Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(28): e2322577121, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38968104

RESUMO

Multiple sclerosis (MS) is a demyelinating central nervous system (CNS) disorder that is associated with functional impairment and accruing disability. There are multiple U.S. Food and Drug Administration (FDA)-approved drugs that effectively dampen inflammation and slow disability progression. However, these agents do not work well for all patients and are associated with side effects that may limit their use. The vagus nerve (VN) provides a direct communication conduit between the CNS and the periphery, and modulation of the inflammatory reflex via electrical stimulation of the VN (VNS) shows efficacy in ameliorating pathology in several CNS and autoimmune disorders. We therefore investigated the impact of VNS in a rat experimental autoimmune encephalomyelitis (EAE) model of MS. In this study, VNS-mediated neuroimmune modulation is demonstrated to effectively decrease EAE disease severity and duration, infiltration of neutrophils and pathogenic lymphocytes, myelin damage, blood-brain barrier disruption, fibrinogen deposition, and proinflammatory microglial activation. VNS modulates expression of genes that are implicated in MS pathogenesis, as well as those encoding myelin proteins and transcription factors regulating new myelin synthesis. Together, these data indicate that neuroimmune modulation via VNS may be a promising approach to treat MS, that not only ameliorates symptoms but potentially also promotes myelin repair (remyelination).


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Estimulação do Nervo Vago , Nervo Vago , Animais , Encefalomielite Autoimune Experimental/terapia , Encefalomielite Autoimune Experimental/imunologia , Ratos , Esclerose Múltipla/terapia , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Estimulação do Nervo Vago/métodos , Inflamação/terapia , Inflamação/patologia , Modelos Animais de Doenças , Feminino , Bainha de Mielina/metabolismo , Barreira Hematoencefálica
2.
Proc Natl Acad Sci U S A ; 117(35): 21527-21535, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817520

RESUMO

Given the known neuroreparative actions of IL-33 in experimental models of central nervous system (CNS) injury, we predicted that compounds which induce IL-33 are likely to promote remyelination. We found anacardic acid as a candidate molecule to serve as a therapeutic agent to promote remyelination. Addition of anacardic acid to cultured oligodendrocyte precursor cells (OPCs) rapidly increased expression of myelin genes and myelin proteins, suggesting a direct induction of genes involved in myelination by anacardic acid. Also, when added to OPCs, anacardic acid resulted in the induction of IL-33. In vivo, treatment of with anacardic acid in doses which ranged from 0.025 mg/kg to 2.5 mg/kg, improved pathologic scores in experimental allergic encephalitis (EAE) and in the cuprizone model of demyelination/remyelination. Electron microscopic studies performed in mice fed with cuprizone and treated with anacardic acid showed lower g-ratio scores when compared to controls, suggesting increased remyelination of axons. In EAE, improvement in paralytic scores was seen when the drug was given prior to or following the onset of paralytic signs. In EAE and in the cuprizone model, areas of myelin loss, which are likely to remyelinate, was associated with a greater recruitment of IL-33-expressing OPCs in mice which received anacardic acid when compared to controls.


Assuntos
Ácidos Anacárdicos/farmacologia , Interleucina-33/biossíntese , Remielinização/efeitos dos fármacos , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Feminino , Interleucina-33/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Básica da Mielina/metabolismo , Proteínas da Mielina/metabolismo , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Remielinização/fisiologia , Células-Tronco/metabolismo
3.
Mol Pharmacol ; 83(1): 206-16, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23087260

RESUMO

Unlike the majority of G protein-coupled receptors, the prostaglandin E(2) (PGE(2)) E-prostanoid 3 (EP3) receptor binds agonist with high affinity that is insensitive to the presence of guanosine 5[prime]-O-(3-thio)triphosphate (GTPγS). We report the identification of mutations that confer GTPγS sensitivity to agonist binding. Seven point mutations were introduced into the conserved motif in the second extracellular loop (ECII) of EP3, resulting in acquisition of GTP-sensitive agonist binding. One receptor mutation W203A was studied in detail. Loss of agonist binding was observed on intact human embryonic kidney 293 cells expressing the W203A receptor, conditions where high GTP levels are present; however, high affinity binding [(3)H]PGE(2) was observed in broken cell preparations washed free of GTP. The [(3)H]PGE(2) binding of W203A in broken cell membrane fractions was inhibited by addition of GTPγS (IC(50) 21 ± 1.8 nM). Taken together, these results suggest that the wild-type EP3 receptor displays unusual characteristics of the complex coupled equilibria between agonist-receptor and receptor-G protein interaction. Moreover, mutation of ECII can alter this coupled equilibrium from GTP-insensitive agonist binding to more conventional GTP-sensitive binding. This suggests that for the mutant receptors, ECII plays a critical role in linking the agonist bound receptor conformation to the G protein nucleotide bound state.


Assuntos
Dinoprostona/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Receptores de Prostaglandina E Subtipo EP3/agonistas , Animais , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Mutação Puntual , Conformação Proteica , Coelhos , Ensaio Radioligante , Receptores de Prostaglandina E Subtipo EP3/genética , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Transdução de Sinais
4.
PLoS One ; 13(12): e0208755, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30562364

RESUMO

OBJECTIVE: We tested the hypothesis that the expression of IL-33 in MS is dynamic and is likely to reflect the clinical and radiological changes during the course of RRMS. METHODS: MS with either clinical or radiological relapses were recruited for the study and followed for one year. IL-33 and a panel of genes was measured by q PCR and flow cytometry at different time points. RESULTS: Among 22 RRMS patients, 4 patients showed highest levels of IL-33 at the time they were recruited to the study (Month 0); in 14 patients highest levels of IL-33 were seen between 6-11 months after relapse and in 4 patients maximal levels of IL-33 were seen 12 months after relapse. A similar pattern of IL-33 kinetics was seen when IL-33 was measured by flow cytometry in an additional cohort of 12 patients. The timing of the improvement clinically did not correlate with IL-33 expression with highest expression levels either preceding or following clinical recovery. From our whole genome RNA-sequencing data we found a strong correlation between expression levels of IL-33 and a ~2000 mRNA genes. However, none of these genes encoded proteins involved in either innate or adaptive immunity. Rather, many of the genes that correlated highly with IL-33 encoded to proteins involved in DNA repair or mitochondrial function and mRNA splicing pathways. INTERPRETATION: Given the neuro-reparative and remodeling functions attributed to IL-33, it is likely that some of the novel genes we have uncovered may be involved in repair and recovery of the CNS in MS.


Assuntos
Interleucina-33/sangue , Esclerose Múltipla Recidivante-Remitente/sangue , Adulto , Biomarcadores/sangue , Estudos Transversais , Progressão da Doença , Feminino , Expressão Gênica , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/diagnóstico por imagem , Esclerose Múltipla Recidivante-Remitente/terapia , RNA Mensageiro/sangue , Adulto Jovem
5.
PLoS One ; 11(3): e0152163, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022724

RESUMO

BACKGROUND: Impaired remyelination of demyelinated axons is a major cause of neurological disability. In inflammatory demyelinating disease of the central nervous system (CNS), although remyelination does happen, it is often incomplete, resulting in poor clinical recovery. Poly-IC a known TLR3 agonist and IL-33, a cytokine which is induced by poly-IC are known to influence recovery and promote repair in experimental models of CNS demyelination. METHODOLOGY AND PRINCIPAL FINDINGS: We examined the effect of addition of poly-IC and IL-33 on the differentiation and maturation of oligodendrocyte precursor cells (OPC) cultured in vitro. Both Poly-IC and IL-33 induced transcription of myelin genes and the differentiation of OPC to mature myelin forming cells. Poly-IC induced IL-33 in OPC and addition of IL-33 to in vitro cultures, amplified further, IL-33 expression suggesting an autocrine regulation of IL-33. Poly-IC and IL-33 also induced phosphorylation of p38MAPK, a signaling molecule involved in myelination. Following the induction of gliotoxic injury with lysolecithin to the corpus callosum (CC), treatment of animals with poly-IC resulted in greater recruitment of OPC and increased staining for myelin in areas of demyelination. Also, poly-IC treated animals showed greater expression of IL-33 and higher expression of M2 phenotype macrophages in the CC. CONCLUSION/SIGNIFICANCE: Our studies suggest that poly-IC and IL-33 play a role in myelin repair by enhancing expression of myelin genes and are therefore attractive therapeutic agents for use as remyelinating agents in human demyelinating disease.


Assuntos
Interleucina-33/metabolismo , Bainha de Mielina/metabolismo , Poli I-C/farmacologia , Receptor 3 Toll-Like/agonistas , Cicatrização/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/metabolismo , Ativação Enzimática/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Lisofosfatidilcolinas/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Modelos Biológicos , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fenótipo , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Receptor 3 Toll-Like/metabolismo , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Neuroimmunol ; 265(1-2): 51-60, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24169446

RESUMO

We examined the activation of innate immune pathway mediated by nucleotide-binding oligomerization domain-containing protein 2 (NOD2) in oligodendrocyte precursor cells (OPCs). We show that activation of NOD2 by ligand peptidoglycan (PGN) leads to the recruitment and phosphorylation of receptor-interacting serine/threonine kinase 2 (RIPK2). Phosphorylation of RIPK2 is followed by phosphorylation of neuronal nitric oxide synthase (nNOS), increase in NOS activity and subsequent accumulation of nitric oxide (NO) mediated N-tyrosinylated compounds in OPCs. The reversal of NOS activity by the nNOS inhibitor 7-nitroindazole (7-NI), but not by the iNOS inhibitor L-canavanine, supported the conclusion that the increased NOS activity was due to the selective activation of nNOS in OPCs. In addition, NO mediated injury to OPC was reflected in reduction in activity of respiratory enzymes such as complex I and IV, decrease in mitochondrial membrane potential and release of cytochrome-C from mitochondria. Furthermore, intracerebral injection of PGN into corpus callosum (CC) of rats led to the development of demyelination, which appeared as early as by day 3 post-injection, and involved the trunk of the CC by day 14. Accumulation of N-tyrosinylated proteins was seen in oligodendrocytes in regions of the CC which were in close proximity to the injection site. Taken together, these results suggest that PGN induced formation of NO, mitochondrial dysfunction and accumulation of N-tyrosinylated proteins in oligodendrocytes are likely mediators of central nervous system demyelination.


Assuntos
Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/terapia , Proteína Adaptadora de Sinalização NOD2/metabolismo , Oligodendroglia/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/metabolismo , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Indazóis/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/ultraestrutura , Peptidoglicano/toxicidade , Ratos , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Transdução de Sinais/efeitos dos fármacos , Células-Tronco , Fatores de Tempo
7.
Mol Neurobiol ; 47(2): 673-98, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23224996

RESUMO

Peripheral myelin protein-22 (PMP22) is primarily expressed in the compact myelin of the peripheral nervous system. Levels of PMP22 have to be tightly regulated since alterations of PMP22 levels by mutations of the PMP22 gene are responsible for >50 % of all patients with inherited peripheral neuropathies, including Charcot-Marie-Tooth type-1A (CMT1A) with trisomy of PMP22, hereditary neuropathy with liability to pressure palsies (HNPP) with heterozygous deletion of PMP22, and CMT1E with point mutations of PMP22. While overexpression and point-mutations of the PMP22 gene may produce gain-of-function phenotypes, deletion of PMP22 results in a loss-of-function phenotype that reveals the normal physiological functions of the PMP22 protein. In this article, we will review the basic genetics, biochemistry and molecular structure of PMP22, followed by discussion of the current understanding of pathogenic mechanisms involving in the inherited neuropathies with mutations in PMP22 gene.


Assuntos
Mutação/genética , Proteínas da Mielina/genética , Doenças do Sistema Nervoso Periférico/genética , Sequência de Aminoácidos , Animais , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Humanos , Dados de Sequência Molecular , Proteínas da Mielina/fisiologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia
8.
PLoS One ; 7(5): e33518, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22590492

RESUMO

BACKGROUND: Prostaglandin I(2) (PGI(2)), a lipid mediator currently used in treatment of human disease, is a critical regulator of adaptive immune responses. Although PGI(2) signaling suppressed Th1 and Th2 immune responses, the role of PGI(2) in Th17 differentiation is not known. METHODOLOGY/PRINCIPAL FINDINGS: In mouse CD4(+)CD62L(+) naïve T cell culture, the PGI(2) analogs iloprost and cicaprost increased IL-17A and IL-22 protein production and Th17 differentiation in vitro. This effect was augmented by IL-23 and was dependent on PGI(2) receptor IP signaling. In mouse bone marrow-derived CD11c(+) dendritic cells (BMDCs), PGI(2) analogs increased the ratio of IL-23/IL-12, which is correlated with increased ability of BMDCs to stimulate naïve T cells for IL-17A production. Moreover, IP knockout mice had delayed onset of a Th17-associated neurological disease, experimental autoimmune encephalomyelitis (EAE), and reduced infiltration of IL-17A-expressing mononuclear cells in the spinal cords compared to wild type mice. These results suggest that PGI(2) promotes in vivo Th17 responses. CONCLUSION: The preferential stimulation of Th17 differentiation by IP signaling may have important clinical implications as PGI(2) and its analogs are commonly used to treat human pulmonary hypertension.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Epoprostenol/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Epoprostenol/imunologia , Células Th17/imunologia , Animais , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Diferenciação Celular/imunologia , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Epoprostenol/análogos & derivados , Epoprostenol/genética , Epoprostenol/imunologia , Feminino , Humanos , Iloprosta/imunologia , Iloprosta/farmacologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Inibidores da Agregação Plaquetária/imunologia , Receptores de Epoprostenol/genética , Medula Espinal/imunologia , Medula Espinal/patologia , Células Th17/patologia
9.
J Biol Chem ; 281(6): 3321-8, 2006 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-16338931

RESUMO

COX-2-dependent prostaglandin (PG) E2 synthesis regulates macrophage MMP expression, which is thought to destabilize atherosclerotic plaques. However, the administration of selective COX-2 inhibitors paradoxically increases the frequency of adverse cardiovascular events potentially through the loss of anti-inflammatory prostanoids and/or disturbance in the balance of pro- and anti-thrombotic prostanoids. To avoid these collateral effects of COX-2 inhibition, a strategy to identify and block specific prostanoid-receptor interactions may be required. We previously reported that macrophage engagement of vascular extracellular matrix (ECM) triggers proteinase expression through a MAPKerk1/2-dependent increase in COX-2 expression and PGE2 synthesis. Here we demonstrate that elicited macrophages express the PGE2 receptors EP1-4. When plated on ECM, their expression of EP2 and EP4, receptors linked to PGE2-induced activation of adenylyl cyclase, is strongly stimulated. Forskolin and dibutryl cyclic-AMP stimulate macrophage matrix metalloproteinase (MMP)-9 expression in a dose-dependent manner. However, an EP2 agonist (butaprost) has no effect on MMP-9 expression, and macrophages from EP2 null mice exhibited enhanced COX-2 and MMP-9 expression when plated on ECM. In contrast, the EP4 agonist (PGE1-OH) stimulated macrophage MMP-9 expression, which was inhibited by the EP4 antagonist ONO-AE3-208. When compared with COX-2 silencing by small interfering RNA or inhibition by celecoxib, the EP4 antagonist was as effective in inhibiting ECM-induced proteinase expression. In addition, ECM-induced MMP-9 expression was blocked in macrophages in which EP4 was silenced by small interfering RNA. Thus, COX-2-dependent ECM-induced proteinase expression is effectively blocked by selective inhibition of EP4, a member of the PGE2 family of receptors.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Matriz Extracelular/enzimologia , Macrófagos/enzimologia , Metaloproteinase 9 da Matriz/biossíntese , Receptores de Prostaglandina E/metabolismo , Animais , Western Blotting , Bucladesina/metabolismo , Celecoxib , Linhagem Celular , Colforsina/farmacologia , Matriz Extracelular/metabolismo , Inativação Gênica , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Naftalenos/farmacologia , Oligonucleotídeos/química , Peritônio/metabolismo , Fenilbutiratos/farmacologia , Fosforilação , Pirazóis/farmacologia , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia , Fatores de Tempo , Transfecção
10.
J Biol Chem ; 280(41): 35028-37, 2005 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-16096282

RESUMO

PGF(2alpha) is the most abundant prostaglandin detected in urine; however, its renal effects are poorly characterized. The present study cloned a PGF-prostanoid receptor (FP) from the rabbit kidney and determined the functional consequences of its activation. Nuclease protection assay showed that FP mRNA expression predominates in rabbit ovary and kidney. In situ hybridization revealed that renal FP expression predominates in the cortical collecting duct (CCD). Although FP receptor activation failed to increase intracellular Ca(2+), it potently inhibited vasopressin-stimulated osmotic water permeability (L(p), 10(-7) cm/(atm.s)) in in vitro microperfused rabbit CCDs. Inhibition of L(p) by the FP selective agonist latanoprost was additive to inhibition of vasopressin action by the EP selective agonist sulprostone. Inhibition of L(p) by latanoprost was completely blocked by pertussis toxin, consistent with a G(i)-coupled mechanism. Heterologous transfection of the rabbit FPr into HEK293 cells also showed that latanoprost inhibited cAMP generation via a pertussis toxin-sensitive mechanism but did not increase cell Ca(2+). These studies demonstrate a functional FP receptor on the basolateral membrane of rabbit CCDs. In contrast to the Ca(2+) signal transduced by other FP receptors, this renal FP receptor signals via a PT-sensitive mechanism that is not coupled to cell Ca(2+).


Assuntos
Dinoprosta/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Água/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Cálcio/metabolismo , Linhagem Celular , Clonagem Molecular , DNA Complementar/metabolismo , Dinoprosta/química , Feminino , Vetores Genéticos , Humanos , Hibridização In Situ , Óperon Lac , Latanoprosta , Ligantes , Dados de Sequência Molecular , Ovário/metabolismo , Perfusão , Toxina Pertussis/farmacologia , Prostaglandinas/metabolismo , Prostaglandinas F Sintéticas/farmacologia , Ligação Proteica , RNA Mensageiro/metabolismo , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleases/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Distribuição Tecidual , Transfecção
11.
J Immunol ; 168(12): 6506-13, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12055272

RESUMO

Experimental allergic encephalomyelitis (EAE) is a CD4(+) Th1 cell-mediated inflammatory demyelinating autoimmune disease of the CNS that serves as an animal model for multiple sclerosis (MS). IL-12 is a proinflammatory cytokine that plays a crucial role in the induction of neural Ag-specific Th1 differentiation and pathogenesis of CNS demyelination in EAE and MS. Curcumin (1,7-Bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) is a naturally occurring polyphenolic phytochemical isolated from the rhizome of the medicinal plant Curcuma longa. It has profound anti-inflammatory activity and been traditionally used to treat inflammatory disorders. In this study we have examined the effect and mechanism of action of curcumin on the pathogenesis of CNS demyelination in EAE. In vivo treatment of SJL/J mice with curcumin significantly reduced the duration and clinical severity of active immunization and adoptive transfer EAE. Curcumin inhibited EAE in association with a decrease in IL-12 production from macrophage/microglial cells and differentiation of neural Ag-specific Th1 cells. In vitro treatment of activated T cells with curcumin inhibited IL-12-induced tyrosine phosphorylation of Janus kinase 2, tyrosine kinase 2, and STAT3 and STAT4 transcription factors. The inhibition of Janus kinase-STAT pathway by curcumin resulted in a decrease in IL-12-induced T cell proliferation and Th1 differentiation. These findings highlight the fact that curcumin inhibits EAE by blocking IL-12 signaling in T cells and suggest its use in the treatment of MS and other Th1 cell-mediated inflammatory diseases.


Assuntos
Curcumina/uso terapêutico , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Interleucina-12/antagonistas & inibidores , Interleucina-12/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/imunologia , Linfócitos T/enzimologia , Transferência Adotiva , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/uso terapêutico , Células Cultivadas , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Curcumina/administração & dosagem , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Doenças Desmielinizantes/enzimologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/prevenção & controle , Encefalomielite Autoimune Experimental/enzimologia , Encefalomielite Autoimune Experimental/patologia , Epitopos de Linfócito T/imunologia , Feminino , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Injeções Intraperitoneais , Interleucina-12/biossíntese , Janus Quinase 1 , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , Proteína Básica da Mielina/administração & dosagem , Proteína Básica da Mielina/imunologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/fisiologia , Fator de Transcrição STAT3 , Fator de Transcrição STAT4 , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Tirosina/metabolismo , Vacinação
12.
Glia ; 45(2): 188-96, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14730712

RESUMO

Microglia, the resident macrophage of the brain, mediates immune and inflammatory responses in the central nervous system (CNS). Activation of microglia and secretion of inflammatory cytokines associate with the pathogenesis of CNS diseases, including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease, prion disease, and AIDS dementia. Microbial pathogens, cytokines, chemokines, and costimulatory molecules are potent inducers of microglial activation in the CNS. Signaling through its receptor, IL-3 induces the activation of JAK-STAT and MAP kinase pathways in microglial cells. In this study, we found that in vitro treatment of EOC-20 microglial cells with tyrphostin AG490 blocked IL-3-induced tyrosine phosphorylation of JAK2, STAT5A, and STAT5B signaling proteins. Stable transfection of EOC-20 cells with a dominant negative JAK2 mutant also blocked IL-3-induced tyrosine phosphorylation of JAK2, STAT5A, and STAT5B in microglia. The blockade of JAK2-STAT5 pathway resulted in a decrease in IL-3-induced proliferation and expression of CD40 and major histocompatibility complex class II molecules in microglia. These findings highlight the fact that JAK2-STAT5 signaling pathway plays a critical role in mediating IL-3-induced activation of microglia.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Encefalite/enzimologia , Interleucina-3/metabolismo , Microglia/enzimologia , Proteínas do Leite , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Animais , Antígenos CD40/efeitos dos fármacos , Antígenos CD40/imunologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Linhagem Celular , Proteínas de Ligação a DNA/imunologia , Encefalite/imunologia , Inibidores Enzimáticos/farmacologia , Gliose/enzimologia , Gliose/imunologia , Antígenos de Histocompatibilidade Classe II/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Interleucina-3/antagonistas & inibidores , Interleucina-3/imunologia , Janus Quinase 2 , Camundongos , Microglia/efeitos dos fármacos , Microglia/imunologia , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/imunologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/imunologia , Fator de Transcrição STAT5 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Transativadores/imunologia , Transfecção , Proteínas Supressoras de Tumor , Tirfostinas/farmacologia
13.
J Immunol ; 171(11): 5743-50, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14634082

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a nuclear receptor transcription factor that regulates cell growth, differentiation, and homeostasis. PPARgamma agonists are potent therapeutic agents for type 2 diabetes, obesity, and inflammation. Experimental allergic encephalomyelitis (EAE) is a Th1 cell-mediated inflammatory demyelinating autoimmune disease model of multiple sclerosis. We have shown recently that PPARgamma agonists inhibit EAE by blocking IL-12 production, IL-12 signaling, and neural Ag-induced Th1 differentiation. In this study, we show that the PPARgamma-deficient heterozygous mice develop an exacerbated EAE with prolonged clinical symptoms than the wild-type littermates, following immunization with myelin oligodendrocyte glycoprotein (MOG) p35-55 peptide. The exacerbation of EAE in PPARgamma(+/-) mice associates with an increased expansion of CD4(+) and CD8(+) T cells and expression of CD40 and MHC class II molecules in response to MOGp35-55 Ag. The PPARgamma(+/-) mice also showed an increase in T cell proliferation and Th1 response to MOGp35-55 Ag than the wild-type littermates. These findings suggest that PPARgamma be a critical physiological regulator of CNS inflammation and demyelination in EAE and perhaps multiple sclerosis and other Th1 cell-mediated autoimmune diseases.


Assuntos
Encefalomielite Autoimune Experimental/genética , Triagem de Portadores Genéticos , Glicoproteínas/imunologia , Proteínas do Tecido Nervoso/imunologia , Fragmentos de Peptídeos/imunologia , Peroxissomos/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Células Th1/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Antígenos CD40/biossíntese , Divisão Celular/genética , Divisão Celular/imunologia , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Glicoproteínas/administração & dosagem , Antígenos de Histocompatibilidade Classe II/biossíntese , Injeções Intramusculares , Interferon gama/biossíntese , Interleucina-12/biossíntese , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito , Proteínas do Tecido Nervoso/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Receptores Citoplasmáticos e Nucleares/fisiologia , Índice de Gravidade de Doença , Baço/citologia , Baço/imunologia , Baço/metabolismo , Células Th1/metabolismo , Células Th1/patologia , Fatores de Transcrição/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA