Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Psychiatry ; 27(5): 2563-2579, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33931727

RESUMEN

Heightened aggressive behavior is considered as one of the central symptoms of many neuropsychiatric disorders including autism, schizophrenia, and dementia. The consequences of aggression pose a heavy burden on patients and their families and clinicians. Unfortunately, we have limited treatment options for aggression and lack mechanistic insight into the causes of aggression needed to inform new efforts in drug discovery and development. Levels of proinflammatory cytokines in the periphery or cerebrospinal fluid were previously reported to correlate with aggressive traits in humans. However, it is still unknown whether cytokines affect brain circuits to modulate aggression. Here, we examined the functional role of interleukin 1ß (IL-1ß) in mediating individual differences in aggression using a resident-intruder mouse model. We found that nonaggressive mice exhibit higher levels of IL-1ß in the dorsal raphe nucleus (DRN), the major source of forebrain serotonin (5-HT), compared to aggressive mice. We then examined the effect of pharmacological antagonism and viral-mediated gene knockdown of the receptors for IL-1 within the DRN and found that both treatments consistently increased aggressive behavior of male mice. Aggressive mice also exhibited higher c-Fos expression in 5-HT neurons in the DRN compared to nonaggressive mice. In line with these findings, deletion of IL-1 receptor in the DRN enhanced c-Fos expression in 5-HT neurons during aggressive encounters, suggesting that modulation of 5-HT neuronal activity by IL-1ß signaling in the DRN controls expression of aggressive behavior.


Asunto(s)
Agresión , Núcleo Dorsal del Rafe , Interleucina-1beta , Serotonina , Agresión/fisiología , Animales , Núcleo Dorsal del Rafe/metabolismo , Humanos , Individualidad , Interleucina-1beta/metabolismo , Masculino , Ratones , Serotonina/metabolismo
2.
Nature ; 534(7609): 688-92, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27357796

RESUMEN

Maladaptive aggressive behaviour is associated with a number of neuropsychiatric disorders and is thought to result partly from the inappropriate activation of brain reward systems in response to aggressive or violent social stimuli. Nuclei within the ventromedial hypothalamus, extended amygdala and limbic circuits are known to encode initiation of aggression; however, little is known about the neural mechanisms that directly modulate the motivational component of aggressive behaviour. Here we established a mouse model to measure the valence of aggressive inter-male social interaction with a smaller subordinate intruder as reinforcement for the development of conditioned place preference (CPP). Aggressors develop a CPP, whereas non-aggressors develop a conditioned place aversion to the intruder-paired context. Furthermore, we identify a functional GABAergic projection from the basal forebrain (BF) to the lateral habenula (lHb) that bi-directionally controls the valence of aggressive interactions. Circuit-specific silencing of GABAergic BF-lHb terminals of aggressors with halorhodopsin (NpHR3.0) increases lHb neuronal firing and abolishes CPP to the intruder-paired context. Activation of GABAergic BF-lHb terminals of non-aggressors with channelrhodopsin (ChR2) decreases lHb neuronal firing and promotes CPP to the intruder-paired context. Finally, we show that altering inhibitory transmission at BF-lHb terminals does not control the initiation of aggressive behaviour. These results demonstrate that the BF-lHb circuit has a critical role in regulating the valence of inter-male aggressive behaviour and provide novel mechanistic insight into the neural circuits modulating aggression reward processing.


Asunto(s)
Agresión/fisiología , Prosencéfalo Basal/fisiología , Habénula/fisiología , Vías Nerviosas/fisiología , Recompensa , Potenciales de Acción , Animales , Prosencéfalo Basal/citología , Condicionamiento Psicológico/fisiología , Neuronas GABAérgicas/metabolismo , Habénula/citología , Halorrodopsinas/metabolismo , Individualidad , Masculino , Ratones , Modelos Neurológicos , Motivación , Inhibición Neural , Refuerzo en Psicología , Rodopsina/metabolismo , Conducta Social
3.
Annu Rev Pharmacol Toxicol ; 58: 411-428, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28992428

RESUMEN

Mood disorders such as depression are among the most prevalent psychiatric disorders in the United States, but they are inadequately treated in a substantial proportion of patients. Accordingly, neuropsychiatric research has pivoted from investigation of monoaminergic mechanisms to exploration of novel mediators, including the role of inflammatory processes. Subsets of mood disorder patients exhibit immune-related abnormalities, including elevated levels of proinflammatory cytokines, monocytes, and neutrophils in the peripheral circulation; dysregulation of neuroglia and blood-brain barrier function; and disruption of gut microbiota. The field of psychoneuroimmunology is one of great therapeutic opportunity, yielding experimental therapeutics for mood disorders, such as peripheral cytokine targeting antibodies, microglia and astrocyte targeting therapies, and probiotic treatments for gut dysbiosis, and producing findings that identify therapeutic targets for future development.


Asunto(s)
Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Trastornos del Humor/metabolismo , Animales , Encéfalo/metabolismo , Citocinas/metabolismo , Humanos
4.
Proc Natl Acad Sci U S A ; 115(5): 1111-1116, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339486

RESUMEN

Behavioral coping strategies are critical for active resilience to stress and depression; here we describe a role for neuroligin-2 (NLGN-2) in the nucleus accumbens (NAc). Neuroligins (NLGN) are a family of neuronal postsynaptic cell adhesion proteins that are constituents of the excitatory and inhibitory synapse. Importantly, NLGN-3 and NLGN-4 mutations are strongly implicated as candidates underlying the development of neuropsychiatric disorders with social disturbances such as autism, but the role of NLGN-2 in neuropsychiatric disease states is unclear. Here we show a reduction in NLGN-2 gene expression in the NAc of patients with major depressive disorder. Chronic social defeat stress in mice also decreases NLGN-2 selectively in dopamine D1-positive cells, but not dopamine D2-positive cells, within the NAc of stress-susceptible mice. Functional NLGN-2 knockdown produces bidirectional, cell-type-specific effects: knockdown in dopamine D1-positive cells promotes subordination and stress susceptibility, whereas knockdown in dopamine D2-positive cells mediates active defensive behavior. These findings establish a behavioral role for NAc NLGN-2 in stress and depression; provide a basis for targeted, cell-type specific therapy; and highlight the role of active behavioral coping mechanisms in stress susceptibility.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Trastorno Depresivo Mayor/fisiopatología , Dominación-Subordinación , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/metabolismo , Estrés Psicológico/fisiopatología , Agresión , Animales , Antidepresivos/farmacología , Conducta Animal , Línea Celular , Modelos Animales de Enfermedad , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Conducta Social , Sinapsis/metabolismo
5.
J Neurophysiol ; 123(6): 2382-2389, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374202

RESUMEN

Parkinson's disease (PD) risk is increased by stress and certain gene mutations, including the most prevalent PD-linked mutation LRRK2-G2019S. Both PD and stress increase risk for psychiatric symptoms, yet it is unclear how PD-risk genes alter neural circuitry in response to stress that may promote psychopathology. Here we show significant differences between adult G2019S knockin and wild-type (wt) mice in stress-induced behaviors, with an unexpected uncoupling of depression-like and hedonia-like responses in G2019S mice. Moreover, mutant spiny projection neurons in nucleus accumbens (NAc) lack an adaptive, stress-induced change in excitability displayed by wt neurons, and instead show stress-induced changes in synaptic properties that wt neurons lack. Some synaptic alterations in NAc are already evident early in postnatal life. Thus G2019S alters the magnitude and direction of behavioral responses to stress that may reflect unique modifications of adaptive plasticity in cells and circuits implicated in psychopathology in humans.NEW & NOTEWORTHY Depression is associated with Parkinson's disease (PD), and environmental stress is a risk factor for both. We investigated how LRRK2-G2019S PD mutation affects depression-like behaviors, synaptic function, and intrinsic neuronal excitability following stress. In response to stress, the mutation drives abnormal synaptic changes, prevents adaptive changes in intrinsic excitability, and leads to aberrant behaviors, thus defining new ways in which PD mutations derail adaptive plasticity in response to stress that may contribute to disease onset.


Asunto(s)
Conducta Animal , Depresión , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Núcleo Accumbens , Enfermedad de Parkinson , Estrés Psicológico , Animales , Conducta Animal/fisiología , Depresión/etiología , Depresión/genética , Depresión/fisiopatología , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleo Accumbens/fisiopatología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Estrés Psicológico/complicaciones , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología
6.
J Neurosci ; 38(26): 5913-5924, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29891732

RESUMEN

A growing number of studies implicate the brain's reward circuitry in aggressive behavior. However, the cellular and molecular mechanisms within brain reward regions that modulate the intensity of aggression as well as motivation for it have been underexplored. Here, we investigate the cell-type-specific influence of ΔFosB, a transcription factor known to regulate a range of reward and motivated behaviors, acting in the nucleus accumbens (NAc), a key reward region, in male aggression in mice. We show that ΔFosB is specifically increased in dopamine D1 receptor (Drd1)-expressing medium spiny neurons (D1-MSNs) in NAc after repeated aggressive encounters. Viral-mediated induction of ΔFosB selectively in D1-MSNs of NAc intensifies aggressive behavior without affecting the preference for the aggression-paired context in a conditioned place preference (CPP) assay. In contrast, ΔFosB induction selectively in D2-MSNs reduces the time spent exploring the aggression-paired context during CPP without affecting the intensity of aggression per se. These data strongly support a dissociable cell-type-specific role for ΔFosB in the NAc in modulating aggression and aggression reward.SIGNIFICANCE STATEMENT Aggressive behavior is associated with several neuropsychiatric disorders and can be disruptive for affected individuals as well as their victims. Studies have shown a positive reinforcement mechanism underlying aggressive behavior that shares many common features with drug addiction. Here, we explore the cell-type-specific role of the addiction-associated transcription factor ΔFosB in the nucleus accumbens in aggression. We found that ΔFosB expression promotes aggressive behavior, effects that are dissociable from its effects on aggression reward. This finding is a significant first step in identifying therapeutic targets for the reduction of aggressive behavior across a range of neuropsychiatric illnesses.


Asunto(s)
Agresión/fisiología , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Conducta Animal/fisiología , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Recompensa
7.
Proc Natl Acad Sci U S A ; 111(45): 16136-41, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25331895

RESUMEN

Depression and anxiety disorders are associated with increased release of peripheral cytokines; however, their functional relevance remains unknown. Using a social stress model in mice, we find preexisting individual differences in the sensitivity of the peripheral immune system that predict and promote vulnerability to social stress. Cytokine profiles were obtained 20 min after the first social stress exposure. Of the cytokines regulated by stress, IL-6 was most highly up-regulated only in mice that ultimately developed a susceptible behavioral phenotype following a subsequent chronic stress, and levels remained elevated for at least 1 mo. We confirmed a similar elevation of serum IL-6 in two separate cohorts of patients with treatment-resistant major depressive disorder. Before any physical contact in mice, we observed individual differences in IL-6 levels from ex vivo stimulated leukocytes that predict susceptibility versus resilience to a subsequent stressor. To shift the sensitivity of the peripheral immune system to a pro- or antidepressant state, bone marrow (BM) chimeras were generated by transplanting hematopoietic progenitor cells from stress-susceptible mice releasing high IL-6 or from IL-6 knockout (IL-6(-/-)) mice. Stress-susceptible BM chimeras exhibited increased social avoidance behavior after exposure to either subthreshold repeated social defeat stress (RSDS) or a purely emotional stressor termed witness defeat. IL-6(-/-) BM chimeric and IL-6(-/-) mice, as well as those treated with a systemic IL-6 monoclonal antibody, were resilient to social stress. These data establish that preexisting differences in stress-responsive IL-6 release from BM-derived leukocytes functionally contribute to social stress-induced behavioral abnormalities.


Asunto(s)
Trastornos de Ansiedad/inmunología , Conducta Animal , Interleucina-6/inmunología , Estrés Psicológico/inmunología , Aloinjertos , Animales , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/patología , Trasplante de Médula Ósea , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/patología , Interleucina-6/genética , Ratones , Ratones Noqueados , Estrés Psicológico/genética , Estrés Psicológico/patología , Factores de Tiempo , Quimera por Trasplante/genética , Quimera por Trasplante/inmunología
8.
J Neurosci ; 35(50): 16362-76, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26674863

RESUMEN

Depression and anxiety disorders are more prevalent in females, but the majority of research in animal models, the first step in finding new treatments, has focused predominantly on males. Here we report that exposure to subchronic variable stress (SCVS) induces depression-associated behaviors in female mice, whereas males are resilient as they do not develop these behavioral abnormalities. In concert with these different behavioral responses, transcriptional analysis of nucleus accumbens (NAc), a major brain reward region, by use of RNA sequencing (RNA-seq) revealed markedly different patterns of stress regulation of gene expression between the sexes. Among the genes displaying sex differences was DNA methyltransferase 3a (Dnmt3a), which shows a greater induction in females after SCVS. Interestingly, Dnmt3a expression levels were increased in the NAc of depressed humans, an effect seen in both males and females. Local overexpression of Dnmt3a in NAc rendered male mice more susceptible to SCVS, whereas Dnmt3a knock-out in this region rendered females more resilient, directly implicating this gene in stress responses. Associated with this enhanced resilience of female mice upon NAc knock-out of Dnmt3a was a partial shift of the NAc female transcriptome toward the male pattern after SCVS. These data indicate that males and females undergo different patterns of transcriptional regulation in response to stress and that a DNA methyltransferase in NAc contributes to sex differences in stress vulnerability. SIGNIFICANCE STATEMENT: Women have a higher incidence of depression than men. However, preclinical models, the first step in developing new diagnostics and therapeutics, have been performed mainly on male subjects. Using a stress-based animal model of depression that causes behavioral effects in females but not males, we demonstrate a sex-specific transcriptional profile in brain reward circuitry. This transcriptional profile can be altered by removal of an epigenetic mechanism, which normally suppresses DNA transcription, creating a hybrid male/female transcriptional pattern. Removal of this epigenetic mechanism also induces behavioral resilience to stress in females. These findings shed new light onto molecular factors controlling sex differences in stress response.


Asunto(s)
Núcleo Accumbens/fisiopatología , Resiliencia Psicológica , Estrés Psicológico/genética , Estrés Psicológico/psicología , Transcriptoma/genética , Animales , Ansiedad/genética , Ansiedad/psicología , Enfermedad Crónica , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Conducta Alimentaria , Femenino , Regulación Enzimológica de la Expresión Génica/genética , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Represión Psicológica , Caracteres Sexuales , Natación/psicología
9.
J Neurosci ; 34(34): 11461-9, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25143625

RESUMEN

Stable changes in neuronal gene expression have been studied as mediators of addicted states. Of particular interest is the transcription factor ΔFosB, a truncated and stable FosB gene product whose expression in nucleus accumbens (NAc), a key reward region, is induced by chronic exposure to virtually all drugs of abuse and regulates their psychomotor and rewarding effects. Phosphorylation at Ser(27) contributes to ΔFosB's stability and accumulation following repeated exposure to drugs, and our recent work demonstrates that the protein kinase CaMKIIα phosphorylates ΔFosB at Ser(27) and regulates its stability in vivo. Here, we identify two additional sites on ΔFosB that are phosphorylated in vitro by CaMKIIα, Thr(149) and Thr(180), and demonstrate their regulation in vivo by chronic cocaine. We show that phosphomimetic mutation of Thr(149) (T149D) dramatically increases AP-1 transcriptional activity while alanine mutation does not affect transcriptional activity when compared with wild-type (WT) ΔFosB. Using in vivo viral-mediated gene transfer of ΔFosB-T149D or ΔFosB-T149A in mouse NAc, we determined that overexpression of ΔFosB-T149D in NAc leads to greater locomotor activity in response to an initial low dose of cocaine than does WT ΔFosB, while overexpression of ΔFosB-T149A does not produce the psychomotor sensitization to chronic low-dose cocaine seen after overexpression of WT ΔFosB and abrogates the sensitization seen in control animals at higher cocaine doses. We further demonstrate that mutation of Thr(149) does not affect the stability of ΔFosB overexpressed in mouse NAc, suggesting that the behavioral effects of these mutations are driven by their altered transcriptional properties.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Núcleo Accumbens/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Treonina/metabolismo , Adenosina Trifosfato/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/farmacología , Línea Celular Tumoral , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Núcleo Accumbens/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Treonina/genética , Factor de Transcripción AP-1/metabolismo
10.
Neurology ; 98(1): e40-e50, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34649873

RESUMEN

BACKGROUND AND OBJECTIVES: To report final, 36-month safety and clinical outcomes from the PD-1101 trial of NBIb-1817 (VY-AADC01) in participants with moderately advanced Parkinson disease (PD) and motor fluctuations. METHODS: PD-1101 was a phase 1b, open-label, dose escalation trial of VY-AADC01, an experimental AAV2 gene therapy encoding the human aromatic l-amino acid decarboxylase (AADC) enzyme. VY-AADC01 was delivered via bilateral, intraoperative MRI-guided putaminal infusions to 3 cohorts (n = 5 participants per cohort): cohort 1, ≤7.5 × 1011 vector genomes (vg); cohort 2, ≤1.5 × 1012 vg; cohort 3, ≤4.7 × 1012 vg. RESULTS: No serious adverse events (SAEs) attributed to VY-AADC01 were reported. All 4 non-vector-related SAEs (atrial fibrillation and pulmonary embolism in 1 participant and 2 events of small bowel obstruction in another participant) resolved. Requirements for PD medications were reduced by 21%-30% in the 2 highest dose cohorts at 36 months. Standard measures of motor function (PD diary, Unified Parkinson's Disease Rating Scale III "off"-medication and "on"-medication scores), global impressions of improvement (Clinical Global Impression of Improvement, Patient Global Impression of Improvement), and quality of life (39-item Parkinson's Disease Questionnaire) were stable or improved compared with baseline at 12, 24, and 36 months following VY-AADC01 administration across cohorts. DISCUSSIONS: VY-AADC01 and the surgical administration procedure were well-tolerated and resulted in stable or improved motor function and quality of life across cohorts, as well as reduced PD medication requirements in cohorts 2 and 3 over 3 years. TRIAL REGISTRATION INFORMATION: NCT01973543. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that, in patients with moderately advanced PD and motor fluctuations, putaminal infusion of VY-AADC01 is well tolerated and may improve motor function.


Asunto(s)
Carboxiliasas , Enfermedad de Parkinson , Aminoácidos/genética , Aminoácidos/uso terapéutico , Antiparkinsonianos/efectos adversos , Carboxiliasas/uso terapéutico , Terapia Genética/métodos , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Calidad de Vida , Resultado del Tratamiento
11.
Nat Neurosci ; 23(5): 638-650, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284606

RESUMEN

Heightened aggression is characteristic of multiple neuropsychiatric disorders and can have various negative effects on patients, their families and the public. Recent studies in humans and animals have implicated brain reward circuits in aggression and suggest that, in subsets of aggressive individuals, domination of subordinate social targets is reinforcing. In this study, we showed that, in male mice, orexin neurons in the lateral hypothalamus activated a small population of glutamic acid decarboxylase 2 (GAD2)-expressing neurons in the lateral habenula (LHb) via orexin receptor 2 (OxR2) and that activation of these GAD2 neurons promoted male-male aggression and conditioned place preference for aggression-paired contexts. Moreover, LHb GAD2 neurons were inhibitory within the LHb and dampened the activity of the LHb as a whole. These results suggest that the orexin system is important for the regulation of inter-male aggressive behavior and provide the first functional evidence of a local inhibitory circuit within the LHb.


Asunto(s)
Agresión/fisiología , Neuronas GABAérgicas/metabolismo , Habénula/metabolismo , Orexinas/metabolismo , Animales , Masculino , Ratones , Transducción de Señal/fisiología
12.
Biol Psychiatry ; 86(6): 474-482, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31101319

RESUMEN

BACKGROUND: Clinical studies suggest that heightened peripheral inflammation contributes to the pathogenesis of stress-related disorders, including major depressive disorder. However, the molecular mechanisms within peripheral immune cells that mediate enhanced stress vulnerability are not well known. Because microRNAs (miRs) are important regulators of immune response, we sought to examine their role in mediating inflammatory and behavioral responses to repeated social defeat stress (RSDS), a mouse model of stress vulnerability that produces susceptible and resilient phenotypes. METHODS: We isolated Ly6chigh monocytes via fluorescence-activated cell sorting in the blood of susceptible and resilient mice following RSDS and profiled miR expression via quantitative real-time polymerase chain reaction. Bone marrow chimeric mice were generated to confirm a causal role of the miR-106b∼25 cluster in bone marrow-derived leukocytes in mediating stress resilience versus susceptibility. RESULTS: We found that RSDS produces an increase in circulating Ly6chigh inflammatory monocytes in both susceptible and resilient mice. We next investigated whether intrinsic leukocyte posttranscriptional mechanisms contribute to individual differences in stress response and the resilient phenotype. Of the miRs profiled in our panel, eight were significantly regulated by RSDS within Ly6chigh monocytes, including miR-25-3p, a member of the miR-106b∼25 cluster. Selective knockout of the miR-106b∼25 cluster in peripheral leukocytes promoted behavioral resilience to RSDS. CONCLUSIONS: Our results identify the miR-106b∼25 cluster as a key regulator of stress-induced inflammation and depression that may represent a novel therapeutic target for drug development.


Asunto(s)
Conducta Animal , Depresión/metabolismo , MicroARNs/metabolismo , Resiliencia Psicológica , Estrés Psicológico/metabolismo , Animales , Trasplante de Médula Ósea , Depresión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Monocitos/metabolismo , Estrés Psicológico/patología , Quimera por Trasplante
13.
Biol Psychiatry ; 85(3): 226-236, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30336931

RESUMEN

BACKGROUND: Homeostatic plasticity in mesolimbic dopamine (DA) neurons plays an essential role in mediating resilience to social stress. Recent evidence implicates an association between stress resilience and projections from the locus coeruleus (LC) to the ventral tegmental area (VTA) (LC→VTA) DA system. However, the precise circuitry and molecular mechanisms of the homeostatic plasticity in mesolimbic DA neurons mediated by the LC→VTA circuitry, and its role in conferring resilience to social defeat stress, have not been described. METHODS: In a well-established chronic social defeat stress model of depression, using projection-specific electrophysiological recordings and optogenetic, pharmacological, and molecular profiling techniques, we investigated the functional role and molecular basis of an LC→VTA circuit in conferring resilience to social defeat stress. RESULTS: We found that LC neurons projecting to the VTA exhibit enhanced firing activity in resilient, but not susceptible, mice. Optogenetically mimicking this firing adaptation in susceptible mice reverses their depression-related behaviors, and induces reversal of cellular hyperactivity and homeostatic plasticity in VTA DA neurons projecting to the nucleus accumbens. Circuit-specific molecular profiling studies reveal that α1- and ß3-adrenergic receptors are highly expressed in VTA→nucleus accumbens DA neurons. Pharmacologically activating these receptors induces similar proresilient effects at the ion channel and cellular and behavioral levels, whereas antagonizing these receptors blocks the proresilient effect of optogenetic activation of LC→VTA circuit neurons in susceptible mice. CONCLUSIONS: These findings reveal a key role of the LC→VTA circuit in mediating homeostatic plasticity in stress resilience and reveal α1- and ß3-adrenergic receptors as new molecular targets for therapeutically promoting resilience.


Asunto(s)
Locus Coeruleus/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos beta 3/fisiología , Resiliencia Psicológica , Área Tegmental Ventral/fisiología , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Antagonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Conducta Animal/fisiología , Neuronas Dopaminérgicas/fisiología , Homeostasis/fisiología , Locus Coeruleus/efectos de los fármacos , Masculino , Ratones , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Resiliencia Psicológica/efectos de los fármacos , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/efectos de los fármacos
14.
Nat Commun ; 9(1): 1116, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549264

RESUMEN

Most people exposed to stress do not develop depression. Animal models have shown that stress resilience is an active state that requires broad transcriptional adaptations, but how this homeostatic process is regulated remains poorly understood. In this study, we analyze upstream regulators of genes differentially expressed after chronic social defeat stress. We identify estrogen receptor α (ERα) as the top regulator of pro-resilient transcriptional changes in the nucleus accumbens (NAc), a key brain reward region implicated in depression. In accordance with these findings, nuclear ERα protein levels are altered by stress in male and female mice. Further, overexpression of ERα in the NAc promotes stress resilience in both sexes. Subsequent RNA-sequencing reveals that ERα overexpression in NAc reproduces the transcriptional signature of resilience in male, but not female, mice. These results indicate that NAc ERα is an important regulator of pro-resilient transcriptional changes, but with sex-specific downstream targets.


Asunto(s)
Adaptación Psicológica/fisiología , Conducta Animal/fisiología , Depresión/fisiopatología , Receptor alfa de Estrógeno/metabolismo , Núcleo Accumbens/metabolismo , Estrés Psicológico/fisiopatología , Animales , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Factores Sexuales , Transcriptoma/genética
15.
Nat Commun ; 9(1): 477, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396460

RESUMEN

Major depressive disorder is associated with abnormalities in the brain and the immune system. Chronic stress in animals showed that epigenetic and inflammatory mechanisms play important roles in mediating resilience and susceptibility to depression. Here, through a high-throughput screening, we identify two phytochemicals, dihydrocaffeic acid (DHCA) and malvidin-3'-O-glucoside (Mal-gluc) that are effective in promoting resilience against stress by modulating brain synaptic plasticity and peripheral inflammation. DHCA/Mal-gluc also significantly reduces depression-like phenotypes in a mouse model of increased systemic inflammation induced by transplantation of hematopoietic progenitor cells from stress-susceptible mice. DHCA reduces pro-inflammatory interleukin 6 (IL-6) generations by inhibiting DNA methylation at the CpG-rich IL-6 sequences introns 1 and 3, while Mal-gluc modulates synaptic plasticity by increasing histone acetylation of the regulatory sequences of the Rac1 gene. Peripheral inflammation and synaptic maladaptation are in line with newly hypothesized clinical intervention targets for depression that are not addressed by currently available antidepressants.


Asunto(s)
Antocianinas/farmacología , Ácidos Cafeicos/farmacología , Epigénesis Genética , Glucósidos/farmacología , Inflamación/genética , Plasticidad Neuronal/genética , Estrés Psicológico/genética , Animales , Antocianinas/administración & dosificación , Ácidos Cafeicos/administración & dosificación , Islas de CpG/efectos de los fármacos , Depresión/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Glucósidos/administración & dosificación , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Antígenos Comunes de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Polifenoles/farmacología , Conducta Social , Estrés Psicológico/tratamiento farmacológico , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
17.
Neuropsychopharmacology ; 42(1): 62-80, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27291462

RESUMEN

Diagnostic criteria for mood disorders including major depressive disorder (MDD) largely ignore biological factors in favor of behavioral symptoms. Compounding this paucity of psychiatric biomarkers is a need for therapeutics to adequately treat the 30-50% of MDD patients who are unresponsive to traditional antidepressant medications. Interestingly, MDD is highly prevalent in patients suffering from chronic inflammatory conditions, and MDD patients exhibit higher levels of circulating pro-inflammatory cytokines. Together, these clinical findings suggest a role for the immune system in vulnerability to stress-related psychiatric illness. A growing body of literature also implicates the immune system in stress resilience and coping. In this review, we discuss the mechanisms by which peripheral and central immune cells act on the brain to affect stress-related neurobiological and neuroendocrine responses. We specifically focus on the roles of pro-inflammatory cytokine signaling, peripheral monocyte infiltration, microglial activation, and hypothalamic-pituitary-adrenal axis hyperactivity in stress vulnerability. We also highlight recent evidence suggesting that adaptive immune responses and treatment with immune modulators (exogenous glucocorticoids, humanized antibodies against cytokines) may decrease depressive symptoms and thus represent an attractive alternative to the current antidepressant treatments.


Asunto(s)
Citocinas/inmunología , Sistema Hipotálamo-Hipofisario/inmunología , Inflamación/inmunología , Microglía/inmunología , Monocitos/inmunología , Trastornos del Humor/inmunología , Sistema Hipófiso-Suprarrenal/inmunología , Estrés Psicológico/inmunología , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Psicológico/metabolismo
18.
Neuroscience ; 350: 180-189, 2017 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-28323008

RESUMEN

Men and women manifest different symptoms of depression and under current diagnostic criteria, depression is twice as prevalent in woman. However, little is known of the mechanisms contributing to these important sex differences. Sub-chronic variable stress (SCVS), a rodent model of depression, induces depression-like behaviors in female mice only, modeling clinical evidence of higher susceptibility to mood disorders in women. Accumulating evidence indicates that altered neuroplasticity of excitatory synapses in the nucleus accumbens (NAc) is a key pathophysiological feature of susceptibility to social stress in males. Here we investigated the effects of SCVS on pre- and post-synaptic protein levels and morphology of glutamatergic synapses of medium spiny neurons (MSNs) in the NAc of female and male mice. Animals underwent six-day exposure to alternating stressors including shock, tail suspension and restraint. MSNs from the NAc were filled with a Lucifer yellow dye and spine density and type were examined using NeuronStudio. In a separate group of animals, immunofluorescence staining was performed for vesicular glutamate transporter 1 (VGLUT1) and vesicular glutamate transporter 2 (VGLUT2), in order to label cortical and subcortical glutamatergic terminals. Immunostaining for post-synaptic density 95 (PSD95) was employed to evaluate post-synaptic density. Females demonstrated circuit-specific pre-synaptic alterations in VGLUT1 and VGLUT2 containing synapses that may contribute to stress susceptibility in the absence of post-synaptic alterations in PSD95 puncta, spine density or type. These data indicate that susceptibility to stress in females is associated with changes in the frequency of distinct glutamatergic inputs to the NAc.


Asunto(s)
Plasticidad Neuronal/fisiología , Núcleo Accumbens/metabolismo , Densidad Postsináptica/metabolismo , Caracteres Sexuales , Estrés Fisiológico , Sinapsis/metabolismo , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
19.
Sci Rep ; 7(1): 12838, 2017 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-28993631

RESUMEN

Numerous studies have employed repeated social defeat stress (RSDS) to study the neurobiological mechanisms of depression in rodents. An important limitation of RSDS studies to date is that they have been conducted exclusively in male mice due to the difficulty of initiating attack behavior directed toward female mice. Here, we establish a female mouse model of RSDS by inducing male aggression toward females through chemogenetic activation of the ventrolateral subdivision of the ventromedial hypothalamus (VMHvl). We demonstrate that females susceptible to RSDS display social avoidance, anxiety-like behavior, reduction of body weight, and elevated levels of circulating interleukin 6. In contrast, a subset of mice we term resilient only display anxiety-like behaviors after RSDS. This model allows for investigation of sex differences in the neurobiological mechanisms of defeat‒induced depression‒like behaviors. A robust female social defeat model is a critical first step in the identification and development of novel therapeutic compounds to treat depression and anxiety disorders in women.


Asunto(s)
Conducta Social , Estrés Psicológico/patología , Agresión , Animales , Reacción de Prevención , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Sensación
20.
Nat Med ; 23(9): 1102-1111, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28825715

RESUMEN

Major depressive disorder (MDD) is a leading cause of disease burden worldwide. While the incidence, symptoms and treatment of MDD all point toward major sex differences, the molecular mechanisms underlying this sexual dimorphism remain largely unknown. Here, combining differential expression and gene coexpression network analyses, we provide a comprehensive characterization of male and female transcriptional profiles associated with MDD across six brain regions. We overlap our human profiles with those from a mouse model, chronic variable stress, and capitalize on converging pathways to define molecular and physiological mechanisms underlying the expression of stress susceptibility in males and females. Our results show a major rearrangement of transcriptional patterns in MDD, with limited overlap between males and females, an effect seen in both depressed humans and stressed mice. We identify key regulators of sex-specific gene networks underlying MDD and confirm their sex-specific impact as mediators of stress susceptibility. For example, downregulation of the female-specific hub gene Dusp6 in mouse prefrontal cortex mimicked stress susceptibility in females, but not males, by increasing ERK signaling and pyramidal neuron excitability. Such Dusp6 downregulation also recapitulated the transcriptional remodeling that occurs in prefrontal cortex of depressed females. Together our findings reveal marked sexual dimorphism at the transcriptional level in MDD and highlight the importance of studying sex-specific treatments for this disorder.


Asunto(s)
Encéfalo/metabolismo , Trastorno Depresivo Mayor/genética , Estrés Psicológico/genética , Transcriptoma , Adulto , Anciano , Animales , Western Blotting , Estudios de Casos y Controles , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Persona de Mediana Edad , Núcleo Accumbens/metabolismo , Técnicas de Placa-Clamp , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Análisis de Secuencia de ARN , Caracteres Sexuales , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA