Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci Res ; 98(11): 2317-2332, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32799373

RESUMO

Interaction between autoreactive immune cells and astroglia is an important part of the pathologic processes that fuel neurodegeneration in multiple sclerosis. In this inflammatory disease, immune cells enter into the central nervous system (CNS) and they spread through CNS parenchyma, but the impact of these autoreactive immune cells on the activity pattern of astrocytes has not been defined. By exploiting naïve astrocytes in culture and CNS-infiltrated immune cells (CNS IICs) isolated from rat with experimental autoimmune encephalomyelitis (EAE), here we demonstrate previously unrecognized properties of immune cell-astrocyte interaction. We show that CNS IICs but not the peripheral immune cell application, evokes a rapid and vigorous intracellular Ca2+ increase in astrocytes by promoting glial release of ATP. ATP propagated Ca2+ elevation through glial purinergic P2X7 receptor activation by the hemichannel-dependent nucleotide release mechanism. Astrocyte Ca2+ increase is specifically triggered by the autoreactive CD4+ T-cell application and these two cell types exhibit close spatial interaction in EAE. Therefore, Ca2+ signals may mediate a rapid astroglial response to the autoreactive immune cells in their local environment. This property of immune cell-astrocyte interaction may be important to consider in studies interrogating CNS autoimmune disease.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Imunidade Celular , Receptores Purinérgicos/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Neuroglia/metabolismo , Ratos , Receptores Purinérgicos P2X7/imunologia , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais , Medula Espinal/citologia , Medula Espinal/imunologia
2.
J Immunol ; 194(6): 2493-503, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681336

RESUMO

Dimethyl fumarate (DMF), a new drug for multiple sclerosis (MS) treatment, acts against neuroinflammation via mechanisms that are triggered by adduct formation with thiol redox switches. Ethyl pyruvate (EP), an off-the-shelf agent, appears to be a redox analog of DMF, but its immunomodulatory properties have not been put into the context of MS therapy. In this article, we examined and compared the effects of EP and DMF on MS-relevant activity/functions of T cells, macrophages, microglia, and astrocytes. EP efficiently suppressed the release of MS signature cytokines, IFN-γ and IL-17, from human PBMCs. Furthermore, the production of these cytokines was notably decreased in encephalitogenic T cells after in vivo application of EP to rats. Production of two other proinflammatory cytokines, IL-6 and TNF, and NO was suppressed by EP in macrophages and microglia. Reactive oxygen species production in macrophages, microglia activation, and the development of Ag-presenting phenotype in microglia and macrophages were constrained by EP. The release of IL-6 was reduced in astrocytes. Finally, EP inhibited the activation of transcription factor NF-κB in microglia and astrocytes. Most of these effects were also found for DMF, implying that EP and DMF share common targets and mechanisms of action. Importantly, EP had in vivo impact on experimental autoimmune encephalomyelitis, an animal model of MS. Treatment with EP resulted in delay and shortening of the first relapse, and lower clinical scores, whereas the second attack was annihilated. Further studies on the possibility to use EP as an MS therapeutic are warranted.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Fumaratos/farmacologia , Esclerose Múltipla/tratamento farmacológico , Piruvatos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Fumarato de Dimetilo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/farmacologia , Linfonodos/citologia , Linfonodos/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , NF-kappa B/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo
3.
Nature ; 462(7269): 94-8, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19829296

RESUMO

The tissues of the central nervous system are effectively shielded from the blood circulation by specialized vessels that are impermeable not only to cells, but also to most macromolecules circulating in the blood. Despite this seemingly absolute seclusion, central nervous system tissues are subject to immune surveillance and are vulnerable to autoimmune attacks. Using intravital two-photon imaging in a Lewis rat model of experimental autoimmune encephalomyelitis, here we present in real-time the interactive processes between effector T cells and cerebral structures from their first arrival to manifest autoimmune disease. We observed that incoming effector T cells successively scanned three planes. The T cells got arrested to leptomeningeal vessels and immediately monitored the luminal surface, crawling preferentially against the blood flow. After diapedesis, the cells continued their scan on the abluminal vascular surface and the underlying leptomeningeal (pial) membrane. There, the T cells encountered phagocytes that effectively present antigens, foreign as well as myelin proteins. These contacts stimulated the effector T cells to produce pro-inflammatory mediators, and provided a trigger to tissue invasion and the formation of inflammatory infiltrations.


Assuntos
Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Meninges/irrigação sanguínea , Meninges/imunologia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos/imunologia , Movimento Celular , Células Cultivadas , Meninges/patologia , Camundongos , Ovalbumina/imunologia , Fagócitos/imunologia , Ratos , Ratos Endogâmicos Lew
4.
Immunol Cell Biol ; 91(6): 427-34, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23732617

RESUMO

Chemokine CXCL12 (C-X-C motif chemokine ligand 12) restricts immune cell invasion of the central nervous system (CNS) and limits neuroinflammation in experimental autoimmune encephalomyelitis (EAE), an animal model of inflammatory and demyelinating disease of the CNS, multiple sclerosis (MS). Nitric oxide (NO), by contrast, predominantly contributes to CNS tissue destruction in MS and EAE. Thus, the influence of NO on CXCL12 in the inflamed CNS was investigated. Excess expression of inducible NO synthase was inversely correlated to CXCL12 gene expression in spinal cord homogenates of rats immunized to develop EAE. NO inhibited gene expression of CXCL12 in astrocytes and endothelial cells in vitro. The inhibition was paralleled with reduction of p38 mitogen-activated protein kinase (MAPK) phosphorylation and it was mimicked with inhibitors of p38 MAPK activation in astrocytes. In vivo suppression of nitric generation recovered CXCL12 expression in the CNS and attenuated EAE in Dark Agouti rats. On the contrary, in vivo NO donation decreased CXCL12 expression in the CNS of EAE-resistant Albino Oxford (AO) rats. However, the effect was not paralleled with induction of EAE in AO rats. It is suggested that NO acting through suppression of p38 MAPK inhibits CXCL12 expression in neuroinflammation. These results imply that downregulation of NO release and protection of CXCL12 expression within the CNS might present the potential approaches in MS therapy.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Encefalomielite Autoimune Experimental/imunologia , Células Endoteliais/imunologia , Esclerose Múltipla/imunologia , Óxido Nítrico/imunologia , Animais , Astrócitos/imunologia , Quimiocina CXCL12/biossíntese , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/terapia , Humanos , Imunoterapia/métodos , Imunoterapia/tendências , Esclerose Múltipla/terapia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Endogâmicos , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Amino Acids ; 44(4): 1115-27, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23269478

RESUMO

In vitro and in vivo studies on the role of tenascins have shown that the two paradigmatic glycoproteins of the tenascin family, tenascin-C (TnC) and tenascin-R (TnR) play important roles in cell proliferation and migration, fate determination, axonal pathfinding, myelination, and synaptic plasticity. As components of the extracellular matrix, both molecules show distinct, but also overlapping dual functions in inhibiting and promoting cell interactions depending on the cell type, developmental stage and molecular microenvironment. They are expressed by neurons and glia as well as, for TnC, by cells of the immune system. The functional relationship between neural and immune cells becomes relevant in acute and chronic nervous system disorders, in particular when the blood brain and blood peripheral nerve barriers are compromised. In this review, we will describe the functional parameters of the two molecules in cell interactions during development and, in the adult, in synaptic activity and plasticity, as well as regeneration after injury, with TnC being conducive for regeneration and TnR being inhibitory for functional recovery. Although not much is known about the role of tenascins in neuroinflammation, we will describe emerging knowledge on the interplay between neural and immune cells in autoimmune diseases, such as multiple sclerosis and polyneuropathies. We will attempt to point out the directions of experimental approaches that we envisage would help gaining insights into the complex interplay of TnC and TnR with the cells that express them in pathological conditions of nervous and immune systems.


Assuntos
Doenças do Sistema Nervoso/imunologia , Tenascina/imunologia , Animais , Humanos , Doenças do Sistema Nervoso/genética , Tenascina/genética
6.
Acta Pharmacol Sin ; 34(3): 424-31, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23377550

RESUMO

AIM: To investigate the influences of betulinic acid (BA), a triterpenoid isolated from birch bark, on neuroinflammatory mediators involved in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis in vitro. METHODS: Encephalitogenic T cells were prepared from draining lymph nodes and spinal cords of Dark Agouti rats 8 to 10 d after immunization with myelin basic protein (MBP) and complete Freund's adjuvant. Macrophages were isolated from the peritoneal cavity of adult untreated rats. Astrocytes were isolated from neonatal rat brains. The cells were cultured and then treated with different agents. IFN-γ, IL-17, iNOS and CXCL12 mRNA levels in the cells were analyzed with RT-PCR. iNOS and CXCL12 protein levels were detected using immunoblot. NO and ROS generation was measured using Griess reaction and flow cytometry, respectively. RESULTS: In encephalitogenic T cells stimulated with MBP (10 µg/mL), addition of BA inhibited IL-17 and IFN-γ production in a dose-dependent manner. The estimated IC(50) values for IL-17 and IFN γ were 11.2 and 63.8 µmol/L, respectively. When the macrophages were stimulated with LPS (10 ng/mL), addition of BA (50 µmol/L) significantly increased ROS generation, and suppressed NO generation. The astrocytes were stimulated with ConASn containing numerous inflammatory mediators, which mimicked the inflammatory milieu within CNS; addition of BA (50 µmol/L) significantly increased ROS generation, and blocked ConASn-induced increases in iNOS and CXCL12 mRNA levels, but did not affect iNOS and CXCL12 protein levels. Importantly, in both the macrophages and astrocytes, addition of BA (50 µmol/L) inhibited lipid peroxidation. CONCLUSION: Besides inhibiting encephalitogenic T cell cytokines and reducing NO generation, BA induces tissue-damaging ROS generation within CNS.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Encefalomielite Autoimune Experimental/imunologia , Mediadores da Inflamação/metabolismo , Esclerose Múltipla/imunologia , Triterpenos/farmacologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/isolamento & purificação , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Encefalomielite Autoimune Experimental/patologia , Ensaio de Imunoadsorção Enzimática , Immunoblotting , Mediadores da Inflamação/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Esclerose Múltipla/patologia , Óxido Nítrico/metabolismo , Triterpenos Pentacíclicos , Ratos , Ratos Endogâmicos , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Triterpenos/administração & dosagem , Triterpenos/isolamento & purificação , Ácido Betulínico
7.
Mol Pharmacol ; 82(4): 700-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22798453

RESUMO

We have examined the influence of the nitric oxide (NO)-modified anti-inflammatory drug (S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid (VGX-1027) named GIT-27NO or the NO-modified antiviral drug saquinavir (Saq) named Saq-NO on two colon cancer cell lines, mouse CT26CL25 and human HCT116. The effects of the drugs on cell viability, apoptosis, proliferation, and metastatic potential were analyzed. The release of NO and oxygen and nitrogen species was also determined. The efficacy of the drugs was evaluated in vivo in BALB/c mice injected with CT26CL25 cells. Both agents suppressed the growth of colon cancer cells in vitro and reduced tumor volume in syngeneic BALB/c mice. However, their mechanisms of action were different because GIT-27NO released larger amounts of nitrite than Saq-NO in cell cultures and its antitumor action depended on the intracellular NO release inside the cells. On the contrary, Saq-NO released barely detectable amounts of NO and its antitumor action was NO-independent. In fact, cotreatment with an NO-peroxynitrite scavenger revealed that GIT-27NO but not Saq-NO acts through peroxynitrite-mediated cell destruction. At the cellular level, GIT-27NO prevalently induced proapoptotic signals followed by caspase-dependent apoptosis. In contrast, Saq-NO blocked cell proliferation, changed the adhesive, migratory, and invasive properties of the cells, and decreased metastatic potential in vivo. In conclusion, differences in NO release and oxidative stress generation between GIT-27NO and Saq-NO resulted in different mechanisms that caused cell death.


Assuntos
Acetatos/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Óxido Nítrico/química , Oxazóis/farmacologia , Saquinavir/farmacologia , Acetatos/química , Acetatos/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Óxido Nítrico/fisiologia , Oxazóis/química , Oxazóis/uso terapêutico , Saquinavir/química , Saquinavir/uso terapêutico
8.
J Cell Physiol ; 226(7): 1803-12, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21506111

RESUMO

We have recently shown that covalent attachment of the NO moiety to the HIV protease inhibitor Saquinavir (Saq) produced a qualitatively new chemical entity, named Saquinavir-NO (Saq-NO), with enhanced anticancer properties and reduced toxicity. In this study we evaluated the impact of Saq-NO on the growth of A375 human melanoma cells, as a prototype of NO-dependent cancer model. The novel compound strongly affected the in vitro and in vivo progression of A375 melanoma cell growth. The mechanism of antimelanoma action comprised dual drug activity-induction of apoptotic cell death and acquisition of melanoma cell responsiveness to TRAIL. Saq-NO-triggered apoptosis was dependent on transient AKT up-regulation and reduced pERK and iNOS expression that were observed within the first 12 h of exposure to the drug. Thereafter, however, Saq-NO up-regulated both iNOS transcription and NO endogenous synthesis and sensitized A375 cells to TRAIL. Furthermore, reduced YY1 expression was observed after 24 h of Saq-NO exposure, which correlated with increased expression of DR5. The biological relevance of this complex and powerful action of Saq-NO was consistent with the marked drug-induced inhibition of the growth of A375 xenotransplants in nude mice.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Melanoma/tratamento farmacológico , Óxido Nítrico Sintase Tipo II/metabolismo , Saquinavir/análogos & derivados , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Masculino , Melanoma/enzimologia , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Nus , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Saquinavir/farmacologia , Fatores de Tempo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Fator de Transcrição YY1/metabolismo
9.
J Neurochem ; 118(2): 205-14, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21554322

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is a model of multiple sclerosis. Dark Agouti rats immunized with spinal cord homogenate (SCH) and carbonyl iron (CI), as an adjuvant, develop severe hyperacute form of EAE. They succumb to EAE earlier and have higher clinical scores and lethality rate in comparison to counterparts immunized with SCH + complete Freund's adjuvant. There is no difference in the number of cells or in histological presentation of the CNS infiltrates of rats immunized with the two adjuvants. However, there are more granulocytes, NK and NKT cells, and less CD4(+) T cells in the spinal cord infiltrates of SCH + CI-immunized animals. Nitric oxide (NO)-generating enzyme inducible NO synthase have higher expression in spinal cord of SCH + CI-immunized rats, and this corresponds to more intensive nitrotyrosine formation in the CNS tissue of these rats. Abundant infiltration of granulocytes and NK cells into the CNS and excessive generation of peroxynitrite within the CNS of SCH + CI-immunized rats might account for the severe neurological deficits induced by immunization with CI. These factors should be closely examined in the fulminant forms of multiple sclerosis and acute disseminated encephalomyelitis, as they could represent a promising targets for therapy.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Adjuvante de Freund/toxicidade , Compostos Carbonílicos de Ferro/toxicidade , Compostos de Ferro/toxicidade , Animais , Encefalomielite Autoimune Experimental/induzido quimicamente , Feminino , Ratos , Índice de Gravidade de Doença
10.
Int J Cancer ; 128(8): 1955-65, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20568104

RESUMO

Various constituents of the olive tree (Olea europaea) have been traditionally used in the treatment of infection, inflammation, prevention of chronic diseases, cardiovascular disorders and cancer. The anticancer potential of dry olive leaf extract (DOLE) represents the net effect of multilevel interactions between different biologically active compounds from the extract, cancer cells and conventional therapy. In this context, it was of primary interest to evaluate the influence of DOLE on progression of the highly malignant, immuno- and chemoresistant type of skin cancer-melanoma. DOLE significantly inhibited proliferation and subsequently restricted clonogenicity of the B16 mouse melanoma cell line in vitro. Moreover, late phase tumor treatment with DOLE significantly reduced tumor volume in a syngeneic strain of mice. DOLE-treated B16 cells were blocked in the G(0) /G(1) phase of the cell cycle, underwent early apoptosis and died by late necrosis. At the molecular level, the dying process started as caspase dependent, but finalized as caspase independent. In concordance, overexpression of antiapoptotic members of the Bcl-2 family, Bcl-2 and Bcl-XL, and diminished expression of their natural antagonists, Bim and p53, were observed. Despite molecular suppression of the proapoptotic process, DOLE successfully promoted cell death mainly through disruption of cell membrane integrity and late caspase-independent fragmentation of genetic material. Taken together, the results of this study indicate that DOLE possesses strong antimelanoma potential. When DOLE was applied in combination with different chemotherapeutics, various outcomes, including synergy and antagonism, were observed. This requires caution in the use of the extract as a supplementary antitumor therapeutic.


Assuntos
Melanoma Experimental/prevenção & controle , Olea/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , Neoplasias Cutâneas/prevenção & controle , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Western Blotting , Caspases/genética , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas
11.
Br J Nutr ; 103(10): 1413-24, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20025835

RESUMO

The health-promoting effects of various constituents of the olive tree (Olea europaea) are mainly associated with hypoglycaemic and insulin-sensitising activities and have been widely demonstrated in the metabolic syndrome and type 2 diabetes. However, their biological activity in autoimmune type 1 diabetes (T1D) is poorly characterised. Therefore, the influence of O. europaea-derived components present in dry olive leaf extract (DOLE) was examined in two established preclinical models of human T1D, which differ in some aspects of diabetogenesis: multiple low-dose streptozotocin-induced diabetes in susceptible C57BL/6 and CBA/H mouse strains; cyclophosphamide-accelerated diabetes in non-obese diabetic mice. In both T1D models, in vivo administration of DOLE significantly reduced clinical signs of diabetes (hyperglycaemia and body weight loss) and led to complete suppression of histopathological changes in pancreatic islets. In line with these, insulin expression and release were restored in DOLE-treated mice. Interestingly, inducible NO synthase expression and NO production were significantly elevated in peripheral tissues but were down-regulated within the local environment of the endocrine pancreas. This interference was reflected in NO-mediated suppression of T lymphocyte proliferation and lower production of the proinflammatory cytokines interferon-gamma, IL-17 and TNF-alpha in the spleen, with subsequent blockade of beta-cell destruction. The results suggest that DOLE interferes with development of autoimmune diabetes by down-regulating production of proinflammatory and cytotoxic mediators. Therefore, the potential use of a DOLE-enriched diet for prophylaxis/treatment of human T1D, and possibly other autoimmune diseases, is worthy of further investigation.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/imunologia , Olea/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , Animais , Autoimunidade/efeitos dos fármacos , Ciclofosfamida , Diabetes Mellitus Tipo 1/imunologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/genética , Interferon gama/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos NOD , Óxido Nítrico/metabolismo , Extratos Vegetais/química
12.
Immunology ; 126(1): 74-83, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18624729

RESUMO

Interleukin (IL)-17 is a pro-inflammatory cytokine produced by recently described T helper type 17 (Th17) cells, which have critical role in immunity to extracellular bacteria and the pathogenesis of several autoimmune disorders. IL-6 and transforming growth factor (TGF)-beta are crucial for the generation of Th17 cells in mice, while the production of IL-17 is supported by various cytokines, including IL-23, IL-1beta, IL-21, IL-15 and tumour necrosis factor (TNF)-alpha. In this study, the influence of a multifunctional cytokine, macrophage migration inhibitory factor (MIF), on IL-17 production in mice was investigated. Treatment of lymph node cells (LNCs) with recombinant MIF up-regulated mitogen-stimulated IL-17 expression and secretion. Additionally, LNCs from MIF knockout mice (mif(-/-)) had severely impaired production of IL-17, as well as of IL-1beta, IL-6, IL-23 and TGF-beta. When stimulated with recombinant IL-1beta, IL-23 or TNF-alpha, mitogen-triggered mif(-/-) LNCs were fully able to achieve the IL-17 production seen in wild-type (WT) LNCs, while the addition of IL-6 and TGF-beta had no effect. Finally, after injection of mice with complete Freund's adjuvant, secretion of IL-17 as well as the number of IL-17-positive cells was significantly lower in the draining lymph nodes of mif(-/-) mice in comparison with WT mice. The effect of MIF on IL-17 production was dependent on p38, extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK) and Janus kinase 2/signal transducer and activator of transcription 3 (Jak2/STAT3), and not on nuclear factor (NF)-kappaB and nuclear factor of activated T cells (NFAT) signalling. Bearing in mind the contribution of MIF and IL-17 to the pathology of inflammatory and autoimmune disorders, from the results presented here it seems plausible that targeting MIF biological activity could be a valid therapeutic approach for the treatment of such diseases.


Assuntos
Interleucina-17/biossíntese , Oxirredutases Intramoleculares/imunologia , Linfonodos/imunologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Animais , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática/métodos , Adjuvante de Freund , Inflamação/imunologia , Interleucina-17/imunologia , Oxirredutases Intramoleculares/deficiência , Ativação Linfocitária/imunologia , Fatores Inibidores da Migração de Macrófagos/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/imunologia , Transdução de Sinais/imunologia , Regulação para Cima/imunologia
13.
BMC Immunol ; 10: 29, 2009 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-19463182

RESUMO

BACKGROUND: Diabetes is characterized by progressive failure of insulin producing beta cells. It is well known that both saturated fatty acids and various products of immune cells can contribute to the reduction of beta cell viability and functionality during diabetes pathogenesis. However, their joint action on beta cells has not been investigated, so far. Therefore, we explored the possibility that leukocytes and saturated fatty acids cooperate in beta cell destruction. RESULTS: Rat pancreatic islets or insulinoma cells (RIN) were co-cultivated with concanavalin A (ConA)-stimulated rat lymph node cells (LNC), or they were treated with cell-free supernatants (Sn) obtained from ConA-stimulated spleen cells or from activated CD3+ cells, in the absence or presence of palmitic acid (PA). ConA-stimulated LNC or Sn and PA cooperated in inducing caspase-3-dependent RIN cell apoptosis. The observed effect of PA and Sn on RIN cell viability was mediated by p38 mitogen-activated protein kinase (MAPK)-signaling and was achieved through auto-destructive nitric oxide (NO) production. The cooperative effect of Sn was mimicked with the combination of interleukin-1beta, interleukin-2, interleukin-6, interleukin-17, interferon-gamma and tumor necrosis factor-alpha. CONCLUSION: These results imply that stimulated T cells produce cytokines that cooperate with saturated free fatty acids in beta cell destruction during diabetes pathogenesis.


Assuntos
Apoptose/imunologia , Comunicação Celular , Células Secretoras de Insulina/imunologia , Insulinoma/imunologia , Ácido Palmítico/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/imunologia , Citocinas/metabolismo , Células Secretoras de Insulina/patologia , Insulinoma/patologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Ácido Palmítico/imunologia , Ratos , Transdução de Sinais , Linfócitos T/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Neuroinflammation ; 6: 37, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-20003332

RESUMO

BACKGROUND: Glucocorticoids have been shown to be effective in the treatment of autoimmune diseases of the CNS such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the mechanisms and the site of glucocorticoids' actions are still not completely defined. The aim of this study was to investigate the in vivo effect of the synthetic glucocorticoid methylprednisolone (MP) on the expression and production of proinflammatory cytokines interferon (IFN)-gamma and interleukin (IL)-17 by cells infiltrating CNS tissue. METHODS: Experimental autoimmune encephalomyelitis was induced in Dark Agouti (DA) rats by immunization with rat spinal cord homogenate mixed with adjuvants. Commencing on the day when the first EAE signs appeared, DA rats were injected daily for 3 days with MP and/or RU486, an antagonist of glucocorticoid receptor. Cytokine production and gene expression in CNS-infiltrating cells and lymph node cells were measured using ELISA and real time PCR, respectively. RESULTS: Treatment of rats with MP ameliorated EAE, and the animals recovered without relapses. Further, MP inhibited IFN-gamma and IL-17 expression and production in cells isolated from the CNS of DA rats with EAE after the last injection of MP. The observed effect of MP in vivo treatment was not mediated through depletion of CD4+ T cells among CNS infiltrating cells, or through induction of their apoptosis within the CNS. Finally, the glucocorticoid receptor-antagonist RU486 prevented the inhibitory effect of MP on IFN-gamma and IL-17 production both in vitro and in vivo, thus indicating that the observed effects of MP were mediated through glucocorticoid receptor-dependent mechanisms. CONCLUSION: Taken together, these results demonstrate that amelioration of EAE by exogenous glucocorticoids might be, at least partly, ascribed to the limitation of effector cell functions in the target tissue.


Assuntos
Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Glucocorticoides/uso terapêutico , Interferon gama/imunologia , Interleucina-17/imunologia , Metilprednisolona/uso terapêutico , Animais , Antígenos CD/imunologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/imunologia , Antagonistas de Hormônios/metabolismo , Mifepristona/metabolismo , Ratos , Ratos Endogâmicos , Receptores de Glucocorticoides/metabolismo
15.
Mol Cancer Ther ; 7(3): 510-20, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18347138

RESUMO

Preclinical studies have shown that nitric oxide (NO)-donating nonsteroidal anti-inflammatory drugs possess anticancer activities. Here, we report in vitro and in vivo studies showing the antitumor effect of the NO-donating isoxazole derivative (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid (GIT-27NO). GIT-27NO, but not the NO-deprived parental compound VGX-1027, significantly affected viability of both rodent (L929, B16, and C6) and human (U251, BT20, HeLa, and LS174) tumor cell lines. GIT-27NO triggered either apoptotic cell death (e.g., L929 cells) or autophagic cell death (C6 and B16 cells). Moreover, GIT-27NO hampered the viability of cisplatin-resistant B16 cells. NO scavenger hemoglobin completely prevented GIT-27NO-induced death, indicating that NO release mediated the tumoricidal effect of the compound. Increase in intracellular NO upon on the treatment was associated with intensified production of reactive oxygen species, whereas their neutralization by antioxidant N-acetylcysteine resulted in partial recovery of cell viability. The antitumor activity of the drug was mediated by the selective activation of mitogen-activated protein kinases in a cell-specific manner and was neutralized by their specific inhibitors. In vivo treatment with GIT-27NO significantly reduced the B16 melanoma growth in syngeneic C57BL/6 mice. The therapeutic effect occurred at dose (0.5 mg/mouse) up to 160 times lower than those needed to induce acute lethality (80 mg/mouse). In addition, a dose of GIT-27NO five times higher than that found effective in the melanoma model was well tolerated by the mice when administered for 4 consecutive weeks. These data warrant additional studies to evaluate the possible translation of these findings to the clinical setting.


Assuntos
Acetatos/farmacologia , Antineoplásicos/farmacologia , Doadores de Óxido Nítrico/farmacologia , Oxazóis/farmacologia , Animais , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Doadores de Óxido Nítrico/toxicidade
16.
J Cell Physiol ; 215(3): 665-75, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18064633

RESUMO

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine of the innate immune system that plays a major role in the induction of immunoinflammatory responses. To examine the role of endogenous MIF in the pathogenesis of type 1 diabetes (TID) we evaluated the effects of administration of neutralizing anti-MIF antibodies to NOD mice with accelerated forms of diabetes induced by injection of cyclophosphamide or by transfer of diabetogenic spleen cells. Both accelerated forms of diabetes were markedly reduced by anti-MIF antibody. Furthermore, MIF-deficient (MIF(-/-)) mice were less susceptible to the induction of immunoinflammatory diabetes, insulitis and apoptosis within the endocrine pancreas by multiple low doses of streptozotocin (MLD-STZ) than genetically matched wild type (WT) mice. MIF deficiency resulted in lower proliferation and lymphocyte adhesion, as well as reduced production from the spleens and peritoneal cells of a variety of inflammatory mediators typically associated with development of the disease including IL-12, IL-23, TNF-alpha, and IL-1beta. Furthermore, MIF deletion affected the production of IL-18, TNF-alpha, IL-1beta, and iNOS in the islets of Langerhans. These data, along with the higher expression of IL-4 and TGF-beta observed in the periphery and in the pancreas of MLD-STZ-challenged MIF(-/-) mice as compared to WT controls suggest that MIF deficiency has induced an immune deviation towards protective type 2/3 response. These results suggest that MIF participates in T1D by controlling the functional activity of monocytes/macrophages and T cells and modulating their secretory capacity of pro- and anti-inflammatory molecules.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Transferência Adotiva , Animais , Anticorpos Monoclonais/farmacologia , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclofosfamida/administração & dosagem , Ciclofosfamida/farmacologia , Citocinas/biossíntese , Citocinas/genética , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Tipo 1/induzido quimicamente , Progressão da Doença , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Oxirredutases Intramoleculares/deficiência , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/patologia , Leucócitos Mononucleares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/deficiência , Camundongos , Camundongos Endogâmicos NOD , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Baço/efeitos dos fármacos , Baço/patologia , Estreptozocina , Tirosina/análogos & derivados , Tirosina/metabolismo
17.
BMC Immunol ; 9: 47, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18700009

RESUMO

BACKGROUND: Interleukin-17 (IL-17)-producing cells are increasingly considered to be the major pathogenic population in various autoimmune disorders. The effects of glucocorticoids, widely used as therapeutics for inflammatory and autoimmune disorders, on IL-17 generation have not been thoroughly investigated so far. Therefore, we have explored the influence of methylprednisolone (MP) on IL-17 expression in rat lymphocytes, and compared it to the effect of the drug on interferon (IFN)-gamma. RESULTS: Production of IL-17 in mitogen-stimulated lymph node cells (LNC) from non-treated rats, as well as in myelin basic protein (MBP)-stimulated draining LNC from rats immunized with spinal cord homogenate and complete Freund's adjuvant was significantly reduced by MP. The reduction was dose-dependent, sustained through the follow-up period of 48 hours, and was not achieved through anti-proliferative effect. Additionally, MP inhibited IL-17 production in purified T cells as well, but to less extent than in LNC. In its influence on IL-17 production MP inhibited Ror-gammaT transcription factor expression, as well as Jun phosphorylation, but not ERK or p38 activation in mitogen-stimulated LNC. Importantly, MP collaborated with IFN-gamma in inhibiting IL-17 generation in LNC. CONCLUSION: The observed difference in the effect of MP on IL-17 and IFN-gamma could be important for the understanding of the variability in the efficiency of glucocorticoids in the treatment of autoimmune diseases.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Interferon gama/antagonistas & inibidores , Interleucina-17/antagonistas & inibidores , Metilprednisolona/uso terapêutico , Linfócitos T/efeitos dos fármacos , Animais , Doenças Autoimunes/imunologia , Concanavalina A/farmacologia , Relação Dose-Resposta Imunológica , Cobaias , Imunização , Interferon gama/metabolismo , Interleucina-17/metabolismo , Ativação Linfocitária , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Proteína Básica da Mielina/imunologia , Proteína Básica da Mielina/metabolismo , Ratos , Medula Espinal/química , Medula Espinal/imunologia , Medula Espinal/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Extratos de Tecidos/química , Extratos de Tecidos/imunologia , Extratos de Tecidos/metabolismo
18.
Nitric Oxide ; 19(2): 177-83, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18460348

RESUMO

In this study we evaluated the effects of the new NO donating compound (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid-nitric oxide (GIT-27NO) on the A375 human melanoma cell line. Treatment with the drug led to concentration-dependent reduction of mitochondrial respiration and number of viable cells in cultures. Decreased cell viability correlated with release and internalization of NO and was neutralized by the extracellular scavenger hemoglobin. GIT-27NO neither influenced cell division nor induced accidental or autophagic cell death. Early signs of apoptosis were observed upon coculture with the drug, and resulting in marked accumulation of hypodiploid cells, suggesting that the induction of apoptosis is one primary mode of action of the compound in A375 cells. GIT-27NO significantly inhibited the expression of the transcription repressor and apoptotic resistant factor YY1 and, in parallel, augmented the presence of total p53. The capacity of GIT-27NO to induce p53-mediated apoptosis along with inhibition of YY1 repressor in A375 melanoma cells indicates that GIT-27NO possesses an important anti-cancer pharmacological profile. The findings suggest the potential therapeutic use of GIT-27NO in the clinical setting.


Assuntos
Apoptose/efeitos dos fármacos , Melanoma/tratamento farmacológico , Doadores de Óxido Nítrico/farmacologia , Proteína Supressora de Tumor p53/efeitos dos fármacos , Acetatos , Antineoplásicos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Melanoma/patologia , Doadores de Óxido Nítrico/uso terapêutico , Oxazóis , Fator de Transcrição YY1/antagonistas & inibidores
19.
Neurosci Lett ; 447(2-3): 148-52, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18852028

RESUMO

Interferon-gamma (IFN-gamma) and interleukin-17 (IL-17) have been involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). We have carried out a follow-up study of the expression and production of these cytokines, as well as of cells expressing these cytokines during the course of active EAE in Dark Agouti (DA) rats. As a result, IL-17, but not IFN-gamma expression and production had the peak value in draining lymph nodes (DLN) during the induction phase of the disease, and in spinal cords (SC) at the onset of clinical signs of the disease, and then declined toward the resolution of the disease. Also, a significant proportion of IFN-gamma/IL-17 double-positive cells was observed in SC of DA rats in active EAE. Importantly, the highest proportion of IL-17 single positive and double-positive cells, but not of IFN-gamma single positive cells, was observed at the onset of the disease. The observed difference in the kinetics of IFN-gamma and IL-17 expression during active EAE in DA rats suggests different roles these cytokines might have in the pathogenesis of the disease.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Regulação da Expressão Gênica/fisiologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Tecido Linfoide/metabolismo , Animais , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Contagem de Células , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Linfócitos/imunologia , Linfócitos/metabolismo , Tecido Linfoide/citologia , Proteína Básica da Mielina/imunologia , Ratos , Medula Espinal/patologia , Fatores de Tempo
20.
Cytokine Growth Factor Rev ; 15(1): 21-32, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14746811

RESUMO

Interleukin-17 (IL-17) is a proinflammatory T cell cytokine presumably involved in physiological responses to infection, but also in immunopathology of autoimmune disorders such as rheumatoid arthritis. The proinflammatory action of IL-17 depends considerably on its ability to trigger the expression of inducible nitric oxide (NO) synthase (iNOS), an enzyme responsible for the generation of cytotoxic and immunoregulatory free radical NO. Here we discuss the role of IL-17 in the cytokine network controlling iNOS expression, and analyze signaling pathways employed by IL-17 for the initiation of iNOS gene transcription. We also propose biological consequences of IL-17-mediated NO release that could be relevant for the mechanisms or therapy of autoimmune and inflammatory disorders.


Assuntos
Interleucina-17/fisiologia , Óxido Nítrico Sintase/metabolismo , Animais , Condrócitos/metabolismo , Citocinas/biossíntese , Ativação Enzimática , Radicais Livres , Humanos , Inflamação , Interleucina-17/metabolismo , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Óxido Nítrico , Óxido Nítrico Sintase Tipo II , Osteoblastos/metabolismo , Linfócitos T/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA