Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Lakartidningen ; 1192022 05 10.
Artigo em Sueco | MEDLINE | ID: mdl-35535651

RESUMO

In recent years several previously unknown autoimmune disorders that affect the central nervous system have been described. Antibodies against glial fibrillary acidic protein (GFAp) were identified in a new form of encephalomyelitis, where the clinical phenotype differs from neuromyelitis optica spectrum disorders. The clinical picture includes headache, encephalopathy, optic neuritis and myelitis. GFAp is a structural component of the cytoskeleton of astrocytes, and an elevated concentration of GFAp in the cerebrospinal fluid, together with antibodies, is a marker of astrocytopathy. The prognosis is good with immunosuppressive treatment.


Assuntos
Encefalomielite , Mielite , Neuromielite Óptica , Anticorpos , Astrócitos/metabolismo , Autoanticorpos , Encefalomielite/diagnóstico , Encefalomielite/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Neuromielite Óptica/diagnóstico , Neuromielite Óptica/tratamento farmacológico
2.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34400495

RESUMO

Although most patients recover from acute COVID-19, some experience postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection (PASC). One subgroup of PASC is a syndrome called "long COVID-19," reminiscent of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS is a debilitating condition, often triggered by viral and bacterial infections, leading to years-long debilitating symptoms including profound fatigue, postexertional malaise, unrefreshing sleep, cognitive deficits, and orthostatic intolerance. Some are skeptical that either ME/CFS or long COVID-19 involves underlying biological abnormalities. However, in this review, we summarize the evidence that people with acute COVID-19 and with ME/CFS have biological abnormalities including redox imbalance, systemic inflammation and neuroinflammation, an impaired ability to generate adenosine triphosphate, and a general hypometabolic state. These phenomena have not yet been well studied in people with long COVID-19, and each of them has been reported in other diseases as well, particularly neurological diseases. We also examine the bidirectional relationship between redox imbalance, inflammation, energy metabolic deficits, and a hypometabolic state. We speculate as to what may be causing these abnormalities. Thus, understanding the molecular underpinnings of both PASC and ME/CFS may lead to the development of novel therapeutics.


Assuntos
COVID-19/metabolismo , Encefalomielite/metabolismo , Síndrome de Fadiga Crônica/metabolismo , Animais , COVID-19/complicações , COVID-19/etiologia , COVID-19/imunologia , Encefalomielite/imunologia , Síndrome de Fadiga Crônica/imunologia , Humanos , Oxirredução , Síndrome de COVID-19 Pós-Aguda
3.
Neuropathol Appl Neurobiol ; 47(2): 316-327, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32910464

RESUMO

AIMS: Progressive encephalomyelitis with rigidity and myoclonus (PERM) is a life-threatening condition often associated with highly raised serum antibodies to glycine receptors (GlyRs); these bind to the surface of large neurons and interneurons in rodent brain and spinal cord sections and, in vitro, inhibit function and reduce surface expression of the GlyRs. The effects in vivo have not been reported. METHODS: Purified plasma IgG from a GlyR antibody-positive patient with PERM, and a healthy control (HC), was injected daily into the peritoneal cavity of mice for 12 days; lipopolysaccharide (LPS) to open the blood-brain barrier, was injected on days 3 and 8. Based on preliminary data, behavioural tests were only performed 48 h post-LPS on days 5-7 and 10-12. RESULTS: The GlyR IgG injected mice showed impaired ability on the rotarod from days 5 to 10 but this normalized by day 12. There were no other behavioural differences but, at termination (d13), the GlyR IgG-injected mice had IgG deposits on the neurons that express GlyRs in the brainstem and spinal cord. The IgG was not only on the surface but also inside these large GlyR expressing neurons, which continued to express surface GlyR. CONCLUSIONS: Despite the partial clinical phenotype, not uncommon in passive transfer studies, the results suggest that the antibodies had accessed the GlyRs in relevant brain regions, led to antibody-mediated internalization and increased GlyR synthesis, compatible with the temporary loss of function.


Assuntos
Autoanticorpos/farmacologia , Encefalomielite/imunologia , Imunoglobulina G/farmacologia , Neurônios Motores/metabolismo , Rigidez Muscular/imunologia , Receptores de Glicina/metabolismo , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Autoantígenos/metabolismo , Tronco Encefálico/imunologia , Tronco Encefálico/metabolismo , Encefalomielite/metabolismo , Humanos , Imunoglobulina G/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/imunologia , Rigidez Muscular/metabolismo , Mioclonia/imunologia , Mioclonia/metabolismo , Receptores de Glicina/imunologia , Medula Espinal/imunologia , Medula Espinal/metabolismo
4.
Ann Neurol ; 88(3): 544-561, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32588476

RESUMO

OBJECTIVE: Impairment of glycinergic neurotransmission leads to complex movement and behavioral disorders. Patients harboring glycine receptor autoantibodies suffer from stiff-person syndrome or its severe variant progressive encephalomyelitis with rigidity and myoclonus. Enhanced receptor internalization was proposed as the common molecular mechanism upon autoantibody binding. Although functional impairment of glycine receptors following autoantibody binding has recently been investigated, it is still incompletely understood. METHODS: A cell-based assay was used for positive sample evaluation. Glycine receptor function was assessed by electrophysiological recordings and radioligand binding assays. The in vivo passive transfer of patient autoantibodies was done using the zebrafish animal model. RESULTS: Glycine receptor function as assessed by glycine dose-response curves showed significantly decreased glycine potency in the presence of patient sera. Upon binding of autoantibodies from 2 patients, a decreased fraction of desensitized receptors was observed, whereas closing of the ion channel remained fast. The glycine receptor N-terminal residues 29 A to 62 G were mapped as a common epitope of glycine receptor autoantibodies. An in vivo transfer into the zebrafish animal model generated a phenotype with disturbed escape behavior accompanied by a reduced number of glycine receptor clusters in the spinal cord of affected animals. INTERPRETATION: Autoantibodies against the extracellular domain mediate alterations of glycine receptor physiology. Moreover, our in vivo data demonstrate that the autoantibodies are a direct cause of the disease, because the transfer of human glycine receptor autoantibodies to zebrafish larvae generated impaired escape behavior in the animal model compatible with abnormal startle response in stiff-person syndrome or progressive encephalitis with rigidity and myoclonus patients. ANN NEUROL 2020;88:544-561.


Assuntos
Autoanticorpos/imunologia , Encefalomielite/imunologia , Rigidez Muscular/imunologia , Receptores de Glicina/metabolismo , Rigidez Muscular Espasmódica/imunologia , Adulto , Idoso , Animais , Autoanticorpos/farmacologia , Autoantígenos/imunologia , Comportamento Animal/efeitos dos fármacos , Encefalomielite/metabolismo , Epitopos de Linfócito B/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Rigidez Muscular/metabolismo , Receptores de Glicina/imunologia , Rigidez Muscular Espasmódica/metabolismo , Peixe-Zebra
5.
Proc Natl Acad Sci U S A ; 116(51): 25860-25869, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31796589

RESUMO

Environmental triggers acting at the intestinal barrier are thought to contribute to the initiation of autoimmune disorders. The transforming growth factor beta inhibitor Smad7 determines the phenotype of CD4+ T cells. We hypothesized that Smad7 in intestinal CD4+ T cells controls initiation of opticospinal encephalomyelitis (OSE), a murine model of multiple sclerosis (MS), depending on the presence of gut microbiota. Smad7 was overexpressed or deleted in OSE CD4+ T cells to determine the effect on clinical progression, T cell differentiation, and T cell migration from the intestine to the central nervous system (CNS). Smad7 overexpression worsened the clinical course of OSE and increased CNS inflammation and demyelination. It favored expansion of intestinal CD4+ T cells toward an inflammatory phenotype and migration of intestinal CD4+ T cells to the CNS. Intestinal biopsies from MS patients revealed decreased transforming growth factor beta signaling with a shift toward inflammatory T cell subtypes. Smad7 in intestinal T cells might represent a valuable therapeutic target for MS to achieve immunologic tolerance in the intestine and suppress CNS inflammation.


Assuntos
Autoimunidade/fisiologia , Linfócitos T CD4-Positivos/imunologia , Sistema Nervoso Central/metabolismo , Esclerose Múltipla/metabolismo , Proteína Smad7/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Encefalomielite/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Humanos , Tolerância Imunológica , Inflamação , Intestinos/patologia , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/patologia , Transdução de Sinais , Proteína Smad7/genética , Medula Espinal/patologia , Fator de Crescimento Transformador beta/metabolismo
6.
Neurology ; 92(21): e2406-e2420, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31028126

RESUMO

OBJECTIVE: To characterize the full spectrum, relative frequency, and prognosis of the neurologic manifestations in Zika virus (ZIKV) postnatal infection. METHODS: We conducted an observational study in consecutive ZIKV-infected patients presenting with neurologic manifestations during the French West Indies 2016 outbreak. RESULTS: Eighty-seven patients, including 6 children, were enrolled. Ninety-five percent of all cases required hospitalization. Guillain-Barré syndrome was the most frequent manifestation (46.0%) followed by encephalitis or encephalomyelitis (20.7%), isolated single or multiple cranial nerve palsies (9.2%), other peripheral manifestations (6.9%), and stroke (1.1%). Fourteen patients (16.1%), including one child, developed a mixed disorder involving both the central and peripheral nervous system. Mechanical ventilation was required in 21 cases, all of whom had ZIKV RNA in at least one biological fluid. Two adult patients died due to neuroZika. Clinical follow-up (median 14 months; interquartile range, 13-17 months) was available for 76 patients. Residual disability (modified Rankin Scale score ≥2) was identified in 19 (25.0%) patients; in 6 cases (7.9%), disability was severe (modified Rankin Scale score ≥4). Among patients with ZIKV RNA detected in one biological fluid, the risk of residual disability or death was higher (odds ratio 9.19; confidence interval 1.12-75.22; p = 0.039). CONCLUSIONS: NeuroZika spectrum represents a heterogeneous group of clinical neurologic manifestations. During an outbreak, clinicians should consider neuroZika in patients presenting with cranial nerve palsies and a mixed neurologic disorder. Long-term sequelae are frequent in NeuroZika. ZIKV reverse-transcription PCR status at admission can inform prognosis and should therefore be taken into consideration in the management of hospitalized patients.


Assuntos
Doenças dos Nervos Cranianos/terapia , Encefalite Viral/terapia , Encefalomielite/terapia , Síndrome de Guillain-Barré/fisiopatologia , Infecção por Zika virus/terapia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Doenças dos Nervos Cranianos/metabolismo , Doenças dos Nervos Cranianos/fisiopatologia , Encefalite Viral/metabolismo , Encefalite Viral/fisiopatologia , Encefalomielite/metabolismo , Encefalomielite/fisiopatologia , Feminino , Hospitalização , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Viral/sangue , RNA Viral/líquido cefalorraquidiano , RNA Viral/urina , Respiração Artificial , Resultado do Tratamento , Índias Ocidentais , Infecção por Zika virus/metabolismo , Infecção por Zika virus/fisiopatologia
7.
Med Hypotheses ; 118: 59-67, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30037616

RESUMO

It is thought that an ill defined biochemical cascade may lead to protracted withdrawal symptoms subsequent to discontinuance of routine use of benzodiazepine class drugs and establish chronic illness in some patients. In this review, published findings are presented that support the novel concept that withdrawal from benzodiazepine class drugs can trigger elevated and sustained levels of a potent oxidant called peroxynitrite via potentiation of the L-type voltage-gated calcium channels, and in the later stages of withdrawal, via excessive N-methyl-D-aspartate receptor activity, as well. Potentiation of L-type voltage-gated calcium channels and excessive N-methyl-D-aspartate receptor activity both result in calcium influx into the cell that triggers nitric oxide synthesis. In pathophysiological conditions, such increased nitric oxide synthesis leads to peroxynitrite formation. The downstream effects of peroxynitrite formation that may occur during withdrawal ultimately lead to further peroxynitrite production in a system of overlapping vicious cycles collectively referred to as the NO/ONOO(-) cycle. Once triggered, the elements of the NO/ONOO(-) cycle perpetuate pathophysiology, perhaps including reduced GABAA receptor functioning, that may explain protracted withdrawal associated symptoms while the vicious cycle nature of the NO/ONOO(-) cycle may explain how withdrawal becomes a chronic state. Suboptimal levels of tetrahydrobiopterin may be one risk factor for the development of the protracted withdrawal syndrome as this will lead to partial nitric oxide uncoupling and resultant peroxynitrite formation. Nitric oxide uncoupling results in superoxide production as calcium-dependent nitric oxide synthases attempt to produce nitric oxide in response to L-type voltage-gated calcium channel-mediated calcium influx that is known to occur during withdrawal. The combination of nitric oxide and superoxide produced, as when partial uncoupling occurs, react together in a very rapid, diffusion limited reaction to form peroxynitrite and thereby trigger the NO/ONOO(-) cycle. The NO/ONOO(-) cycle may explain the nature of the protracted withdrawal syndrome and the related constellation of symptoms that are also common in other illnesses characterized as NO/ONOO(-) disorders such as myalgic encephalomyelitis/chronic fatigue syndrome and fibromyalgia.


Assuntos
Benzodiazepinas/administração & dosagem , Benzodiazepinas/efeitos adversos , Síndrome de Abstinência a Substâncias , Animais , Cálcio/metabolismo , Doença Crônica , Encefalomielite/metabolismo , Síndrome de Fadiga Crônica/metabolismo , Fibromialgia/metabolismo , Humanos , Modelos Teóricos , Nitratos/química , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Oxidantes/metabolismo , Ácido Peroxinitroso/química , Receptores de N-Metil-D-Aspartato/metabolismo , Fatores de Risco , Superóxidos/química
8.
Nat Commun ; 9(1): 251, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343683

RESUMO

Hypoxia-inducible factors (HIFs) are key elements for controlling immune cell metabolism and functions. While HIFs are known to be involved in T cells and macrophages activation, their functions in B lymphocytes are poorly defined. Here, we show that hypoxia-inducible factor-1α (HIF-1α) contributes to IL-10 production by B cells. HIF-1α regulates IL-10 expression, and HIF-1α-dependent glycolysis facilitates CD1dhiCD5+ B cells expansion. Mice with B cell-specific deletion of Hif1a have reduced number of IL-10-producing B cells, which result in exacerbated collagen-induced arthritis and experimental autoimmune encephalomyelitis. Wild-type CD1dhiCD5+ B cells, but not Hif1a-deficient CD1dhiCD5+ B cells, protect recipient mice from autoimmune disease, while the protective function of Hif1a-deficient CD1dhiCD5+ B cells is restored when their defective IL-10 expression is genetically corrected. Taken together, this study demonstrates the key function of the hypoxia-associated transcription factor HIF-1α in driving IL-10 expression in CD1dhiCD5+ B cells, and in controlling their protective activity in autoimmune disease.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos B/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Interleucina-10/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Doenças Autoimunes/metabolismo , Encefalomielite/imunologia , Encefalomielite/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos
9.
J Biol Chem ; 292(35): 14532-14543, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28684423

RESUMO

In both multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), the C-C chemokine receptor 6 (CCR6) is critical for pathogenic T helper 17 (Th17) cell migration to the central nervous system (CNS). Whereas many cytokines and their receptors are potently regulated via post-transcriptional mechanisms in response to various stimuli, how CCR6 expression is post-transcriptionally regulated in Th17 cells is unknown. Here, using RNA-binding protein HuR conditional knock-out (KO) and wild-type (WT) mice, we present evidence that HuR post-transcriptionally regulates CCR6 expression by binding to and stabilizing Ccr6 mRNA and by promoting CCR6 translation. We also found that HuR down-regulates several microRNA expressions, which could target the 3'-UTR of Ccr6 mRNA for decay. Accordingly, knock-out of HuR reduced CCR6 expression on Th17 cells and impaired their migration to CNS compared with the response of WT Th17 cells and thereby ameliorated EAE. Together, these findings highlight how HuR contributes to Th17 cell-mediated autoimmune neuroinflammation and support the notion that targeting HuR might be a potential therapeutic intervention for managing autoimmune disorders of the CNS.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Regulação da Expressão Gênica , RNA Mensageiro/metabolismo , Receptores CCR6/agonistas , Linfócitos T Auxiliares-Indutores/metabolismo , Regiões 3' não Traduzidas , Animais , Doenças Autoimunes do Sistema Nervoso/imunologia , Doenças Autoimunes do Sistema Nervoso/metabolismo , Doenças Autoimunes do Sistema Nervoso/patologia , Linhagem Celular , Movimento Celular , Células Cultivadas , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Proteína Semelhante a ELAV 1/antagonistas & inibidores , Proteína Semelhante a ELAV 1/genética , Encefalomielite/imunologia , Encefalomielite/metabolismo , Encefalomielite/patologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/metabolismo , Biossíntese de Proteínas , Interferência de RNA , Estabilidade de RNA , Receptores CCR6/antagonistas & inibidores , Receptores CCR6/genética , Receptores CCR6/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia
10.
J Virol ; 90(20): 9251-62, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27489275

RESUMO

UNLABELLED: Inflammation is a necessary part of the response to infection but can also cause neuronal injury in both infectious and autoimmune diseases of the central nervous system (CNS). A neurovirulent strain of Sindbis virus (NSV) causes fatal paralysis in adult C57BL/6 mice during clearance of infectious virus from the CNS, and the virus-specific immune response is implicated as a mediator of neuronal damage. Previous studies have shown that survival is improved in T-cell-deficient mice and in mice with pharmacological inhibition of the inflammatory response and glutamate excitotoxicity. Because glutamine metabolism is important in the CNS for the generation of glutamate and in the immune system for lymphocyte proliferation, we tested the effect of the glutamine antagonist DON (6-diazo-5-oxo-l-norleucine) on the outcome of NSV infection in mice. DON treatment for 7 days from the time of infection delayed the onset of paralysis and death. Protection was associated with reduced lymphocyte proliferation in the draining cervical lymph nodes, decreased leukocyte infiltration into the CNS, lower levels of inflammatory cytokines, and delayed viral clearance. In vitro studies showed that DON inhibited stimulus-induced proliferation of lymphocytes. When in vivo treatment with DON was stopped, paralytic disease developed along with the inflammatory response and viral clearance. These studies show that fatal NSV-induced encephalomyelitis is immune mediated and that antagonists of glutamine metabolism can modulate the immune response and protect against virus-induced neuroinflammatory disease. IMPORTANCE: Encephalomyelitis due to infection with mosquito-borne alphaviruses is an important cause of death and of long-term neurological disability in those who survive infection. This study demonstrates the role of the virus-induced immune response in the generation of neurological disease. DON, a glutamine antagonist, inhibited the proliferation of lymphocytes in response to infection, prevented the development of brain inflammation, and protected mice from paralysis and death during treatment. However, because DON inhibited the immune response to infection, clearance of the virus from the brain was also prevented. When treatment was stopped, the immune response was generated, brain inflammation occurred, virus was cleared, and mice developed paralysis and died. Therefore, more definitive treatment for alphaviral encephalomyelitis should inhibit virus replication as well as neuroinflammatory damage.


Assuntos
Infecções por Alphavirus/tratamento farmacológico , Alphavirus/efeitos dos fármacos , Diazo-Oxo-Norleucina/farmacologia , Encefalomielite/tratamento farmacológico , Encefalomielite/virologia , Glutamina/antagonistas & inibidores , Infecções por Alphavirus/virologia , Animais , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Citocinas/metabolismo , Encefalite/metabolismo , Encefalite/virologia , Encefalite Viral/tratamento farmacológico , Encefalite Viral/virologia , Encefalomielite/metabolismo , Feminino , Linfócitos/metabolismo , Linfócitos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Paralisia/metabolismo , Paralisia/virologia , Sindbis virus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
11.
Brain Pathol ; 26(1): 102-19, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25959295

RESUMO

High dietary fat and/or cholesterol intake is a risk factor for multiple diseases and has been debated for multiple sclerosis. However, cholesterol biosynthesis is a key pathway during myelination and disturbances are described in demyelinating diseases. To address the possible interaction of dyslipidemia and demyelination, cholesterol biosynthesis gene expression, composition of the body's major lipid repositories and Paigen diet-induced, systemic hypercholesterolemia were examined in Theiler's murine encephalomyelitis (TME) using histology, immunohistochemistry, serum clinical chemistry, microarrays and high-performance thin layer chromatography. TME-virus (TMEV)-infected mice showed progressive loss of motor performance and demyelinating leukomyelitis. Gene expression associated with cholesterol biosynthesis was overall down-regulated in the spinal cord of TMEV-infected animals. Spinal cord levels of galactocerebroside and sphingomyelin were reduced on day 196 post TMEV infection. Paigen diet induced serum hypercholesterolemia and hepatic lipidosis. However, high dietary fat and cholesterol intake led to no significant differences in clinical course, inflammatory response, astrocytosis, and the amount of demyelination and remyelination in the spinal cord of TMEV-infected animals. The results suggest that down-regulation of cholesterol biosynthesis is a transcriptional marker for demyelination, quantitative loss of myelin-specific lipids, but not cholesterol occurs late in chronic demyelination, and serum hypercholesterolemia exhibited no significant effect on TMEV infection.


Assuntos
Sistema Nervoso Central/patologia , Colesterol/sangue , Doenças Desmielinizantes/etiologia , Encefalomielite , Recuperação de Função Fisiológica/fisiologia , Animais , Antígenos CD , Vias Biossintéticas/genética , Cromatografia em Camada Fina , Dieta com Restrição de Gorduras , Modelos Animais de Doenças , Encefalomielite/complicações , Encefalomielite/metabolismo , Encefalomielite/virologia , Feminino , Regulação Viral da Expressão Gênica/fisiologia , Fígado/metabolismo , Camundongos , Análise em Microsséries , Theilovirus/fisiologia , Fatores de Tempo
12.
Brain Pathol ; 25(6): 712-23, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25495532

RESUMO

Microglia and macrophages play a central role for demyelination in Theiler's murine encephalomyelitis (TME) virus infection, a commonly used infectious model for chronic-progressive multiple sclerosis. In order to determine the dynamic changes of microglia/macrophage polarization in TME, the spinal cord of Swiss Jim Lambert (SJL) mice was investigated by gene expression profiling and immunofluorescence. Virus persistence and demyelinating leukomyelitis were confirmed by immunohistochemistry and histology. Electron microscopy revealed continuous myelin loss together with abortive myelin repair during the late chronic infection phase indicative of incomplete remyelination. A total of 59 genes out of 151 M1- and M2-related genes were differentially expressed in TME virus-infected mice over the study period. The onset of virus-induced demyelination was associated with a dominating M1 polarization, while mounting M2 polarization of macrophages/microglia together with sustained prominent M1-related gene expression was present during the chronic-progressive phase. Molecular results were confirmed by immunofluorescence, showing an increased spinal cord accumulation of CD16/32(+) M1-, arginase-1(+) M2- and Ym1(+) M2-type cells associated with progressive demyelination. The present study provides a comprehensive database of M1-/M2-related gene expression involved in the initiation and progression of demyelination supporting the hypothesis that perpetuating interaction between virus and macrophages/microglia induces a vicious circle with persistent inflammation and impaired myelin repair in TME.


Assuntos
Infecções por Cardiovirus/metabolismo , Encefalomielite/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Theilovirus , Animais , Infecções por Cardiovirus/patologia , Encefalomielite/patologia , Feminino , Imunofluorescência , Imuno-Histoquímica , Macrófagos/ultraestrutura , Macrófagos/virologia , Camundongos , Análise em Microsséries , Microglia/ultraestrutura , Microglia/virologia , Microscopia Eletrônica , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , Neuroimunomodulação/fisiologia , Medula Espinal/metabolismo , Medula Espinal/ultraestrutura , Medula Espinal/virologia
13.
Arthritis Rheumatol ; 66(2): 327-39, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24504805

RESUMO

OBJECTIVE: Rheumatoid arthritis (RA) is an autoimmune disease caused by loss of immunologic self tolerance and characterized by chronic joint inflammation. Cells isolated from human amniotic membrane (HAMCs) were recently found to display immunosuppressive properties. The aim of this study was to characterize the effect of HAMCs on antigen-specific T cell responses in RA patients and to evaluate their therapeutic potential in a preclinical experimental model of RA. METHODS: We investigated the effects of HAMCs on collagen-reactive T cell proliferation and cytokine production, on the production of mediators of inflammation by synoviocytes, and on the generation of Treg cells in peripheral blood mononuclear cells and synovial membrane cells isolated from RA patients. Mice with collagen-induced arthritis (CIA) were treated with HAMCs after disease onset, and clinical scores and joint levels of mediators of inflammation were evaluated. We determined Th1/Th17-mediated autoreactive responses in the mice by measuring the proliferation and the cytokine profile of lymph node cells restimulated with collagen. RESULTS: Treatment with HAMCs suppressed synovial inflammatory responses and antigen-specific Th1/Th17 activation in cells isolated from RA patients. Moreover, HAMCs stimulated the generation of human CD4+CD25+FoxP3+ Treg cells with a capacity to suppress collagen-specific T cell responses. Systemic infusion of HAMCs significantly reduced the incidence and severity of CIA by down-regulating the 2 deleterious components of disease: Th1-driven autoimmunity and inflammation. In mice with CIA, HAMC treatment decreased the production of various inflammatory cytokines and chemokines in the joints, impaired antigen-specific Th1/Th17 cell expansion in the lymph nodes, and generated peripheral antigen-specific Treg cells. HAMCs also protected the mice from experimental sepsis, inflammatory bowel disease, and autoimmune encephalomyelitis. CONCLUSION: HAMCs have emerged as attractive candidates for a cell-based therapy for RA.


Assuntos
Âmnio/citologia , Artrite Experimental/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Colite/terapia , Encefalomielite/terapia , Sepse/terapia , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Proliferação de Células , Células Cultivadas , Colite/metabolismo , Colite/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite/metabolismo , Encefalomielite/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Sepse/metabolismo , Sepse/patologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Linfócitos T/patologia , Resultado do Tratamento
14.
Glia ; 60(10): 1555-66, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22736486

RESUMO

Type I interferons (IFNα/ß) provide a primary defense against infection. Nevertheless, the dynamics of IFNα/ß induction and responsiveness by central nervous system (CNS) resident cells in vivo in response to viral infections are poorly understood. Mice were infected with a neurotropic coronavirus with tropism for oligodendroglia and microglia to probe innate antiviral responses during acute encephalomyelitis. Expression of genes associated with the IFNα/ß pathways was monitored in microglia and oligodendroglia purified from naïve and infected mice by fluorescent activated cell sorting. Compared with microglia, oligodendroglia were characterized by low basal expression of mRNA encoding viral RNA sensing pattern recognition receptors (PRRs), IFNα/ß receptor chains, interferon sensitive genes (ISG), as well as kinases and transcription factors critical in IFNα/ß signaling. Although PRRs and ISGs were upregulated by infection in both cell types, the repertoire and absolute mRNA levels were more limited in oligodendroglia. Furthermore, although oligodendroglia harbored higher levels of viral RNA compared with microglia, Ifnα/ß was only induced in microglia. Stimulation with the double stranded RNA analogue poly I:C also failed to induce Ifnα/ß in oligodendroglia, and resulted in reduced and delayed induction of ISGs compared with microglia. The limited antiviral response by oligodendroglia was associated with a high threshold for upregulation of Ikkε and Irf7 transcripts, both central to amplifying IFNα/ß responses. Overall, these data reveal that oligodendroglia from the adult CNS are poor sensors of viral infection and suggest they require exogenous IFNα/ß to establish an antiviral state.


Assuntos
Interferon Tipo I/metabolismo , Oligodendroglia/metabolismo , Animais , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Encefalomielite/genética , Encefalomielite/metabolismo , Encefalomielite/patologia , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Indutores de Interferon/farmacologia , Helicase IFIH1 Induzida por Interferon , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Oligodendroglia/efeitos dos fármacos , Poli I-C/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
Immunol Rev ; 248(1): 23-35, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22725952

RESUMO

In multiple sclerosis, type I interferon (IFN) is considered immune-modulatory, and recombinant forms of IFN-ß are the most prescribed treatment for this disease. This is in contrast to most other autoimmune disorders, because type I IFN contributes to the pathologies. Even within the relapsing-remitting multiple sclerosis (RRMS) population, 30-50% of MS patients are non-responsive to this treatment, and it consistently worsens neuromyelitis optica, a disease similar to RRMS. In this article, we discuss the recent advances in the field of autoimmunity and introduce the theory explain how type I IFNs can be pro-inflammatory in disease that is predominantly driven by a Th17 response and are therapeutic when disease is predominantly Th1.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Interferon Tipo I/uso terapêutico , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Modelos Animais de Doenças , Encefalomielite/tratamento farmacológico , Encefalomielite/imunologia , Encefalomielite/metabolismo , Humanos , Interferon Tipo I/efeitos adversos , Interferon Tipo I/metabolismo , Interferon beta/efeitos adversos , Interferon beta/metabolismo , Interferon beta/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
16.
J Nutr Biochem ; 23(11): 1498-507, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22402368

RESUMO

Nutraceuticals and phytochemicals are important regulators of human health and diseases. Curcumin is a polyphenolic phytochemical isolated from the rhizome of the plant Curcuma longa (turmeric) that has been traditionally used for the treatment of inflammation and wound healing for centuries. Systematic analyses have shown that curcumin exerts its beneficial effects through antioxidant, antiproliferative and anti-inflammatory properties. We and others have shown earlier that curcumin ameliorates experimental autoimmune encephalomyelitis (EAE) model for multiple sclerosis. In this study, we show that C57BL/6 mice induced to develop EAE express elevated levels of interferon (IFN) γ and interleukin (IL)-17 in the central nervous system (CNS) and lymphoid organs that decreased significantly following in vivo treatment with curcumin. The EAE mice also showed elevated expression of IL-12 and IL-23 that decreased after treatment with curcumin. Ex vivo and in vitro treatment with curcumin resulted in a dose-dependent decrease in the secretion of IFNγ, IL-17, IL-12 and IL-23 in culture. The inhibition of EAE by curcumin was also associated with an up-regulation of IL-10, peroxisome proliferator activated receptor γ and CD4(+)CD25(+-)Foxp3(+) Treg cells in the CNS and lymphoid organs. These findings highlight that curcumin differentially regulates CD4(+) T helper cell responses in EAE.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Curcumina/farmacologia , Encefalomielite/tratamento farmacológico , Encefalomielite/imunologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalomielite/metabolismo , Feminino , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Doença Autoimune do Sistema Nervoso Experimental/tratamento farmacológico , Baço/citologia , Baço/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
17.
PLoS One ; 6(9): e24538, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931744

RESUMO

Radial glia (RG) are primarily embryonic neuroglial progenitors that express Brain Lipid Binding Protein (Blbp a.k.a. Fabp7) and Glial Fibrillary Acidic Protein (Gfap). We used these transcripts to demarcate the distribution of spinal cord radial glia (SCRG) and screen for SCRG gene expression in the Allen Spinal Cord Atlas (ASCA). We reveal that neonatal and adult SCRG are anchored in a non-ventricular niche at the spinal cord (SC) pial boundary, and express a "signature" subset of 122 genes, many of which are shared with "classic" neural stem cells (NSCs) of the subventricular zone (SVZ) and SC central canal (CC). A core expressed gene set shared between SCRG and progenitors of the SVZ and CC is particularly enriched in genes associated with human disease. Visualizing SCRG in a Fabp7-EGFP reporter mouse reveals an extensive population of SCRG that extend processes around the SC boundary and inwardly (through) the SC white matter (WM), whose abundance increases in a gradient from cervical to lumbar SC. Confocal analysis of multiple NSC-enriched proteins reveals that postnatal SCRG are a discrete and heterogeneous potential progenitor population that become activated by multiple SC lesions, and that CC progenitors are also more heterogeneous than previously appreciated. Gene ontology analysis highlights potentially unique regulatory pathways that may be further manipulated in SCRG to enhance repair in the context of injury and SC disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neuroglia/patologia , Medula Espinal/patologia , Células-Tronco/citologia , Animais , Doenças Autoimunes/patologia , Encefalomielite/metabolismo , Proteína 7 de Ligação a Ácidos Graxos , Proteínas de Ligação a Ácido Graxo/metabolismo , Perfilação da Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Fenótipo , Medula Espinal/citologia , Traumatismos da Medula Espinal/patologia
18.
Acta Neuropathol ; 122(1): 21-34, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21468722

RESUMO

Inflammatory lesions in the central nervous system of patients with neuromyelitis optica are characterized by infiltration of T cells and deposition of aquaporin-4-specific antibodies and complement on astrocytes at the glia limitans. Although the contribution of aquaporin-4-specific autoantibodies to the disease process has been recently elucidated, a potential role of aquaporin-4-specific T cells in lesion formation is unresolved. To address this issue, we raised aquaporin-4-specific T cell lines in Lewis rats and characterized their pathogenic potential in the presence and absence of aquaporin-4-specific autoantibodies of neuromyelitis optica patients. We show that aquaporin-4-specific T cells induce brain inflammation with particular targeting of the astrocytic glia limitans and permit the entry of pathogenic anti-aquaporin-4-specific antibodies to induce NMO-like lesions in spinal cord and brain. In addition, transfer of aquaporin-4-specific T cells provoked mild (subclinical) myositis and interstitial nephritis. We further show that the expression of the conformational epitope, recognized by NMO patient-derived aquaporin-4-specific antibodies is induced in kidney cells by the pro-inflammatory cytokine gamma-interferon. Our data provide further support for the view that NMO lesions may be induced by a complex interplay of T cell mediated and humoral immune responses against aquaporin-4.


Assuntos
Aquaporina 4/imunologia , Autoanticorpos/imunologia , Encefalomielite/imunologia , Encefalomielite/fisiopatologia , Linfócitos T/imunologia , Linfócitos T/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Autoanticorpos/metabolismo , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Encefalomielite/metabolismo , Epitopos , Humanos , Imunoglobulina G/metabolismo , Interferon gama/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Neuromielite Óptica/imunologia , Ratos , Ratos Endogâmicos Lew , Linfócitos T/metabolismo
19.
Curr Drug Targets ; 11(12): 1605-13, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20735353

RESUMO

Prostanoids, consisting of prostaglandins and thromboxane, are cyclooxygenase metabolites of arachidonic acid released in various pathophysiological conditions which exert a range of actions mediated through their respective receptors expressed on target cells. Although it has been difficult to analyze the physiological role of prostanoids, recent developments in both the disruption of the respective gene and receptor selective compounds have enabled us to investigate the physiological roles for each receptor. It has been demonstrated that each prostanoid receptor has multiple functions, and that their expression is regulated in a context-dependent manner that sometimes results in opposite, excitatory and inhibitory, outcomes. The balance of prostanoid production and receptor expression has been revealed to be important for homeostasis of the human body. Here, we review new findings on the roles of prostanoids in allergic and immune diseases, focusing on contact dermatitis, atopic dermatitis, asthma, rheumatoid arthritis, and encephalomyelitis, and also discuss the clinical potentials of receptor-selective drugs.


Assuntos
Antialérgicos/farmacologia , Desenho de Fármacos , Hipersensibilidade/tratamento farmacológico , Prostaglandinas/fisiologia , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Tromboxanos/antagonistas & inibidores , Tromboxanos/fisiologia , Animais , Antialérgicos/uso terapêutico , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Encefalomielite/tratamento farmacológico , Encefalomielite/imunologia , Encefalomielite/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Imunização , Terapia de Alvo Molecular , Antagonistas de Prostaglandina/farmacologia , Antagonistas de Prostaglandina/uso terapêutico , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/fisiologia , Receptores de Tromboxanos/genética , Receptores de Tromboxanos/fisiologia , Pele/efeitos dos fármacos , Pele/imunologia , Pele/metabolismo , Tromboxanos/antagonistas & inibidores
20.
J Biol Chem ; 285(38): 29039-43, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20667820

RESUMO

Development of Foxp3(+) regulatory T cells and pro-inflammatory Th17 cells from naive CD4(+) T cells requires transforming growth factor-ß (TGF-ß) signaling. Although Smad4 and Smad3 have been previously shown to regulate Treg cell induction by TGF-ß, they are not required in the development of Th17 cells. Thus, how TGF-ß regulates Th17 cell differentiation remains unclear. In this study, we found that TGF-ß-induced Foxp3 expression was significantly reduced in the absence of Smad2. More importantly, Smad2 deficiency led to reduced Th17 differentiation in vitro and in vivo. In the experimental autoimmune encephalomyelitis model, Smad2 deficiency in T cells significantly ameliorated disease severity and reduced generation of Th17 cells. Furthermore, we found that Smad2 associated with retinoid acid receptor-related orphan receptor-γt (RORγt) and enhanced RORγt-induced Th17 cell generation. These results demonstrate that Smad2 positively regulates the generation of inflammatory Th17 cells.


Assuntos
Interleucina-17/metabolismo , Proteína Smad2/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Encefalomielite/genética , Encefalomielite/imunologia , Encefalomielite/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Ligação Proteica/genética , Ligação Proteica/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad2/genética , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA