Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Science ; 384(6698): eadh3707, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38781393

RESUMEN

The molecular pathology of stress-related disorders remains elusive. Our brain multiregion, multiomic study of posttraumatic stress disorder (PTSD) and major depressive disorder (MDD) included the central nucleus of the amygdala, hippocampal dentate gyrus, and medial prefrontal cortex (mPFC). Genes and exons within the mPFC carried most disease signals replicated across two independent cohorts. Pathways pointed to immune function, neuronal and synaptic regulation, and stress hormones. Multiomic factor and gene network analyses provided the underlying genomic structure. Single nucleus RNA sequencing in dorsolateral PFC revealed dysregulated (stress-related) signals in neuronal and non-neuronal cell types. Analyses of brain-blood intersections in >50,000 UK Biobank participants were conducted along with fine-mapping of the results of PTSD and MDD genome-wide association studies to distinguish risk from disease processes. Our data suggest shared and distinct molecular pathology in both disorders and propose potential therapeutic targets and biomarkers.


Asunto(s)
Trastorno Depresivo Mayor , Estudio de Asociación del Genoma Completo , Corteza Prefrontal , Trastornos por Estrés Postraumático , Biología de Sistemas , Humanos , Trastorno Depresivo Mayor/genética , Trastornos por Estrés Postraumático/genética , Corteza Prefrontal/metabolismo , Masculino , Encéfalo , Femenino , Adulto , Redes Reguladoras de Genes , Persona de Mediana Edad , Neuronas/metabolismo , Biomarcadores/sangre , Amígdala del Cerebelo
2.
Am J Psychiatry ; 180(10): 739-754, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37491937

RESUMEN

OBJECTIVE: Multidisciplinary studies of posttraumatic stress disorder (PTSD) and major depressive disorder (MDD) implicate the dorsolateral prefrontal cortex (DLPFC) in disease risk and pathophysiology. Postmortem brain studies have relied on bulk-tissue RNA sequencing (RNA-seq), but single-cell RNA-seq is needed to dissect cell-type-specific mechanisms. The authors conducted the first single-nucleus RNA-seq postmortem brain study in PTSD to elucidate disease transcriptomic pathology with cell-type-specific resolution. METHOD: Profiling of 32 DLPFC samples from 11 individuals with PTSD, 10 with MDD, and 11 control subjects was conducted (∼415K nuclei; >13K cells per sample). A replication sample included 15 DLPFC samples (∼160K nuclei; >11K cells per sample). RESULTS: Differential gene expression analyses identified significant single-nucleus RNA-seq differentially expressed genes (snDEGs) in excitatory (EX) and inhibitory (IN) neurons and astrocytes, but not in other cell types or bulk tissue. MDD samples had more false discovery rate-corrected significant snDEGs, and PTSD samples had a greater replication rate. In EX and IN neurons, biological pathways that were differentially enriched in PTSD compared with MDD included glucocorticoid signaling. Furthermore, glucocorticoid signaling in induced pluripotent stem cell (iPSC)-derived cortical neurons demonstrated greater relevance in PTSD and opposite direction of regulation compared with MDD, especially in EX neurons. Many snDEGs were from the 17q21.31 locus and are particularly interesting given causal roles in disease pathogenesis and DLPFC-based neuroimaging (PTSD: ARL17B, LINC02210-CRHR1, and LRRC37A2; MDD: LRRC37A and LRP4), while others were regulated by glucocorticoids in iPSC-derived neurons (PTSD: SLC16A6, TAF1C; MDD: CDH3). CONCLUSIONS: The study findings point to cell-type-specific mechanisms of brain stress response in PTSD and MDD, highlighting the importance of examining cell-type-specific gene expression and indicating promising novel biomarkers and therapeutic targets.


Asunto(s)
Trastorno Depresivo Mayor , Trastornos por Estrés Postraumático , Humanos , Corteza Prefontal Dorsolateral , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/metabolismo , Trastornos por Estrés Postraumático/genética , Glucocorticoides/metabolismo , Perfilación de la Expresión Génica , Transcriptoma/genética , Neuronas/metabolismo , Corteza Prefrontal/metabolismo
3.
Neurobiol Stress ; 23: 100530, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36891528

RESUMEN

Hypothalamic-pituitary adrenal (HPA)axis dysregulation has long been implicated in stress-related disorders such as major depression and post-traumatic stress disorder. Glucocorticoids (GCs) are released from the adrenal glands as a result of HPA-axis activation. The release of GCs is implicated with several neurobiological changes that are associated with negative consequences of chronic stress and the onset and course of psychiatric disorders. Investigating the underlying neurobiological effects of GCs may help to better understand the pathophysiology of stress-related psychiatric disorders. GCs impact a plethora of neuronal processes at the genetic, epigenetic, cellular, and molecular levels. Given the scarcity and difficulty in accessing human brain samples, 2D and 3D in vitro neuronal cultures are becoming increasingly useful in studying GC effects. In this review, we provide an overview of in vitro studies investigating the effects of GCs on key neuronal processes such as proliferation and survival of progenitor cells, neurogenesis, synaptic plasticity, neuronal activity, inflammation, genetic vulnerability, and epigenetic alterations. Finally, we discuss the challenges in the field and offer suggestions for improving the use of in vitro models to investigate GC effects.

5.
Int J Mol Sci ; 22(19)2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34639084

RESUMEN

Exposure to trauma is one of the most important and prevalent risk factors for mental and physical ill-health. Excessive or prolonged stress exposure increases the risk of a wide variety of mental and physical symptoms. However, people differ strikingly in their susceptibility to develop signs and symptoms of mental illness after traumatic stress. Post-traumatic stress disorder (PTSD) is a debilitating disorder affecting approximately 8% of the world's population during their lifetime, and typically develops after exposure to a traumatic event. Despite that exposure to potentially traumatizing events occurs in a large proportion of the general population, about 80-90% of trauma-exposed individuals do not develop PTSD, suggesting an inter-individual difference in vulnerability to PTSD. While the biological mechanisms underlying this differential susceptibility are unknown, epigenetic changes have been proposed to underlie the relationship between exposure to traumatic stress and the susceptibility to develop PTSD. Epigenetic mechanisms refer to environmentally sensitive modifications to DNA and RNA molecules that regulate gene transcription without altering the genetic sequence itself. In this review, we provide an overview of various molecular biological, biochemical and physiological alterations in PTSD, focusing on changes at the genomic and epigenomic level. Finally, we will discuss how current knowledge may aid us in early detection and improved management of PTSD patients.


Asunto(s)
Epigénesis Genética , Epigenómica , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/patología , Humanos , Factores de Riesgo
6.
Clin Epigenetics ; 12(1): 11, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31931860

RESUMEN

BACKGROUND: Epigenetic mechanisms have been suggested to play a role in the development of post-traumatic stress disorder (PTSD). Here, blood-derived DNA methylation data (HumanMethylation450 BeadChip) collected prior to and following combat exposure in three cohorts of male military members were analyzed to assess whether DNA methylation profiles are associated with the development of PTSD. A total of 123 PTSD cases and 143 trauma-exposed controls were included in the analyses. The Psychiatric Genomics Consortium (PGC) PTSD EWAS QC pipeline was used on all cohorts, and results were combined using a sample size weighted meta-analysis in a two-stage design. In stage one, we jointly analyzed data of two new cohorts (N = 126 and 78) for gene discovery, and sought to replicate significant findings in a third, previously published cohort (N = 62) to assess the robustness of our results. In stage 2, we aimed at maximizing power for gene discovery by combining all three cohorts in a meta-analysis. RESULTS: Stage 1 analyses identified four CpG sites in which, conditional on pre-deployment DNA methylation, post-deployment DNA methylation was significantly associated with PTSD status after epigenome-wide adjustment for multiple comparisons. The most significant (intergenic) CpG cg05656210 (p = 1.0 × 10-08) was located on 5q31 and significantly replicated in the third cohort. In addition, 19 differentially methylated regions (DMRs) were identified, but failed replication. Stage 2 analyses identified three epigenome-wide significant CpGs, the intergenic CpG cg05656210 and two additional CpGs located in MAD1L1 (cg12169700) and HEXDC (cg20756026). Interestingly, cg12169700 had an underlying single nucleotide polymorphism (SNP) which was located within the same LD block as a recently identified PTSD-associated SNP in MAD1L1. Stage 2 analyses further identified 12 significant differential methylated regions (DMRs), 1 of which was located in MAD1L1 and 4 were situated in the human leukocyte antigen (HLA) region. CONCLUSIONS: This study suggests that the development of combat-related PTSD is associated with distinct methylation patterns in several genomic positions and regions. Our most prominent findings suggest the involvement of the immune system through the HLA region and HEXDC, and MAD1L1 which was previously associated with PTSD.


Asunto(s)
Islas de CpG/genética , Epigénesis Genética/genética , Personal Militar/psicología , Trastornos por Estrés Postraumático/genética , Adulto , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Estudios de Cohortes , Metilación de ADN/genética , Epigenoma/genética , Genómica/métodos , Humanos , Estudios Longitudinales , Masculino , Polimorfismo de Nucleótido Simple/genética , Trastornos por Estrés Postraumático/etnología
7.
Mol Psychiatry ; 25(5): 965-976, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31142820

RESUMEN

Disruption of persistent, stress-associated memories is relevant for treating posttraumatic stress disorder (PTSD) and related syndromes, which develop in a subset of individuals following a traumatic event. We previously developed a stress-enhanced fear learning (SEFL) paradigm in inbred mice that produces PTSD-like characteristics in a subset of mice, including persistently enhanced memory and heightened cFos in the basolateral amygdala complex (BLC) with retrieval of the remote (30-day-old) stress memory. Here, the contribution of BLC microRNAs (miRNAs) to stress-enhanced memory was investigated because of the molecular complexity they achieve through their ability to regulate multiple targets simultaneously. We performed small-RNA sequencing (smRNA-Seq) and quantitative proteomics on BLC tissue collected from mice 1 month after SEFL and identified persistently changed microRNAs, including mir-135b-5p, and proteins associated with PTSD-like heightened fear expression. Viral-mediated overexpression of mir-135b-5p in the BLC of stress-resilient animals enhanced remote fear memory expression and promoted spontaneous renewal 14 days after extinction. Conversely, inhibition of BLC mir-135b-5p in stress-susceptible animals had the opposite effect, promoting a resilient-like phenotype. mir-135b-5p is highly conserved across mammals and was detected in post mortem human amygdala, as well as human serum samples. The mir-135b passenger strand, mir-135b-3p, was significantly elevated in serum from PTSD military veterans, relative to combat-exposed control subjects. Thus, miR-135b-5p may be an important therapeutic target for dampening persistent, stress-enhanced memory and its passenger strand a potential biomarker for responsivity to a mir-135-based therapeutic.


Asunto(s)
Miedo/fisiología , Memoria/fisiología , MicroARNs/genética , Animales , Complejo Nuclear Basolateral/fisiología , Femenino , Humanos , Masculino , Ratones , MicroARNs/análisis , MicroARNs/sangre
9.
Front Genet ; 10: 1042, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31824554

RESUMEN

Posttraumatic stress disorder (PTSD) is a psychiatric disorder that can develop upon exposure to a traumatic event. While most people are able to recover promptly, others are at increased risk of developing PTSD. However, the exact underlying biological mechanisms of differential susceptibility are unknown. Identifying biomarkers of PTSD could assist in its diagnosis and facilitate treatment planning. Here, we identified serum microRNAs (miRNAs) of subjects that underwent a traumatic event and aimed to assess their potential to serve as diagnostic biomarkers of PTSD. Next-generation sequencing was performed to examine circulating miRNA profiles of 24 members belonging to the Dutch military cohort Prospective Research in Stress-Related Military Operations (PRISMO). Three groups were selected: "susceptible" subjects who developed PTSD after combat exposure, "resilient" subjects without PTSD, and nonexposed control subjects (N = 8 per group). Differential expression analysis revealed 22 differentially expressed miRNAs in PTSD subjects compared to controls and 1 in PTSD subjects compared to resilient individuals (after multiple testing correction and a log2 fold-change cutoff of ≥|1|). Weighted Gene Coexpression Network Analysis (WGCNA) identified a module of coexpressed miRNAs which could distinguish between the three groups. In addition, receiver operating characteristic curve analyses suggest that the miRNAs with the highest module memberships could have a strong diagnostic accuracy as reflected by high areas under the curves. Overall, the results of our pilot study suggest that serum miRNAs could potentially serve as diagnostic biomarkers of PTSD, both individually or grouped within a cluster of coexpressed miRNAs. Larger studies are now needed to validate and build upon these preliminary findings.

10.
Twin Res Hum Genet ; 22(6): 460-466, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31708010

RESUMEN

Meta-analyses suggest that clinical psychopathology is preceded by dimensional behavioral and cognitive phenotypes such as psychotic experiences, executive functioning, working memory and affective dysregulation that are determined by the interplay between genetic and nongenetic factors contributing to the severity of psychopathology. The liability to mental ill health can be psychometrically measured using experimental paradigms that assess neurocognitive processes such as salience attribution, sensitivity to social defeat and reward sensitivity. Here, we describe the TwinssCan, a longitudinal general population twin cohort, which comprises 1202 individuals (796 adolescent/young adult twins, 43 siblings and 363 parents) at baseline. The TwinssCan is part of the European Network of National Networks studying Gene-Environment Interactions in Schizophrenia project and recruited from the East Flanders Prospective Twin Survey. The main objective of this project is to understand psychopathology by evaluating the contribution of genetic and nongenetic factors on subclinical expressions of dimensional phenotypes at a young age before the onset of disorder and their association with neurocognitive processes, such as salience attribution, sensitivity to social defeat and reward sensitivity.


Asunto(s)
Trastorno Depresivo/epidemiología , Enfermedades en Gemelos/epidemiología , Interacción Gen-Ambiente , Trastornos Neurocognitivos/epidemiología , Gemelos Dicigóticos/genética , Gemelos Monocigóticos/genética , Adolescente , Adulto , Bélgica/epidemiología , Trastorno Depresivo/genética , Trastorno Depresivo/patología , Enfermedades en Gemelos/genética , Enfermedades en Gemelos/patología , Femenino , Humanos , Incidencia , Estudios Longitudinales , Masculino , Trastornos Neurocognitivos/genética , Trastornos Neurocognitivos/patología , Estudios Prospectivos , Factores Protectores , Factores de Riesgo , Medio Social , Adulto Joven
11.
Psychoneuroendocrinology ; 100: 237-245, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30390522

RESUMEN

BACKGROUND: Acute and chronic stress can lead to a dysregulation of the immune response. Growing evidence suggests peripheral immune dysregulation and low-grade systemic inflammation in posttraumatic stress disorder (PTSD), with numerous reports of elevated plasma interleukin-6 (IL-6) levels. However, only a few studies have assessed IL-6 levels in the cerebrospinal fluid (CSF). Most of those have used single time-point measurements, and thus cannot take circadian level variability and CSF-plasma IL-6 correlations into account. METHODS: This study used time-matched, sequential 24-h plasma and CSF measurements to investigate the effects of combat stress and PTSD on physiologic levels and biorhythmicity of IL-6 in 35 male study volunteers, divided in 3 groups: (PTSD = 12, combat controls, CC = 12, and non-deployed healthy controls, HC = 11). RESULTS: Our findings show no differences in diurnal mean concentrations of plasma and CSF IL-6 across the three comparison groups. However, a significantly blunted circadian rhythm of plasma IL-6 across 24 h was observed in all combat-zone deployed participants, with or without PTSD, in comparison to HC. CSF IL-6 rhythmicity was unaffected by combat deployment or PTSD. CONCLUSIONS: Although no significant group differences in mean IL-6 concentration in either CSF or plasma over a 24-h timeframe was observed, we provide first evidence for a disrupted peripheral IL-6 circadian rhythm as a sequel of combat deployment, with this disruption occurring in both PTSD and CC groups. The plasma IL-6 circadian blunting remains to be replicated and its cause elucidated in future research.


Asunto(s)
Ritmo Circadiano/fisiología , Trastornos de Combate/sangre , Trastornos de Combate/líquido cefalorraquídeo , Interleucina-6 , Trastornos por Estrés Postraumático/sangre , Trastornos por Estrés Postraumático/líquido cefalorraquídeo , Adulto , Estudios de Casos y Controles , Trastornos de Combate/psicología , Humanos , Interleucina-6/análisis , Interleucina-6/sangre , Interleucina-6/líquido cefalorraquídeo , Masculino , Personal Militar/psicología , Veteranos/psicología , Adulto Joven
12.
Prog Mol Biol Transl Sci ; 158: 129-157, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30072051

RESUMEN

The Wnt signaling pathway has been recognized as an important pathway, extending its function throughout the lifespan. Evidence suggests that dysfunctional Wnt signaling in the adult brain leads to aberrant neurogenesis, synaptogenesis, modulation of mature synapses and neurotransmitter release in the hippocampus. Due to the involvement of Wnt proteins in hippocampal functioning, altered Wnt signaling has been suggested to be an important factor in the pathophysiology of mood disorders. Interestingly, the effects of mood-stabilizing drugs are believed to work through interactions with Wnt molecules, and epigenetic mechanisms have been shown to interact with components of the Wnt pathway and impact mechanisms such as synaptic plasticity. This can affect learning and memory formation, in addition to various behavioral outcomes in individuals, when they are faced with stressful or conflict situations. This review will discuss the integrated role of Wnt signaling in the context of appropriate stress response, which is believed to be mediated by adult hippocampal neurogenesis and plasticity. Current knowledge regarding the role of Wnt signaling in mood disorders and antidepressant medication effect will be covered. Finally, the interplay between Wnt signaling and epigenetic mechanisms will be discussed along with their combined potential to impact neuroplasticity.


Asunto(s)
Epigénesis Genética , Hipocampo/metabolismo , Neurogénesis/genética , Plasticidad Neuronal/genética , Estrés Psicológico/genética , Vía de Señalización Wnt/genética , Animales , Humanos
13.
Prog Mol Biol Transl Sci ; 158: 15-27, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30072052

RESUMEN

Over the last years, interest in epigenetic mechanisms has strongly increased in the field of neuroscience. Neuroepigenetics has intensely evolved and now refers to the assessment of a variety of epigenetic marks which can be found across several regions of the healthy or diseased brain. These marks include DNA (hydroxy)methylation, a large diversity of post-translational histone modifications and an increasing number of non-coding RNAs. The present chapter aims to concisely summarize the techniques used to study these mechanisms in the brain and provides an overview of their current challenges along with future perspectives that will allow the field to move forward.


Asunto(s)
Epigénesis Genética , Sistema Nervioso/metabolismo , Proyectos de Investigación , Metilación de ADN/genética , Humanos , Especificidad de Órganos/genética
14.
Front Psychiatry ; 9: 676, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30631285

RESUMEN

Given the high prevalence of stress-related mental disorders, their impact on person, family, and society and the paucity of treatment options for most of these disorders, there is currently a pressing need for innovative approaches to deal with these issues and enhance well-being. One approach which has received increasing attention over the last decade is to shift our scientific and clinical focus from risk factors for psychopathology to factors promoting resilience and mental well-being. In order to summarize and evaluate the current state of scientific affairs on the biological basis of resilience, we provide an overview of the literature on animal and human studies of resilience. Because resilience can only truly be operationalized through longitudinal data collection and analyses, we focus primarily on longitudinal studies. This review shows that the concept of resilience is currently being operationalized, measured and even defined in widely variable manners, both within animal and human studies. We further provide an overview of existing and new strategies that could help promote resilience and which are proposed to be implemented more often in clinical situations. Finally, we summarize the challenges the field is facing and provide recommendations for future research.

15.
Curr Top Behav Neurosci ; 38: 23-46, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29063484

RESUMEN

Post-traumatic stress disorder (PTSD) is a psychiatric disorder that can develop following exposure to or witnessing of a (potentially) threatening event. A critical issue is to pinpoint the (neuro)biological mechanisms underlying the susceptibility to stress-related disorder such as PTSD, which develops in the minority of ~15% of individuals exposed to trauma. Over the last few years, a first wave of epigenetic studies has been performed in an attempt to identify the molecular underpinnings of the long-lasting behavioral and mental effects of trauma exposure. The potential roles of non-coding RNAs (ncRNAs) such as microRNAs (miRNAs) in moderating or mediating the impact of severe stress and trauma are increasingly gaining attention. To date, most studies focusing on the roles of miRNAs in PTSD have, however, been completed in animals, using cross-sectional study designs and focusing almost exclusively on subjects with susceptible phenotypes. Therefore, there is a strong need for new research comprising translational and cross-species approaches that use longitudinal designs for studying trajectories of change contrasting susceptible and resilient subjects. The present review offers a comprehensive overview of available studies of miRNAs in PTSD and discusses the current challenges, pitfalls, and future perspectives of this field.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Estudios Transversales , Epigenómica , MicroARNs , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...