Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 212(7): 1094-1104, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38426888

RESUMO

Type 1 diabetes (T1D) is a prototypic T cell-mediated autoimmune disease. Because the islets of Langerhans are insulated from blood vessels by a double basement membrane and lack detectable lymphatic drainage, interactions between endocrine and circulating T cells are not permitted. Thus, we hypothesized that initiation and progression of anti-islet immunity required islet neolymphangiogenesis to allow T cell access to the islet. Combining microscopy and single cell approaches, the timing of this phenomenon in mice was situated between 5 and 8 wk of age when activated anti-insulin CD4 T cells became detectable in peripheral blood while peri-islet pathology developed. This "peri-insulitis," dominated by CD4 T cells, respected the islet basement membrane and was limited on the outside by lymphatic endothelial cells that gave it the attributes of a tertiary lymphoid structure. As in most tissues, lymphangiogenesis seemed to be secondary to local segmental endothelial inflammation at the collecting postcapillary venule. In addition to classic markers of inflammation such as CD29, V-CAM, and NOS, MHC class II molecules were expressed by nonhematopoietic cells in the same location both in mouse and human islets. This CD45- MHC class II+ cell population was capable of spontaneously presenting islet Ags to CD4 T cells. Altogether, these observations favor an alternative model for the initiation of T1D, outside of the islet, in which a vascular-associated cell appears to be an important MHC class II-expressing and -presenting cell.


Assuntos
Diabetes Mellitus Tipo 1 , Ilhotas Pancreáticas , Humanos , Camundongos , Animais , Células Endoteliais , Antígenos de Histocompatibilidade Classe II , Inflamação/patologia , Camundongos Endogâmicos NOD
2.
J Telemed Telecare ; : 1357633X231174488, 2023 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-37221865

RESUMO

INTRODUCTION: Given that chronic, long-term conditions are increasingly common in older patients, the impact of telesurveillance program on clinical outcomes is uncertain. This study aimed to evaluate the feasibility and effectiveness of a 12-month remote monitoring program in preventing rehospitalizations in older patients with two or more chronic diseases returning home after hospitalization. METHODS: We conducted a multicenter randomized controlled trial in two parallel groups to evaluate the remote monitoring system. Elderly patients with chronic diseases (at least two comorbidities) aged 65 years or older and discharged home after acute hospital care for a chronic disease were randomized to receive a home telemonitoring program (intervention group, n = 267) or conventional care (control group, n = 267). The remote home monitoring program was an online biometric home life analysis technology (e-COBAHLT) with tele-homecare/automation and biometric sensors. The eCOBALTH intervention group received the automation sensors containing chronic disease clinical factor trackers to monitor their biometric parameters and detect any abnormal prodromal disease decompensation by remote monitoring and providing geriatric expertise to general practitioners. The usual care group received no eCOBALTH program. In both groups, baseline visits were conducted at baseline and the final visit at 12 months. The primary outcome was the incidence of unplanned hospitalizations for decompensation during the 12-month period. RESULTS: Among 534 randomized participants (mean [SD] age, 80.3 [8.1] years; 280 [52.4%] women), 492 (92.1%) completed the 12-month follow-up; 182 (34.1) had chronic heart failure, 115 (21.5%) had stroke, and 77 (14.4%) had diabetes. During the 12-month follow-up period, 238 patients had at least one unplanned hospitalization for decompensation of a chronic disease: 108 (40.4%) in the intervention group versus 130 (48.7%) in the control group (P = 0.04). The risk of rehospitalization was significantly reduced in the intervention group (age- and sex-adjusted relative risk: 0.72, 95% 95% confidence intervals 0.51-0.94). CONCLUSION: A 12-month home telemonitoring program with online biometric analysis using Home life technology combining telecare and biometric sensors is feasible and effective in preventing unplanned hospitalizations for chronic disease decompensation in elderly patients with chronic diseases at high risk for hospitalizations.

3.
Front Microbiol ; 14: 1148319, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36998410

RESUMO

Objectives: The study aimed to describe the dynamics and risk factors of Gram-negative bacteria (GNB) acquisition in preterm infants. Methods: This prospective multicenter French study included mothers hospitalized for preterm delivery and their newborns, followed until hospital discharge. Maternal feces and vaginal fluids at delivery, and neonatal feces from birth to discharge were tested for cultivable GNB, potential acquired resistance, and integrons. The primary outcome was the acquisition of GNB and integrons in neonatal feces, and their dynamics, evaluated by survival analysis using the actuarial method. Risk factors were analyzed using Cox models. Results: Two hundred thirty-eight evaluable preterm dyads were included by five different centers over 16 months. GNB were isolated in 32.6% of vaginal samples, with 15.4% of strains producing extended-spectrum beta-lactamase (ESBL) or hyperproducing cephalosporinase (HCase), and in 96.2% of maternal feces, with 7.8% ESBL-GNB or HCase-GNB. Integrons were detected in 40.2% of feces and 10.6% of GNB strains. The mean (SD) length of stay of newborns was 39.5 (15.9) days; 4 died in the hospital. At least one infection episode occurred in 36.1% of newborns. The acquisition of GNB and integrons was progressive from birth to discharge. At discharge, half of newborns had ESBL-GNB or HCase-GNB, independently favored by a premature rupture of membranes (Hazard Ratio (HR), 3.41, 95% confidence interval (CI), 1.71; 6.81), and 25.6% had integrons (protective factor: multiple gestation, HR, 0.367, 95% CI, 0.195; 0.693). Conclusion: In preterm newborns, the acquisitions of GNB, including resistant ones, and integrons are progressive from birth to discharge. A premature rupture of membranes favored the colonization by ESBL-GNB or Hcase-GNB.

4.
BMC Geriatr ; 22(1): 991, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36550496

RESUMO

OBJECTIVE: The GERONTACCESS trial evaluated the utility and cost-effectiveness of a gerontological telemedicine (TLM) programme for preventing unplanned hospitalisation of residents living in nursing homes (NHs) in regions lacking medical facilities and/or qualified medical providers ("medical deserts"). DESIGN: GERONTACCESS was a 12-month, multicentre, prospective cluster-randomised trial conducted in NHs. The intervention group underwent TLM assessments every 3 months. The control group received the usual care. In both groups, comprehensive on-site assessments were conducted at baseline and the final visit. Care requirements were documented throughout the study. SETTING AND PARTICIPANTS: NH residents aged ≥ 60 years with multiple chronic diseases. METHODS: The study outcomes were the proportion of patients who experienced avoidable and unplanned hospitalisation, and the incremental cost savings per quality-adjusted life years from baseline to the 12-month follow-up. RESULTS: Of the 426 randomised participants (mean ± standard deviation age, 87.2 ± 7.6 years; 311 [73.0%] women), 23.4% in the intervention group and 32.5% in the control group experienced unplanned hospitalisation (odds ratio [OR] = 0.73, 95% confidence interval [CI] 0.43 to 0.97; p = 0.034). Each avoided hospitalisation in the intervention group saved $US 3,846. CONCLUSIONS AND IMPLICATIONS: The results of GERONTACCESS revealed that our gerontological, preventative TLM program significantly reduced unplanned hospitalisations. This innovative intervention limited disease progression and promoted a healthy lifestyle among NH residents. TRIAL REGISTRATION: Clinicaltrials.gov, NCT02816177, registered June 28, 2016.


Assuntos
Casas de Saúde , Telemedicina , Humanos , Feminino , Idoso , Idoso de 80 Anos ou mais , Masculino , Análise Custo-Benefício , Estudos Prospectivos , Hospitalização , Qualidade de Vida
5.
Front Endocrinol (Lausanne) ; 13: 933965, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35937815

RESUMO

Type 1 diabetes (T1D) is a chronic autoimmune disease in which pancreatic islet ß cells are destroyed by immune cells, ultimately leading to overt diabetes. The progressive increase in T1D incidence over the years points to the role of environmental factors in triggering or accelerating the disease process which develops on a highly multigenic susceptibility background. Evidence that environmental factors induce T1D has mostly been obtained in animal models. In the human, associations between viruses, dietary habits or changes in the microbiota and the development of islet cell autoantibodies or overt diabetes have been reported. So far, prediction of T1D development is mostly based on autoantibody detection. Future work should focus on identifying a causality between the different environmental risk factors and T1D development to improve prediction scores. This should allow developing preventive strategies to limit the T1D burden in the future.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Autoanticorpos , Autoimunidade , Diabetes Mellitus Tipo 1/etiologia , Humanos
6.
Front Endocrinol (Lausanne) ; 13: 883000, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35498419

RESUMO

Type 1 Diabetes (T1D) is an autoimmune disease that results from the destruction of pancreatic islet ß-cells by auto-reactive T cells. The clinical management of T1D faces the lack of fully predictive biomarkers in its preclinical stage and of antigen-specific therapies to induce or re-induce immune tolerance to ß-cell autoantigens and prevent its development. From a therapeutic standpoint, preclinical models of T1D have fallen short of directly translating into humans. To circumvent this limitation, preclinical models are being optimized to allow defining autoantigen epitopes that are presented to T cells and directly apply to the human. In this review, we propose to make a point on the latest available models such as humanized immunodeficient NOD mice models and HLA and autoantigen transgenic mice and their application in the context of T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Animais , Autoantígenos , Diabetes Mellitus Tipo 1/tratamento farmacológico , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos NOD
7.
Front Immunol ; 12: 748679, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721418

RESUMO

To circumvent the limitations of available preclinical models for the study of type 1 diabetes (T1D), we developed a new humanized model, the YES-RIP-hB7.1 mouse. This mouse is deficient of murine major histocompatibility complex class I and class II, the murine insulin genes, and expresses as transgenes the HLA-A*02:01 allele, the diabetes high-susceptibility HLA-DQ8A and B alleles, the human insulin gene, and the human co-stimulatory molecule B7.1 in insulin-secreting cells. It develops spontaneous T1D along with CD4+ and CD8+ T-cell responses to human preproinsulin epitopes. Most of the responses identified in these mice were validated in T1D patients. This model is amenable to characterization of hPPI-specific epitopes involved in T1D and to the identification of factors that may trigger autoimmune response to insulin-secreting cells in human T1D. It will allow evaluating peptide-based immunotherapy that may directly apply to T1D in human and complete preclinical model availability to address the issue of clinical heterogeneity of human disease.


Assuntos
Antígeno B7-1/genética , Diabetes Mellitus Tipo 1/imunologia , Antígenos HLA-DQ/genética , Insulina/genética , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Feminino , Antígenos H-2/genética , Antígeno HLA-A2/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Adulto Jovem
8.
Nat Immunol ; 22(3): 322-335, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33531712

RESUMO

Immune system dysfunction is paramount in coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-associated invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in cohorts totaling 208 patients with various stages of disease. MAIT cell frequency is strongly reduced in blood. They display a strong activated and cytotoxic phenotype that is more pronounced in lungs. Blood MAIT cell alterations positively correlate with the activation of other innate cells, proinflammatory cytokines, notably interleukin (IL)-18, and with the severity and mortality of severe acute respiratory syndrome coronavirus 2 infection. We also identified a monocyte/macrophage interferon (IFN)-α-IL-18 cytokine shift and the ability of infected macrophages to induce the cytotoxicity of MAIT cells in an MR1-dependent manner. Together, our results suggest that altered MAIT cell functions due to IFN-α-IL-18 imbalance contribute to disease severity, and their therapeutic manipulation may prevent deleterious inflammation in COVID-19 aggravation.


Assuntos
COVID-19/imunologia , Interferon-alfa/imunologia , Interleucina-18/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Lavagem Broncoalveolar , Estudos de Casos e Controles , Chlorocebus aethiops , Estudos de Coortes , Feminino , França , Humanos , Imunofenotipagem , Interleucina-10/imunologia , Interleucina-15/imunologia , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Interleucina-8/imunologia , Masculino , Pessoa de Meia-Idade , RNA-Seq , SARS-CoV-2 , Índice de Gravidade de Doença , Análise de Célula Única , Células Vero , Adulto Jovem
9.
Diabetes ; 67(9): 1816-1829, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29967002

RESUMO

Key requirements in type 1 diabetes (T1D) are in setting up new assays as diagnostic biomarkers that will apply to prediabetes, likely T-cell assays, and in designing antigen-specific therapies to prevent T1D development. New preclinical models of T1D will be required to help with advancing both aims. By crossing mouse strains that lack either murine MHC class I and class II genes and insulin genes, we developed YES mice that instead express human HLA-A*02:01, HLA-DQ8, and insulin genes as transgenes. The metabolic and immune phenotype of YES mice is basically identical to that of the parental strains. YES mice remain insulitis and diabetes free up to 1 year of follow-up, maintain normoglycemia to an intraperitoneal glucose challenge in the long-term range, have a normal ß-cell mass, and show normal immune responses to conventional antigens. This new model has been designed to evaluate adaptive immune responses to human insulin on a genetic background that recapitulates a human high-susceptibility HLA-DQ8 genetic background. Although insulitis free, YES mice develop T1D when challenged with polyinosinic-polycytidylic acid. They allow the characterization of preproinsulin epitopes recognized by CD8+ and CD4+ T cells upon immunization against human preproinsulin or during diabetes development.


Assuntos
Imunidade Adaptativa , Envelhecimento , Doenças Autoimunes/fisiopatologia , Diabetes Mellitus Tipo 1/fisiopatologia , Modelos Animais de Doenças , Ilhotas Pancreáticas/fisiopatologia , Estado Pré-Diabético/fisiopatologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Progressão da Doença , Feminino , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Antígenos HLA-DQ/sangue , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/metabolismo , Humanos , Insulina/sangue , Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos , Poli I-C/toxicidade , Estado Pré-Diabético/imunologia , Estado Pré-Diabético/metabolismo , Estado Pré-Diabético/patologia , Precursores de Proteínas/sangue , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Organismos Livres de Patógenos Específicos
10.
J Transl Med ; 14: 10, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26791256

RESUMO

BACKGROUND: Colorectal cancer (CRC) remains a major public concern. While conventional chemotherapeutic regimens have proved useful against advanced/metastatic diseases, progresses are to be made to effectively cure the large portion of patients not benefiting from these treatments. One direction to improve response rates is to develop chemosensitivity and resistance assays (CSRAs) efficiently assisting clinicians in treatment selection process, an already long preoccupation of oncologists and researchers. Several methods have been described to this day, none achieving yet sufficient reliability for recommended use in the clinical routine. METHODS: We led a pilot study on 19 metastatic CRC patients evaluating capacity of the Oncogramme, a standardized process using tumor ex vivo models, to provide chemosensitivity profiles and predict clinical outcome of patients receiving standard CRC chemotherapeutics. Oncogramme responses were categorized according to the method of percentiles to assess sensitivity, specificity and concordance. RESULTS: We report from a primary analysis a success rate of 97.4 %, a very good sensitivity (84.6 %), a below-average specificity (33.3 %), along with a global agreement of 63.6 % and a concordance between Oncogramme results and patients' responses (Kappa coefficient) of 0.193. A supplementary analysis, focusing on CRC patients with no treatment switch over a longer time course, demonstrated improvement in specificity and concordance. CONCLUSIONS: Results establish feasibility and usefulness of the Oncogramme, prelude to a larger-scale trial. Advantages and drawbacks of the procedure are discussed, as well as the place of CSRAs within the future arsenal of methods available to clinicians to individualize treatments and improve patient prognosis. TRIAL REGISTRATION: ClinicalTrials.gov database, registration number: NCT02305368.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Seleção de Pacientes , Projetos Piloto , Resultado do Tratamento
11.
Diabetes ; 64(10): 3532-42, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25918233

RESUMO

The first signs of autoimmune activation leading to ß-cell destruction in type 1 diabetes (T1D) appear during the first months of life. Thus, the perinatal period offers a suitable time window for disease prevention. Moreover, thymic selection of autoreactive T cells is most active during this period, providing a therapeutic opportunity not exploited to date. We therefore devised a strategy by which the T1D-triggering antigen preproinsulin fused with the immunoglobulin (Ig)G Fc fragment (PPI-Fc) is delivered to fetuses through the neonatal Fc receptor (FcRn) pathway, which physiologically transfers maternal IgGs through the placenta. PPI-Fc administered to pregnant PPIB15-23 T-cell receptor-transgenic mice efficiently accumulated in fetuses through the placental FcRn and protected them from subsequent diabetes development. Protection relied on ferrying of PPI-Fc to the thymus by migratory dendritic cells and resulted in a rise in thymic-derived CD4(+) regulatory T cells expressing transforming growth factor-ß and in increased effector CD8(+) T cells displaying impaired cytotoxicity. Moreover, polyclonal splenocytes from nonobese diabetic (NOD) mice transplacentally treated with PPI-Fc were less diabetogenic upon transfer into NOD.scid recipients. Transplacental antigen vaccination provides a novel strategy for early T1D prevention and, further, is applicable to other immune-mediated conditions.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Antígenos de Histocompatibilidade Classe I/metabolismo , Insulina/metabolismo , Troca Materno-Fetal/fisiologia , Precursores de Proteínas/metabolismo , Receptores Fc/metabolismo , Animais , Autoimunidade , Proliferação de Células , Células Dendríticas/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Insulina/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Placenta/metabolismo , Gravidez , Precursores de Proteínas/administração & dosagem , Receptores Fc/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Timo/fisiologia
12.
PLoS One ; 9(4): e96151, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24788601

RESUMO

We explored in this study the status and potential role of IL-17-producing iNKT cells (iNKT17) in type 1 diabetes (T1D) by analyzing these cells in patients with T1D, and in NOD mice, a mouse model for T1D. Our analysis in mice showed an increase of iNKT17 cells in NOD vs control C57BL/6 mice, partly due to a better survival of these cells in the periphery. We also found a higher frequency of these cells in autoimmune-targeted organs with the occurrence of diabetes, suggesting their implication in the disease development. In humans, though absent in fresh PMBCs, iNKT17 cells are detected in vitro with a higher frequency in T1D patients compared to control subjects in the presence of the proinflammatory cytokine IL-1ß, known to contribute to diabetes occurrence. These IL-1ß-stimulated iNKT cells from T1D patients keep their potential to produce IFN-γ, a cytokine that drives islet ß-cell destruction, but not IL-4, with a reverse picture observed in healthy volunteers. On the whole, our results argue in favour of a potential role of IL-17-producing iNKT cells in T1D and suggest that inflammation in T1D patients could induce a Th1/Th17 cytokine secretion profile in iNKT cells promoting disease development.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Interleucina-17/biossíntese , Células Matadoras Naturais/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD
13.
Diabetes ; 61(10): 2546-55, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22997432

RESUMO

The cartography of ß-cell epitopes targeted by CD8(+) T cells in type 1 diabetic (T1D) patients remains largely confined to the common HLA-A2 restriction. We aimed to identify ß-cell epitopes restricted by the HLA-B7 (B*07:02) molecule, which is associated with mild T1D protection. Using DNA immunization on HLA-B7-transgenic mice and prediction algorithms, we identified GAD and preproinsulin candidate epitopes. Interferon-γ (IFN-γ) enzyme-linked immunospot assays on peripheral blood mononuclear cells showed that most candidates were recognized by new-onset T1D patients, but not by type 2 diabetic and healthy subjects. Some epitopes were highly immunodominant and specific to either T1D children (GAD(530-538); 44% T cell-positive patients) or adults (GAD(311-320); 38%). All epitopes displayed weak binding affinity and stability for HLA-B7 compared with HLA-A2-restricted ones, a general feature of HLA-B7. Single-cell PCR analysis on ß-cell-specific (HLA-B7 tetramer-positive) T cells revealed uniform IFN-γ and transforming growth factor-ß (TGF-ß) mRNA expression, different from HLA-A2-restricted T cells. We conclude that HLA-B7-restricted islet epitopes display weak HLA-binding profiles, are different in T1D children and adults, and are recognized by IFN-γ(+)TGF-ß(+)CD8(+) T cells. These features may explain the T1D-protective effect of HLA-B7. The novel epitopes identified should find valuable applications for immune staging of HLA-B7(+) individuals.


Assuntos
Diabetes Mellitus Tipo 1/genética , Epitopos/genética , Antígeno HLA-B7/genética , Células Secretoras de Insulina/metabolismo , Adolescente , Adulto , Idoso , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Antígeno HLA-B7/metabolismo , Humanos , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Fator de Crescimento Transformador beta/metabolismo
14.
Diabetes ; 60(12): 3289-99, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21998398

RESUMO

OBJECTIVE: Both the early steps and the high recurrence of autoimmunity once the disease is established are unexplained in human type 1 diabetes. Because CD8(+) T cells are central and insulin is a key autoantigen in the disease process, our objective was to characterize HLA class I-restricted autoreactive CD8(+) T cells specific for preproinsulin (PPI) in recent-onset and long-standing type 1 diabetic patients and healthy control subjects. RESEARCH DESIGN AND METHODS: We used HLA-A*02:01 tetramers complexed to PPI peptides to enumerate circulating PPI-specific CD8(+) T cells in patients and characterize them using membrane markers and single-cell PCR. RESULTS: Most autoreactive CD8(+) T cells detected in recent-onset type 1 diabetic patients are specific for leader sequence peptides, notably PPI(6-14), whereas CD8(+) T cells in long-standing patients recognize the B-chain peptide PPI(33-42) (B(9-18)). Both CD8(+) T-cell specificities are predominantly naïve, central, and effector memory cells, and their gene expression profile differs from cytomegalovirus-specific CD8(+) T cells. PPI(6-14)-specific CD8(+) T cells detected in one healthy control displayed Il-10 mRNA expression, which was not observed in diabetic patients. CONCLUSIONS: PPI-specific CD8(+) T cells in type 1 diabetic patients include central memory and target different epitopes in new-onset versus long-standing disease. Our data support the hypothesis that insulin therapy may contribute to the expansion of autoreactive CD8(+) T cells in the long term.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Insulina/metabolismo , Adulto , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Humanos , Insulina/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Diabetes ; 58(2): 394-402, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19011169

RESUMO

OBJECTIVE: A restricted region of proinsulin located in the B chain and adjacent region of C-peptide has been shown to contain numerous candidate epitopes recognized by CD8(+) T-cells. Our objective is to characterize HLA class I-restricted epitopes located within the preproinsulin leader sequence. RESEARCH DESIGN AND METHODS: Seven 8- to 11-mer preproinsulin peptides carrying anchoring residues for HLA-A1, -A2, -A24, and -B8 were selected from databases. HLA-A2-restricted peptides were tested for immunogenicity in transgenic mice expressing a chimeric HLA-A*0201/beta2-microglobulin molecule. The peptides were studied for binding to purified HLA class I molecules, selected for carrying COOH-terminal residues generated by proteasome digestion in vitro and tested for recognition by human lymphocytes using an ex vivo interferon-gamma (IFN-gamma) ELISpot assay. RESULTS: Five HLA-A2-restricted peptides were immunogenic in transgenic mice. Murine T-cell clones specific for these peptides were cytotoxic against cells transfected with the preproinsulin gene. They were recognized by peripheral blood mononuclear cells (PBMCs) from 17 of 21 HLA-A2 type 1 diabetic patients. PBMCs from 25 of 38 HLA-A1, -A2, -A24, or -B8 patients produced IFN-gamma in response to six preproinsulin peptides covering residues 2-25 within the preproinsulin region. In most patients, the response was against several class I-restricted peptides. T-cells recognizing preproinsulin peptide were characterized as CD8(+) T-cells by staining with peptide/HLA-A2 tetramers. CONCLUSIONS: We defined class I-restricted epitopes located within the leader sequence of human preproinsulin through in vivo (transgenic mice) and ex vivo (diabetic patients) assays, illustrating the possible role of preproinsulin-specific CD8(+) T-cells in human type 1 diabetes.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Antígenos HLA/imunologia , Proinsulina/imunologia , Animais , Linhagem Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Feminino , Antígenos HLA/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo
16.
Biochem Biophys Res Commun ; 348(2): 653-61, 2006 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-16890191

RESUMO

Hypokalemic periodic paralysis type 2 (hypoPP2) is an inherited skeletal muscle disorder caused by missense mutations in the SCN4A gene encoding the alpha subunit of the skeletal muscle Na+ channel (Nav1.4). All hypoPP2 mutations reported so far target an arginine residue of the voltage sensor S4 of domain II (R672/G/H/S). We identified a novel hypoPP2 mutation that neutralizes an arginine residue in DIII-S4 (R1132Q), and studied its functional consequences in HEK cells transfected with the human SCN4A cDNA. Whole-cell current recordings revealed an enhancement of both fast and slow inactivation, as well as a depolarizing shift of the activation curve. The unitary Na+ conductance remained normal in R1132Q and in R672S mutants, and cannot therefore account for the reduction of Na+ current presumed in hypoPP2. Altogether, our results provide a clear evidence for the role of R1132 in channel activation and inactivation, and confirm loss of function effects of hypoPP2 mutations leading to muscle hypoexcitability.


Assuntos
Paralisia Periódica Hipopotassêmica/genética , Ativação do Canal Iônico/genética , Canais de Sódio/genética , Canais de Sódio/fisiologia , Adulto , Sequência de Aminoácidos , Arginina/genética , Glutamina/genética , Humanos , Paralisia Periódica Hipopotassêmica/fisiopatologia , Masculino , Canal de Sódio Disparado por Voltagem NAV1.4 , Linhagem , Mutação Puntual
17.
J Physiol ; 565(Pt 2): 415-27, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15790667

RESUMO

Missense mutations in the human skeletal muscle Na+ channel alpha subunit (hSkM1) are responsible for a number of muscle excitability disorders. Among them, paramyotonia congenita (PC) is characterized by episodes of muscle stiffness induced by cold and aggravated by exercise. We have identified a new PC-associated mutation, which substitutes aspartic acid for a conserved alanine in the S4-S5 linker of domain III (A1152D). This residue is of particular interest since its homologue in the rat brain type II Na+ channel has been suggested as an essential receptor site for the fast inactivation particle. To identify the biophysical changes induced by the A1152D mutation, we stably expressed hSkM1 mutant or wild-type (WT) channels in HEK293 (human embryonic kidney) cells, and recorded whole-cell Na+ currents with the patch-clamp technique. Experiments were performed both at 21 and 11 degrees C to better understand the sensitivity to cold of paramyotonia. The A1152D mutation disrupted channel fast inactivation. In comparison to the WT, mutant channels inactivated with slower kinetics and displayed a 5 mV depolarizing shift in the voltage dependence of the steady-state. The other noticeable defect of A1152D mutant channels was an accelerated rate of deactivation from the inactivated state. Decreasing temperature by 10 degrees C amplified the differences in channel gating kinetics between mutant and WT, and unveiled differences in both the sustained current and channel deactivation from the open state. Overall, cold-exacerbated mutant defects may result in a sufficient excess of Na+ influx to produce repetitive firing and myotonia. In the light of previous reports, our data point to functional as well as phenotypic differences between mutations of conserved S4-S5 residues in domains II and III of the human skeletal muscle Na+ channel.


Assuntos
Ativação do Canal Iônico/fisiologia , Mutação de Sentido Incorreto , Transtornos Miotônicos/genética , Canais de Sódio/genética , Idoso , Sequência de Aminoácidos , Temperatura Baixa , Feminino , Humanos , Cinética , Masculino , Potenciais da Membrana/fisiologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Linhagem , Estrutura Terciária de Proteína , Subunidades Proteicas/genética , Canais de Sódio/química , Canais de Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...