Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 10: 1038, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31156627

RESUMO

Type I diabetes (T1D) is a T cell-driven autoimmune disease that results in the killing of pancreatic ß-cells and, consequently, loss of insulin production. Using the multiple low-dose streptozotocin (MLD-STZ) model of experimental autoimmune diabetes, we previously reported that pretreatment with a specific acetylcholinesterase inhibitor (AChEI), paraoxon, prevented the development of hyperglycemia in C57BL/6 mice. This correlated with an inhibition of T cell infiltration into the pancreatic islets and a reduction in pro-inflammatory cytokines. The cholinergic anti-inflammatory pathway utilizes nicotinic and muscarinic acetylcholine receptors (nAChRs and mAChRs, respectively) expressed on a variety of cell types. In this study, we carried out a comparative analysis of the effect of specific antagonists of nAChRs or mAChRs on the development of autoimmune diabetes. Co-administration of mecamylamine, a non-selective antagonist of nAChRs maintained the protective effect of AChEI on the development of hyperglycemia. In contrast, co-administration of atropine, a non-selective antagonist of mAChRs, mitigated AChEI-mediated protection. Mice pretreated with mecamylamine had an improved response in glucose tolerance test (GTT) than mice pretreated with atropine. These differential effects of nAChR and mAChR antagonists correlated with the extent of islet cell infiltration and with the structure and functionality of the ß-cells. Taken together, our data suggest that mAChRs are essential for the protective effect of cholinergic stimulation in autoimmune diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolinesterase/sangue , Animais , Atropina/farmacologia , Diabetes Mellitus Experimental/induzido quimicamente , Modelos Animais de Doenças , Feminino , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/sangue , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Mecamilamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas Muscarínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Paraoxon/farmacologia , Paraoxon/uso terapêutico , Estreptozocina/farmacologia
3.
Front Immunol ; 9: 551, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29616040

RESUMO

Inflammation is a crucial defense mechanism that protects the body from the devastating effects of invading pathogens. However, an unrestrained inflammatory reaction may result in systemic manifestations with dire consequences to the host. The extent of activation of the inflammatory response is tightly regulated through immunological and neural pathways. Previously, we demonstrated that cholinergic stimulation confers enhanced protection in experimental animals orally infected with virulent Salmonella enterica serovar Typhimurium. In this study, we investigated the mechanism by which this enhanced protection takes place. Cholinergic stimulation was induced by a 3-week pretreatment with paraoxon, a highly specific acetylcholinesterase (AChE) inhibitor. This treatment enhanced host survival following oral-route infection and this correlated with significantly reduced bacterial load in systemic target organs. Enhanced protection was not due to increased gut motility or rapid bacterial clearance from the gastrointestinal tract. Moreover, protection against bacterial infection was not evident when the animals were infected systemically, suggesting that acetylcholine-mediated protective effect was mostly confined to the gut mucosal tissue. In vivo imaging demonstrated a more localized infection and delay in bacterial dissemination into systemic organs in mice pretreated with paraoxon. Morphological analysis of the small intestine (ileum) showed that AChE inhibition induced the degranulation of goblet cells and Paneth cells, two specialized secretory cells involved in innate immunity. Our findings demonstrate a crucial pathway between neural and immune systems that acts at the mucosal interface to protect the host against oral pathogens.


Assuntos
Agonistas Colinérgicos/uso terapêutico , Inibidores da Colinesterase/uso terapêutico , Íleo/patologia , Mucosa Intestinal/imunologia , Paraoxon/uso terapêutico , Salmonella typhi/fisiologia , Febre Tifoide/tratamento farmacológico , Acetilcolinesterase/metabolismo , Administração Oral , Animais , Carga Bacteriana/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Imunidade Inata/efeitos dos fármacos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuroimunomodulação
4.
Emerg Radiol ; 25(1): 7-11, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28861635

RESUMO

PURPOSE: The purpose of this study is to measure the performance of restricted diffusion of the pericholecystic hepatic parenchyma for distinguishing between acute and chronic cholecystitis. METHODS: The institutional review board approved this HIPAA-compliant retrospective study. Two hundred sixty-six patients presenting with acute right upper quadrant pain between 10/3/2010 and 11/28/2012 undergoing MR within 48 h of equivocal utility of ultrasound (US) were included. Diffusion-weighted imaging (DWI) sequences (b = 0, 600 s/mm2, apparent diffusion coefficient (ADC) maps) were reviewed and graded in a blinded fashion by two abdominal fellowship-trained radiologists for the presence of restricted diffusion in the pericholecystic hepatic parenchyma and the gallbladder wall. Sensitivity, specificity, and inter-observer agreement for individual imaging signs were calculated using surgical pathology as the reference standard for acute cholecystitis. RESULTS: Of the 266 patients, 113 were treated conservatively and 153 underwent cholecystectomy. Restricted diffusion of the pericholecystic hepatic parenchyma showed low sensitivity (reviewer 1, 40%; reviewer 2, 30%) and moderately high specificity (reviewer 1, 84%; reviewer 2, 75%) for distinguishing acute and chronic cholecystitis. Restricted diffusion in the gallbladder wall showed low sensitivity (reviewer 1, 30%; reviewer 2, 7%) and high specificity (reviewer 1, 93%; reviewer 2, 97%) for distinguishing acute and chronic cholecystitis. CONCLUSION: Diffusion-weighted imaging of the pericholecystic hepatic parenchyma shows moderately high specificity for distinguishing between acute and chronic cholecystitis.


Assuntos
Colecistite/diagnóstico por imagem , Imagem de Difusão por Ressonância Magnética/métodos , Doença Aguda , Adulto , Colecistectomia , Colecistite/terapia , Doença Crônica , Tratamento Conservador , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Estudos Retrospectivos , Sensibilidade e Especificidade
5.
Front Immunol ; 7: 419, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27790217

RESUMO

Type I diabetes (T1D) results from T cell-mediated damage of pancreatic ß-cells and loss of insulin production. The cholinergic anti-inflammatory pathway represents a physiological link connecting the central nervous and immune systems via vagus nerve, and functions to control the release of proinflammatory cytokines. Using the multiple low-dose streptozotocin (MLD-STZ) model to induce experimental autoimmune diabetes, we investigated the potential of regulating the development of hyperglycemia through administration of paraoxon, a highly specific acetylcholinesterase inhibitor (AChEI). We demonstrate that pretreatment with paraoxon prevented hyperglycemia in STZ-treated C57BL/6 mice. This correlated with a reduction in T cell infiltration into pancreatic islets and preservation of the structure and functionality of ß-cells. Gene expression analysis of pancreatic tissue revealed that increased peripheral cholinergic activity prevented STZ-mediated loss of insulin production, this being associated with a reduction in IL-1ß, IL-6, and IL-17 proinflammatory cytokines. Intracellular cytokine analysis in splenic T cells demonstrated that inhibition of AChE led to a shift in STZ-induced immune response from a predominantly disease-causing IL-17-expressing Th17 cells to IFNγ-positive Th1 cells. Consistent with this conclusion, inhibition of AChE failed to prevent STZ-induced hyperglycemia in IFNγ-deficient mice. Our results provide mechanistic evidence for the prevention of murine T1D by inhibition of AChE and suggest a promising strategy for modulating disease severity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...