Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 134(11): e150-e175, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38781298

RESUMO

HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.


Assuntos
Infecções por HIV , Hipertensão , Humanos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , Infecções por HIV/complicações , Hipertensão/tratamento farmacológico , Hipertensão/epidemiologia , Fatores de Risco , HIV-1/patogenicidade , Animais
2.
Int J Mol Sci ; 22(9)2021 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-34065088

RESUMO

Loss of function KCNK3 mutation is one of the gene variants driving hereditary pulmonary arterial hypertension (PAH). KCNK3 is expressed in several cell and tissue types on both membrane and endoplasmic reticulum and potentially plays a role in multiple pathological process associated with PAH. However, the role of various stressors driving the susceptibility of KCNK3 mutation to PAH is unknown. Hence, we exposed kcnk3fl/fl animals to hypoxia, metabolic diet and low dose lipopolysaccharide (LPS) and performed molecular characterization of their tissue. We also used tissue samples from KCNK3 patients (skin fibroblast derived inducible pluripotent stem cells, blood, lungs, peripheral blood mononuclear cells) and performed microarray, immunohistochemistry (IHC) and mass cytometry time of flight (CyTOF) experiments. Although a hypoxic insult did not alter vascular tone in kcnk3fl/fl mice, RNASeq study of these lungs implied that inflammatory and metabolic factors were altered, and the follow-up diet study demonstrated a dysregulation of bone marrow cells in kcnk3fl/fl mice. Finally, a low dose LPS study clearly showed that inflammation could be a possible second hit driving PAH in kcnk3fl/fl mice. Multiplex, IHC and CyTOF immunophenotyping studies on human samples confirmed the mouse data and strongly indicated that cell mediated, and innate immune responses may drive PAH susceptibility in these patients. In conclusion, loss of function KCNK3 mutation alters various physiological processes from vascular tone to metabolic diet through inflammation. Our data suggests that altered circulating immune cells may drive PAH susceptibility in patients with KCNK3 mutation.


Assuntos
Imunomodulação/genética , Mutação , Proteínas do Tecido Nervoso/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/imunologia , Animais , Biomarcadores , Estudos de Casos e Controles , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Monócitos/imunologia , Monócitos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Hipertensão Arterial Pulmonar/complicações , Hipertensão Arterial Pulmonar/fisiopatologia , Transcriptoma
3.
Life Sci ; 110(1): 1-7, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-24880075

RESUMO

AIMS: The molecular mechanisms for the loss of 3,4-dihydroxyphenylalanine (l-dopa) efficacy during the treatment of Parkinson's disease (PD) are unknown. Modifications related to catecholamine metabolism such as changes in l-dopa and dopamine (DA) metabolism, the modulation of catecholamine enzymes and the production of interfering metabolites are the primary concerns of this study. MAIN METHODS: Normal (saline) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) pre-treated mice were primed with 100mg/kg of l-dopa twice a day for 14 days, and a matching group remained l-dopa naïve. l-dopa naive and primed mice received a challenge dose of 100mg/kg of l-dopa and were sacrificed 30 min later. Striatal catecholamine levels and the expression and activity of catechol-O-methyltransferase (COMT) were determined. KEY FINDINGS: Normal and MPTP pre-treated animals metabolize l-dopa and DA similarly during l-dopa therapy. Administration of a challenge dose of l-dopa increased l-dopa and DA metabolism in l-dopa naïve animals, and this effect was enhanced in l-dopa primed mice. The levels of 3-OMD in MPTP pre-treated animals were almost identical to those in normal mice, which we found are likely due to increased COMT activity in MPTP pre-treated mice. SIGNIFICANCE: The results of this comparative study provide evidence that sub-chronic administration of l-dopa decreases the ability of the striatum to accumulate l-dopa and DA, due to increased metabolism via methylation and oxidation. This data supports evidence for the metabolic adaptation of the catecholamine pathway during long-term treatment with l-dopa, which may explain the causes for the loss of l-dopa efficacy.


Assuntos
Antiparkinsonianos/farmacologia , Catecol O-Metiltransferase/metabolismo , Dopamina/metabolismo , Levodopa/farmacologia , Transtornos Parkinsonianos/tratamento farmacológico , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Animais , Antiparkinsonianos/administração & dosagem , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Esquema de Medicação , Levodopa/administração & dosagem , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Transtornos Parkinsonianos/fisiopatologia , Fatores de Tempo
4.
Life Sci ; 91(19-20): 921-7, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23000099

RESUMO

AIMS: In this study, the effects of prenatal exposure to methanol (MeOH) on the nigrostriatal dopamine (NSDA) system were examined to determine if the interaction could sensitize this system, and serve as an underpinning for Parkinson's disease (PD) like changes that occur later in life. Methanol was studied because its toxicity resembles the symptoms of PD and the symptoms are relieved by L-dopa meaning that MeOH targets the NSDA system. Since fermentation and wood combustion are major sources for MeOH, the incidence of human encounters with MeOH is high. As a superior solvent and the precursor for formaldehyde, MeOH has a powerful and sometimes, irreversible impact on chemical processes, such as cross-linking proteins and nucleic acids. It may cause subthreshold changes that sensitizes the NSDA system to PD, that occur during aging. MAIN METHODS: To study the prenatal effects of MeOH, pregnant C57BL/6J mice were administered 40 mg/kg MeOH by oral gavage during gestation days 8-12, twice daily. Twelve weeks after birth, behavior impairments were recorded. The striatum was dissected for the determination of tyrosine hydroxylase (TH), L-aromatic amino acid decarboxylase (LAAD), α-synuclein and levels of dopamine (DA) and its metabolites. KEY FINDINGS: MeOH reduced striatal TH and LAAD protein by 47% and 57% respectively and DA by 32%. SIGNIFICANCE: The results mean that in utero exposure to toxins similar to MeOH could sensitize the striatal system to changes that cause PD. This study may help identify strategies to block this type of in utero toxicity.


Assuntos
Dopamina/metabolismo , Metanol/toxicidade , Doença de Parkinson/etiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Metanol/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/fisiopatologia , Gravidez , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/metabolismo
5.
Life Sci ; 89(17-18): 638-43, 2011 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-21871902

RESUMO

AIMS: l-3,4-Dihydroxyphenylalanine (L-DOPA) is the most effective symptomatic treatment for Parkinson's disease (PD), but PD patients usually experience a successful response to L-DOPA therapy followed by a progressive loss of response. L-DOPA efficacy relies on its decarboxylation by aromatic l-amino acid decarboxylase (AAAD) to form dopamine (DA). So exogenous L-DOPA drives the reaction and AAAD becomes the rate limiting enzyme in the supply of DA. In turn, exogenous L-DOPA regulates the expression and activity of AAAD as well as the synthesis of DA and its metabolites, changes that may be linked to the efficacy and side-effects of L-DOPA. MAIN METHODS: One-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse PD model was utilized to study the effects of L-DOPA on the steady-state level and activity of AAAD, tyrosine hydroxylase (TH), DA and the metabolites of DA. The MPTP and control mice were treated twice daily with PBS or with 100mg/kg of L-DOPA for 14days and the expression and activity of AAAD, the expression of TH and the levels of DA and its metabolites were determined 24h after L-DOPA or PBS treatment, when exogenous L-DOPA is eliminated. KEY FINDINGS: In the MPTP model, L-DOPA reduced the steady-state expression and the activity of striatal AAAD by 52% and 50%, respectively, DA and metabolites were also significantly decreased. SIGNIFICANCE: The outcome shows that while L-DOPA replenishes striatal DA it also down-regulates AAAD and the steady-state synthesis and metabolic capability of the dopaminergic system. These findings are important in the precipitation of L-DOPA induced side effects and the management of L-DOPA therapy.


Assuntos
Antiparkinsonianos/farmacologia , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Levodopa/farmacologia , Doença de Parkinson/tratamento farmacológico , Tirosina 3-Mono-Oxigenase/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/análogos & derivados , Animais , Antiparkinsonianos/efeitos adversos , Descarboxilases de Aminoácido-L-Aromático/genética , Corpo Estriado/enzimologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Levodopa/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/enzimologia , Doença de Parkinson/metabolismo , Doença de Parkinson Secundária/induzido quimicamente
6.
J Neuroimmunol ; 178(1-2): 76-86, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16844232

RESUMO

Peroxisome proliferator-activated receptor-gamma is a nuclear receptor transcription factor that regulates cell growth, differentiation and homeostasis. PPARgamma agonists have been used to treat obesity, diabetes, cancer and inflammation and recent studies have shown the protective effects of PPARgamma agonists on experimental allergic encephalomyelitis (EAE), a Th1 cell-mediated autoimmune disease model of multiple sclerosis (MS). Our studies have further demonstrated that the PPARgamma agonists, 15d-PGJ2 and Ciglitazone, inhibit EAE through blocking IL-12 signaling leading to Th1 differentiation and the PPARgamma deficient heterozygous mice (PPARgamma+/-) or those treated with PPARgamma antagonists develop an exacerbated EAE in association with an augmented Th1 response. In this study, we show that the PPARgamma antagonists, Bisphenol A diglycidyl ether (BADGE) and 2-chloro-5-nitro-N-(4-pyridyl)benzamide (T0070907), reverse the inhibition of EAE by the PPARgamma agonists, Ciglitazone and 15-Deoxy-Delta(12,14)-Prostaglandin J2, in C57BL/6 wild-type and PPARgamma+/- mice. The reversal of EAE by BADGE and T0070907 was associated with restoration of neural antigen-induced T cell proliferation, IFNgamma production and Th1 differentiation inhibited by Ciglitazone and 15d-PGJ2. These results suggest that Ciglitazone and 15d-PGJ2 ameliorate EAE through PPARgamma-dependent mechanisms and further confirm a physiological role for PPARgamma in the regulation of CNS inflammation and demyelination in EAE.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Glicoproteína Associada a Mielina/imunologia , PPAR gama/antagonistas & inibidores , Prostaglandina D2/análogos & derivados , Células Th1/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Animais , Antígenos/imunologia , Benzamidas/farmacologia , Compostos Benzidrílicos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Compostos de Epóxi/farmacologia , Feminino , Fatores Imunológicos/farmacologia , Inflamação/imunologia , Interferon gama/biossíntese , Interferon gama/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas da Mielina , Glicoproteína Mielina-Oligodendrócito , PPAR gama/agonistas , PPAR gama/metabolismo , Prostaglandina D2/farmacologia , Piridinas/farmacologia , Células Th1/imunologia
7.
Exp Parasitol ; 114(3): 193-203, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16647056

RESUMO

Filariasis is a debilitating parasitic disease in many tropical countries. Despite the highly evolved immune system, the filarial parasites successfully evade host immunity to persist for a sustained period of time. Earlier studies have shown that the filarial parasites achieve this long-term survival through release of immunosuppressive materials in the host. In this study, we show that the secreted filarial lipids (SFL) isolated from Setaria digitata suppress Th1 immune response. While immunization with myelin antigen induces Th1 response in mice, in vitro treatment with SFL resulted in a dose-dependent decrease in myelin antigen-induced proliferation and secretion of IL-12 and IFNgamma. The SFL also inhibited IL-12-induced T cell proliferation and Th1 differentiation in vitro. The inhibition of T cell responses by SFL associates with the blockade of IL-12-induced activation of JAK-STAT signaling pathway in T cells. These findings suggest that the SFL modulates Th1 immune response by blocking IL-12 signaling in T cells and thus play a role in host immune evasion of filarial parasites.


Assuntos
Interleucina-12/imunologia , Lipídeos/imunologia , Setaria (Nematoide)/imunologia , Setaríase/imunologia , Células Th1/imunologia , Animais , Western Blotting , Bovinos , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Imunoprecipitação , Interferon gama/análise , Interleucina-12/análise , Interleucina-12/fisiologia , Janus Quinases/metabolismo , Metabolismo dos Lipídeos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Básica da Mielina/imunologia , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Fatores de Transcrição STAT/fisiologia , Setaria (Nematoide)/metabolismo , Baço/citologia , Baço/imunologia
8.
J Neurosci Res ; 83(7): 1299-309, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16547967

RESUMO

Experimental allergic encephalomyelitis (EAE) is a Th1 cell-mediated autoimmune disease model of multiple sclerosis (MS). Vitamin D deficiency is commonly observed in MS patients and vitamin D supplements reduce the clinical symptoms of EAE and MS. Earlier studies have shown that in vivo treatment with vitamin D analogs ameliorates EAE in association with the inhibition of IL-12 production and Th1 differentiation. The mechanisms in the regulation of Th1 response by vitamin D in EAE/MS are, however, not known. We show that in vivo treatment of C57BL/6 and SJL/J mice (i.p.) with 100 ng of 1,25 dihydroxyvitamin D3, on every other day from Day 0-30, ameliorates EAE in association with the inhibition of IL-12 production and neural antigen-specific Th1 response. In vitro treatment with 1,25(OH)2D3 inhibited IFNgamma-induced tyrosine phosphorylation of STAT1, without affecting JAK2, in EOC-20 microglial cells. Treatment of activated T cells with 1,25(OH)2D3 also inhibited the IL-12-induced tyrosine phosphorylation of JAK2, TYK2, STAT3, and STAT4 in association with a decrease in T cell proliferation in vitro. These findings highlight the fact that vitamin D modulates JAK-STAT signaling pathway in IL-12/IFNgamma axis leading to Th1 differentiation and further suggest its use in the treatment of MS and other Th1 cell-mediated autoimmune diseases.


Assuntos
Calcitriol/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Imunidade Celular/imunologia , Interleucina-12/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células Th1/metabolismo , Animais , Calcitriol/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Imunidade Celular/efeitos dos fármacos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-12/imunologia , Janus Quinase 2 , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia
9.
J Clin Immunol ; 26(1): 73-85, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16418805

RESUMO

Experimental allergic encephalomyelitis (EAE) is a Th1 cell-mediated autoimmune disease model of multiple sclerosis (MS). IL-12 plays a crucial role in the pathogenesis of EAE/MS and inhibition of IL-12 production or IL-12 signaling was effective in preventing EAE. Cyclooxygenase (COX-2) is a key enzyme promoting inflammation in rheumatoid arthritis and tumor induced angiogenesis. Recent studies have shown that COX-2 inhibitors prevent EAE, however, their mechanism of action is not fully understood. In this study, we show that in vivo treatment (i.p.) with 100 mug COX-2 selective inhibitors (LM01, LM08, LM11, and NS398), on every other day from day 0 to 30, significantly reduced the incidence and severity of EAE in SJL/J and C57BL/6 mice. Further analyses showed that the COX-2 inhibitors reduced neural antigen-induced IL-12 production, T cell proliferation and Th1 differentiation ex vivo and in vitro. The COX-2 inhibitors also decreased IL-12-induced T cell responses through blocking tyrosine phosphorylation of JAK2, TYK2, STAT3, and STAT4 proteins in T cells. These results demonstrate that COX-2 inhibitors ameliorate EAE in association with the modulation of IL-12 signaling through JAK-STAT pathway leading to Th1 differentiation and suggest their use in the treatment of MS and other Th1 cell-mediated autoimmune diseases.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Encefalomielite Autoimune Experimental/metabolismo , Interleucina-12/fisiologia , Células Th1/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Doenças Desmielinizantes/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Inflamação , Interferon gama/metabolismo , Janus Quinase 2 , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla Crônica Progressiva/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais
10.
Biochem Biophys Res Commun ; 340(2): 359-68, 2006 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-16364242

RESUMO

Adult T cell leukemia is an aggressive and frequently fatal malignancy that expressess constitutively activated growth-signaling pathways in association with deregulated growth and resistance to apoptosis. Curcumin (diferuloylmethane) is a naturally occurring yellow pigment, isolated from the rhizomes of the plant Curcuma longa that has traditionally been used in the treatment of injury and inflammation. But the effect and mechanism of action of curcumin on T cell leukemia is not known. To investigate the antitumor activity of curcumin in T cell leukemia, we examined its effect on constitutive phosphorylation of JAK and STAT proteins, proliferation, and apoptosis in HTLV-I-transformed T cell lines. HTLV-I-transformed T cell leukemia lines, MT-2, HuT-102, and SLB-1, express constitutively phosphorylated JAK3, TYK2, STAT3, and STAT5 signaling proteins. In vitro treatment with curcumin induced a dose-dependent decrease in JAK and STAT phosphorylation resulting in the induction of growth-arrest and apoptosis in T cell leukemia. The induction of growth-arrest and apoptosis in association with the blockade of constitutively active JAK-STAT pathway suggests this be a mechanism by which curcumin induces antitumor activity in T cell leukemia.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/farmacologia , Inibidores do Crescimento/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Leucemia de Células T/enzimologia , Leucemia de Células T/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Janus Quinase 3 , Leucemia de Células T/genética , Sistema de Sinalização das MAP Quinases/genética , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/biossíntese , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT5/antagonistas & inibidores , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/genética , TYK2 Quinase
11.
J Neuroimmunol ; 167(1-2): 99-107, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16091293

RESUMO

Peroxisome proliferator-activated receptor-gamma is a nuclear receptor transcription factor that regulates cell growth, differentiation and homeostasis. PPARgamma agonists have been used in the treatment of obesity, diabetes, cancer and inflammation. We and others have shown recently that PPARgamma agonists ameliorate experimental allergic encephalomyelitis (EAE), a Th1 cell-mediated autoimmune disease model of multiple sclerosis (MS). We have further shown that PPARgamma agonists inhibit EAE through blocking IL-12 signaling leading to Th1 differentiation and the PPARgamma-deficient heterozygous mice (PPARgamma(+/-)) develop an exacerbated EAE. In this study, we show that in vivo treatment (i.p.) with 100 mug PPARgamma antagonists, Bisphenol A diglycidyl ether (BADGE) or 2-Chloro-5-nitro-N-(4-pyridyl)benzamide (T0070907), on every other day from day 0 to 30, increased the severity and duration of EAE in C57BL/6 wild-type and PPARgamma(+/-) mice. The exacerbation of EAE by PPARgamma antagonists associates with an augmented neural antigen-induced T cell proliferation, IFNgamma production or Th1 differentiation. These results further suggest that PPARgamma is a critical physiological regulator of CNS inflammation and demyelination in EAE.


Assuntos
Benzamidas/administração & dosagem , Encefalomielite Autoimune Experimental/fisiopatologia , Compostos de Epóxi/administração & dosagem , PPAR gama/antagonistas & inibidores , Piridinas/administração & dosagem , Células Th1/efeitos dos fármacos , Animais , Compostos Benzidrílicos , Proliferação de Células/efeitos dos fármacos , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/patologia , Relação Dose-Resposta Imunológica , Esquema de Medicação , Interações Medicamentosas , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Glicoproteínas/imunologia , Inflamação/etiologia , Inflamação/genética , Inflamação/patologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito , PPAR gama/deficiência , Fragmentos de Peptídeos/imunologia , Timidina/farmacocinética , Fatores de Tempo , Trítio/farmacocinética
12.
J Clin Immunol ; 24(5): 542-52, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15359113

RESUMO

Experimental allergic encephalomyelitis (EAE) is a Th1 cell-mediated inflammatory demyelinating autoimmune disease model of multiple sclerosis (MS). Quercetin (3,3'4',5,7-pentahydroxy flavone) is a flavonoid phytoestrogen that has profound anticancer and anti-inflammatory activities. In this study, we show that in vivo treatment of SJL/J mice with quercetin (i.p. 50 or 100 microg every other day) ameliorates EAE in association with the inhibition of IL-12 production and neural antigen-specific Th1 differentiation. In vitro treatment of activated T cells with quercetin blocks IL-12-induced tyrosine phosphorylation of JAK2, TYK2, STAT3, and STAT4, resulting in a decrease in IL-12-induced T cell proliferation and Th1 differentiation. These findings highlight the fact that quercetin ameliorates EAE by blocking IL-12 signaling and Th1 differentiation and suggest its use in the treatment of MS and other Th1 cell-mediated autoimmune diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Interleucina-12/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Quercetina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Janus Quinase 2 , Macrófagos/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Fator de Transcrição STAT3 , Fator de Transcrição STAT4 , Baço/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Transativadores/metabolismo
13.
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
14.
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