Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ethnopharmacol ; 285: 114895, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34875348

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Datura metel L. has been used as an anesthetic in clinic for more than 1800 years in China, and the main efficacy of D. metel L. flower is relieving asthma and cough, relieving spasm and relieving pain. From 1978 to 1980, Datura metel L. was used as an anesthetic agent and occasionally cured psoriasis patients during anesthesia clinically, and our group confirmed that the effective portion is total withanolides (YWS). Moreover, the new drug "Datura metel L. capsule" composed of YWS has since been approved and used for the treatment of more than 3,000 psoriasis patients, with efficacy and cure rates greater than 90% and 65%. However, the immunological mechanism has not been elucidated. AIM OF THE STUDY: Nowadays, although total withanolides from Datura metel L. have a better clinical efficacy in the treatment of psoriasis, there is a lack of overall understanding of the mechanism of their treatment, especially about some immune cells and proteins closely related to psoriasis and their relationship in executive function and biological significance. This study focused on investigating the mechanism of psoriasis treatment by YWS and determined the biochemical processes in the treatment of psoriasis based on Treg/Th17 axis cell-mediated bidirectional immunoregulatory functions, which provides an important scientific basis for understanding the mechanism underlying the treatment of psoriasis by YWS. MATERIALS AND METHODS: The effects of YWS on the lesion pathology of IMQ-induced psoriasis mice and the underlying molecular mechanism were assessed directly using HE staining, the PASI score and the animal body mass. We also investigated the effects of YWS on the Treg/Th17 axis and their critical functions in psoriasis pathogenesis via molecular biological methods. Finally, we performed differential proteomics analysis on skin in IMQ-induced psoriasis mice to clarify the effect of YWS by incorporates mass spectrometry-bioinformatics and annotated the functions and pathways associated with the differential proteins through GO enrichment, KEGG pathway analysis and PPI networks analysis, respectively. RESULTS: YWS regulated the imbalance of the Treg/Th17 axis. And proteomic analysis showed that YWS up-regulated 46 and down-regulated 37 proteins. According to the bioinformatics analysis, the improvement of Treg/Th17 imbalance may be the key immunological mechanism of YWS in the treatment of psoriasis by up-regulating the butyrate metabolism pathway, down-regulating leukocyte migration, inhibiting the phagocytic function of natural killer cells, suppressing osteoclast differentiation and interfering with chemokine activity, and the critical proteins involved are Lyn, HMGCS2, ABAT, ITGß2, PRKCß, MMP9, NCF1, JUNß, and Hck. CONCLUSION: This research clarified that the improvement of the imbalance of the Treg/Th17 axis may be the key immunological mechanism of YWS in the treatment of psoriasis through metabolic pathways and influencing key proteins. The results not only expand the therapeutic targets and approaches for the treatment of psoriasis, which is a challenging and complex disease, but also deepens the understanding of the mechanism of YWS in the treatment of psoriasis and other important conditions to open up a new way of thinking for research on YWS in the treatment of psoriasis.


Assuntos
Imiquimode/toxicidade , Inflamação/tratamento farmacológico , Psoríase/tratamento farmacológico , Vitanolídeos/uso terapêutico , Animais , Biologia Computacional , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Indutores de Interferon/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mapas de Interação de Proteínas , Psoríase/induzido quimicamente , Distribuição Aleatória , Transdução de Sinais , Regulação para Cima/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
2.
J Neurosci ; 41(48): 9971-9987, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34607967

RESUMO

Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Transtornos do Neurodesenvolvimento/etiologia , Complicações Infecciosas na Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Feminino , Indutores de Interferon/toxicidade , Macaca mulatta , Masculino , Transtornos do Neurodesenvolvimento/patologia , Neurogênese/fisiologia , Poli I-C/toxicidade , Gravidez , Complicações Infecciosas na Gravidez/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
3.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-34639115

RESUMO

Psoriasis is a chronic inflammatory skin disease. Recently, lysophosphatidic acid (LPA)/LPAR5 signaling has been reported to be involved in both NLRP3 inflammasome activation in macrophages and keratinocyte activation to produce inflammatory cytokines, contributing to psoriasis pathogenesis. However, the effect and molecular mechanisms of LPA/LPAR signaling in keratinocyte proliferation in psoriasis remain unclear. In this study, we investigated the effects of LPAR1/3 inhibition on imiquimod (IMQ)-induced psoriasis-like mice. Treatment with the LPAR1/3 antagonist, ki16425, alleviated skin symptoms in IMQ-induced psoriasis-like mouse models and decreased keratinocyte proliferation in the lesion. It also decreased LPA-induced cell proliferation and cell cycle progression via increased cyclin A2, cyclin D1, cyclin-dependent kinase (CDK)2, and CDK4 expression and decreased p27Kip1 expression in HaCaT cells. LPAR1 knockdown in HaCaT cells reduced LPA-induced proliferation, suppressed cyclin A2 and CDK2 expression, and restored p27Kip1 expression. LPA increased Rho-associated protein kinase 2 (ROCK2) expression and PI3K/AKT activation; moreover, the pharmacological inhibition of ROCK2 and PI3K/AKT signaling suppressed LPA-induced cell cycle progression. In conclusion, we demonstrated that LPAR1/3 antagonist alleviates IMQ-induced psoriasis-like symptoms in mice, and in particular, LPAR1 signaling is involved in cell cycle progression via ROCK2/PI3K/AKT pathways in keratinocytes.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica/efeitos dos fármacos , Imiquimode/toxicidade , Queratinócitos/citologia , Lisofosfolipídeos/farmacologia , Psoríase/tratamento farmacológico , Animais , Apoptose , Biomarcadores/metabolismo , Ciclo Celular , Células Cultivadas , Humanos , Indutores de Interferon/toxicidade , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Psoríase/induzido quimicamente , Psoríase/metabolismo , Psoríase/patologia , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
4.
J Leukoc Biol ; 110(6): 1047-1055, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34494306

RESUMO

Tumor necrosis factor alpha (TNF) has been implicated in the pathogenesis of psoriasis and anti-TNF therapeutics are used in the treatment of psoriasis in the clinic. However, considerable proportion of patients fail to respond to anti-TNF treatment. Furthermore, anti-TNF therapy induces de novo development of psoriasis in some patients with other type of autoimmune disorders. Therefore, further understanding of the role of TNF-TNFR signaling in pathogenesis of psoriasis remains a critical to devise safer and more effective treatment. In this study, it is shown that in imiquimod-induced mouse psoriasis model, TNF receptor type 1 (TNFR1) deficiency inhibited the development of skin diseases. In sharp contrast, TNF receptor type 2 (TNFR2) deficiency led to more severe psoriasis that was associated with increased Th1 and Th17 responses and reduced number of CD4+ Foxp3+ regulatory T cells (Tregs). Importantly, adoptive transfer of WT Tregs was able to attenuate inflammatory responses in imiquimod-treated TNFR2-/- mice, suggestive of a role of malfunctioned Tregs in mice deficient in TNFR2. RNA sequencing data revealed that Tregs deficient in TNFR2 exhibited down-regulation of different biological processes linked to proliferative expansion. Taken together, our study clearly indicated that TNFR1 was pathogenic in mouse psoriasis. In contrast, through boosting the proliferative expansion of Tregs, TNFR2 was protective in this model. The data thus suggest that TNFR1-specific antagonist or TNFR2-specific agonist may be useful in the treatment of patients with psoriasis.


Assuntos
Imiquimode/toxicidade , Psoríase/induzido quimicamente , Psoríase/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Animais , Modelos Animais de Doenças , Feminino , Indutores de Interferon/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Psoríase/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/imunologia
5.
J Neurosci ; 41(32): 6954-6965, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34253630

RESUMO

Episodic memory requires information to be stored and recalled in sequential order, and these processes are disrupted in schizophrenia. Hippocampal phase precession and theta sequences are thought to provide a biological mechanism for sequential ordering of experience at timescales suitable for plasticity. These phenomena have not previously been examined in any models of schizophrenia risk. Here, we examine these phenomena in a maternal immune activation (MIA) rodent model. We show that while individual pyramidal cells in the CA1 region continue to precess normally in MIA animals, the starting phase of precession as an animal enters a new place field is considerably more variable in MIA animals than in controls. A critical consequence of this change is a disorganization of the ordered representation of experience via theta sequences. These results provide the first evidence of a biological-level mechanism that, if it occurs in schizophrenia, may explain aspects of disorganized sequential processing that contribute to the cognitive symptoms of the disorder.SIGNIFICANCE STATEMENT Hippocampal phase precession and theta sequences have been proposed as biophysical mechanisms by which the sequential structure of cognition might be ordered. Disturbances of sequential processing have frequently been observed in schizophrenia. Here, we show for the first time that phase precession and theta sequences are disrupted in a maternal immune activation (MIA) model of schizophrenia risk. This is a result of greater variability in the starting phase of precession, indicating that the mechanisms that coordinate precession at the assembly level are disrupted. We propose that this disturbance in phase precession underlies some of the disorganized cognitive symptoms that occur in schizophrenia. These findings could have important preclinical significance for the identification and treatment of schizophrenia risk factors.


Assuntos
Hipocampo/fisiopatologia , Memória Episódica , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Esquizofrenia/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Inflamação/induzido quimicamente , Indutores de Interferon/toxicidade , Masculino , Exposição Materna/efeitos adversos , Poli I-C/toxicidade , Gravidez , Ratos Sprague-Dawley , Esquizofrenia/etiologia
6.
Brain Res ; 1759: 147370, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33600830

RESUMO

Genes and environment interact during intrauterine life, and potentially alter the developmental trajectory of the brain. This can result in life-long consequences on brain function. We have previously developed two transgenic mouse lines that suppress Gad1 expression in parvalbumin (PVALB) and neuropeptide Y (NPY) expressing interneuron populations using a bacterial artificial chromosome (BAC)-driven miRNA-based silencing technology. We were interested to assess if maternal immune activation (MIA), genetic interneuronal inhibition, and the combination of these two factors disrupt and result in long-term changes in neuroinflammatory gene expression, sterol biosynthesis, and acylcarnitine levels in the brain of maternally exposed offspring. Pregnant female WT mice were given a single intraperitoneal injection of saline or polyinosinic-polycytidilic acid [poly(I:C)] at E12.5. Brains of offspring were analyzed at postnatal day 90. We identified complex and persistent neuroinflammatory gene expression changes in the hippocampi of MIA-exposed offspring, as well in the hippocampi of Npy/Gad1 and Pvalb/Gad1 mice. In addition, both MIA and genetic inhibition altered the post-lanosterol sterol biosynthesis in the neocortex and disrupted the typical acylcarnitine profile. In conclusion, our findings suggest that both MIA and inhibition of interneuronal function have long-term consequences on critical homeostatic mechanisms of the brain, including immune function, sterol levels, and energy metabolism.


Assuntos
Mediadores da Inflamação/imunologia , Interneurônios/imunologia , Neuroimunomodulação/fisiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/imunologia , Animais , Feminino , Glutamato Descarboxilase/deficiência , Glutamato Descarboxilase/genética , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Hipocampo/metabolismo , Mediadores da Inflamação/metabolismo , Indutores de Interferon/toxicidade , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/efeitos dos fármacos , Neocórtex/imunologia , Neocórtex/metabolismo , Neuroimunomodulação/efeitos dos fármacos , Poli I-C/toxicidade , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo
7.
FEBS J ; 288(14): 4249-4266, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33452755

RESUMO

Viral infection is a significant burden to health care worldwide. Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors, are widely used as cholesterol-lowering drugs. Recently, long-term statin therapy was shown to reduce the antiviral immune response; however, the underlying molecular mechanisms are unclear. Here, we found that simvastatin decreased polyinosinic-polycytidylic acid [poly(I:C)]-induced expression of antiviral interferon (IFN)-ß and IFN-stimulated genes (ISGs) in the bronchoalveolar lavage fluid (BALF) and lungs of mice with high-fat diet-induced hyperlipidemia. Macrophages were the dominant cell type in the BALF of poly(I:C)-treated mice. We examined the effects of simvastatin in primary lung macrophages and found that simvastatin suppressed poly(I:C)-induced expression of IFN-ß and ISGs. We examined the molecular mechanisms of statin-mediated inhibition of antiviral gene expression using murine macrophage-like cell line, J774.1/JA-4. Simvastatin and pitavastatin decreased poly(I:C)-induced expression of IFN-ß and ISGs. Moreover, they repressed poly(I:C)-induced phosphorylation of IFN regulatory factor (IRF) 3 and signal transducers and activators of transcription (STAT) 1, which is involved in Janus kinase (JAK)/STAT signaling. Mevalonate and geranylgeranyl pyrophosphate (GGPP), but not cholesterol, counteracted the negative effect of statins on IFN-ß and ISG expression and phosphorylation of IRF3 and STAT1. The geranylgeranyltransferase inhibitor suppressed poly(I:C)-induced expression of IFN-ß and ISGs and phosphorylation of IRF3 and STAT1. These results suggest that statins suppressed the expression of IFN-ß and ISGs in poly(I:C)-treated hyperlipidemic mice and murine macrophages and that these effects occurred through the inhibition of IRF3 and JAK/STAT signaling in macrophages. Furthermore, GGPP recovered the statin-suppressed IRF3 and JAK/STAT signaling in poly(I:C)-treated macrophages.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hiperlipidemias/tratamento farmacológico , Fatores Reguladores de Interferon/metabolismo , Interferon beta/metabolismo , Macrófagos/efeitos dos fármacos , Poli I-C/toxicidade , Animais , Hiperlipidemias/induzido quimicamente , Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Indutores de Interferon/toxicidade , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Interferon beta/genética , Janus Quinases/antagonistas & inibidores , Janus Quinases/genética , Janus Quinases/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
8.
Alcohol Clin Exp Res ; 44(9): 1728-1733, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32583876

RESUMO

BACKGROUND: A relationship between alcohol consumption and psoriasis has been reported, but it is unclear whether alcohol consumption aggravates psoriasis. Here, we studied the effect of chronic ethanol (EtOH) consumption in the murine model of Aldara-induced psoriasiform dermatitis. METHODS: C57BL/6 mice received 5% EtOH in their drinking water for 10 weeks. Dermatitis was induced from weeks 9 to 10, by applying Aldara to the shaved patch of skin on the back. Inflammation was characterized by histological and transcriptomic analyses. RESULTS: EtOH consumption aggravated Aldara-induced dermatitis. The scales were more severe, epidermal thickening was more pronounced, and cutaneous expression of Th17-related cytokines was exacerbated. Control mice simply receiving EtOH displayed minimal cutaneous inflammation, characterized by epidermal infiltrates of T lymphocytes and the overexpression of IL-17A and the Th17-recruiting chemokine CCL20. In vitro studies showed that low concentrations of EtOH induce the expression of CCL20 by murine epidermal keratinocytes. CONCLUSION: Alcohol consumption leads to subliminar skin inflammation, which is revealed by the exacerbation of Aldara-induced experimental psoriasiform dermatitis, likely through Th17-type minimal skin inflammation. These results favor the systematic management of alcohol consumption in psoriatic patients.


Assuntos
Consumo de Bebidas Alcoólicas , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Psoríase/patologia , Pele/efeitos dos fármacos , Animais , Quimiocina CCL20/efeitos dos fármacos , Quimiocina CCL20/metabolismo , Perfilação da Expressão Gênica , Imiquimode/toxicidade , Indutores de Interferon/toxicidade , Interleucina-17/genética , Interleucina-23/genética , Interleucinas/genética , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Psoríase/genética , Pele/metabolismo , Pele/patologia , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Interleucina 22
9.
Int J Mol Sci ; 21(10)2020 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-32456211

RESUMO

Interferon regulatory factors (IRFs) play diverse roles in the regulation of the innate and adaptive immune responses in various diseases. In psoriasis, IRF2 is known to be involved in pathogenesis, while studies on other IRFs are limited. In this study, we investigated the role of IRF5 in psoriasis using imiquimod-induced psoriasis-like dermatitis. Although IRF5 is known to play a critical role in the induction of proinflammatory cytokines by immune cells, such as dendritic cells (DCs), macrophages, and monocytes, IRF5 deficiency unexpectedly exacerbated psoriasiform skin inflammation. The interferon-α and tumor necrosis factor-α mRNA expression levels were decreased, while levels of Th17 cytokines including IL-17, IL-22, and IL-23 were increased in IRF5-deficient mice. Furthermore, IL-23 expression in DCs from IRF5-deficient mice was upregulated both in steady state and after toll-like receptor 7/8 agonist stimulation. Importantly, the expression of IRF4, which is also important for the IL-23 production in DCs, was augmented in DCs from IRF5-deficient mice. Taken together, our results suggest that IRF5 deficiency induces the upregulation of IRF4 in DCs followed by augmented IL-23 production, resulting in the amplification of Th17 responses and the exacerbation of imiquimod-induced psoriasis-like skin inflammation. The regulation of IRF4 or IRF5 expression may be a novel therapeutic approach to psoriasis.


Assuntos
Fatores Reguladores de Interferon/genética , Interleucinas/metabolismo , Psoríase/metabolismo , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Feminino , Imiquimode/toxicidade , Indutores de Interferon/toxicidade , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/metabolismo , Interferons/genética , Interferons/metabolismo , Interleucinas/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Psoríase/etiologia , Psoríase/genética , Pele/efeitos dos fármacos , Pele/metabolismo , Células Th17/metabolismo
10.
Eur Neuropsychopharmacol ; 29(5): 577-589, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30948194

RESUMO

Accumulating data point to heme involvement in neuropsychiatric disorders. Heme plays a role in major cellular processes such as signal transduction, protein complex assembly and regulation of transcription and translation. Its synthesis involves the mitochondria, which dysfunction, specifically that of the complex I (Co-I) of the electron transport chain is involved in the pathophysiology of schizophrenia (SZ). Here we aimed to demonstrate that deficits in Co-I affect heme metabolism. We show a significant decrease in heme levels in Co-I deficient SZ-derived EBV transformed lymphocytes (lymphoblastoid cell lines - LCLs) as compared to healthy subjects-derived cells (n = 9/cohort). Moreover, protein levels assessed by immunoblotting and mRNA levels assessed by qRT-PCR of heme catabolic enzyme, heme Oxygenase 1 (HO-1), and protein levels of heme downstream target phosphorylated eukaryotic initiation factor 2-alpha (Peif2a/eif2a) were significantly elevated in SZ-derived cells. In contrast, protein and mRNA levels of heme synthesis rate limiting enzyme aminolevulinic acid synthase-1 (ALAS1) were unchanged in SZ derived LCLs. In addition, inhibition of Co-I by rotenone in healthy subjects-derived LCLs (n = 4/cohort) exhibited an initial increase followed by a later decrease in heme levels. These findings were associated with opposite changes in heme's downstream target and HO-1 level, similar to our findings in SZ-derived cells. We also show a brain region specific pattern of impairment in Co-I subunits and in HO-1 and PeIF2α/eIF2α in the Poly-IC rat model of SZ (n = 6/cohort). Our results provide evidence for a link between CoI and heme metabolism both in-vitro and in-vivo suggesting its contribution to SZ pathophysiology.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Hemeproteínas/metabolismo , Mitocôndrias/metabolismo , Esquizofrenia/metabolismo , Adulto , Animais , Linhagem Celular , Feminino , Humanos , Indutores de Interferon/toxicidade , Masculino , Pessoa de Meia-Idade , Poli I-C/toxicidade , Ratos , Esquizofrenia/induzido quimicamente , Adulto Jovem
11.
J Neuroinflammation ; 16(1): 18, 2019 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-30691477

RESUMO

BACKGROUND: Important insight into the mechanisms through which gene-environmental interactions cause schizophrenia can be achieved through preclinical studies combining prenatal immune stimuli with disease-related genetic risk modifications. Accumulating evidence associates JNK signalling molecules, including MKK7/MAP2K7, with genetic risk. We tested the hypothesis that Map2k7 gene haploinsufficiency in mice would alter the prenatal immune response to the viral mimetic polyriboinosinic-polyribocytidylic acid (polyI:C), specifically investigating the impact of maternal versus foetal genetic variants. METHODS: PolyI:C was administered to dams (E12.5), and cytokine/chemokine levels were measured 6 h later, in maternal plasma, placenta and embryonic brain. RESULTS: PolyI:C dramatically elevated maternal plasma levels of most cytokines/chemokines. Induction of IL-1ß, IL-2, IL-10, IL-12, TNF-α and CXCL3 was enhanced, while CCL5 was suppressed, in Map2k7 hemizygous (Hz) dams relative to controls. Maternal polyI:C administration also increased embryonic brain chemokines, influenced by both maternal and embryonic genotype: CCL5 and CXCL10 levels were higher in embryonic brains from Map2k7 dams versus control dams; for CCL5, this was more pronounced in Map2k7 Hz embryos. Placental CXCL10 and CXCL12 levels were also elevated by polyI:C, the former enhanced and the latter suppressed, in placentae from maternal Map2k7 Hzs relative to control dams receiving polyI:C. CONCLUSIONS: The results demonstrate JNK signalling as a mediator of MIA effects on the foetus. Since both elevated CXCL10 and supressed CXCL12 compromise developing GABAergic interneurons, the results support maternal immune challenge contributing to schizophrenia-associated neurodevelopmental abnormalities. The influence of Map2k7 on cytokine/chemokine induction converges the genetic and environmental aspects of schizophrenia, and the overt influence of maternal genotype offers an intriguing new insight into modulation of embryonic neurodevelopment by genetic risk.


Assuntos
Indutores de Interferon/toxicidade , Sistema de Sinalização das MAP Quinases/fisiologia , Poli I-C/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Esquizofrenia/etiologia , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Esquizofrenia/sangue , Fator A de Crescimento do Endotélio Vascular
12.
Br J Dermatol ; 179(1): 101-109, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29150843

RESUMO

BACKGROUND: Psoriasis exhibits several extracutaneous manifestations. Little is known about hepatic parameters specifically associated with psoriasis. OBJECTIVES: To study whether psoriasiform dermatitis is associated with liver injury. METHODS: We studied liver parameters of inflammation and fibrosis in a murine model of psoriasiform dermatitis induced by topical application of imiquimod for 9 weeks. RESULTS: Topical treatment with imiquimod induced a form of psoriasiform dermatitis reminiscent of the human disorder, characterized by thickened and scaly skin, psoriasiform epidermal hyperplasia, altered keratinocyte differentiation and cutaneous overexpression of interleukin-17A. Mice with dermatitis displayed hepatitis, as shown by elevation of plasma transaminase levels, as well as portal and periportal hepatitis, characterized by T-lymphocyte (CD3ε+ ) and polymorphonuclear cell (Gr1+ ) infiltrates. The hepatitis progressed towards liver fibrogenesis, as shown by excessive Sirius red staining, which is consistent with the expression of α-smooth muscle actin by hepatic stellate cells. CONCLUSIONS: These results indicate that liver inflammation and fibrosis are associated with experimental psoriasiform dermatitis. Our results suggest that psoriatic inflammation may be associated with specific liver injury.


Assuntos
Toxidermias/etiologia , Imiquimode/toxicidade , Indutores de Interferon/toxicidade , Cirrose Hepática/etiologia , Psoríase/complicações , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Modelos Animais de Doenças , Imiquimode/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL
13.
PLoS One ; 12(8): e0182646, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28777803

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pleiotropic, Th17-derived cytokine thought to critically contribute to the pathogenesis of diverse autoimmune diseases, including rheumatoid arthritis and psoriasis. Treatment with monoclonal antibodies that block GM-CSF activity is associated with favorable therapeutic effects in patients with rheumatoid arthritis. We evaluated the role of GM-CSF as a potential target for therapeutic interference in psoriasis using a combined pharmacologic and genetic approach and the mouse model of imiquimod-induced psoriasiform dermatitis (IMQPD). Neutralization of murine GM-CSF by an anti-GM-CSF antibody ameliorated IMQPD. In contrast, genetic deficiency in GM-CSF did not alter the course of IMQPD, suggesting the existence of mechanisms compensating for chronic, but not acute, absence of GM-CSF. Further investigation uncovered an alternative pathogenic pathway for IMQPD in the absence of GM-CSF characterized by an expanded plasmacytoid dendritic cell population and release of IFNα and IL-22. This pathway was not activated in wild-type mice during short-term anti-GM-CSF treatment. Our investigations support the potential value of GM-CSF as a therapeutic target in psoriatic disease. The discovery of an alternative pathogenic pathway for psoriasiform dermatitis in the permanent absence of GM-CSF, however, suggests the need for monitoring during therapeutic use of long-term GM-CSF blockade.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Psoríase/tratamento farmacológico , Aminoquinolinas/toxicidade , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imiquimode , Indutores de Interferon/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Psoríase/induzido quimicamente , Psoríase/imunologia , Psoríase/patologia
14.
Eur Arch Psychiatry Clin Neurosci ; 267(5): 479-483, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28417199

RESUMO

Brain-derived neurotrophic factor (BDNF) and its high-affinity receptor, tropomyosin receptor kinase B (TrkB) signaling plays a key role in the brain neurodevelopment. The exposure of pregnant mice to polyinosinic-polycytidylic acid [poly(I:C)] causes cognitive deficits in adult offspring. Supplementation with a TrkB agonist, 7,8-dihydroxyflavone, in poly(I:C)-treated pregnant mice from pregnancy to weaning could prevent the onset of cognitive deficits and reduced BDNF-TrkB signaling in the prefrontal cortex of their adult offspring. These findings suggest that supplementation with a TrkB agonist in pregnant women with an ultra-high risk of psychosis may reduce the development of psychosis in their offspring.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Flavonas/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Indutores de Interferon/toxicidade , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Poli I-C/toxicidade , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Receptor trkB/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
15.
Int J Dev Neurosci ; 58: 1-8, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28122258

RESUMO

Maternal immune activation (MIA) during pregnancy in rodents increases the risk of the offspring to develop schizophrenia-related behaviors, suggesting a relationship between the immune system and the brain development. Here we tested the hypothesis that MIA induced by the viral mimetic polyinosinic-polycytidylic acid (poly I:C) in early or late gestation of mice leads to behavioral and neuroanatomical disorders in the adulthood. On gestational days (GDs) 9 or 17 pregnant dams were treated with poly I:C or saline via intravenous route and the offspring behaviors were measured during adulthood. Considering the progressive structural neuroanatomical alterations in the brain of individuals with schizophrenia, we used magnetic resonance imaging (MRI) to perform brain morphometric analysis of the offspring aged one year. MIA on GD9 or GD17 led to increased basal locomotor activity, enhanced motor responses to ketamine, a psychotomimetic drug, and reduced time spent in the center of the arena, suggesting an increased anxiety-like behavior. In addition, MIA on GD17 reduced glucose preference in the offspring. None of the treatments altered the relative volume of the lateral ventricles. However, a decrease in brain volume, especially for posterior structures, was observed for one-year-old animals treated with poly I:C compared with control groups. Thus, activation of the maternal immune system at different GDs lead to neuroanatomical and behavioral alterations possibly related to the positive and negative symptoms of schizophrenia. These results provide insights on neuroimmunonological and neurodevelopmental aspects of certain psychopathologies, such as schizophrenia.


Assuntos
Encéfalo/patologia , Transtornos Mentais/etiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Esquizofrenia/complicações , Esquizofrenia/etiologia , Esquizofrenia/patologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Encéfalo/diagnóstico por imagem , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Preferências Alimentares , Indutores de Interferon/toxicidade , Ketamina/toxicidade , Locomoção/fisiologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/toxicidade , Gravidez , Esquizofrenia/diagnóstico por imagem , Sacarose/administração & dosagem
16.
Cereb Cortex ; 27(6): 3397-3413, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27797829

RESUMO

Prenatal exposure to maternal infection increases the risk of neurodevelopmental disorders, including schizophrenia and autism. The molecular processes underlying this pathological association, however, are only partially understood. Here, we combined unbiased genome-wide transcriptional profiling with follow-up epigenetic analyses and structural magnetic resonance imaging to explore convergent molecular and neuromorphological alterations in corticostriatal areas of adult offspring exposed to prenatal immune activation. Genome-wide transcriptional profiling revealed that prenatal immune activation caused a differential expression of 116 and 251 genes in the medial prefrontal cortex and nucleus accumbens, respectively. A large part of genes that were commonly affected in both brain areas were related to myelin functionality and stability. Subsequent epigenetic analyses indicated that altered DNA methylation of promoter regions might contribute to the differential expression of myelin-related genes. Quantitative relaxometry comparing T1, T2, and myelin water fraction revealed sparse increases in T1 relaxation times and consistent reductions in T2 relaxation times. Together, our multi-system approach demonstrates that prenatal viral-like immune activation causes myelin-related transcriptional and epigenetic changes in corticostriatal areas. Even though these abnormalities do not seem to be associated with overt white matter reduction, they may provide a molecular mechanism whereby prenatal infection can impair myelin functionality and stability.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Regulação da Expressão Gênica/fisiologia , Imageamento por Ressonância Magnética , Transtornos do Neurodesenvolvimento , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Estudos de Coortes , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Idade Gestacional , Indutores de Interferon/toxicidade , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Básica da Mielina/metabolismo , Proteínas da Mielina/metabolismo , Transtornos do Neurodesenvolvimento/diagnóstico por imagem , Transtornos do Neurodesenvolvimento/etiologia , Transtornos do Neurodesenvolvimento/metabolismo , Poli I-C/toxicidade , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia
17.
Gastroenterology ; 152(5): 1100-1113.e12, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27993525

RESUMO

BACKGROUND & AIMS: Wheat amylase-trypsin inhibitors (ATIs) are nutritional activators of innate immunity, via activation of the toll-like receptor 4 (TLR4) on myeloid cells. We aimed to characterize the biologic activity of ATIs in various foods and their effect on intestinal inflammation. METHODS: We selected 38 different gluten-containing and gluten-free products, either unprocessed (such as wheat, rye, barley, quinoa, amaranth, soya, lentils, and rice) or processed (such as pizza, pasta, bread, and biscuits). ATIs were extracted and their biological activities determined in TLR4-responsive mouse and human cell lines. Effects of oral ATIs on intestinal inflammation were determined in healthy C57BL/6 mice on a gluten-free or ATI-free diet and in mice given low-level polyinosinic:polycytidylic acid or dextran sodium sulfate to induce colitis. Parameters of innate and adaptive immune activation were determined in duodenum, ileum, colon, and mesenteric lymph nodes. RESULTS: Modern gluten-containing staples had levels of TLR4-activating ATIs that were as much as 100-fold higher than in most gluten-free foods. Processed or baked foods retained ATI bioactivity. Most older wheat variants (such as Emmer or Einkorn) had lower bioactivity than modern (hexaploid) wheat. ATI species CM3 and 0.19 were the most prevalent activators of TLR4 in modern wheat and were highly resistant to intestinal proteolysis. Their ingestion induced modest intestinal myeloid cell infiltration and activation, and release of inflammatory mediators-mostly in the colon, then in the ileum, and then in the duodenum. Dendritic cells became prominently activated in mesenteric lymph nodes. Concentrations of ATIs found in a normal daily gluten-containing diet increased low-level intestinal inflammation. CONCLUSIONS: Gluten-containing cereals have by far the highest concentrations of ATIs that activate TLR4. Orally ingested ATIs are largely resistant to proteases and heat, and increase intestinal inflammation by activating gut and mesenteric lymph node myeloid cells.


Assuntos
Amilases/antagonistas & inibidores , Doença Celíaca/imunologia , Colite/imunologia , Glutens/imunologia , Intestinos/imunologia , Células Mieloides/imunologia , Receptor 4 Toll-Like/imunologia , Inibidores da Tripsina/imunologia , Imunidade Adaptativa , Animais , Linhagem Celular , Colite/induzido quimicamente , Colo/imunologia , Sulfato de Dextrana/toxicidade , Dieta Livre de Glúten , Duodeno/imunologia , Humanos , Íleo/imunologia , Imunidade Inata/imunologia , Inflamação , Indutores de Interferon/toxicidade , Linfonodos/imunologia , Mesentério , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Plantas/imunologia , Poli I-C/toxicidade , Triticum/imunologia
18.
J Biol Chem ; 292(1): 51-63, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27895126

RESUMO

Viral infection is an exacerbating factor contributing to chronic airway diseases, such as asthma, via mechanisms that are still unclear. Polyinosine-polycytidylic acid (poly(I:C)), a Toll-like receptor 3 (TLR3) agonist used as a mimetic to study viral infection, has been shown to elicit inflammatory responses in lungs and to exacerbate pulmonary allergic reactions in animal models. Previously, we have shown that poly(I:C) stimulates lung fibroblasts to accumulate an extracellular matrix (ECM), enriched in hyaluronan (HA) and its binding partner versican, which promotes monocyte adhesion. In the current study, we aimed to determine the in vivo role of versican in mediating inflammatory responses in poly(I:C)-induced lung inflammation using a tamoxifen-inducible versican-deficient mouse model (Vcan-/- mice). In C57Bl/6 mice, poly(I:C) instillation significantly increased accumulation of versican and HA, especially in the perivascular and peribronchial regions, which were enriched in infiltrating leukocytes. In contrast, versican-deficient (Vcan-/-) lungs did not exhibit increases in versican or HA in these regions and had strikingly reduced numbers of leukocytes in the bronchoalveolar lavage fluid and lower expression of inflammatory chemokines and cytokines. Poly(I:C) stimulation of lung fibroblasts isolated from control mice generated HA-enriched cable structures in the ECM, providing a substrate for monocytic cells in vitro, whereas lung fibroblasts from Vcan-/- mice did not. Moreover, increases in proinflammatory cytokine expression were also greatly attenuated in the Vcan-/- lung fibroblasts. These findings provide strong evidence that versican is a critical inflammatory mediator during poly(I:C)-induced acute lung injury and, in association with HA, generates an ECM that promotes leukocyte infiltration and adhesion.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Indutores de Interferon/toxicidade , Pneumonia/prevenção & controle , Poli I-C/toxicidade , Versicanas/fisiologia , Animais , Líquido da Lavagem Broncoalveolar/química , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia
19.
Cereb Cortex ; 26(11): 4265-4281, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27613435

RESUMO

Cognitive deficits represent a major burden of neuropsychiatric disorders and result in part from abnormal communication within hippocampal-prefrontal circuits. While it has been hypothesized that this network dysfunction arises during development, long before the first clinical symptoms, experimental evidence is still missing. Here, we show that pre-juvenile mice mimicking genetic and environmental risk factors of disease (dual-hit GE mice) have poorer recognition memory that correlates with augmented coupling by synchrony and stronger directed interactions between prefrontal cortex and hippocampus. The network dysfunction emerges already during neonatal development, yet it initially consists in a diminished hippocampal theta drive and consequently, a weaker and disorganized entrainment of local prefrontal circuits in discontinuous oscillatory activity in dual-hit GE mice when compared with controls. Thus, impaired maturation of functional communication within hippocampal-prefrontal networks switching from hypo- to hyper-coupling may represent a mechanism underlying the pathophysiology of cognitive deficits in neuropsychiatric disorders.


Assuntos
Transtornos Cognitivos , Deficiências do Desenvolvimento , Interação Gene-Ambiente , Hipocampo/fisiologia , Vias Neurais/fisiologia , Córtex Pré-Frontal/fisiopatologia , Animais , Animais Recém-Nascidos , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Deficiências do Desenvolvimento/induzido quimicamente , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/genética , Feminino , Hipocampo/efeitos dos fármacos , Indutores de Interferon/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/efeitos dos fármacos , Poli I-C/toxicidade , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
20.
Exp Neurol ; 283(Pt A): 142-50, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27302677

RESUMO

Schizophrenia is a debilitating psychiatric disorder with a significant number of patients not adequately responding to treatment. Deep brain stimulation (DBS) is a surgical technique currently investigated for medically-refractory psychiatric disorders. Here, we use the poly I:C rat model of schizophrenia to study the effects of medial prefrontal cortex (mPFC) and nucleus accumbens (Nacc) DBS on two behavioral schizophrenia-like deficits, i.e. sensorimotor gating, as reflected by disrupted prepulse inhibition (PPI), and attentional selectivity, as reflected by disrupted latent inhibition (LI). In addition, the neurocircuitry influenced by DBS was studied using FDG PET. We found that mPFC- and Nacc-DBS alleviated PPI and LI abnormalities in poly I:C offspring, whereas Nacc- but not mPFC-DBS disrupted PPI and LI in saline offspring. In saline offspring, mPFC-DBS increased metabolism in the parietal cortex, striatum, ventral hippocampus and Nacc, while reducing it in the brainstem, cerebellum, hypothalamus and periaqueductal gray. Nacc-DBS, on the other hand, increased activity in the ventral hippocampus and olfactory bulb and reduced it in the septal area, brainstem, periaqueductal gray and hypothalamus. In poly I:C offspring changes in metabolism following mPFC-DBS were similar to those recorded in saline offspring, except for a reduced activity in the brainstem and hypothalamus. In contrast, Nacc-DBS did not induce any statistical changes in brain metabolism in poly I:C offspring. Our study shows that mPFC- or Nacc-DBS delivered to the adult progeny of poly I:C treated dams improves deficits in PPI and LI. Despite common behavioral responses, stimulation in the two targets induced different metabolic effects.


Assuntos
Encéfalo/diagnóstico por imagem , Estimulação Encefálica Profunda , Transtornos Neurológicos da Marcha/etiologia , Transtornos Mentais/etiologia , Transtornos Mentais/terapia , Esquizofrenia/complicações , Esquizofrenia/patologia , Estimulação Acústica , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fluordesoxiglucose F18/farmacocinética , Indutores de Interferon/toxicidade , Masculino , Transtornos Mentais/diagnóstico por imagem , Núcleo Accumbens/fisiologia , Poli I-C/toxicidade , Tomografia por Emissão de Pósitrons , Córtex Pré-Frontal/fisiologia , Inibição Pré-Pulso/fisiologia , Ratos , Ratos Wistar , Reflexo de Sobressalto/fisiologia , Esquizofrenia/induzido quimicamente , Esquizofrenia/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...