Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Rheumatology (Oxford) ; 62(10): 3469-3479, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36802235

RESUMO

OBJECTIVE: Trained immunity (TI) is a de facto memory program of innate immune cells, characterized by immunometabolic and epigenetic changes sustaining enhanced production of cytokines. TI evolved as a protective mechanism against infections; however, inappropriate activation can cause detrimental inflammation and might be implicated in the pathogenesis of chronic inflammatory diseases. In this study, we investigated the role of TI in the pathogenesis of giant cell arteritis (GCA), a large-vessel vasculitis characterized by aberrant macrophage activation and excess cytokine production. METHODS: Monocytes from GCA patients and from age- and sex-matched healthy donors were subjected to polyfunctional studies, including cytokine production assays at baseline and following stimulation, intracellular metabolomics, chromatin immunoprecipitation-qPCR, and combined ATAC/RNA sequencing. Immunometabolic activation (i.e. glycolysis) was assessed in inflamed vessels of GCA patients with FDG-PET and immunohistochemistry (IHC), and the role of this pathway in sustaining cytokine production was confirmed with selective pharmacologic inhibition in GCA monocytes. RESULTS: GCA monocytes exhibited hallmark molecular features of TI. Specifically, these included enhanced IL-6 production upon stimulation, typical immunometabolic changes (e.g. increased glycolysis and glutaminolysis) and epigenetic changes promoting enhanced transcription of genes governing pro-inflammatory activation. Immunometabolic changes of TI (i.e. glycolysis) were a feature of myelomonocytic cells in GCA lesions and were required for enhanced cytokine production. CONCLUSIONS: Myelomonocytic cells in GCA activate TI programs sustaining enhanced inflammatory activation with excess cytokine production.


Assuntos
Arterite de Células Gigantes , Humanos , Arterite de Células Gigantes/patologia , Monócitos/metabolismo , Imunidade Treinada , Inflamação , Citocinas
2.
Blood ; 138(17): 1554-1569, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34077954

RESUMO

Trained immunity (TI) is a proinflammatory program induced in monocyte/macrophages upon sensing of specific pathogens and is characterized by immunometabolic and epigenetic changes that enhance cytokine production. Maladaptive activation of TI (ie, in the absence of infection) may result in detrimental inflammation and development of disease; however, the exact role and extent of inappropriate activation of TI in the pathogenesis of human diseases is undetermined. In this study, we uncovered the oncogene-induced, maladaptive induction of TI in the pathogenesis of a human inflammatory myeloid neoplasm (Erdheim-Chester disease, [ECD]), characterized by the BRAFV600E oncogenic mutation in monocyte/macrophages and excess cytokine production. Mechanistically, myeloid cells expressing BRAFV600E exhibit all molecular features of TI: activation of the AKT/mammalian target of rapamycin signaling axis; increased glycolysis, glutaminolysis, and cholesterol synthesis; epigenetic changes on promoters of genes encoding cytokines; and enhanced cytokine production leading to hyperinflammatory responses. In patients with ECD, effective therapeutic strategies combat this maladaptive TI phenotype; in addition, pharmacologic inhibition of immunometabolic changes underlying TI (ie, glycolysis) effectively dampens cytokine production by myeloid cells. This study revealed the deleterious potential of inappropriate activation of TI in the pathogenesis of human inflammatory myeloid neoplasms and the opportunity for inhibition of TI in conditions characterized by maladaptive myeloid-driven inflammation.


Assuntos
Doença de Erdheim-Chester/genética , Inflamação/genética , Proteínas Proto-Oncogênicas B-raf/genética , Células Cultivadas , Epigênese Genética , Doença de Erdheim-Chester/imunologia , Doença de Erdheim-Chester/patologia , Humanos , Imunidade , Inflamação/imunologia , Inflamação/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Oncogenes , Mutação Puntual , Proteínas Proto-Oncogênicas B-raf/imunologia
3.
Cell Mol Neurobiol ; 42(4): 1225-1240, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-33259004

RESUMO

One of the most substantial and established environmental risk factors for neurological and psychiatric disorders is stress exposure, whose detrimental consequences hinge on several variables including time. In this regard the gestational period is known to present an intrinsic vulnerability to environmental insults and thus stressful events during pregnancy can lead to severe consequences on the offspring's brain development with long-term repercussions throughout adulthood. On this basis, we investigated the long-lasting impact of prenatal stress exposure on the susceptibility to the experimental autoimmune encephalomyelitis (EAE), a well-established murine model of multiple sclerosis. Although stress is considered a triggering factor for this chronic, progressive, autoimmune disease, little is known about the underlying mechanisms. To this end, EAE was induced by immunization with MOG35-55/CFA and pertussis toxin administration in adult female C57BL/6 mice born from control or stressed dams exposed to restraint stress during the last days of gestation. Our results demonstrate that gestational stress induces a marked increase in the severity of EAE symptoms in adulthood. Further, we highlight an altered maturation of oligodendrocytes in the spinal cord of prenatally stressed EAE mice, as indicated by the higher levels of GPR17, a marker of immature oligodendrocyte precursor cells. These behavioral and molecular alterations are paralleled by changes in the expression and signaling of the neurotrophin BDNF, an important mediator of neural plasticity that may contribute to stress-induced impaired remyelination. Since several already marketed drugs are able to modulate BDNF levels, these results pave the way to the possibility of repositioning these drugs in multiple sclerosis.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Medula Espinal/metabolismo
4.
Int J Mol Sci ; 23(20)2022 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-36293308

RESUMO

Neuroinflammation has emerged as an important factor in the molecular underpinnings of major depressive disorder (MDD) pathophysiology and in the mechanism of action of antidepressants. Among the inflammatory mediators dysregulated in depressed patients, interleukin (IL)-6 has recently been proposed to play a crucial role. IL-6 activates a signaling pathway comprising the JAK/STAT proteins and characterized by a specific negative feedback loop exerted by the cytoplasmic protein suppressor of cytokine signalling-3 (SOCS3). On these bases, here, we explored the potential involvement of IL-6 signaling in the ability of the antidepressant drug agomelatine to normalize the anhedonic-like phenotype induced in the rat by chronic stress exposure. To this aim, adult male Wistar rats were subjected to the chronic mild stress (CMS) paradigm and chronically treated with vehicle or agomelatine. The behavioral evaluation was assessed by the sucrose consumption test, whereas molecular analyses were performed in the prefrontal cortex. We found that CMS was able to stimulate IL-6 production and signaling, including SOCS3 gene and protein expression, but the SOCS3-mediated feedback-loop inhibition failed to suppress the IL-6 cascade in stressed animals. Conversely, agomelatine treatment normalized the stress-induced decrease in sucrose consumption and restored the negative modulation of the IL-6 signaling via SOCS3 expression and activity. Our results provide additional information about the pleiotropic mechanisms that contribute to agomelatine's therapeutic effects.


Assuntos
Transtorno Depressivo Maior , Interleucina-6 , Animais , Ratos , Masculino , Interleucina-6/genética , Interleucina-6/metabolismo , Depressão/tratamento farmacológico , Depressão/etiologia , Depressão/metabolismo , Ratos Wistar , Transtorno Depressivo Maior/tratamento farmacológico , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Transdução de Sinais , Mediadores da Inflamação/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Sacarose
5.
Pharmacol Res ; 163: 105330, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33276101

RESUMO

Psychiatric disorders represent a critical challenge to our society, given their high global prevalence, complex symptomatology, elusive etiology and the variable effectiveness of pharmacological therapies. Recently, there has been a shift in investigating and redefining these diseases by integrating behavioral observations and multilevel neurobiological measures. Accordingly, endophenotype-oriented studies are needed to develop new therapeutic strategies, with the idea of targeting shared symptoms instead of one defined disease. With these premises, here we investigated the therapeutic properties of chronic treatment with the second-generation antipsychotic blonanserin in counteracting the alterations caused by 7 weeks of Chronic Mild Stress (CMS) in the rat. CMS is a well-established preclinical model able to induce depressive and anxiety-like alterations, which are shared by different psychiatric disorders. Our results demonstrated that the antipsychotic treatment normalizes the CMS-induced emotionality deficits, an effect that may be due to its ability in modulating, within the prefrontal cortex, redox mechanisms, a molecular dysfunction associated with several psychiatric disorders. These evidences provide new insights into the therapeutic properties and potential use of blonanserin as well as in its mechanisms of action and provide further support for the role of oxidative stress in the pathophysiology of psychiatric disorders.


Assuntos
Antipsicóticos/uso terapêutico , Piperazinas/uso terapêutico , Piperidinas/uso terapêutico , Estresse Psicológico/tratamento farmacológico , Animais , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Oxirredutases/genética , Piperazinas/farmacologia , Piperidinas/farmacologia , Ratos Wistar , Estresse Psicológico/genética
6.
Int J Mol Sci ; 21(17)2020 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-32872446

RESUMO

Depression is a recurrent disorder, with about 50% of patients experiencing relapse. Exposure to stressful events may have an adverse impact on the long-term course of the disorder and may alter the response to a subsequent stressor. Indeed, not all the systems impaired by stress may normalize during symptoms remission, facilitating the relapse to the pathology. Hence, we investigated the long-lasting effects of chronic restraint stress (CRS) and its influence on the modifications induced by the exposure to a second hit on brain-derived neurotrophic factor (BDNF) signaling in the prefrontal cortex (PFC). We exposed adult male Sprague Dawley rats to 4 weeks of CRS, we left them undisturbed for the subsequent 3 weeks, and then we exposed animals to one hour of acute restraint stress (ARS). We found that CRS influenced the release of corticosterone induced by ARS and inhibited the ability of ARS to activate mature BDNF, its receptor Tropomyosin receptor kinase B (TRKB), and their associated intracellular cascades: the TRKB-PI3K-AKT), the MEK-MAPK/ERK, and the Phospholipase C γ (PLCγ) pathways, positively modulated by ARS in non-stressed animals. These results suggest that CRS induces protracted and detrimental consequences that interfere with the ability of PFC to cope with a challenging situation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Corticosterona/metabolismo , Córtex Pré-Frontal/metabolismo , Restrição Física/psicologia , Estresse Psicológico/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Masculino , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkB/genética , Receptor trkB/metabolismo , Transdução de Sinais , Estresse Psicológico/etiologia , Estresse Psicológico/genética
8.
Brain Behav Immun ; 82: 422-431, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31525509

RESUMO

Vincristine (VCR) treatment is often associated to painful neuropathy. Its development is independent from antitumoral mechanism and involves neuroinflammation. We investigated the role of the chemokine prokineticin (PK)2 in a mouse model of VCR induced neuropathy using a PK-receptors (PK-R) antagonist to counteract its development. We also evaluated emotional like deficits in VCR mice. VCR (0,1 mg/kg) was i.p. injected in C57BL/6J male mice once a day for 14 consecutive days. Pain, anxiety and depressive like behaviors were assessed in animals. PK2, PK-Rs, cytokines, neuroinflammatory markers (CD68, CD11b, GFAP, TLR4) and ATF3 were evaluated in DRG, spinal cord, prefrontal cortex and hippocampus. The PK-Rs antagonist PC1, was s.c. injected (150 µg/kg) twice a day from day 7 (hypersensitivity state) until day 14. Its effect on pain and neuroinflammation was evaluated. VCR mice developed neuropathic pain but not mood alterations. After 7 days of VCR treatment we observed a neuroinflammatory condition in DRG with high levels of PK-Rs, TLR4, CD68, ATF3 and IL-1ß without relevant alterations in spinal cord. At day 14, an upregulation of PK system and a marked neuroinflammation was evident also in spinal cord. Moreover, at the same time, we observed initial alterations in supraspinal brain areas. PC1 treatment significantly counteracted neuropathic pain and blunted neuroinflammation.


Assuntos
Hormônios Gastrointestinais/metabolismo , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Neuropeptídeos/metabolismo , Vincristina/toxicidade , Animais , Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Comportamento Animal/efeitos dos fármacos , Citocinas/metabolismo , Depressão/induzido quimicamente , Depressão/metabolismo , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroimunomodulação/efeitos dos fármacos , Distribuição Aleatória , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
9.
Int J Neuropsychopharmacol ; 21(9): 883-893, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29788232

RESUMO

Background: Psychiatric disorders are associated with altered function of inhibitory neurotransmission within the limbic system, which may be due to the vulnerability of selective neuronal subtypes to challenging environmental conditions, such as stress. In this context, parvalbumin-positive GABAergic interneurons, which are critically involved in processing complex cognitive tasks, are particularly vulnerable to stress exposure, an effect that may be the consequence of dysregulated redox mechanisms. Methods: Adult Male Wistar rats were subjected to the chronic mild stress procedure for 7 weeks. After 2 weeks, both control and stress groups were further divided into matched subgroups to receive chronic administration of vehicle or lurasidone (3 mg/kg/d) for the subsequent 5 weeks. Using real-time RT-PCR and western blot, we investigated the expression of GABAergic interneuron markers and the levels of key mediators of the oxidative balance in the dorsal and ventral hippocampus. Results: Chronic mild stress induced a specific decrease of parvalbumin expression in the dorsal hippocampus, an effect normalized by lurasidone treatment. Interestingly, the regulation of parvalbumin levels was correlated to the modulation of the antioxidant master regulator NRF2 and its chaperon protein KEAP1, which were also modulated by pharmacological intervention. Conclusions: Our findings suggest that the susceptibility of parvalbumin neurons to stress may represent a key mechanism contributing to functional and structural impairments in specific brain regions relevant for psychiatric disorders. Moreover, we provide new insights on the mechanism of action of lurasidone, demonstrating that its chronic treatment normalizes chronic mild stress-induced parvalbumin alterations, possibly by potentiating antioxidant mechanisms, which may ameliorate specific functions that are deteriorated in psychiatric patients.


Assuntos
Antipsicóticos/farmacologia , Hipocampo/metabolismo , Cloridrato de Lurasidona/farmacologia , Parvalbuminas/metabolismo , Estresse Psicológico/metabolismo , Animais , Doença Crônica , Modelos Animais de Doenças , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Masculino , NADPH Oxidase 2/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Oxirredução , RNA Mensageiro/metabolismo , Ratos Wistar , Estresse Psicológico/tratamento farmacológico
11.
Pharmacol Res ; 104: 140-50, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26742719

RESUMO

Disruptions of biological rhythms are known to be associated with depressive disorders, suggesting that abnormalities in the molecular clock may contribute to the development of these disorders. These mechanisms have been extensively characterized in the suprachiasmatic nucleus, but little is know about the role exerted by individual clock genes in brain structures that are important for depressive disorders. Using the chronic mild stress model we found a significant reduction of BMAL1 and CLOCK protein levels in the nuclear compartment of the prefrontal cortex of CMS rats, which was paralleled by a down-regulation of the expression of several target genes, including Pers and Crys but also Reverbß and Pparα. Interestingly, chronic treatment with the multi receptor modulator lurasidone (3mg/kg for 5 weeks) was able to normalize the molecular changes induced by CMS exposure in prefrontal cortex, but it was also able to regulate some of these genes within the hippocampus. We believe that changes in clock genes expression after CMS exposure may contribute to the disturbances associated with depressive disorders and that the ability of chronic lurasidone to normalize such alterations may be relevant for its therapeutic properties in ameliorating functions that are deteriorated in patients with major depression and other stress-related disorders.


Assuntos
Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Cloridrato de Lurasidona/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Estresse Psicológico/genética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Antipsicóticos/farmacologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Antagonistas dos Receptores de Dopamina D2/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Estresse Psicológico/metabolismo
12.
Pharmacol Res ; 103: 1-12, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26535964

RESUMO

Major depression is a complex disease that originates from the interaction between a genetic background of susceptibility and environmental factors such as stress. At molecular level, it is characterized by dysfunctions of multiple systems including neurotransmitters, hormones, signalling pathways, neurotrophic and neuroplastic molecules and - more recently - inflammatory mediators. Accordingly, in the present study we used the chronic mild stress (CMS) paradigm in the rat to elucidate to what extent brain inflammation may contribute to the development and/or the maintenance of an anhedonic phenotype and how pharmacological intervention may interfere with such behavioral and molecular stress-induced alterations. To this aim, adult male rats were exposed to CMS for 2 weeks and the cerebral expression of several mediators of the inflammatory system was evaluated in the hippocampus and prefrontal cortex of both stressed and control animals in parallel with the sucrose intake. Next, the animals that showed a decreased sucrose consumption were exposed to five further weeks of CMS and treated with the antidepressants imipramine or agomelatine, or the antipsychotic lurasidone. Our results demonstrate that only the stressed animals that were characterized by a deficit in sucrose intake showed increased expression of the pro-inflammatory cytokines IL-1ß, IL-6 and up-regulation of markers and mediators of microglia activation such as CD11b, CX3CL1 and its receptor CX3CR1 in comparison with stress-resilient animals. Some of these molecular alterations persisted also after longer stress exposure and were modulated, similarly to the behavioral effects of CMS, by chronic pharmacological treatment. These data suggest that neuroinflammation may have a key role in the pathological consequences of stress exposure, thus contributing to the subject's vulnerability for depression.


Assuntos
Anedonia/fisiologia , Encéfalo/metabolismo , Estresse Fisiológico/fisiologia , Acetamidas/farmacologia , Animais , Antidepressivos/farmacologia , Antipsicóticos/farmacologia , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1/genética , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Imipramina/farmacologia , Interleucina-1beta/genética , Interleucina-6/genética , Cloridrato de Lurasidona/farmacologia , Masculino , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores de Quimiocinas/genética , Fator de Crescimento Transformador beta/genética
13.
Proc Natl Acad Sci U S A ; 110(21): 8708-13, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23650397

RESUMO

Stress and glucocorticoid hormones regulate hippocampal neurogenesis, but the molecular mechanisms mediating these effects are poorly understood. Here we identify the glucocorticoid receptor (GR) target gene, serum- and glucocorticoid-inducible kinase 1 (SGK1), as one such mechanism. Using a human hippocampal progenitor cell line, we found that a small molecule inhibitor for SGK1, GSK650394, counteracted the cortisol-induced reduction in neurogenesis. Moreover, gene expression and pathway analysis showed that inhibition of the neurogenic Hedgehog pathway by cortisol was SGK1-dependent. SGK1 also potentiated and maintained GR activation in the presence of cortisol, and even after cortisol withdrawal, by increasing GR phosphorylation and GR nuclear translocation. Experiments combining the inhibitor for SGK1, GSK650394, with the GR antagonist, RU486, demonstrated that SGK1 was involved in the cortisol-induced reduction in progenitor proliferation both downstream of GR, by regulating relevant target genes, and upstream of GR, by increasing GR function. Corroborating the relevance of these findings in clinical and rodent settings, we also observed a significant increase of SGK1 mRNA in peripheral blood of drug-free depressed patients, as well as in the hippocampus of rats subjected to either unpredictable chronic mild stress or prenatal stress. Our findings identify SGK1 as a mediator for the effects of cortisol on neurogenesis and GR function, with particular relevance to stress and depression.


Assuntos
Depressão/enzimologia , Glucocorticoides/metabolismo , Hipocampo/enzimologia , Hidrocortisona/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Glucocorticoides/metabolismo , Estresse Fisiológico , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Adulto , Animais , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Depressão/patologia , Feminino , Proteínas Hedgehog/metabolismo , Hipocampo/patologia , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Masculino , Pessoa de Meia-Idade , Mifepristona/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
14.
Neural Plast ; 2016: 2592319, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881097

RESUMO

Although activity-dependent transcription represents a crucial mechanism for long-lasting experience-dependent changes in the hippocampus, limited data exist on its contribution to pathological conditions. We aim to investigate the influence of chronic stress on the activity-dependent transcription of brain-derived neurotrophic factor (BDNF). The ex vivo methodology of acute stimulation of hippocampal slices obtained from rats exposed to chronic mild stress (CMS) was used to evaluate whether the adverse experience may alter activity-dependent BDNF gene expression. CMS reduces BDNF expression and that acute depolarization significantly upregulates total BDNF mRNA levels only in control animals, showing that CMS exposure may alter BDNF transcription under basal conditions and during neuronal activation. Moreover, while the basal effect of CMS on total BDNF reflects parallel modulations of all the transcripts examined, isoform-specific changes were found after depolarization. This different effect was also observed in the activation of intracellular signaling pathways related to the neurotrophin. In conclusion, our study discloses a functional alteration of BDNF transcription as a consequence of stress. Being the activity-regulated transcription a critical process in synaptic and neuronal plasticity, the different regulation of individual BDNF promoters may contribute to long-lasting changes, which are fundamental for the vulnerability of the hippocampus to stress-related diseases.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Hipocampo/metabolismo , Estresse Psicológico/metabolismo , Transcrição Gênica/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/genética , Estresse Psicológico/psicologia
15.
Int J Neuropsychopharmacol ; 18(4)2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25522402

RESUMO

BACKGROUND: Major depression is associated with several alterations, including reduced neuronal plasticity and impaired synaptic function, which represent an important target of pharmacological intervention. METHODS: In the present study, we have investigated the ability of the antipsychotic drug lurasidone to modulate behavioral and neuroplastic alterations in the chronic mild stress model of depression. RESULTS: Rats that show reduced sucrose consumption after 2 weeks of chronic mild stress have reduced expression of the pool of Bdnf transcripts with the long 3' untranslated region (3'-UTR) that may be targeted to the synaptic compartment, suggesting the contribution of the neurotrophin to the behavioral dysfunction produced by chronic mild stress. The downregulation of Bdnf expression persisted also after 7 weeks of chronic mild stress, whereas chronic lurasidone treatment improved anhedonia in chronic mild stress rats and restored Bdnf mRNA levels in the prefrontal cortex. Moreover, chronic lurasidone treatment was able to normalize chronic mild stress-induced defects of Psd95 and Gfap as well as changes in molecular regulators of protein translation at the synapse, including mTOR and eEF2. CONCLUSIONS: These results demonstrate that lurasidone shows antidepressant properties in the chronic mild stress model through the modulation of synaptic and neuroplastic proteins. Such changes may contribute to the amelioration of functional capacities, which are deteriorated in patients with major depression and stress-related disorders.


Assuntos
Antidepressivos/farmacologia , Transtorno Depressivo/tratamento farmacológico , Cloridrato de Lurasidona/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Doença Crônica , Transtorno Depressivo/metabolismo , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Quinase do Fator 2 de Elongação/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Wistar , Estresse Psicológico , Serina-Treonina Quinases TOR/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
16.
Curr Neuropharmacol ; 22(2): 323-338, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37475559

RESUMO

The development of new antipsychotics with pro-cognitive properties and less side effects represents a priority in schizophrenia drug research. In this study, we present for the first time a preclinical exploration of the effects of the promising natural atypical antipsychotic Methyl-2-Amino-3- Methoxybenzoate (MAM), a brain-penetrable protoalkaloid from the seed of the plant Nigella damascena. Using animal models related to hyperdopaminergic activity, namely the pharmacogenetic apomorphine (D2/D1 receptor agonist)-susceptible (APO-SUS) rat model and pharmacologically induced mouse and rat models of schizophrenia, we found that MAM reduced gnawing stereotypy and climbing behaviours induced by dopaminergic agents. This predicts antipsychotic activity. In line, MAM antagonized apomorphine-induced c-Fos and NPAS4 mRNA levels in post-mortem brain nucleus accumbens and dorsolateral striatum of APO-SUS rats. Furthermore, phencyclidine (PCP, an NMDA receptor antagonist) and 2,5-Dimethoxy-4-iodoamphetamine (DOI, a 5HT2A/2C receptor agonist) induced prepulse inhibition deficits, reflecting the positive symptoms of schizophrenia, which were rescued by treatment with MAM and atypical antipsychotics alike. Post-mortem brain immunostaining revealed that MAM blocked the strong activation of both PCP- and DOI-induced c-Fos immunoreactivity in a number of cortical areas. Finally, during a 28-day subchronic treatment regime, MAM did not induce weight gain, hyperglycemia, hyperlipidemia or hepato- and nephrotoxic effects, side effects known to be induced by atypical antipsychotics. MAM also did not show any cataleptic effects. In conclusion, its brain penetrability, the apparent absence of preclinical side effects, and its ability to antagonize positive and cognitive symptoms associated with schizophrenia make MAM an exciting new antipsychotic drug that deserves clinical testing.


Assuntos
Antipsicóticos , Esquizofrenia , Ratos , Camundongos , Animais , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Esquizofrenia/induzido quimicamente , Esquizofrenia/tratamento farmacológico , Apomorfina/farmacologia , Apomorfina/uso terapêutico , Éteres de Hidroxibenzoatos/uso terapêutico , Modelos Animais de Doenças , Cognição
17.
J Neuroinflammation ; 10: 116, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24050835

RESUMO

BACKGROUND: Growing evidence suggests that alterations of the inflammatory/immune system contribute to the pathogenesis of depression. Indeed, depressed patients exhibit increased levels of inflammatory markers in both the periphery and the brain, and high comorbidity exists between major depression and diseases associated with inflammatory alterations. In order to characterize the link between depression and inflammation, we aimed to investigate whether an altered inflammatory system is present in a genetic model of vulnerability for depression, namely rats with partial or total deletion of the serotonin transporter (SERT) gene. METHODS: Wild-type, heterozygous and homozygous SERT rats were analyzed under basal condition or following a challenge with an acute injection of lipopolysaccharide (LPS) and killed 24 h or 5 days later. RESULTS: We found that SERT mutant rats show altered cytokine expression in the dorsal and ventral hippocampus at basal conditions, and they also display an exacerbated cytokine response to the LPS challenge. Moreover, mutant rats exhibit differences in the expression of markers for microglia activation. CONCLUSION: Based on these data, we suggest that basal or functional alterations of immune/inflammatory systems might contribute to the phenotype of SERT rats and to their heightened susceptibility to depressive-like behavior.


Assuntos
Depressão/imunologia , Hipocampo/imunologia , Inflamação/imunologia , Neuroimunomodulação/imunologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/imunologia , Animais , Citocinas/biossíntese , Depressão/genética , Depressão/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Lipopolissacarídeos/toxicidade , Neuroimunomodulação/genética , Ratos , Ratos Mutantes , Reação em Cadeia da Polimerase em Tempo Real , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
18.
Cell Tissue Res ; 354(1): 51-60, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23708527

RESUMO

Gene expression changes are well documented in depression and schizophrenia and might contribute to the pathologic phenotype associated with these disorders. On this basis, the investigation of transcriptional changes is extensively employed at the preclinical level in order to identify and characterize genes causally related to a pathologic condition. Key information can be achieved with respect to functional alterations in selected brain structures and to anatomical networks and systems that are altered in mental disorders. Furthermore, such analyses are used to investigate the impact of pharmacologic intervention in mechanisms that are significantly impaired in the disease or that might contribute to their therapeutic effectiveness. In this review, we will primarily discuss target approach analyses as a valuable instrument to address key questions in experimental work. We will focus on a few paradigmatic examples, such as the modulation of the neurotrophin brain-derived neurotrophic factor and the regulation of inducible early genes and the functional implication of such analyses. We will also briefly discuss genome-wide approaches that aim at identifying, in an unbiased manner, all the genes differentially affected under a given experimental setting.


Assuntos
Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Transtornos Mentais/genética , Animais , Encéfalo/fisiologia , Humanos , Transtornos Mentais/metabolismo , Camundongos , Ratos , Roedores , Ácido gama-Aminobutírico/genética , Ácido gama-Aminobutírico/metabolismo
19.
Curr Opin Cell Biol ; 18(5): 491-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16904306

RESUMO

Unlike most somatic cells, leukocytes are constitutively non-adherent. However, adhesive interactions are not only a required step in essentially all effector functions performed by leukocytes, but they also relay increasingly well-defined intracellular signals that affect the leukocyte as well as the surrounding tissues. Dissecting such signals in leukocytes has provided a wealth of information that contributes to our understanding of how adhesion controls higher-order biological responses, ranging from cell migration to proliferation, differentiation and survival.


Assuntos
Adesão Celular/fisiologia , Endotélio/metabolismo , Leucócitos/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Matriz Extracelular/metabolismo , Inflamação/metabolismo , Integrinas/metabolismo , Transdução de Sinais/fisiologia
20.
J Immunol ; 184(8): 4497-509, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20231695

RESUMO

Inhibitor of NF-kappaB kinases beta (IKKbeta) and alpha (IKKalpha) activate distinct NF-kappaB signaling modules. The IKKbeta/canonical NF-kappaB pathway rapidly responds to stress-like conditions, whereas the IKKalpha/noncanonical pathway controls adaptive immunity. Moreover, IKKalpha can attenuate IKKbeta-initiated inflammatory responses. High mobility group box 1 (HMGB1), a chromatin protein, is an extracellular signal of tissue damage-attracting cells in inflammation, tissue regeneration, and scar formation. We show that IKKalpha and IKKbeta are each critically important for HMGB1-elicited chemotaxis of fibroblasts, macrophages, and neutrophils in vitro and neutrophils in vivo. By time-lapse microscopy we dissected different parameters of the HMGB1 migration response and found that IKKalpha and IKKbeta are each essential to polarize cells toward HMGB1 and that each kinase also differentially affects cellular velocity in a time-dependent manner. In addition, HMGB1 modestly induces noncanonical IKKalpha-dependent p52 nuclear translocation and p52/RelB target gene expression. Akin to IKKalpha and IKKbeta, p52 and RelB are also required for HMGB1 chemotaxis, and p52 is essential for cellular orientation toward an HMGB1 gradient. RAGE, a ubiquitously expressed HMGB1 receptor, is required for HMGB1 chemotaxis. Moreover, IKKbeta, but not IKKalpha, is required for HMGB1 to induce RAGE mRNA, suggesting that RAGE is at least one IKKbeta target involved in HMGB1 migration responses, and in accord with these results enforced RAGE expression rescues the HMGB1 migration defect of IKKbeta, but not IKKalpha, null cells. Thus, proinflammatory HMGB1 chemotactic responses mechanistically require the differential collaboration of both IKK-dependent NF-kappaB signaling pathways.


Assuntos
Quimiotaxia/imunologia , Proteína HMGB1/fisiologia , Quinase I-kappa B/fisiologia , Animais , Células Cultivadas , Quimiotaxia/genética , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibroblastos/imunologia , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Macrófagos/citologia , Macrófagos/enzimologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neutrófilos/citologia , Neutrófilos/enzimologia , Neutrófilos/imunologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA