Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 200(11): 3697-3710, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669783

RESUMO

Vasoactive intestinal peptide (VIP) is a neuropeptide that exerts various vascular and cardioprotective functions and regulates immune function and inflammatory response at multiple levels. However, its role in inflammatory cardiovascular disorders is largely unknown. Myocarditis and atherosclerosis are two inflammatory and autoimmune cardiovascular diseases that cause important adverse circulatory events. In this study, we investigate the therapeutic effects of VIP in various well-established preclinical models of experimental autoimmune myocarditis and atherosclerosis. Intraperitoneal injection of VIP during the effector phase of experimental autoimmune myocarditis in susceptible BALB/c mice significantly reduced its prevalence, ameliorated signs of heart hypertrophy and injury, attenuated myocardial inflammatory infiltration, and avoided subsequent profibrotic cardiac remodeling. This effect was accompanied by a reduction of Th17-driven cardiomyogenic responses in peripheral lymphoid organs and in the levels of myocardial autoantibodies. In contrast, acute and chronic atherosclerosis was induced in apolipoprotein E-deficient mice fed a hyperlipidemic diet and subjected to partial carotid ligation. Systemic VIP treatment reduced the number and size of atherosclerotic plaques in carotid, aorta, and sinus in hypercholesterolemic mice. VIP reduced Th1-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and their draining lymph nodes. VIP also regulated cholesterol efflux in macrophages and reduced the formation of foam cells and their presence in atherosclerotic plaques. Finally, VIP inhibited proliferation and migration of smooth muscle cells and neointima formation in a mouse model of complete carotid ligation. These findings encourage further studies aimed to assess whether VIP can be used as a pharmaceutical agent to treat heart inflammation and atherosclerosis.


Assuntos
Aterosclerose/imunologia , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Inflamação/imunologia , Miocardite/imunologia , Peptídeo Intestinal Vasoativo/imunologia , Animais , Apolipoproteínas E/imunologia , Autoanticorpos/imunologia , Modelos Animais de Doenças , Feminino , Linfonodos/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Músculo Liso/imunologia , Miocárdio/imunologia , Neuropeptídeos/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
2.
Sci Rep ; 7: 46444, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28406244

RESUMO

Atherosclerosis is a chronic inflammatory cardiovascular disease that is responsible of high mortality worldwide. Evidence indicates that maladaptive autoimmune responses in the arterial wall play critical roles in the process of atherosclerosis. Cortistatin is a neuropeptide expressed in the vascular system and atherosclerotic plaques that regulates vascular calcification and neointimal formation, and inhibits inflammation in different experimental models of autoimmune diseases. Its role in inflammatory cardiovascular disorders is largely unexplored. The aim of this study is to investigate the potential therapeutic effects of cortistatin in two well-established preclinical models of atherosclerosis, and the molecular and cellular mechanisms involved. Systemic treatment with cortistatin reduced the number and size of atherosclerotic plaques in carotid artery, heart, aortic arch and aorta in acute and chronic atherosclerosis induced in apolipoprotein E-deficient mice fed a high-lipid diet. This effect was exerted at multiple levels. Cortistatin reduced Th1/Th17-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and lymphoid organs. Moreover, cortistatin reduced the capacity of endothelial cells to bind and recruit immune cells to the plaque and impaired the formation of foam cells by enhancing cholesterol efflux from macrophages. Cortistatin emerges as a new candidate for the treatment of the clinical manifestations of atherosclerosis.


Assuntos
Anti-Inflamatórios/administração & dosagem , Células Espumosas/efeitos dos fármacos , Hiperlipidemias/complicações , Neuropeptídeos/administração & dosagem , Placa Aterosclerótica/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Células Espumosas/metabolismo , Hiperlipidemias/genética , Camundongos , Camundongos Knockout para ApoE , Neuropeptídeos/farmacologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
3.
Br J Pharmacol ; 174(3): 267-280, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27922195

RESUMO

BACKGROUND AND PURPOSE: Myocarditis is an inflammatory and autoimmune cardiovascular disease that causes dilated myocardiopathy and is responsible for high morbidity and mortality worldwide. Cortistatin is a neuropeptide produced by neurons and cells of the immune and vascular systems. Besides its action in locomotor activity and sleep, cortistatin inhibits inflammation in different experimental models of autoimmune diseases. However, its role in inflammatory cardiovascular disorders is unexplored. Here, we investigated the therapeutic effects of cortistatin in a well-established preclinical model of experimental autoimmune myocarditis (EAM). EXPERIMENTAL APPROACH: We induced EAM by immunization with a fragment of cardiac myosin in susceptible Balb/c mice. Cortistatin was administered i.p. starting 7, 11 or 15 days after EAM induction. At day 21, we evaluated heart hypertrophy, myocardial injury, cardiac inflammatory infiltration and levels of serum and cardiac inflammatory cytokines, cortistatin and autoantibodies. We determined proliferation and cytokine production by heart draining lymph node cells in response to cardiac myosin restimulation. KEY RESULTS: Systemic injection of cortistatin during the effector phase of the disease significantly reduced its prevalence and signs of heart hypertrophy and injury (decreased the levels of brain natriuretic peptide) and impaired myocardial inflammatory cell infiltration. This effect was accompanied by a reduction in self-antigen-specific T-cell responses in lymph nodes and in the levels of cardiomyogenic antibodies and inflammatory cytokines in serum and myocardium. Finally, we found a positive correlation between cardiac and systemic cortistatin levels and EAM severity. CONCLUSIONS AND IMPLICATIONS: Cortistatin emerges as a new candidate to treat inflammatory dilated cardiomyopathy.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Cardiomiopatia Dilatada/tratamento farmacológico , Miocardite/tratamento farmacológico , Neuropeptídeos/farmacologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/fisiopatologia , Linfonodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miocardite/imunologia , Miocardite/fisiopatologia , Neuropeptídeos/administração & dosagem , Índice de Gravidade de Doença , Linfócitos T/imunologia , Fatores de Tempo
4.
Ann N Y Acad Sci ; 1351: 89-98, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25951888

RESUMO

Cortistatin is a neuropeptide isolated from cortical brain regions, showing high structural homology and sharing many functions with somatostatin. However, cortistatin exerts unique functions in the central nervous and immune systems, including decreasing locomotor activity, inducing sleep-promoting effects, and deactivating inflammatory and T helper (TH )1/TH 17-driven responses in preclinical models of sepsis, arthritis, multiple sclerosis, and colitis. Besides its release by cortical and hippocampal interneurons, cortistatin is produced by macrophages, lymphocytes, and peripheral nociceptive neurons in response to inflammatory stimuli, supporting a physiological role of cortistatin in the immune and nociceptive systems. Cortistatin-deficient mice have been shown to have exacerbated nociceptive responses to neuropathic and inflammatory pain sensitization. However, a paradoxical effect has been observed in studies of immune disorders, in which, despite showing competent inflammatory/autoreactive responses, cortistatin-deficient mice were partially resistant to systemic autoimmunity and inflammation. This unexpected phenotype was associated with elevated circulating glucocorticoids and anxiety-like behavior. These findings support cortistatin as a novel multimodal therapeutic approach to treat autoimmunity and clinical pain and identify it as a key endogenous component of the neuroimmune system related to stress responses.


Assuntos
Anti-Inflamatórios/metabolismo , Doenças Autoimunes/imunologia , Córtex Cerebral/metabolismo , Neuropeptídeos/metabolismo , Sono/fisiologia , Animais , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Camundongos , Atividade Motora/fisiologia , Nociceptores/imunologia , Somatostatina/metabolismo , Células Th1/imunologia , Células Th17/imunologia
5.
J Immunol ; 191(5): 2144-54, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23918980

RESUMO

Cortistatin is a cyclic-neuropeptide produced by brain cortex and immune cells that shows potent anti-inflammatory activity. In this article, we investigated the effect of cortistatin in two models of experimental autoimmune encephalomyelitis (EAE) that mirror chronic and relapsing-remitting multiple sclerosis. A short-term systemic treatment with cortistatin reduced clinical severity and incidence of EAE, the appearance of inflammatory infiltrates in spinal cord, and the subsequent demyelination and axonal damage. This effect was associated with a reduction of the two deleterious components of the disease, namely, the autoimmune and inflammatory response. Cortistatin decreased the presence/activation of encephalitogenic Th1 and Th17 cells in periphery and nervous system, and downregulated various inflammatory mediators, whereas it increased the number of regulatory T cells with suppressive effects on the encephalitogenic response. Moreover, cortistatin regulated glial activity and favored an active program of neuroprotection/regeneration. We further used cortistatin-deficient mice to investigate the role of endogenous cortistatin in the control of immune responses. Surprisingly, cortistatin-deficient mice were partially resistant to EAE and other inflammatory disorders, despite showing competent inflammatory/autoreactive responses. This unexpected phenotype was associated with elevated circulating glucocorticoids and an anxiety-like behavior. Our findings provide a powerful rationale for the assessment of the efficacy of cortistatin as a novel multimodal therapeutic approach to treat multiple sclerosis and identify cortistatin as a key endogenous component of neuroimmune system.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Neuropeptídeos/metabolismo , Linfócitos T/efeitos dos fármacos , Animais , Encefalomielite Autoimune Experimental/patologia , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/imunologia , Medula Espinal/patologia , Linfócitos T/imunologia
6.
Circ Res ; 112(11): 1444-55, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23595952

RESUMO

RATIONALE: Proliferation and migration of smooth muscle cells (SMCs) are key steps for the progression of atherosclerosis and restenosis. Cortistatin is a multifunctional neuropeptide belonging to the somatostatin family that exerts unique functions in the nervous and immune systems. Cortistatin is elevated in plasma of patients experiencing coronary heart disease and attenuates vascular calcification. OBJECTIVE: To investigate the occurrence of vascular cortistatin and its effects on the proliferation and migration of SMCs in vitro and in vivo and to delimitate the receptors and signal transduction pathways governing its actions. METHODS AND RESULTS: SMCs from mouse carotid and human aortic arteries and from human atherosclerotic plaques highly expressed cortistatin. Cortistatin expression positively correlated with the progression of arterial intima hyperplasia. Cortistatin inhibited platelet-derived growth factor-stimulated proliferation of human aortic SMCs via binding to somatostatin receptors (sst2 and sst5) and ghrelin receptor, induction of cAMP and p38-mitogen-activated protein kinase, and inhibition of Akt activity. Moreover, cortistatin impaired lamellipodia formation and migration of human aortic SMCs toward platelet-derived growth factor by inhibiting, in a ghrelin-receptor-dependent manner, Rac1 activation and cytosolic calcium increases. These effects on SMC proliferation and migration correlated with an inhibitory action of cortistatin on the neointimal formation in 2 models of carotid arterial ligation. Endogenous cortistatin seems to play a critical role in regulating SMC function because cortistatin-deficient mice developed higher neointimal hyperplasic lesions than wild-type mice. CONCLUSIONS: Cortistatin emerges as a natural endogenous regulator of SMCs under pathological conditions and an attractive candidate for the pharmacological management of vascular diseases that course with neointimal lesion formation.


Assuntos
Aterosclerose/patologia , Lesões das Artérias Carótidas/patologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Neointima/patologia , Neuropeptídeos/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Lesões das Artérias Carótidas/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Humanos , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neointima/tratamento farmacológico , Neointima/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , RNA Mensageiro/metabolismo , Receptores de Grelina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Somatostatina/metabolismo
7.
Brain Behav Immun ; 30: 54-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23376169

RESUMO

Ghrelin is an important gastrointestinal hormone that regulates feeding and metabolism. Moreover, ghrelin is produced by immune cells and shows potent anti-inflammatory activities. Here, we investigated its effect in two models of experimental autoimmune encephalomyelitis (EAE) that mirror chronic and relapsing-remitting multiple sclerosis. A short systemic treatment with ghrelin after the disease onset reduced clinical severity and incidence of both forms of EAE, which was associated with a decrease in inflammatory infiltrates in spinal cord and in the subsequent demyelination. This therapeutic effect was exerted through the reduction of the autoimmune and inflammatory components of the disease. Ghrelin decreased the presence/activation of encephalitogenic Th1 and Th17 cells in periphery and nervous system, down-regulated various inflammatory mediators, and induced regulatory T cells. In summary, our findings provide a powerful rationale for the assessment of the efficacy of ghrelin as a novel therapeutic approach for treating multiple sclerosis through distinct immunomodulatory mechanisms and further support the concept that the neuroendocrine and immune systems crosstalk to finely tune the final immune response of our body.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Grelina/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Grelina/farmacologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/metabolismo , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-23094830

RESUMO

Vasoactive intestinal peptide (VIP) is a well-known immunoregulatory neuropeptide produced by the immune system in response to inflammation, autoimmunity or alloantigens as a natural endogenous mechanism of induction of tolerance. VIP has been proven therapeutically effective in various experimental models of autoimmune disorders and recently in human sarcoidosis. Numerous studies clearly show that VIP exerts its immunomodulatory effects by downregulating both inflammatory and Th1 responses. Recent evidences suggest that new actors enter in scene to play a role in this scenario of tolerance. By inducing antigen-specific regulatory T cells and tolerogenic dendritic cells, VIP seems to reinforce/reinstall immune tolerance, especially under autoimmune conditions. Transplantation is also a condition where VIP-related therapies emerge as promising tools for clinical application. Induction of alloantigen-specific tolerance is critical to achieve organ transplant tolerance and to avoid graft-versus-host responses following allogeneic hematopoietic transplantation. This review will focus on describing the capacity of VIP to induce suppressive/regulatory immune cells and how we can manage this cell-based therapeutic strategy to induce transplant tolerance in subjects free of immunosuppressive drugs.


Assuntos
Transplante de Órgãos/métodos , Imunologia de Transplantes/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/uso terapêutico , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Tolerância Imunológica/efeitos dos fármacos , Linfócitos T/imunologia
9.
Curr Pharm Des ; 16(28): 3158-72, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20687881

RESUMO

Because there are no particular molecular signatures of self, autoimmunity is the inevitable evolutionary price of being able to make effective responses against a wide variety of pathogens by the immune system. Without the various phenomena referred to as immune tolerance, the organism would surely self-destruct. Considerable evidence suggests that various endogenous neuropeptides play a major role in the education of our immune system to be self-tolerant. The fact that neuropeptides regulate various layers involved in maintenance of tolerance, including regulation of the balance between pro-inflammatory and anti-inflammatory responses and between self-reactive Th1/Th17 cells and regulatory T cells, makes them attractive candidates for the development of new therapies for the treatment of autoimmune disorders. Here we use the vasoactive intestinal peptide of a prototype of immunomodulatory neuropeptide to review the most relevant data found for other neuropeptides with similar characteristics, including melanocyte-stimulating hormone, urocortin, adrenomedullin, neuropeptide Y, cortistatin and ghrelin. We also evaluate the challenges that must be overcome before achieving their clinical application and offer our opinion on how a physiologically functional neuropeptide system contributes to general health.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Autoimunidade/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Neuroimunomodulação/efeitos dos fármacos , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , Animais , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/imunologia , Modelos Biológicos , Peptídeo Intestinal Vasoativo/farmacologia , Peptídeo Intestinal Vasoativo/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...