Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Diabetologia ; 61(2): 389-398, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29030662

RESUMO

AIM/HYPOTHESIS: Combination therapy targeting the major actors involved in the immune-mediated destruction of pancreatic beta cells appears to be an indispensable approach to treat type 1 diabetes effectively. We hypothesised that the combination of an orally active pan-histone deacetylase inhibitor (HDACi: givinostat) with subtherapeutic doses of CD3 antibodies may provide ideal synergy to treat ongoing autoimmunity. METHODS: NOD mice transgenic for the human CD3ε (also known as CD3E) chain (NOD-huCD3ε) were treated for recent-onset diabetes with oral givinostat, subtherapeutic doses of humanised CD3 antibodies (otelixizumab, 50 µg/day, 5 days, i.v.) or a combination of both drugs. Disease remission, metabolic profiles and autoreactive T cell responses were analysed in treated mice. RESULTS: We demonstrated that givinostat synergised with otelixizumab to induce durable remission of diabetes in 80% of recently diabetic NOD-huCD3ε mice. Remission was obtained in only 47% of mice treated with otelixizumab alone. Oral givinostat monotherapy did not reverse established diabetes but reduced the in situ production of inflammatory cytokines (IL-1ß, IL-6, TNF-α). Importantly, the otelixizumab + givinostat combination strongly improved the metabolic status of NOD-huCD3ε mice; the mice recovered the capacity to appropriately produce insulin, control hyperglycaemia and sustain glucose tolerance. Finally, diabetes remission induced by the combination therapy was associated with a significant reduction of insulitis and autoantigen-specific CD8+ T cell responses. CONCLUSIONS/INTERPRETATION: HDACi and low-dose CD3 antibodies synergised to abrogate in situ inflammation and thereby improved pancreatic beta cell survival and metabolic function leading to long-lasting diabetes remission. These results support the therapeutic potential of protocols combining these two drugs, both in clinical development, to restore self-tolerance and insulin independence in type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Inibidores de Histona Desacetilases/uso terapêutico , Imunoterapia/métodos , Células Secretoras de Insulina/metabolismo , Linfócitos T/fisiologia , Administração Oral , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Citometria de Fluxo , Inibidores de Histona Desacetilases/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/sangue , Interleucina-10/sangue , Interleucina-1beta/sangue , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Fator de Necrose Tumoral alfa/sangue
2.
J Neuroinflammation ; 11: 27, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24490798

RESUMO

BACKGROUND: Interleukin 1 (IL-1) is implicated in neuroinflammation, an essential component of neurodegeneration. We evaluated the potential anti-inflammatory effect of a novel peptide antagonist of IL-1 signaling, Ilantide. METHODS: We investigated the binding of Ilantide to IL-1 receptor type I (IL-1RI) using surface plasmon resonance, the inhibition of Il-1ß-induced activation of nuclear factor κB (NF-κB) in HEK-Blue cells that contained an IL-1ß-sensitive reporter, the secretion of TNF-α in macrophages, protection against IL-1-induced apoptosis in neonatal pancreatic islets, and the penetration of Ilantide through the blood-brain barrier using competitive enzyme-linked immunosorbent assay (ELISA). We studied the effects of the peptide on social behavior and memory in rat models of lipopolysaccharide (LPS)- and amyloid-induced neuroinflammation, respectively, and its effect in a rat model of experimental autoimmune enchephalomyelitis. RESULTS: Ilantide bound IL-1RI, inhibited the IL-1ß-induced activation of NF-κB, and inhibited the secretion of TNF-α in vitro. Ilantide protected pancreatic islets from apoptosis in vitro and reduced inflammation in an animal model of arthritis. The peptide penetrated the blood-brain barrier. It reduced the deficits in social activity and memory in LPS- and amyloid-treated animals and delayed the development of experimental autoimmune enchephalomyelitis. CONCLUSIONS: These findings indicate that Ilantide is a novel and potent IL-1RI antagonist that is able to reduce inflammatory damage in the central nervous system and pancreatic islets.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Artrite/induzido quimicamente , Células Cultivadas , Cerebelo/citologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Humanos , Proteína Antagonista do Receptor de Interleucina 1/química , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Lipopolissacarídeos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Comportamento Social , Transfecção , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
3.
Arch Biochem Biophys ; 528(2): 171-84, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23063755

RESUMO

Disease is conventionally viewed as the chaotic inappropriate outcome of deranged tissue function resulting from aberrancies in cellular processes. Yet the patho-biology of cellular dysfunction and death encompasses a coordinated network no less sophisticated and regulated than maintenance of homeostatic balance. Cellular demise is far from passive subordination to stress but requires controlled coordination of energy-requiring activities including gene transcription and protein translation that determine the graded transition between defensive mechanisms, cell cycle regulation, dedifferentiation and ultimately to the activation of death programmes. In fact, most stressors stimulate both homeostasis and regeneration on one hand and impairment and destruction on the other, depending on the ambient circumstances. Here we illustrate this bimodal ambiguity in cell response by reviewing recent progress in our understanding of how the pancreatic ß cell copes with inflammatory stress by changing gene transcription and protein translation by the differential and interconnected action of reactive oxygen and nitric oxide species, microRNAs and posttranslational protein modifications.


Assuntos
Citocinas/genética , Citocinas/fisiologia , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Histona Desacetilases/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transcrição Gênica
4.
J Interferon Cytokine Res ; 35(1): 63-70, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25062500

RESUMO

Histone deacetylase (HDAC) inhibition protects pancreatic beta-cells against apoptosis induced by the combination of the proinflammatory cytokines interleukin (IL)-1ß and interferon (IFN)-γ. Decreased expression of cell damage-related genes is observed on the transcriptional level upon HDAC inhibition using either IL-1ß or IFN-γ alone. Whereas HDAC inhibition has been shown to regulate NFκB-activity, related primarily to IL-1ß signaling, it is unknown whether the inhibition of HDACs affect IFN-γ signaling in beta-cells. Further, in non-beta-cells, there is a dispute whether HDAC inhibition regulates IFN-γ signaling at the level of STAT1 Tyr701 phosphorylation. Using different small molecule HDAC inhibitors with varying class selectivity, INS-1E wild type and stable HDAC1-3 knockdown pancreatic INS-1 cell lines, we show that IFN-γ-induced Cxcl9 and iNos expression as well as Cxcl9 and GAS reporter activity were decreased by HDAC inhibition in a STAT1 Tyr701 phosphorylation-independent fashion. In fact, knockdown of HDAC1 increased IFN-γ-induced STAT1 phosphorylation.


Assuntos
Histona Desacetilase 1/genética , Histona Desacetilases/genética , Células Secretoras de Insulina/imunologia , Óxido Nítrico Sintase Tipo II/biossíntese , Fator de Transcrição STAT1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carbamatos/farmacologia , Linhagem Celular , Quimiocina CXCL10/biossíntese , Quimiocina CXCL9/biossíntese , Inibidores de Histona Desacetilases/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , NF-kappa B/metabolismo , Fosforilação , Ratos , Fator de Transcrição STAT1/genética
5.
J Clin Endocrinol Metab ; 100(10): E1289-98, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26218753

RESUMO

CONTEXT: Mechanisms explaining exercise-induced ß-cell health are unknown. OBJECTIVE: This study aimed to define the role of muscle contraction and acute exercise-derived soluble humoral mediators on ß-cell health. DESIGN: In vitro models were used. SETTING: University. PARTICIPANTS: Healthy subjects. INTERVENTION(S): Conditioned media (CM) were collected from human skeletal muscle (HSkM) cells treated with or without electrical pulse stimulation (EPS). Antecubital and femoral venous blood serum were collected before and after an exercise bout. CM and sera with or without IL-6 neutralization were used to incubate insulin-producing INS-1 cells and rat islets for 24 h in the presence or absence of proinflammatory cytokines (IL-1ß+IFN-γ). MAIN OUTCOME MEASURE(S): INS-1 and islet apoptosis and accumulated insulin secretion. RESULTS: IL-1ß+IFN-γ increased INS-1 and islet apoptosis and decreased insulin secretion. EPS-treated HSkM cell CM did not affect these variables. Exercise-conditioned antecubital but not femoral sera prevented IL-1ß+IFN-γ-induced INS-1 and islet apoptosis. Femoral sera reduced insulin secretion under normal and proinflammatory conditions in INS-1 but not islet cells. EPS increased HSkM cell IL-6 secretion and exercise increased circulating IL-6 levels in antecubital and femoral serum. IL-6 neutralization demonstrated that muscle-derived IL-6 prevents INS-1 and islet apoptosis in the absence of IL-1ß+IFN-γ, but augments apoptosis under proinflammatory conditions, and that muscle-derived IL-6 supports islet insulin secretion in the absence of IL-1ß+IFN-γ. CONCLUSIONS: Unidentified circulating humoral mediators released during exercise prevent proinflammatory cytokine-induced ß-cell apoptosis. Muscle-derived mediators released during exercise suppress ß-cell insulin secretion. Furthermore, muscle-derived IL-6 seems to prevent ß-cell apoptosis under normal conditions but contributes to ß-cell apoptosis under proinflammatory conditions.


Assuntos
Apoptose/fisiologia , Exercício Físico/fisiologia , Células Secretoras de Insulina/metabolismo , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Estimulação Elétrica , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Masculino , Ratos , Ratos Wistar , Adulto Jovem
6.
Islets ; 4(6): 417-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23486342

RESUMO

AIMS: Pro-inflammatory cytokines and chemokines, in particular IL-1ß, IFNγ, and CXCL10, contribute to ß-cell failure and loss in DM via IL-1R, IFNγR, and TLR4 signaling. IL-1 signaling deficiency reduces diabetes incidence, islet IL-1ß secretion, and hyperglycemia in animal models of diabetes. Further, IL-1R antagonism improves normoglycemia and ß-cell function in type 2 diabetic patients. Inhibition of lysine deacetylases (KDACi) counteracts ß-cell toxicity induced by the combination of IL-1 and IFNγ and reduces diabetes incidence in non-obese diabetic (NOD) mice. We hypothesized that KDACi breaks an autoinflammatory circuit by differentially preventing ß-cell expression of the ß-cell toxic inflammatory molecules IL-1ß and CXCL10 induced by single cytokines. RESULTS: CXCL10 did not induce transcription of IL-1ß mRNA. IL-1ß induced ß-cell IL-1ß mRNA and both IL-1ß and IFNγ individually induced Cxcl10 mRNA transcription. Givinostat inhibited IL-1ß-induced IL-1ß mRNA expression in INS-1 and rat islets and IL-1ß processing in INS-1 cells. Givinostat also reduced IFNγ induced Cxcl10 transcription in INS-1 cells but not in rat islets, while IL-1ß induced Cxcl10 transcription was unaffected in both. MATERIALS AND METHODS: INS-1 cells and rat islets of Langerhans were exposed to IL-1ß, IFNγ or CXCL10 in the presence or absence of KDACi (givinostat). Cytokine and chemokine mRNA expressions were quantified by real-time qPCR, and IL-1ß processing by western blotting of cell lysates. CONCLUSION/INTERPRETATION: Inhibition of ß-cell IL-1ß expression and processing and Cxcl10 transcription contributes to the ß-cell protective actions of KDACi. In vitro ß-cell destructive effects of CXCL10 are not mediated via IL-1ß transcription. The differential proinflammatory actions of KDACs may be attractive novel drug targets in DM.


Assuntos
Carbamatos/farmacologia , Diabetes Mellitus/imunologia , Inibidores de Histona Desacetilases/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/biossíntese , Animais , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/genética , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Humanos , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/imunologia , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-1beta/genética , RNA Mensageiro/química , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa