Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Neuroinflammation ; 20(1): 154, 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37380974

RESUMO

Brain vascular integrity is critical for brain health, and its disruption is implicated in many brain pathologies, including psychiatric disorders. Brain-vascular barriers are a complex cellular landscape composed of endothelial, glial, mural, and immune cells. Yet currently, little is known about these brain vascular-associated cells (BVACs) in health and disease. Previously, we demonstrated that 14 days of chronic social defeat (CSD), a mouse paradigm that produces anxiety and depressive-like behaviors, causes cerebrovascular damage in the form of scattered microbleeds. Here, we developed a technique to isolate barrier-related cells from the mouse brain and subjected the isolated cells to single-cell RNA sequencing. Using this isolation technique, we found an enrichment in BVAC populations, including distinct subsets of endothelial and microglial cells. In CSD compared to non-stress, home-cage control, differential gene expression patterns disclosed biological pathways involving vascular dysfunction, vascular healing, and immune system activation. Overall, our work demonstrates a unique technique to study BVAC populations from fresh brain tissue and suggests that neurovascular dysfunction is a key driver of psychosocial stress-induced brain pathology.


Assuntos
Encéfalo , Derrota Social , Animais , Camundongos , Sistema Imunitário , Barreira Hematoencefálica , Expressão Gênica
2.
Brain Behav Immun ; 101: 346-358, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35063606

RESUMO

Immune surveillance of the brain plays an important role in health and disease. Peripheral leukocytes patrol blood-brain barrier interfaces, and after injury, monocytes cross the cerebrovasculature and follow a pattern of pro- and anti-inflammatory activity leading to tissue repair. We have shown that chronic social defeat (CSD) causes scattered vasculature disruptions. Here, we assessed CCR2+ monocyte trafficking to the vascular injury sites in Ccr2wt/rfp reporter mice both during CSD and one week following CSD cessation. We found that CSD for 14 days induced microhemorrhages where plasma fibrinogen leaked into perivascular spaces, but it did not affect the distribution or density of CCR2rfp+ monocytes in the brain. However, after recovery from CSD, many vascularly adhered CCR2+ cells were detected, and gene expression of the CCR2 chemokine receptor ligands CCL7 and CCL12, but not CCL2, was elevated in endothelial cells. Adhered CCR2+ cells were mostly the non-classical, anti-inflammatory Ly6Clo type, and they phagocytosed fibrinogen in perivascular spaces. In CCR2-deficient Ccr2rfp/rfp mice, fibrinogen levels remained elevated in recovery. Fibrinogen infused intracerebroventricularly induced CCR2+ cells to adhere to the vasculature and phagocytose perivascular fibrinogen in Ccr2wt/rfp but not Ccr2rfp/rfp mice. Depletion of monocytes with clodronate liposomes during CSD recovery prevented fibrinogen clearance and blocked behavioral recovery. We hypothesize that peripheral CCR2+ monocytes are not elevated in the brain on day 14 at the end of CSD and do not contribute to its behavioral effects at that time, but in recovery following cessation of stress, they enter the brain and exert restorative functions mediating vascular repair and normalization of behavior.


Assuntos
Monócitos , Receptores CCR2 , Animais , Quimiocina CCL2/metabolismo , Células Endoteliais/metabolismo , Fibrinogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Receptores CCR2/metabolismo , Derrota Social
3.
Brain Behav Immun ; 97: 226-238, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34371135

RESUMO

There is increasing interest in how immune cells, including those within the meninges at the blood-brain interface, influence brain function and mood disorders, but little data on humoral immunity in this context. Here, we show that in mice exposed to psychosocial stress, there is increased splenic B cell activation and secretion of the immunoregulatory cytokine interleukin (IL)-10. Meningeal B cells were prevalent in homeostasis but substantially decreased following stress, whereas Ly6Chi monocytes increased, and meningeal myeloid cells showed augmented expression of activation markers. Single-cell RNA sequencing of meningeal B cells demonstrated the induction of innate immune transcriptional programmes following stress, including genes encoding antimicrobial peptides that are known to alter myeloid cell activation. Cd19-/- mice, that have reduced B cells, showed baseline meningeal myeloid cell activation and decreased exploratory behaviour. Together, these data suggest that B cells may influence behaviour by regulating meningeal myeloid cell activation.


Assuntos
Linfócitos B , Meninges , Animais , Apresentação de Antígeno , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides , Estresse Psicológico
4.
J Neurosci ; 39(28): 5594-5605, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31085604

RESUMO

Chronic social defeat (CSD) in male mice can produce anxiety and aberrant socialization. Animals susceptible to CSD show activation of microglia, which have elevated levels of oxidative stress markers. We hypothesized that microglia and reactive oxygen species (ROS) production contribute to the CSD stress-induced changes in affective behavior. First, we selectively depleted microglia (99%) by administering the CSF1R (colony-stimulating factor 1 receptor) antagonist PLX5622 before and during the 14 d CSD procedure. Microglia-depleted mice in contrast to nondepleted mice were protected from the stress effects measured by light/dark and social interaction tests. ROS production, measured histochemically following dihydroethidium administration, was elevated by CSD, and the production was reduced to basal levels in mice lacking microglia. The deleterious stress effects were also blocked in nondepleted mice by continuous intracerebral administration of N-acetylcysteine (NAC), a ROS inhibitor. In a second experiment, at the end of the CSD period, PLX5622 was discontinued to allow microglial repopulation. After 14 d, the brain had a full complement of newly generated microglia. At this time, the mice that had previously been protected now showed behavioral deficits, and their brain ROS production was elevated, both in all brain cells and in repopulated microglia. NAC administration during repopulation prevented the behavioral decline in the repopulated mice, and it supported behavioral recovery in nondepleted stressed mice. The data suggest that microglia drive elevated ROS production during and after stress exposure. This elevated ROS activity generates a central state supporting dysregulated affect, and it hinders the restoration of behavioral and neurochemical homeostasis after stress cessation.SIGNIFICANCE STATEMENT Chronic psychosocial stress is associated with psychiatric disorders such as depression and anxiety. Understanding the details of CNS cellular contributions to stress effects could lead to the development of intervention strategies. Inflammation and oxidative stress are positively linked to depression severity, but the cellular nature of these processes is not clear. The chronic social defeat (CSD) paradigm in mice produces mood alterations and microglial activation characterized by elevated reactive oxygen species (ROS) production. The depletion of microglia or ROS inhibition prevented adverse stress effects. Microglial repopulation of the brain post-CSD reintroduced adverse stress effects, and ROS inhibition in this phase protected against the effects. The results suggest that stress-induced microglial ROS production drives a central state that supports dysregulated affective behavior.


Assuntos
Microglia/metabolismo , Estresse Oxidativo , Comportamento Social , Estresse Psicológico/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Compostos Orgânicos/toxicidade , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores
5.
J Neuroinflammation ; 17(1): 140, 2020 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-32359360

RESUMO

BACKGROUND: Cyclooxygenase-2 (COX-2), which is rapidly upregulated by inflammation, is a key enzyme catalyzing the rate-limiting step in the synthesis of several inflammatory prostanoids. Successful positron emission tomography (PET) radioligand imaging of COX-2 in vivo could be a potentially powerful tool for assessing inflammatory response in the brain and periphery. To date, however, the development of PET radioligands for COX-2 has had limited success. METHODS: The novel PET tracer [11C]MC1 was used to examine COX-2 expression [1] in the brains of four rhesus macaques at baseline and after injection of the inflammogen lipopolysaccharide (LPS) into the right putamen, and [2] in the joints of two human participants with rheumatoid arthritis and two healthy individuals. In the primate study, two monkeys had one LPS injection, and two monkeys had a second injection 33 and 44 days, respectively, after the first LPS injection. As a comparator, COX-1 expression was measured using [11C]PS13. RESULTS: COX-2 binding, expressed as the ratio of specific to nondisplaceable uptake (BPND) of [11C]MC1, increased on day 1 post-LPS injection; no such increase in COX-1 expression, measured using [11C]PS13, was observed. The day after the second LPS injection, a brain lesion (~ 0.5 cm in diameter) with high COX-2 density and high BPND (1.8) was observed. Postmortem brain analysis at the gene transcript or protein level confirmed in vivo PET results. An incidental finding in an unrelated monkey found a line of COX-2 positivity along an incision in skull muscle, demonstrating that [11C]MC1 can localize inflammation peripheral to the brain. In patients with rheumatoid arthritis, [11C]MC1 successfully imaged upregulated COX-2 in the arthritic hand and shoulder and apparently in the brain. Uptake was blocked by celecoxib, a COX-2 preferential inhibitor. CONCLUSIONS: Taken together, these results indicate that [11C]MC1 can image and quantify COX-2 upregulation in both monkey brain after LPS-induced neuroinflammation and in human peripheral tissue with inflammation. TRIAL REGISTRATION: ClinicalTrials.gov NCT03912428. Registered April 11, 2019.


Assuntos
Ciclo-Oxigenase 2/análise , Inflamação/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Pirimidinas , Compostos Radiofarmacêuticos , Adulto , Animais , Artrite Reumatoide/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Feminino , Humanos , Macaca mulatta , Pessoa de Meia-Idade
6.
Brain Behav Immun ; 88: 735-747, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32413560

RESUMO

Psychological stress and affective disorders are clinically associated with hypertension and vascular disease, but the biological links between the conditions have not been fully explored. To examine this relationship, we used chronic social defeat (CSD) stress, which produces anxiety-like and depressive-like behavioral declines in susceptible mice. In such mice, CSD also produces cerebrovascular microbleeds in scattered locations. Here, we showed further evidence of vascular pathology and blood-brain barrier breakdown by visualizing plasma immunoglobulins and erythrocytes within the parenchyma and perivascular spaces of CSD brains. To further characterize the impact of stress on the cerebrovasculature, brain endothelial cells (bECs) were isolated, and global gene expression profiles were generated. Bioinformatic analysis of CSD-induced transcriptional changes in bECs showed enrichment in pathways that delineate the vascular response to injury. These pathways followed a temporal sequence of inflammation, oxidative stress, growth factor signaling, and wound healing (i.e., platelet aggregation, hemostasis, fibrinogen deposition, and angiogenesis). Immunohistochemical staining for markers of fibrinogen deposition and angiogenesis confirmed the existence of the markers at the sites of vascular disruptions. Recovery after CSD cessation was marked by recruitment of leukocytes perhaps participating in vascular repair. The data suggest that co-morbidity of affective disorders and vascular diseases may be attributed in part to a common link in altered endothelial cell function.


Assuntos
Derrota Social , Animais , Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Células Endoteliais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico
7.
J Neurosci ; 35(4): 1530-8, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25632130

RESUMO

We examined whether cells of the adaptive immune system retain the memory of psychosocial stress and thereby alter mood states and CNS function in the host. Lymphocytes from mice undergoing chronic social defeat stress or from unstressed control mice were isolated and adoptively transferred into naive lymphopenic Rag2(-/-) mice. Changes in affective behavior, hippocampal cell proliferation, microglial activation states, and blood cytokine levels were examined in reconstituted stress-naive mice. The mice receiving lymphocytes from defeated donors showed less anxiety, more social behavior, and increased hippocampal cell proliferation compared with those receiving no cells or cells from unstressed donors. Mice receiving stressed immune cells had reduced pro-inflammatory cytokine levels in the blood relative to the other groups, an effect opposite to the elevated donor pro-inflammatory cytokine profile. Furthermore, mice receiving stressed immune cells had microglia skewed toward an anti-inflammatory, neuroprotective M2-like phenotype, an effect opposite the stressed donors' M1-like pro-inflammatory profile. However, stress had no effect on lymphocyte surface marker profiles in both donor and recipient mice. The data suggest that chronic stress-induced changes in the adaptive immune system, contrary to conferring anxiety and depressive behavior, protect against the deleterious effects of stress. Improvement in affective behavior is potentially mediated by reduced peripheral pro-inflammatory cytokine load, protective microglial activity, and increased hippocampal cell proliferation. The data identify the peripheral adaptive immune system as putatively involved in the mechanisms underlying stress resilience and a potential basis for developing novel rapid-acting antidepressant therapies.


Assuntos
Transferência Adotiva , Antidepressivos/uso terapêutico , Linfócitos/fisiologia , Estresse Psicológico/imunologia , Estresse Psicológico/terapia , Animais , Antidepressivos/farmacologia , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Corticosterona/sangue , Citocinas/sangue , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Adaptação à Escuridão/efeitos dos fármacos , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Psicológico/sangue , Estresse Psicológico/psicologia , Urina/química
8.
J Neuroinflammation ; 13(1): 224, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27581371

RESUMO

BACKGROUND: We are interested in the causal interactions between psychological stress and activity within different compartments of the immune system. Psychosocial stress has been reported to not only alter microglia morphology but also produce anxiety-like and depressive-like effects by triggering CNS infiltration of macrophages from the periphery. We sought to test these phenomena in a somewhat different but standardized model of chronic social defeat (SD) stress. METHODS: We used a paradigm of dyadic home pairing of dominant and subordinate mice that has been validated to induce powerful anxiety-like and depressive-like effects manifested by behavior assessed in social tasks. We administered the SD stress for 3 days (acute SD) or 14 days (chronic SD) and looked for monocyte entry into the brain by three independent means, including CD45 activation states assessed by flow cytometry and tracking fluorescently tagged peripheral cells from Ccr2 (wt/rfp) and Ubc (gfp/gfp) reporter mice. We further characterized the effects of SD stress on microglia using quantitative morphometric analysis, ex vivo phagocytosis assays, flow cytometry, and immunochemistry. RESULTS: We saw no evidence of stress-induced macrophage entry after acute or chronic defeat stress. In comparison, brain infiltration of peripheral cells did occur after endotoxin administration. Furthermore, mutant mice lacking infiltrating macrophages due to CCR2 knockout developed the same degree of chronic SD-induced depressive behavior as wildtype mice. We therefore focused more closely on the intrinsic immune cells, the microglia. Using Cx3cr1 (wt/gpf) microglial reporter mice, we saw by quantitative methods that microglial morphology was not altered by stress at either time point. However, chronic SD mice had elevated numbers of CD68(hi) microglia examined by flow cytometry. CD68 is a marker for phagocytic activity. Indeed, these cells ex vivo showed elevated phagocytosis, confirming the increased activation status of chronic SD microglia. Finally, acute SD but not chronic SD increased microglial proliferation, which occurred selectively in telencephalic stress-related brain areas. CONCLUSIONS: In the SD paradigm, changes in CNS-resident microglia numbers and activation states might represent the main immunological component of the psychosocial stress-induced depressive state.


Assuntos
Depressão/patologia , Macrófagos/patologia , Microglia/patologia , Comportamento Social , Estresse Psicológico/patologia , Animais , Depressão/imunologia , Depressão/metabolismo , Feminino , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/imunologia , Microglia/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia
9.
J Neurosci ; 33(7): 2961-72, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23407954

RESUMO

Both social defeat stress and environmental enrichment stimulate adrenal glucocorticoid secretion, but they have opposing effects on hippocampal neurogenesis and mood. Hypothalamic-pituitary-adrenal axis dysregulation and decreased neurogenesis are consequences of social defeat. These outcomes are correlated with depressive states, but a causal role in the etiology of depression remains elusive. The antidepressant actions of environmental enrichment are neurogenesis-dependent, but the contribution of enrichment-elevated glucocorticoids is unexplored. Importantly, for both social defeat and environmental enrichment, how glucocorticoids interact with neurogenesis to alter mood is unknown. Here, we investigate causal roles of glucocorticoids and neurogenesis in induction of depressive-like behavior and its amelioration by environmental enrichment in mice. By blocking neurogenesis and surgically clamping adrenal hormone secretions, we showed that neurogenesis, via hypothalamic-pituitary-adrenal axis interactions, is directly involved in precipitating the depressive phenotype after social defeat. Mice adrenalectomized before social defeat showed enhanced behavioral resiliency and increased survival of adult-born hippocampal neurons compared with sham-operated defeated mice. However, mice lacking hippocampal neurogenesis did not show protective effects of adrenalectomy. Moreover, glucocorticoids secreted during environmental enrichment promoted neurogenesis and were required for restoration of normal behavior after social defeat. The data demonstrate that glucocorticoid-dependent declines in neurogenesis drive changes in mood after social defeat and that glucocorticoids secreted during enrichment promote neurogenesis and restore normal behavior after defeat. These data provide new evidence for direct involvement of neurogenesis in the etiology of depression, suggesting that treatments promoting neurogenesis can enhance stress resilience.


Assuntos
Afeto/fisiologia , Glucocorticoides/fisiologia , Neurogênese/fisiologia , Adaptação Psicológica/fisiologia , Adrenalectomia , Animais , Antimetabólitos , Comportamento Animal/fisiologia , Bromodesoxiuridina , Corticosterona/metabolismo , Corticosterona/farmacologia , Depressão/psicologia , Meio Ambiente , Abrigo para Animais , Camundongos , Camundongos Endogâmicos C57BL , Resiliência Psicológica , Comportamento Social , Predomínio Social , Estresse Psicológico/psicologia
10.
J Neurosci ; 31(16): 6159-73, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21508240

RESUMO

Enriched environmental (EE) housing dampens stress-induced alterations in neurobiological systems, promotes adaptability, and extinguishes submissive behavioral traits developed during social defeat stress (SD). In the present study, we hypothesized that enrichment before SD can confer stress resiliency and, furthermore, that neuronal activity in the prefrontal cortex (PFC) is requisite for this resiliency. To test these hypotheses, mice were housed in EE, standard (SE), or impoverished (IE) housing and then exposed to SD. EE conferred resilience to SD as measured in several behavioral tasks. EE-housed mice expressed elevated FosB/ΔFosB immunostaining in areas associated with emotional regulation and reward processing, i.e., infralimbic, prelimbic, and anterior cingulate cortices, amygdala, and nucleus accumbens, and this expression was mostly preserved in mice receiving EE followed by SD. In contrast, in SE- or IE-housed animals, SD increased maladaptive behaviors and greatly reduced FosB/ΔFosB staining in the forebrain. We tested the putative involvement of the PFC in mediating resilience by lesioning individual regions of the PFC either before or after EE housing and then exposing the mice to SD. We found that discrete lesions of the infralimbic but not prelimbic or cingulate cortex made before but not after EE abolished the behavioral resiliency to stress afforded by EE and attenuated FosB/ΔFosB expression in the accumbens and amygdala while increasing it in the paraventricular hypothalamic nucleus. These data suggest that pathological ventromedial PFC outputs to downstream limbic targets could predispose an individual to anxiety disorders in stressful situations, whereas enhanced ventromedial PFC outputs could convey stress resilience.


Assuntos
Córtex Cerebral/fisiologia , Dominação-Subordinação , Meio Ambiente , Sistema Límbico/fisiologia , Rede Nervosa/fisiologia , Resiliência Psicológica , Análise de Variância , Animais , Abrigo para Animais , Imuno-Histoquímica , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Psicológico/fisiopatologia
11.
Neuron ; 109(23): 3793-3809.e8, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34614419

RESUMO

Psychosocial stress is a common risk factor for anxiety disorders. The cellular mechanism for the anxiogenic effect of psychosocial stress is largely unclear. Here, we show that chronic social defeat (CSD) stress in mice causes mitochondrial impairment, which triggers the PINK1-Parkin mitophagy pathway selectively in the amygdala. This mitophagy elevation causes excessive mitochondrial elimination and consequent mitochondrial deficiency. Mitochondrial deficiency in the basolateral amygdalae (BLA) causes weakening of synaptic transmission in the BLA-BNST (bed nucleus of the stria terminalis) anxiolytic pathway and increased anxiety. The CSD-induced increase in anxiety-like behaviors is abolished in Pink1-/- and Park2-/- mice and alleviated by optogenetic activation of the BLA-BNST synapse. This study identifies an unsuspected role of mitophagy in psychogenetic-stress-induced anxiety elevation and reveals that mitochondrial deficiency is sufficient to increase anxiety and underlies the psychosocial-stress-induced anxiety increase. Mitochondria and mitophagy, therefore, can be potentially targeted to ameliorate anxiety.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Mitofagia , Animais , Ansiedade , Transtornos de Ansiedade , Complexo Nuclear Basolateral da Amígdala/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
12.
Brain Behav Immun ; 24(6): 1008-17, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20399847

RESUMO

The role of altered activity of nuclear factor kappaB (NF-kappaB) in specific aspects of motivated behavior and learning and memory was examined in mice lacking the p50 subunit of the NF-kappaB/rel transcription factor family. Nfkb1-deficient mice are unable to produce p50 and show specific susceptibilities to infections and inflammatory challenges, but the behavioral phenotype of such mice has been largely unexamined, owing in large part to the lack of understanding of the role of NF-kappaB in nervous system function. Here we show that Nfkb1 (p50) knockout mice more rapidly learned to find the hidden platform in the Morris water maze than did wildtype mice. The rise in plasma corticosterone levels after the maze test was greater in p50 knockout than in wildtype mice. In the less stressful Barnes maze, which tests similar kinds of spatial learning, the p50 knockout mice performed similarly to control mice. Adrenalectomy with corticosterone replacement eliminated the differences between p50 knockout and wildtype mice in the water maze. Knockout mice showed increased levels of basal anxiety in the open-field and light/dark box tests, suggesting that their enhanced escape latency in the water maze was due to activation of the stress (hypothalamic-pituitary-adrenal) axis leading to elevated corticosterone production by strongly but not mildly anxiogenic stimuli. The results suggest that, as in the immune system, p50 in the nervous system normally serves to dampen NF-kappaB-mediated intracellular activities, which are manifested physiologically through elevated stress responses to aversive stimuli and behaviorally in the facilitated escape performance in learning tasks.


Assuntos
Aprendizagem da Esquiva/fisiologia , NF-kappa B/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Adrenalectomia , Animais , Ansiedade/genética , Ansiedade/psicologia , Aprendizagem da Esquiva/efeitos dos fármacos , Quimiocina CXCL1/biossíntese , Corticosterona/sangue , Ensaio de Desvio de Mobilidade Eletroforética , Comportamento Exploratório/fisiologia , Genes Precoces/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/biossíntese , NF-kappa B/fisiologia , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/fisiologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Estresse Psicológico/genética
13.
Mucosal Immunol ; 13(2): 216-229, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31772323

RESUMO

Mononuclear phagocytes are a heterogeneous population of leukocytes essential for immune homeostasis that develop tissue-specific functions due to unique transcriptional programs driven by local microenvironmental cues. Single cell RNA sequencing (scRNA-seq) of colonic myeloid cells from specific pathogen free (SPF) and germ-free (GF) C57BL/6 mice revealed extensive heterogeneity of both colon macrophages (MPs) and dendritic cells (DCs). Modeling of developmental pathways combined with inference of gene regulatory networks indicate two major trajectories from common CCR2+ precursors resulting in colon MP populations with unique transcription factors and downstream target genes. Compared to SPF mice, GF mice had decreased numbers of total colon MPs, as well as selective proportional decreases of two major CD11c+CD206intCD121b+ and CD11c-CD206hiCD121b- colon MP populations, whereas DC numbers and proportions were not different. Importantly, these two major colon MP populations were clearly distinct from other colon MP populations regarding their gene expression profile, localization within the lamina propria (LP) and ability to phagocytose macromolecules from the blood. These data uncover the diversity of intestinal myeloid cell populations at the molecular level and highlight the importance of microbiota on the unique developmental as well as anatomical and functional fates of colon MPs.


Assuntos
Colo/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Células Mieloides/fisiologia , Animais , Antígeno CD11c/metabolismo , Diferenciação Celular , Células Cultivadas , Ontologia Genética , Redes Reguladoras de Genes , Homeostase , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/genética , Fenótipo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma
14.
Sci Rep ; 8(1): 11240, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30050134

RESUMO

An animal's ability to cope with or succumb to deleterious effects of chronic psychological stress may be rooted in the brain's immune responses manifested in microglial activity. Mice subjected to chronic social defeat (CSD) were categorized as susceptible (CSD-S) or resilient (CSD-R) based on behavioral phenotyping, and their microglia were isolated and analyzed by microarray. Microglia transcriptomes from CSD-S mice were enriched for pathways associated with inflammation, phagocytosis, oxidative stress, and extracellular matrix remodeling. Histochemical experiments confirmed the array predictions: CSD-S microglia showed elevated phagocytosis and oxidative stress, and the brains of CSD-S but not CSD-R or non-stressed control mice showed vascular leakage of intravenously injected fluorescent tracers. The results suggest that the inflammatory profile of CSD-S microglia may be precipitated by extracellular matrix degradation, oxidative stress, microbleeds, and entry and phagocytosis of blood-borne substances into brain parenchyma. We hypothesize that these CNS-centric responses contribute to the stress-susceptible behavioral phenotype.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Microglia/imunologia , Microglia/patologia , Estresse Psicológico/fisiopatologia , Animais , Comportamento Animal , Perfilação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Análise em Microsséries
15.
Biochem Pharmacol ; 73(8): 1063-75, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17097068

RESUMO

Central alpha(1)-adrenoceptors are activated by norepinephrine (NE), epinephrine (EPI) and possibly dopamine (DA), and function in two fundamental and opposed types of behavior: (1) positively motivated exploratory and approach activities, and (2) stress reactions and behavioral inhibition. Brain microinjection studies have revealed that the positive-linked receptors are located in eight to nine brain regions spanning the neuraxis including the secondary motor cortex, piriform cortex, nucleus accumbens, preoptic area, lateral hypothalamic area, vermis cerebellum, locus coeruleus, dorsal raphe and possibly the C1 nucleus of the ventrolateral medulla, whereas the stress-linked receptors are present in at least three areas including the paraventricular nucleus of the hypothalamus, central nucleus of the amygdala and bed nucleus of the stria terminalis. Recent studies utilizing c-fos expression and mitogen-activated protein kinase activation have shown that various diverse models of depression in mice produce decreases in positive region-neural activity elicited by motivating stimuli along with increases in neural activity of stress areas. Both types of change are attenuated by various antidepressant agents. This has suggested that the balance of the two networks determines whether an animal displays depressive behavior. A central unresolved question concerns how the alpha(1)-receptors in the positive-activity and stress systems are differentially activated during the appropriate behavioral conditions and to what extent this is related to differences in endogenous ligands or receptor subtype distributions.


Assuntos
Comportamento Animal/fisiologia , Depressão/fisiopatologia , Atividade Motora/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Animais , Encéfalo/metabolismo , Depressão/metabolismo , Camundongos , Motivação , Ratos , Receptores Adrenérgicos alfa 1/metabolismo , Tropismo/fisiologia
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(6): 1196-207, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17513031

RESUMO

A previous study showed that two mouse models of behavioral depression, immune system activation and depletion of brain monoamines, are accompanied by marked reductions in stimulated neural activity in brain regions involved in motivated behavior. The present study tested whether this effect is common to other depression models by examining the effects of repeated forced swimming, chronic subordination stress or acute intraventricular galanin injection - three additional models - on baseline or stimulated c-fos expression in several brain regions known to be involved in motor or motivational processes (secondary motor, M2, anterior piriform cortex, APIR, posterior cingulate gyrus, CG, nucleus accumbens, NAC). Each of the depression models was found to reduce the fos response stimulated by exposure to a novel cage or a swim stress in all four of these brain areas but not to affect the response of a stress-sensitive region (paraventricular hypothalamus, PVH) that was included for control purposes. Baseline fos expression in these structures was either unaffected or affected in an opposite direction to the stimulated response. Pretreatment with either desmethylimipramine (DMI) or tranylcypromine (tranyl) attenuated these changes. It is concluded that the pattern of a reduced neural function of CNS motor/motivational regions with an increased function of stress areas is common to 5 models of behavioral depression in the mouse and is a potential experimental analog of the neural activity changes occurring in the clinical condition.


Assuntos
Depressão/metabolismo , Depressão/patologia , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Análise de Variância , Animais , Antidepressivos/administração & dosagem , Comportamento Animal , Depressão/etiologia , Depressão/prevenção & controle , Desipramina/administração & dosagem , Modelos Animais de Doenças , Dominação-Subordinação , Reação de Congelamento Cataléptica/efeitos dos fármacos , Reação de Congelamento Cataléptica/fisiologia , Galanina , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Estresse Psicológico/complicações , Natação/psicologia , Fatores de Tempo , Tranilcipromina/administração & dosagem
17.
Sci Rep ; 7: 46548, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28418035

RESUMO

The medial prefrontal cortex (mPFC) plays a key role in top-down control of the brain's stress axis, and its structure and function are particularly vulnerable to stress effects, which can lead to depression in humans and depressive-like states in animals. We tested whether chronic social defeat produces structural alterations in the mPFC in mice. We first performed a microarray analysis of mPFC gene expression changes induced by defeat, and biological pathway analysis revealed a dominant pattern of down-regulation of myelin-associated genes. Indeed, 69% of the most significantly down-regulated genes were myelin-related. The down regulation was confirmed by in situ hybridization histochemistry for two strongly down-regulated genes, myelin oligodendrocyte glycoprotein (Mog) and ermin (Ermn), and by immunohistochemistry for myelin basic protein. To test for stress-induced changes in myelin integrity, aurophosphate (Black Gold) myelin staining was performed on mPFC sections. Quantitative stereologic analysis showed reduced myelinated fiber length and density. Behavioral analysis confirmed that the 14-day social defeat sessions resulted in induction of depressive-like states measured in social interaction and light/dark tests. The combined data suggest that chronic social defeat induces molecular changes that reduce myelination of the prefrontal cortex, which may be an underlying basis for stress-induced depressive states.


Assuntos
Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Animais , Masculino , Camundongos
18.
J Neurosci ; 25(38): 8696-703, 2005 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16177038

RESUMO

Female rats modulate the number and interval between the intromissions the female receives during mating. This patterned vaginocervical stimulation (VCS) is critical for triggering long-term changes in prolactin (PRL) secretion necessary for pregnancy or pseudopregnancy (P/PSP). Previous work has shown that NMDA receptor activation in the posterodorsal medial amygdala (MEApd) is required at the time of mating for VCS to induce the twice-daily PRL surges characteristic of P/PSP. The current studies examined whether patterned activation of glutamate receptors within the MEApd induces P/PSP. In anesthetized, cycling females, three 0.27 microg NMDA infusions given at 30 min intervals into the MEApd initiated P/PSP, whereas a single NMDA infusion of the same total dose (0.8 microg) had no effect. In conscious, freely behaving females, three infusions of an excitatory amino acid (EAA) mixture applied at the same interval were more effective in initiating P/PSP and nocturnal PRL surges than were single infusions at the same or higher concentrations. Infusion intervals of 5 and 60 min as well as continuous 1 h infusion did not induce P/PSP. Finally, a synergistic effect was observed between EAA and mating stimulation, because a subthreshold EAA infusion combined with subthreshold numbers of intromissions induced P/PSP. These results demonstrate that repeated, properly spaced, temporally discreet periods of glutamate receptor activation within the MEApd, which mimic mating stimulation, encode for P/PSP. Such findings suggest that single intromissions normally release individually subthreshold quanta of glutamate within the MEApd that summate to induce P/PSP.


Assuntos
Tonsila do Cerebelo/metabolismo , Pseudogravidez/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Comportamento Sexual Animal/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Feminino , Masculino , N-Metilaspartato/farmacologia , Ratos , Ratos Long-Evans , Receptores de N-Metil-D-Aspartato/agonistas , Comportamento Sexual Animal/efeitos dos fármacos , Fatores de Tempo
19.
Biol Psychiatry ; 60(8): 803-11, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16814258

RESUMO

BACKGROUND: Immune stimulation inhibits positively motivated behavior and induces depressive illness. To help clarify the mechanism of these effects, neural activity in response to a positive stimulus was examined in brain regions associated with positively motivated activity defined on the basis of prior behavioral studies of central alpha1-adrenoceptor action. METHODS: Mice pretreated with either lipopolysaccharide or, for comparison, reserpine were exposed to a motivating stimulus (fresh cage) and subsequently assayed for fos expression and mitogen-activated protein kinase (MAPK) phosphorylation, two measures associated with alpha1-adrenoceptor-dependent neural activity, in several positive-activity-related (motor, piriform, cingulate cortex, nucleus accumbens, locus coeruleus) and stress-related brain regions (paraventricular hypothalamus, bed nucleus stria terminalis). RESULTS: Both lipopolysaccharide and reserpine pretreatment abolished fresh cage-induced fos expression and MAPK activation in the positive activity-related brain regions but enhanced these measures in the stress-related areas. CONCLUSIONS: The results support the hypothesis that immune activation reduces alpha1-adrenoceptor-related signaling and neural activity in brain regions associated with positive activity while it increases these functions in stress-associated areas. It is suggested that neural activities of these two types of brain regions are mutually antagonistic and that a reciprocal shift toward the stress regions is a factor in the loss of positively motivated behaviors in sickness behavior and depressive illness.


Assuntos
Adjuvantes Imunológicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Depressão/induzido quimicamente , Depressão/psicologia , Motivação , Neurônios/efeitos dos fármacos , Inibidores da Captação Adrenérgica/farmacologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-fos/biossíntese , Reserpina/farmacologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-27109071

RESUMO

Our group has recently provided novel insights into a poorly understood component of intercommunication between the brain and the immune system by showing that psychological stress can modify lymphocytes in a manner that may boost resilience to psychological stress. To demonstrate the influence of the adaptive immune system on mood states, we previously showed that cells from lymph nodes of socially defeated mice, but not from unstressed mice, conferred anxiolytic and antidepressant-like effects and elevated hippocampal cell proliferation when transferred into naïve lymphopenic Rag2(-/-) mice. In the present study, we asked whether similar transfer could be anxiolytic and antidepressant when done in animals that had been rendered anxious and depressed by chronic psychological stress. First, we demonstrated that lymphopenic Rag2(-/-) mice and their wild-type C57BL/6 mouse counterparts had similar levels of affect normally. Second, we found that following chronic (14days) restraint stress, both groups displayed an anxious and depressive-like phenotype and decreased hippocampal cell proliferation. Third, we showed that behavior in the open field test and light/dark box was normalized in the restraint-stressed Rag2(-/-) mice following adoptive transfer of lymph node cells from green fluorescent protein (GFP) expressing donor mice previously exposed to chronic (14days) of social defeat stress. Cells transferred from unstressed donor mice had no effect on behavior. Immunolabeling of GFP+ cells confirmed that tissue engraftment had occurred at 14days after transfer. We found GFP+ lymphocytes in the spleen, lymph nodes, blood, choroid plexus, and meninges of the recipient Rag2(-/-) mice. The findings suggest that the adaptive immune system may play a key role in promoting recovery from chronic stress. The data support using lymphocytes as a novel therapeutic target for anxiety states.


Assuntos
Transfusão de Linfócitos , Linfócitos/fisiologia , Estresse Psicológico/imunologia , Estresse Psicológico/terapia , Animais , Ansiedade , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/imunologia , Hipocampo/patologia , Linfonodos/imunologia , Linfonodos/patologia , Transfusão de Linfócitos/métodos , Linfócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Neurogênese/fisiologia , Neurônios/imunologia , Neurônios/patologia , Restrição Física , Baço/imunologia , Baço/patologia , Estresse Psicológico/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA