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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 87: 831-839, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32217081

RESUMEN

The prevalence of depression in later life is higher in women than in men. However, the sex difference in the pathophysiology of depression in elderly patients is not fully understood. Here, we performed gene expression profiling in leukocytes of middle-aged and elderly patients with major depressive disorder, termed later-life depression (LLD) in this context, and we characterized the sex-dependent pathophysiology of LLD. A microarray dataset obtained from leukocytes of patients (aged ≥50 years) with LLD (32 males and 39 females) and age-matched healthy individuals (20 males and 24 females) was used. Differentially expressed probes were determined by comparing the expression levels between patients and healthy individuals, and then functional annotation analyses (Ingenuity Pathway Analysis, Reactome pathway analysis, and cell-type enrichment analysis) were performed. A total of 1656 probes were differentially expressed in LLD females, but only 3 genes were differentially expressed in LLD males. The differentially expressed genes in LLD females were relevant to leukocyte extravasation signaling, Tec kinase signaling and the innate immune response. The upregulated genes were relevant to myeloid lineage cells such as CD14+ monocytes. In contrast, the downregulated genes were relevant to CD4+ and CD8+ T cells. Remarkable innate immune signatures are present in the leukocytes of LLD females but not males. Because inflammation is involved in the pathophysiology of depression, the altered inflammatory activity may be involved in the pathophysiology of LLD in women. In contrast, abnormal inflammation may be an uncommon feature in LLD males.


Asunto(s)
Trastorno Depresivo Mayor , Anciano , Linfocitos T CD8-positivos , Trastorno Depresivo Mayor/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Masculino , Análisis por Micromatrices , Persona de Mediana Edad
2.
Psychiatry Clin Neurosci ; 72(4): 212-227, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29154458

RESUMEN

Major depressive disorder is one of the most common mental illnesses as it affects more than 350 million people globally. Major depressive disorder is etiologically complex and disabling. Genetic factors play a role in the etiology of major depression. However, identical twin studies have shown high rates of discordance, indicating non-genetic mechanisms as well. For instance, stressful life events increase the risk of depression. Environmental stressors also induce stable changes in gene expression within the brain that may lead to maladaptive neuronal plasticity in regions implicated in disease pathogenesis. Epigenetic events alter the chromatin structure and thus modulate expression of genes that play a role in neuronal plasticity, behavioral response to stress, depressive behaviors, and response to antidepressants. Here, we review new information regarding current understanding of epigenetic events that may impact depression. In particular, we discuss the roles of histone acetylation, DNA methylation, and non-coding RNA. These novel mechanisms of action may lead to new therapeutic strategies for treating major depression.


Asunto(s)
Trastorno Depresivo Mayor , Epigénesis Genética , Plasticidad Neuronal , Trastorno Depresivo Mayor/etiología , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/metabolismo , Humanos
3.
J Neurosci ; 36(4): 1185-202, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26818507

RESUMEN

Neurogenesis and memory formation are essential features of the dentate gyrus (DG) area of the hippocampus, but to what extent the mechanisms responsible for both processes overlap remains poorly understood. Stathmin protein, whose tubulin-binding and microtubule-destabilizing activity is negatively regulated by its phosphorylation, is prominently expressed in the DG. We show here that stathmin is involved in neurogenesis, spinogenesis, and memory formation in the DG. tTA/tetO-regulated bitransgenic mice, expressing the unphosphorylatable constitutively active Stathmin4A mutant (Stat4A), exhibit impaired adult hippocampal neurogenesis and reduced spine density in the DG granule neurons. Although Stat4A mice display deficient NMDA receptor-dependent memory in contextual discrimination learning, which is dependent on hippocampal neurogenesis, their NMDA receptor-independent memory is normal. Confirming NMDA receptor involvement in the memory deficits, Stat4A mutant mice have a decrease in the level of synaptic NMDA receptors and a reduction in learning-dependent CREB-mediated gene transcription. The deficits in neurogenesis, spinogenesis, and memory in Stat4A mice are not present in mice in which tTA/tetO-dependent transgene transcription is blocked by doxycycline through their life. The memory deficits are also rescued within 3 d by intrahippocampal infusion of doxycycline, further indicating a role for stathmin expressed in the DG in contextual memory. Our findings therefore point to stathmin and microtubules as a mechanistic link between neurogenesis, spinogenesis, and NMDA receptor-dependent memory formation in the DG. SIGNIFICANCE STATEMENT: In the present study, we aimed to clarify the role of stathmin in neuronal and behavioral functions. We characterized the neurogenic, behavioral, and molecular consequences of the gain-of-function stathmin mutation using a bitransgenic mouse expressing a constitutively active form of stathmin. We found that stathmin plays an important role in adult hippocampal neurogenesis and spinogenesis. In addition, stathmin mutation led to impaired NMDA receptor-dependent and neurogenesis-associated memory and did not affect NMDA receptor-independent memory. Moreover, biochemical analysis suggested that stathmin regulates the synaptic transport of NMDA receptors, which in turn influence CREB-mediated gene transcription machinery. Overall, these data suggest that stathmin is an important molecule for neurogenesis, spinogenesis, and NMDA receptor-dependent learning and memory.


Asunto(s)
Espinas Dendríticas/fisiología , Giro Dentado/fisiología , Memoria/fisiología , Neurogénesis/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Factor de Transcripción STAT4/metabolismo , Animales , Proteína de Unión a CREB/metabolismo , Giro Dentado/citología , Aprendizaje Discriminativo/fisiología , Proteínas de Dominio Doblecortina , Conducta Exploratoria/fisiología , Péptido Liberador de Gastrina/genética , Péptido Liberador de Gastrina/metabolismo , Regulación de la Expresión Génica/genética , Masculino , Aprendizaje por Laberinto , Memoria/efectos de los fármacos , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/citología , Neuronas/fisiología , Neuropéptidos/farmacología , Factor de Transcripción STAT4/genética , Fracciones Subcelulares/metabolismo
4.
J Neurosci ; 36(27): 7253-67, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27383599

RESUMEN

UNLABELLED: Chronic stress-induced aberrant gene expression in the brain and subsequent dysfunctional neuronal plasticity have been implicated in the etiology and pathophysiology of mood disorders. In this study, we examined whether altered expression of small, regulatory, noncoding microRNAs (miRNAs) contributes to the depression-like behaviors and aberrant neuronal plasticity associated with chronic stress. Mice exposed to chronic ultra-mild stress (CUMS) exhibited increased depression-like behaviors and reduced hippocampal expression of the brain-enriched miRNA-124 (miR-124). Aberrant behaviors and dysregulated miR-124 expression were blocked by chronic treatment with an antidepressant drug. The depression-like behaviors are likely not conferred directly by miR-124 downregulation because neither viral-mediated hippocampal overexpression nor intrahippocampal infusion of an miR-124 inhibitor affected depression-like behaviors in nonstressed mice. However, viral-mediated miR-124 overexpression in hippocampal neurons conferred behavioral resilience to CUMS, whereas inhibition of miR-124 led to greater behavioral susceptibility to a milder stress paradigm. Moreover, we identified histone deacetylase 4 (HDAC4), HDAC5, and glycogen synthase kinase 3ß (GSK3ß) as targets for miR-124 and found that intrahippocampal infusion of a selective HDAC4/5 inhibitor or GSK3 inhibitor had antidepressant-like actions on behavior. We propose that miR-124-mediated posttranscriptional controls of HDAC4/5 and GSK3ß expressions in the hippocampus have pivotal roles in susceptibility/resilience to chronic stress. SIGNIFICANCE STATEMENT: Depressive disorders are a major public health concern worldwide. Although a clear understanding of the etiology of depression is still lacking, chronic stress-elicited aberrant neuronal plasticity has been implicated in the pathophysiology of depression. We show that the hippocampal expression of microRNA-124 (miR-124), an endogenous small, noncoding RNA that represses gene expression posttranscriptionally, controls resilience/susceptibility to chronic stress-induced depression-like behaviors. These effects on depression-like behaviors may be mediated through regulation of the mRNA or protein expression levels of histone deacetylases HDAC4/5 and glycogen synthase kinase 3ß, all highly conserved miR-124 targets. Moreover, miR-124 contributes to stress-induced dendritic hypotrophy and reduced spine density of dentate gyrus granule neurons. Modulation of hippocampal miR-124 pathways may have potential antidepressant effects.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , MicroARNs/metabolismo , Estrés Psicológico/fisiopatología , Animales , Antidepresivos/uso terapéutico , Depresión/etiología , Modelos Animales de Enfermedad , Fluoroquinolonas/farmacología , Preferencias Alimentarias/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Histona Desacetilasas/metabolismo , Imipramina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/ultraestructura , Oligodesoxirribonucleótidos Antisentido/farmacología , Inhibidores de Topoisomerasa II/farmacología
5.
Neurobiol Learn Mem ; 124: 52-61, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26211874

RESUMEN

Microtubules, one of the major cytoskeletal structures, were previously considered stable and only indirectly involved in synaptic structure and function in mature neurons. However, recent evidence demonstrates that microtubules are dynamic and have an important role in synaptic structure, synaptic plasticity, and memory. In particular, learning induces changes in microtubule turnover and stability, and pharmacological manipulation of microtubule dynamics alters synaptic plasticity and long-term memory. These learning-induced changes in microtubules are controlled by the phosphoprotein stathmin, whose only known cellular activity is to negatively regulate microtubule formation. During the first eight hours following learning, changes in the phosphorylation of stathmin go through two phases causing biphasic shifts in microtubules stability/instability. These shifts, in turn, regulate memory formation by controlling in the second phase synaptic transport of the GluA2 subunit of AMPA receptors. Improper regulation of stathmin and microtubule dynamics has been observed in aged animals and in patients with Alzheimer's disease and depression. Thus, recent work on stathmin and microtubules has identified new molecular players in the early stages of memory encoding.


Asunto(s)
Aprendizaje/fisiología , Memoria/fisiología , Microtúbulos/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Estatmina/metabolismo , Sinapsis/metabolismo , Animales , Humanos , Cinesinas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(4): 1681-6, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21205885

RESUMEN

Aberrant transcriptional regulation in the brain is thought to be one of the key components of the pathogenesis and pathophysiology of neuropsychiatric disorders. Heat shock factors (HSFs) modulate cellular homeostasis through the control of gene expression. However, the roles of HSFs in brain function have yet to be elucidated fully. In the present study, we attempted to clarify the role of HSF1-mediated gene regulation in neuronal and behavioral development using HSF1-deficient (HSF1(-/-)) mice. We found granule neurons of aberrant morphology and impaired neurogenesis in the dentate gyrus of HSF1(-/-) mice. In addition, HSF1(-/-) mice showed aberrant affective behavior, including reduced anxiety and sociability but increased depression-like behavior and aggression. Furthermore, HSF1 deficiency enhanced behavioral vulnerability to repeated exposure to restraint stress. Importantly, rescuing the HSF1 deficiency in the neonatal but not the adult hippocampus reversed the aberrant anxiety and depression-like behaviors. These results indicate a crucial role for hippocampal HSF1 in neuronal and behavioral development. Analysis of the molecular mechanisms revealed that HSF1 directly modulates the expression of polysialyltransferase genes, which then modulate polysialic acid-neural cell adhesion molecule (PSA-NCAM) levels in the hippocampus. Enzymatic removal of PSA from the neonatal hippocampus resulted in aberrant behavior during adulthood, similar to that observed in HSF1(-/-) mice. Thus, these results suggest that one role of HSF1 is to control hippocampal PSA-NCAM levels through the transcriptional regulation of polysialyltransferases, a process that might be involved in neuronal and behavioral development in mice.


Asunto(s)
Conducta Animal/fisiología , Proteínas de Unión al ADN/metabolismo , Hipocampo/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Ansiedad/fisiopatología , Secuencia de Bases , Western Blotting , Proteínas de Unión al ADN/genética , Espinas Dendríticas/fisiología , Conducta Alimentaria/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción del Choque Térmico , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Datos de Secuencia Molecular , Actividad Motora/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácidos Siálicos/genética , Ácidos Siálicos/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Factores de Transcripción/genética
7.
Neuron ; 112(5): 786-804.e8, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38228137

RESUMEN

Chronic stress is a major risk factor for psychiatric disorders, including depression. Although depression is a highly heterogeneous syndrome, it remains unclear how chronic stress drives individual differences in behavioral responses. In this study, we developed a subtyping-based approach wherein stressed male mice were divided into four subtypes based on their behavioral patterns of social interaction deficits and anhedonia, the core symptoms of psychiatric disorders. We identified three prefrontal cortical neuronal projections that regulate repeated stress-induced behavioral phenotypes. Among them, the medial prefrontal cortex (mPFC)→anterior paraventricular thalamus (aPVT) pathway determines the specific behavioral subtype that exhibits both social deficits and anhedonia. Additionally, we identified the circuit-level molecular mechanism underlying this subtype: KDM5C-mediated epigenetic repression of Shisa2 transcription in aPVT projectors in the mPFC led to social deficits and anhedonia. Thus, we provide a set of biological aspects at the cellular, molecular, and epigenetic levels that determine distinctive stress-induced behavioral phenotypes.


Asunto(s)
Anhedonia , Trastornos Mentales , Humanos , Ratones , Masculino , Animales , Neuronas , Corteza Prefrontal/fisiología , Trastornos Mentales/metabolismo , Fenotipo , Estrés Psicológico/metabolismo
8.
Neuron ; 112(8): 1265-1285.e10, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38377990

RESUMEN

Despite the rapid and sustained antidepressant effects of ketamine and its metabolites, their underlying cellular and molecular mechanisms are not fully understood. Here, we demonstrate that the sustained antidepressant-like behavioral effects of (2S,6S)-hydroxynorketamine (HNK) in repeatedly stressed animal models involve neurobiological changes in the anterior paraventricular nucleus of the thalamus (aPVT). Mechanistically, (2S,6S)-HNK induces mRNA expression of extrasynaptic GABAA receptors and subsequently enhances GABAA-receptor-mediated tonic currents, leading to the nuclear export of histone demethylase KDM6 and its replacement by histone methyltransferase EZH2. This process increases H3K27me3 levels, which in turn suppresses the transcription of genes associated with G-protein-coupled receptor signaling. Thus, our findings shed light on the comprehensive cellular and molecular mechanisms in aPVT underlying the sustained antidepressant behavioral effects of ketamine metabolites. This study may support the development of potentially effective next-generation pharmacotherapies to promote sustained remission of stress-related psychiatric disorders.


Asunto(s)
Ketamina , Animales , Humanos , Ketamina/farmacología , Simulación de Dinámica Molecular , Antidepresivos/farmacología , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
9.
Sci Adv ; 9(14): eade5397, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018397

RESUMEN

Chronic stress increases the risk of developing psychiatric disorders, including mood and anxiety disorders. Although behavioral responses to repeated stress vary across individuals, the underlying mechanisms remain unclear. Here, we perform a genome-wide transcriptome analysis of an animal model of depression and patients with clinical depression and report that dysfunction of the Fos-mediated transcription network in the anterior cingulate cortex (ACC) confers a stress-induced social interaction deficit. Critically, CRISPR-Cas9-mediated ACC Fos knockdown causes social interaction deficits under stressful situation. Moreover, two classical second messenger pathways, calcium and cyclic AMP, in the ACC during stress differentially modulate Fos expression and regulate stress-induced changes in social behaviors. Our findings highlight a behaviorally relevant mechanism for the regulation of calcium- and cAMP-mediated Fos expression that has potential as a therapeutic target for psychiatric disorders related to stressful environments.


Asunto(s)
Calcio , Proteínas Proto-Oncogénicas c-fos , Animales , Calcio/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Giro del Cíngulo/metabolismo , AMP Cíclico/metabolismo , Estrés Psicológico
10.
Biol Psychiatry Glob Open Sci ; 3(1): 87-98, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36712563

RESUMEN

Background: A key challenge in the understanding and treatment of depression is identifying cell types and molecular mechanisms that mediate behavioral responses to antidepressant drugs. Because treatment responses in clinical depression are heterogeneous, it is crucial to examine treatment responders and nonresponders in preclinical studies. Methods: We used the large variance in behavioral responses to long-term treatment with multiple classes of antidepressant drugs in different inbred mouse strains and classified the mice into responders and nonresponders based on their response in the forced swim test. Medial prefrontal cortex tissues were subjected to RNA sequencing to identify molecules that are consistently associated across antidepressant responders. We developed and used virus-mediated gene transfer to induce the gene of interest in specific cell types and performed forced swim, sucrose preference, social interaction, and open field tests to investigate antidepressant-like and anxiety-like behaviors. Results: Cartpt expression was consistently upregulated in responders to four types of antidepressants but not in nonresponders in different mice strains. Responder mice given a single dose of ketamine, a fast-acting non-monoamine-based antidepressant, exhibited high CART peptide expression. CART peptide overexpression in the GABAergic (gamma-aminobutyric acidergic) neurons of the anterior cingulate cortex led to antidepressant-like behavior and drove chronic stress resiliency independently of mouse genetic background. Conclusions: These data demonstrate that activation of CART peptide signaling in GABAergic neurons of the anterior cingulate cortex is a common molecular mechanism across antidepressant responders and that this pathway also drives stress resilience.

11.
J Med Chem ; 66(22): 15171-15188, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37847303

RESUMEN

Histone deacetylase 1 and 2 (HDAC1/2) inhibitors are potentially useful as tools for probing the biological functions of the isoforms and as therapeutic agents for cancer and neurodegenerative disorders. To discover potent and selective inhibitors, we screened a focused library synthesized by using click chemistry and obtained KPZ560 as an HDAC1/2-selective inhibitor. Kinetic binding analysis revealed that KPZ560 inhibits HDAC2 through a two-step slow-binding mechanism. In cellular assays, KPZ560 induced a dose- and time-dependent increase of histone acetylation and showed potent breast cancer cell growth-inhibitory activity. In addition, gene expression analyses suggested that the two-step slow-binding inhibition by KPZ560 regulated the expression of genes associated with cell proliferation and DNA damage. KPZ560 also induced neurite outgrowth of Neuro-2a cells and an increase in the spine density of granule neuron dendrites of mice. The unique two-step slow-binding character of o-aminoanilides such as KPZ560 makes them interesting candidates as therapeutic agents.


Asunto(s)
Histona Desacetilasa 1 , Histona Desacetilasas , Ratones , Animales , Histona Desacetilasas/metabolismo , Química Clic , Inhibidores de Histona Desacetilasas/farmacología , Neuronas/metabolismo , Histona Desacetilasa 2
12.
Heliyon ; 9(1): e13059, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36711294

RESUMEN

Only 50% of patients with depression respond to the first antidepressant drug administered. Thus, biomarkers for prediction of antidepressant responses are needed, as predicting which patients will not respond to antidepressants can optimize selection of alternative therapies. We aimed to identify biomarkers that could predict antidepressant responsiveness using a novel data-driven approach based on statistical pattern recognition. We retrospectively divided patients with major depressive disorder into antidepressant responder and non-responder groups. Comprehensive gene expression analysis was performed using peripheral blood without narrowing the genes. We designed a classifier according to our own discrete Bayes decision rule that can handle categorical data. Nineteen genes showed differential expression in the antidepressant non-responder group (n = 15) compared to the antidepressant responder group (n = 15). In the training sample of 30 individuals, eight candidate genes had significantly altered expression according to quantitative real-time polymerase chain reaction. The expression of these genes was examined in an independent test sample of antidepressant responders (n = 22) and non-responders (n = 12). Using the discrete Bayes classifier with the HERC5, IFI6, and IFI44 genes identified in the training set yielded 85% discrimination accuracy for antidepressant responsiveness in the 34 test samples. Pathway analysis of the RNA sequencing data for antidepressant responsiveness identified that hypercytokinemia- and interferon-related genes were increased in non-responders. Disease and biofunction analysis identified changes in genes related to inflammatory and infectious diseases, including coronavirus disease. These results strongly suggest an association between antidepressant responsiveness and inflammation, which may be useful for future treatment strategies for depression.

13.
Artículo en Japonés | MEDLINE | ID: mdl-23012885

RESUMEN

Recent research has raised the notion that epigenetic mechanisms (e.g., DNA methylation and histone modifications), which exert lasting control over gene expression without altering the genetic code, could mediate stable changes in brain function. However, the role of environmental factors along with genetic factors in the epigenetic regulation of the pathogenesis of depression is largely unknown. Two genetically distinct mice strains, BALB/c (BALB) and C57BL/6 (B6), exhibit different behavioral responses to chronic stress. With chronic stress, BALB mice showed depressive-like behaviors, but not B6 mice, and glial cell-derived neurotrophic factor (GDNF) expression level was decreased in the ventral striatum of BALB mice but increased in B6 mice. In BALB mice, depressive-like behaviors and decreased GDNF expression were recovered by chronic antidepressant treatment. Therefore, we used these two mice strains to investigate how the epigenetic status of the GDNF gene in the ventral striatum modulates stress vulnerability. Both mice strains showed increased DNA methylation levels and MeCP2 recruitment in the GDNF promoter region. However, histone H3 acetylation level was decreased in BALB mice, but increased in B6 mice. Furthermore, BALB mice showed increased histone deacetylase2 (HDAC2) expression level and Re-ChIP assay revealed HDAC2-MeCP2 complex in BALB mice. Our results indicate the crucial role of histone modification by HDAC2 and MeCP2 complex for the control of GDNF expression and subsequent behavioral responses to chronic stress, in other words, the susceptibility to stress. In addition, we investigated the effect of antidepressants on the epigenetic regulation of GDNF expression. We found a reduced level of HDAC4 recruitment at the GDNF promoter region with antidepressants. Thus, our data suggest that antidepressants increase transcriptional activity of the GDNF gene through the modulation of histone acetylation by HDAC4. Finally, we examined the expressions of GDNF and epigenetic-related molecules mRNAs with major depressive and bipolar disorder patients by using quantitative real-time PCR. We found the aberrant expression of GDNF and epigenetic-related genes including HDAC2 and HDAC4 in mood disorder patients. Thus, our data provide novel insights suggesting that epigenetic mechanisms of GDNF expression are involved in the pathogenesis or pathophysiology of depression.


Asunto(s)
Depresión/genética , Epigénesis Genética , Animales , Depresión/etiología , Depresión/metabolismo , Depresión/fisiopatología , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos
14.
Seishin Shinkeigaku Zasshi ; 114(7): 812-20, 2012.
Artículo en Japonés | MEDLINE | ID: mdl-22897028

RESUMEN

In order to identify the possible biomarkers of mood disorders, we measured the mRNA levels for a variety of genes in peripheral leukocytes of mood disorder patients in a depressive, as well as in a remissive state, comparing with healthy controls. We selected and measured the levels of genes of interest, which are listed as follows: glucocorticoid receptor, neurotrophic factors, cell adhesion molecules, SR protein splicing factors, transcription factors, epigenetic factors (histone deacetylase, sirtuin, DNA methyltransferase), since these molecules are suggested to be associated with the neural function, synaptic plasticity, and behaviors in animal models, as well as with the pathophysiology and pathogenesis of mood disorders. We found the three different types of biological markers: 1) state markers those revealed alterations of gene expression only in a depressive state of major depressive patients and/or bipolar depressive patients, 2) trait markers those showed altered gene expression both in a depressive and a remissive state of major depressive patients and/or bipolar depressive patients, and 3) markers of the treatment resistance those revealed different alterations of gene expression between treatment resistant and treatment responsive patients in a depressive state. The use of state and trait markers in combination would allow us to put a differential diagnosis between major depressive and bipolar depressive states, as well as between mood disorders and neurotic depressive states. Furthermore, candidate biomarkers of treatment resistance could be used to consider forward of applying the electric convulsive therapy even in an early stage of a depressive state.


Asunto(s)
Biomarcadores/análisis , Diagnóstico Diferencial , Expresión Génica , Leucocitos/metabolismo , Trastornos del Humor/diagnóstico , Humanos , Trastornos del Humor/genética , ARN Mensajero/metabolismo
15.
Front Mol Neurosci ; 15: 844295, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401110

RESUMEN

Maternal behavior is shaped and challenged by the changing developmental needs of offspring and a broad range of environmental factors, with evidence indicating that the maternal brain exhibits a high degree of plasticity. This plasticity is displayed within cellular and molecular systems, including both intra- and intercellular signaling processes as well as transcriptional profiles. This experience-associated plasticity may have significant overlap with the mechanisms controlling memory processes, in particular those that are activity-dependent. While a significant body of work has identified various molecules and intracellular processes regulating maternal care, the role of activity- and experience-dependent processes remains unclear. We discuss recent progress in studying activity-dependent changes occurring at the synapse, in the nucleus, and during the transport between these two structures in relation to maternal behavior. Several pre- and postsynaptic molecules as well as transcription factors have been found to be critical in these processes. This role reflects the principal importance of the molecular and cellular mechanisms of memory formation to maternal and other behavioral adaptations.

16.
J Neurosci ; 30(45): 15007-18, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21068306

RESUMEN

There is growing evidence suggesting that early life events have long-term effects on the neuroendocrine and behavioral developments of rodents. However, little is known about the involvement of early life events in the susceptibility to subsequent stress exposure during adulthood. The present study characterized the effect of maternal separation, an animal model of early life adversity, on the behavioral response to repeated restraint stress in adult rats and investigated the molecular mechanism underlying behavioral vulnerability to chronic stress induced by the maternal separation. Rat pups were separated from the dams for 180 min per day from postnatal day 2 through 14 (HMS180 rats). We found that, as young adults, HMS180 rats showed a greater hypothalamic-pituitary-adrenal axis response to acute restraint stress than nonseparated control rats. In addition, repeatedly restrained HMS180 rats showed increased depression-like behavior and an anhedonic response compared with nonrestrained HMS180 rats. Furthermore, HMS180 rats showed increased expression of REST4, a neuron-specific splicing variant of the transcriptional repressor REST (repressor element-1 silencing transcription factor), and a variety of REST target gene mRNAs and microRNAs in the medial prefrontal cortex (mPFC). Finally, REST4 overexpression in the mPFC of neonatal mice via polyethyleneimine-mediated gene transfer enhanced the expression of its target genes as well as behavioral vulnerability to repeated restraint stress. In contrast, REST4 overexpression in the mPFC of adult mice did not affect depression-like behaviors after repeated stress exposure. These results suggest that the activation of REST4-mediated gene regulation in the mPFC during postnatal development is involved in stress vulnerability.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Represoras/metabolismo , Estrés Psicológico/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Northern Blotting , Western Blotting , Línea Celular , Células Cultivadas , Corticosterona/sangre , Femenino , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Privación Materna , Ratones , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Restricción Física , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología
17.
Front Behav Neurosci ; 15: 749180, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34658809

RESUMEN

Major depressive disorder (MDD) is a debilitating disease characterized by depressed mood, loss of interest or pleasure, suicidal ideation, and reduced motivation or hopelessness. Despite considerable research, mechanisms underlying MDD remain poorly understood, and current advances in treatment are far from satisfactory. The antidepressant effect of ketamine is among the most important discoveries in psychiatric research over the last half-century. Neurobiological insights into the ketamine's effects have shed light on the mechanisms underlying antidepressant efficacy. However, mechanisms underlying the rapid and sustained antidepressant effects of ketamine remain controversial. Elucidating such mechanisms is key to identifying new therapeutic targets and developing therapeutic strategies. Accumulating evidence demonstrates the contribution of the glutamatergic pathway, the major excitatory neurotransmitter system in the central nervous system, in MDD pathophysiology and antidepressant effects. The hypothesis of a connection among the calcium signaling cascade stimulated by the glutamatergic system, neural plasticity, and epigenetic regulation of gene transcription is further supported by its associations with ketamine's antidepressant effects. This review briefly summarizes the potential mechanisms of ketamine's effects with a specific focus on glutamatergic signaling from a multiscale perspective, including behavioral, cellular, molecular, and epigenetic aspects, to provide a valuable overview of ketamine's antidepressant effects.

18.
Front Mol Neurosci ; 14: 708004, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276306

RESUMEN

Major depressive disorder (MDD) is a leading cause of disability worldwide. Although the etiology and pathophysiology of MDD remain poorly understood, aberrant neuroplasticity mediated by the epigenetic dysregulation of gene expression within the brain, which may occur due to genetic and environmental factors, may increase the risk of this disorder. Evidence has also been reported for sex-related differences in the pathophysiology of MDD, with female patients showing a greater severity of symptoms, higher degree of functional impairment, and more atypical depressive symptoms. Males and females also differ in their responsiveness to antidepressants. These clinical findings suggest that sex-dependent molecular and neural mechanisms may underlie the development of depression and the actions of antidepressant medications. This review discusses recent advances regarding the role of epigenetics in stress and depression. The first section presents a brief introduction of the basic mechanisms of epigenetic regulation, including histone modifications, DNA methylation, and non-coding RNAs. The second section reviews their contributions to neural plasticity, the risk of depression, and resilience against depression, with a particular focus on epigenetic modulators that have causal relationships with stress and depression in both clinical and animal studies. The third section highlights studies exploring sex-dependent epigenetic alterations associated with susceptibility to stress and depression. Finally, we discuss future directions to understand the etiology and pathophysiology of MDD, which would contribute to optimized and personalized therapy.

19.
Sci Rep ; 11(1): 22615, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799651

RESUMEN

Postpartum depression is an important mental health issue not only for the mother but also for the child's development, other family members, and the society. An appropriate animal model is desired to elucidate the pathogenesis of postpartum depression. However, methods for stress loading during pregnancy have not been established. Behavioral experiments to investigate postpartum depression-like behaviors should be conducted without stress because behavioral tests affect rearing behaviors such as lactation. Therefore, we developed a new mouse model of postpartum depression using a psychological stress method. Mating partners were made to witness their partners experiencing social defeat stress and then listen to their cries. Emotional stress loading during pregnancy significantly increased postpartum depression-like behaviors. Postpartum depression also affected nurturing behaviors and caused disturbances in pup care. Furthermore, nesting behavior was impaired in the stressed group, suggesting that the observation of nesting behavior may be useful for assessing social dysfunction in postpartum depression. These results demonstrate the utility of this new mouse model of postpartum depression.


Asunto(s)
Conducta Animal , Depresión Posparto/fisiopatología , Depresión Posparto/psicología , Modelos Animales de Enfermedad , Comportamiento de Nidificación , Animales , Ansiedad , Prueba de Esfuerzo , Femenino , Humanos , Lactancia , Masculino , Ratones , Ratones Endogámicos BALB C , Distrés Psicológico , Estrés Psicológico , Natación
20.
Sci Rep ; 11(1): 2296, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504850

RESUMEN

The heterogeneity of major depressive disorder (MDD) is attributed to the fact that diagnostic criteria (e.g., DSM-5) are only based on clinical symptoms. The discovery of blood biomarkers has the potential to change the diagnosis of MDD. The purpose of this study was to identify blood biomarkers of DNA methylation by strategically subtyping patients with MDD by onset age. We analyzed genome-wide DNA methylation of patients with adult-onset depression (AOD; age ≥ 50 years, age at depression onset < 50 years; N = 10) and late-onset depression (LOD; age ≥ 50 years, age at depression onset ≥ 50 years; N = 25) in comparison to that of 30 healthy subjects. The methylation profile of the AOD group was not only different from that of the LOD group but also more homogenous. Six identified methylation CpG sites were validated by pyrosequencing and amplicon bisulfite sequencing as potential markers for AOD in a second set of independent patients with AOD and healthy control subjects (N = 11). The combination of three specific methylation markers achieved the highest accuracy (sensitivity, 64%; specificity, 91%; accuracy, 77%). Taken together, our findings suggest that DNA methylation markers are more suitable for AOD than for LOD patients.


Asunto(s)
Metilación de ADN/fisiología , Depresión/genética , Depresión/fisiopatología , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/fisiopatología , Anciano , Metilación de ADN/genética , Epigénesis Genética/genética , Epigenómica , Femenino , Marcadores Genéticos/genética , Marcadores Genéticos/fisiología , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA