activity affected the O-glycosylation of proteins critical for EV cargo loading and secretion. Here, we hypothesized that placental OGT plays an essential role in maternal homeostatic regulation during pregnancy via its regulation of maternal circulating EV concentrations. Our studies found that changes to key metabolic factors over the circadian cycle, including glucocorticoids, insulin, and glucose, were significantly associated with changes in circulating EV concentration. Targeting placental OGT in mice, we found a novel significant positive relationship between placental OGT and maternal circulating EV concentration that was associated with improving maternal glucose tolerance during pregnancy. Finally, an intravenous elevation in EVs, matching the concentration of EVs during pregnancy, shifted non-pregnant female glucose sensitivity, blunted glucose variance, and improved synchrony of glucose uptake. These data suggest an important and novel role for circulating EVs as homeostatic regulators important in maternal health during pregnancy.


Assuntos
Vesículas Extracelulares , Placenta , Gravidez , Feminino , Animais , Camundongos , Placenta/metabolismo , Vesículas Extracelulares/metabolismo , Feto , Glucose/metabolismo , Homeostase
19.
Sci Rep ; 13(1): 21082, 2023 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-38030664

RESUMO

The genetic material encoded on X and Y chromosomes provides the foundation by which biological sex differences are established. Epigenetic regulators expressed on these sex chromosomes, including Kdm6a (Utx), Kdm5c, and Ddx3x have far-reaching impacts on transcriptional control of phenotypic sex differences. Although the functionality of UTY (Kdm6c, the Y-linked homologue of UTX), has been supported by more recent studies, its role in developmental sex differences is not understood. Here we test the hypothesis that UTY is an important transcriptional regulator during development that could contribute to sex-specific phenotypes and disease risks across the lifespan. We generated a random insertion Uty transgenic mouse (Uty-Tg) to overexpress Uty. By comparing transcriptomic profiles in developmental tissues, placenta and hypothalamus, we assessed potential UTY functional activity, comparing Uty-expressing female mice (XX + Uty) with wild-type male (XY) and female (XX) mice. To determine if Uty expression altered physiological or behavioral outcomes, adult mice were phenotypically examined. Uty expression masculinized female gene expression patterns in both the placenta and hypothalamus. Gene ontology (GO) and gene set enrichment analysis (GSEA) consistently identified pathways including immune and synaptic signaling as biological processes associated with UTY. Interestingly, adult females expressing Uty gained less weight and had a greater glucose tolerance compared to wild-type male and female mice when provided a high-fat diet. Utilizing a Uty-overexpressing transgenic mouse, our results provide novel evidence as to a functional transcriptional role for UTY in developing tissues, and a foundation to build on its prospective capacity to influence sex-specific developmental and health outcomes.


Assuntos
Regulação da Expressão Gênica , Transcriptoma , Masculino , Feminino , Animais , Camundongos , Estudos Prospectivos , Perfilação da Expressão Gênica , Camundongos Transgênicos
20.
Biol Sex Differ ; 14(1): 7, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36803442

RESUMO

BACKGROUND: The mechanisms by which parental early life stress can be transmitted to the next generation, in some cases in a sex-specific manner, are unclear. Maternal preconception stress may increase susceptibility to suboptimal health outcomes via in utero programming of the fetal hypothalamic-pituitary-adrenal (HPA) axis. METHODS: We recruited healthy pregnant women (N = 147), dichotomized into low (0 or 1) and high (2+) adverse childhood experience (ACE) groups based on the ACE Questionnaire, to test the hypothesis that maternal ACE history influences fetal adrenal development in a sex-specific manner. At a mean (standard deviation) of 21.5 (1.4) and 29.5 (1.4) weeks gestation, participants underwent three-dimensional ultrasounds to measure fetal adrenal volume, adjusting for fetal body weight (waFAV). RESULTS: At ultrasound 1, waFAV was smaller in high versus low ACE males (b = - 0.17; z = - 3.75; p < .001), but females did not differ significantly by maternal ACE group (b = 0.09; z = 1.72; p = .086). Compared to low ACE males, waFAV was smaller for low (b = - 0.20; z = - 4.10; p < .001) and high ACE females (b = - 0.11; z = 2.16; p = .031); however, high ACE males did not differ from low (b = 0.03; z = .57; p = .570) or high ACE females (b = - 0.06; z = - 1.29; p = .196). At ultrasound 2, waFAV did not differ significantly between any maternal ACE/offspring sex subgroups (ps ≥ .055). Perceived stress did not differ between maternal ACE groups at baseline, ultrasound 1, or ultrasound 2 (ps ≥ .148). CONCLUSIONS: We observed a significant impact of high maternal ACE history on waFAV, a proxy for fetal adrenal development, but only in males. Our observation that the waFAV in males of mothers with a high ACE history did not differ from the waFAV of females extends preclinical research demonstrating a dysmasculinizing effect of gestational stress on a range of offspring outcomes. Future studies investigating intergenerational transmission of stress should consider the influence of maternal preconception stress on offspring outcomes.


Assuntos
Experiências Adversas da Infância , Masculino , Humanos , Feminino , Gravidez , Feto/diagnóstico por imagem , Sistema Hipotálamo-Hipofisário , Idade Gestacional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA