Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1276255, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37908349

RESUMO

Gold nanoparticles (GNPs) have been used in the development of novel therapies as a way of delivery of both stimulatory and tolerogenic peptide cargoes. Here we report that intradermal injection of GNPs loaded with the proinsulin peptide C19-A3, in patients with type 1 diabetes, results in recruitment and retention of immune cells in the skin. These include large numbers of clonally expanded T-cells sharing the same paired T-cell receptors (TCRs) with activated phenotypes, half of which, when the TCRs were re-expressed in a cell-based system, were confirmed to be specific for either GNP or proinsulin. All the identified gold-specific clones were CD8+, whilst proinsulin-specific clones were both CD8+ and CD4+. Proinsulin-specific CD8+ clones had a distinctive cytotoxic phenotype with overexpression of granulysin (GNLY) and KIR receptors. Clonally expanded antigen-specific T cells remained in situ for months to years, with a spectrum of tissue resident memory and effector memory phenotypes. As the T-cell response is divided between targeting the gold core and the antigenic cargo, this offers a route to improving resident memory T-cells formation in response to vaccines. In addition, our scRNAseq data indicate that focusing on clonally expanded skin infiltrating T-cells recruited to intradermally injected antigen is a highly efficient method to enrich and identify antigen-specific cells. This approach has the potential to be used to monitor the intradermal delivery of antigens and nanoparticles for immune modulation in humans.


Assuntos
Diabetes Mellitus Tipo 1 , Nanopartículas Metálicas , Humanos , Autoantígenos , Proinsulina/genética , Ouro , Injeções Intradérmicas , Análise da Expressão Gênica de Célula Única , Peptídeos/genética , Receptores de Antígenos de Linfócitos T/genética
2.
Front Immunol ; 14: 1227133, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37731505

RESUMO

Introduction: In this study, we report a novel therapeutic approach using B lymphocytes to attract islet-specific T cells in the non-obese diabetic (NOD) mouse model and prevent the development of autoimmune diabetes. Rather than using the antibody receptor of B cells, this approach utilizes their properties as antigen-presenting cells to T cells. Methods: Purified splenic B cells were treated with lipopolysaccharide, which increases regulatory B (Breg) cell function, then electroporated with mRNA encoding either chimeric MHC-I or MHC-II molecules covalently linked to antigenic peptides. Immunoregulatory functions of these engineered B cells (e-B cells) were tested by in vitro assays and in vivo co-transfer experiments with beta-cell-antigen-specific CD8+ or CD4+ T cells in NOD.Scid mice, respectively. Results: The e-B cells expressing chimeric MHC-I-peptide inhibited antigen-specific CD8+ T-cell cytotoxicity in vitro. The e-B cells expressing chimeric MHC-II-peptide induced antigen-specific CD4+ T cells to express the regulatory markers, PD-1, ICOS, CTLA-4, Lag3, and Nrp1. Furthermore, e-B cells encoding the chimeric MHC-I and MHC-II peptide constructs protected NOD.Scid mice from autoimmune diabetes induced by transfer of antigen-specific CD8+ and CD4+ T cells. Discussion: MHC-peptide chimeric e-B cells interacted with pathogenic T cells, and protected the host from autoimmune diabetes, in a mouse model. Thus, we have successfully expressed MHC-peptide constructs in B cells that selectively targeted antigen-specific cells, raising the possibility that this strategy could be used to endow different protective cell types to specifically regulate/remove pathogenic cells.


Assuntos
Linfócitos B Reguladores , Diabetes Mellitus Tipo 1 , Ilhotas Pancreáticas , Imunodeficiência Combinada Severa , Camundongos , Animais , Diabetes Mellitus Tipo 1/prevenção & controle , Camundongos Endogâmicos NOD , Camundongos SCID , Antígenos de Histocompatibilidade Classe II
3.
Front Immunol ; 12: 751701, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659258

RESUMO

In the past few years, huge advances have been made in techniques to analyse cells at an individual level using RNA sequencing, and many of these have precipitated exciting discoveries in the immunology of type 1 diabetes (T1D). This review will cover the first papers to use scRNAseq to characterise human lymphocyte phenotypes in T1D in the peripheral blood, pancreatic lymph nodes and islets. These have revealed specific genes such as IL-32 that are differentially expressed in islet -specific T cells in T1D. scRNAseq has also revealed wider gene expression patterns that are involved in T1D and can predict its development even predating autoantibody production. Single cell sequencing of TCRs has revealed V genes and CDR3 motifs that are commonly used to target islet autoantigens, although truly public TCRs remain elusive. Little is known about BCR repertoires in T1D, but scRNAseq approaches have revealed that insulin binding BCRs commonly use specific J genes, share motifs between donors and frequently demonstrate poly-reactivity. This review will also summarise new developments in scRNAseq technology, the insights they have given into other diseases and how they could be leveraged to advance research in the type 1 diabetes field to identify novel biomarkers and targets for immunotherapy.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Animais , Antígenos/imunologia , Biomarcadores , Progressão da Doença , Humanos , Fenótipo , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Análise de Sequência de RNA , Análise de Célula Única
4.
Diabetes ; 70(4): 955-965, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33531355

RESUMO

The NOD mouse develops spontaneous type 1 diabetes, with some features of disease that are very similar to the human disease. However, a proportion of NOD mice are naturally protected from developing diabetes, and currently, studies characterizing this cohort are very limited. Here, using both immunofluorescence and multiparameter flow cytometry, we focus on the pancreatic islet morphology and immune infiltrate observed in naturally protected NOD mice. We show that naturally protected NOD mice are characterized by an increased frequency of insulin-containing, smaller-sized, pancreatic islets. Although mice remain diabetes free, florid immune infiltrate remains. However, this immune infiltrate is skewed toward a regulatory phenotype in both T- and B-cell compartments. Pancreatic islets have an increased frequency of IL-10-producing B cells and associated cell surface markers. Resident memory CD69+CD8+ T cells show a significant shift toward reduced CD103 expression, while CD4+ T cells have increased FoxP3+CTLA4+ expression. These data indicate that naturally protected NOD mice have a unique islet signature and provide new insight into regulatory mechanisms within pancreatic islets.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Diabetes Mellitus Tipo 1/genética , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Cadeias alfa de Integrinas/metabolismo , Camundongos
5.
Diabetes ; 70(2): 529-537, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33122391

RESUMO

Lymph node stromal cells (LNSC) are essential for providing and maintaining peripheral self-tolerance of potentially autoreactive cells. In type 1 diabetes, proinsulin-specific CD8+ T cells, escaping central and peripheral tolerance, contribute to ß-cell destruction. Using G9Cα-/-CD8+ T cells specific for proinsulin, we studied the mechanisms by which LNSC regulate low-avidity autoreactive cells in the NOD mouse model of type 1 diabetes. Whereas MHC-matched NOD-LNSC significantly reduced G9Cα-/-CD8+ T-cell cytotoxicity and dendritic cell-induced proliferation, they failed to sufficiently regulate T cells stimulated by anti-CD3/CD28. In contrast, non-MHC-matched, control C57BL/6 mouse LNSC suppressed T-cell receptor engagement by anti-CD3/CD28 via MHC-independent mechanisms. This C57BL/6-LNSC suppression was maintained even after removal of the LNSC, demonstrating a direct effect of LNSC on T cells, modifying antigen sensitivity and effector function. Thus, our results suggest that a loss of NOD-LNSC MHC-independent suppressive mechanisms may contribute to diabetes development.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Linfonodos/metabolismo , Proinsulina/metabolismo , Células Estromais/metabolismo , Animais , Células Dendríticas/metabolismo , Camundongos , Camundongos Endogâmicos NOD
6.
Methods Mol Biol ; 2128: 87-92, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180187

RESUMO

There are now a number of different mouse models for type 1 diabetes. The best known is the nonobese diabetic (NOD) mouse which has a genetic susceptibility to autoimmune diabetes with some features that are similar to human type 1 diabetes. The mice also have a propensity to other autoimmune diatheses, including autoimmune thyroid disease and sialadenitis. In addition, it is well known that environmental factors affect the incidence of disease in these mice. While there are other rodent models, including numerous transgenic and knockout models, as well as those that express human proteins, none of these develop spontaneous diabetes over a period of time, when the natural history can be studied. We focus here on the unmanipulated NOD mouse and discuss features of the husbandry and investigation of the mice that allow for use of these long-studied mice in the pathogenesis of an autoimmune type of diabetes.


Assuntos
Criação de Animais Domésticos/métodos , Glicemia/análise , Diabetes Mellitus Tipo 1/diagnóstico , Camundongos Endogâmicos NOD , Urinálise/métodos , Idade de Início , Animais , Autoimunidade/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Glicosúria , Masculino , Camundongos
7.
Methods Mol Biol ; 2128: 269-289, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180199

RESUMO

Type 1 diabetes is an autoimmune disease resulting in the loss of insulin production and, consequently, hyperglycemia. The nonobese diabetic (NOD) mouse develops spontaneous diabetes with considerable similarity to the disease in humans. Immunological studies using the NOD mouse model allow for the investigation of the natural history of the disease and leukocyte and lymphocyte pathogenic and regulatory functions, as well as testing potential therapies for intervention. The analyses of the cellular events leading up to diabetes may utilize different in vitro cellular assays, immunohistochemistry, and in vivo adoptive transfer, to study mechanisms of the disease and the effects of therapeutic intervention. In this chapter, we describe some common techniques for phenotyping and mechanistic analyses of function, particularly of CD8+ T cells.


Assuntos
Transferência Adotiva/métodos , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Citometria de Fluxo/métodos , Imuno-Histoquímica/métodos , Animais , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Secções Congeladas/métodos , Imunidade/imunologia , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos Endogâmicos NOD , Inclusão em Parafina/métodos , Coloração e Rotulagem/métodos
8.
Diabetes ; 65(11): 3429-3439, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27495224

RESUMO

Low-avidity autoreactive CD8 T cells (CTLs) escape from thymic negative selection, and peripheral tolerance mechanisms are essential for their regulation. We report the role of proinsulin (PI) expression on the development and activation of insulin-specific CTLs in the NOD mouse model of type 1 diabetes. We studied insulin B-chain-specific CTL from different T-cell receptor transgenic mice (G9Cα-/-) expressing normal PI1 and PI2 or altered PI expression levels. In the absence of PI2 (Ins2-/-), CTL in pancreatic lymph nodes (PLNs) were more activated, and male G9Cα-/- mice developed T1D. Furthermore, when the insulin-specific CTLs developed in transgenic mice lacking their specific PI epitope, the CTLs demonstrated increased cytotoxicity and proliferation in vitro and in vivo in the PLNs after adoptive transfer into NOD recipients. Dendritic cell-stimulated proliferation of insulin-specific T cells was reduced in the presence of lymph node stromal cells (LNSCs) from NOD mice but not from mice lacking the PI epitope. Our study shows that LNSCs regulate CTL activation and suggests that exposure to PI in the periphery is very important in maintenance of tolerance of autoreactive T cells. This is relevant for human type 1 diabetes and has implications for the use of antigen-specific therapy in tolerance induction.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proinsulina/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Epitopos/genética , Feminino , Citometria de Fluxo , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos
9.
Diabetes ; 65(6): 1679-89, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26953160

RESUMO

NOD mice, a model strain for human type 1 diabetes, express proinsulin (PI) in the thymus. However, insulin-reactive T cells escape negative selection, and subsequent activation of the CD8(+) T-cell clonotype G9C8, which recognizes insulin B15-23 via an αß T-cell receptor (TCR) incorporating TRAV8-1/TRAJ9 and TRBV19/TRBJ2-3 gene rearrangements, contributes to the development of diabetes. In this study, we used fixed TRAV8-1/TRAJ9 TCRα-chain transgenic mice to assess the impact of PI isoform expression on the insulin-reactive CD8(+) T-cell repertoire. The key findings were: 1) PI2 deficiency increases the frequency of insulin B15-23-reactive TRBV19(+)CD8(+) T cells and causes diabetes; 2) insulin B15-23-reactive TRBV19(+)CD8(+) T cells are more abundant in the pancreatic lymph nodes of mice lacking PI1 and/or PI2; 3) overexpression of PI2 decreases TRBV19 usage in the global CD8(+) T-cell compartment; 4) a biased repertoire of insulin-reactive CD8(+) T cells emerges in the periphery regardless of antigen exposure; and 5) low-avidity insulin-reactive CD8(+) T cells are less affected by antigen exposure in the thymus than in the periphery. These findings inform our understanding of the diabetogenic process and reveal new avenues for therapeutic exploitation in type 1 diabetes.


Assuntos
Afinidade de Anticorpos , Linfócitos T CD8-Positivos/metabolismo , Insulina/metabolismo , Proinsulina/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia
10.
Methods Mol Biol ; 1433: 127-34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26791361

RESUMO

Tracking autoreactive cells in vivo is important in the study of autoimmune diseases, such as type 1 diabetes. This method provides a model to study the responses of T cells responding to physiologically relevant and organ-specific antigen. Intracellular fluorescent tracers are useful tools to identify adoptively transferred T cells. Firstly, they provide a unique fluorescent signal to distinguish adoptively transferred from endogenous cells. Secondly, cytoplasmic dyes can be used to evaluate proliferation, as the fluorescent intensity is halved with each round of cell division. This provides an important readout to assess cell activation and function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Rastreamento de Células/métodos , Diabetes Mellitus Tipo 1/imunologia , Imunofluorescência/métodos , Ilhotas Pancreáticas/imunologia , Animais , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos
11.
Biochim Biophys Acta ; 1812(11): 1532-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21839170

RESUMO

NADH-cytochrome b5 oxidoreductase (Ncb5or) is an endoplasmic reticulum (ER)-associated redox enzyme involved in fatty acid metabolism, and phenotypic abnormalities of Ncb5or(-/-) mice include diabetes and lipoatrophy. These mice are lean and insulin-sensitive but become hyperglycemic at age 7 weeks as a result of ß-cell dysfunction and loss. Here we examine early cellular and molecular events associated with manifestations of ß-cell defects in Ncb5or(-/-) mice. We observe lower islet ß-cell content in pancreata at age 4 weeks and prominent ER distention in ß-cells by age 5 weeks. Ultrastructural changes progress rapidly in severity from age 5 to 6 weeks, and their frequency rises from 10% of ß-cells at 5 weeks to 33% at 6 weeks. These changes correlate temporally with the onset of diabetes. ER stress responses and lipid load in Ncb5or(-/-) ß-cells were assessed with isolated islets from mice at age 5 weeks. Expression levels of the stress marker protein Grp78/BiP and of phosphorylated eIF2α protein were found to be reduced, although their transcript levels did not decline. This pattern stands in contrast to the canonical unfolded protein response. Ncb5or(-/-) ß-cells also accumulated higher intracellular levels of palmitate and other free fatty acids and exhibited greater reactive oxygen species production than wild-type cells. An alloxan-susceptible genetic background was found to confer accelerated onset of diabetes in Ncb5or(-/-) mice. These findings provide the first direct evidence that manifestations of diabetes in lean Ncb5or(-/-) mice involve saturated free fatty acid overload of ß-cells and ER and oxidative stress responses.


Assuntos
Citocromo-B(5) Redutase/fisiologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Retículo Endoplasmático/patologia , Células Secretoras de Insulina/patologia , Estresse Oxidativo , Aloxano , Animais , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Ácidos Graxos Insaturados/metabolismo , Feminino , Imunofluorescência , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oxirredução , Palmitatos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Resposta a Proteínas não Dobradas
12.
Diabetes ; 60(8): 2144-51, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21715554

RESUMO

OBJECTIVE: The role of reactive oxygen species (ROS) and their dissipation in type 1 diabetes pathogenesis have garnered considerable controversy. Our recent work has demonstrated the importance of NADPH oxidase (NOX) activity for type 1 diabetes development and modulating T-cell autoreactivity. We previously linked decreased monocyte ROS with diabetes resistance in the alloxan-resistant mouse, and NOD-Ncf1(m1J) mice with a genetic ablation of NOX activity had reduced and delayed type 1 diabetes compared with NOD mice. RESEARCH DESIGN AND METHODS: To determine the required cellular sources of ROS that are necessary for type 1 diabetes initiation, we used antibody depletion and adoptive transfer experiments into NOD and NOD-Scid females, respectively. After receiving treatment, female mice were monitored for hyperglycemia and overt diabetes. RESULTS: Depletion of macrophages and neutrophils fully protected NOD mice from type 1 diabetes. However, elimination of neutrophils alone showed no significant reduction or delay. Type 1 diabetes induction in NOD-Scid mice by adoptive transfer with NOD-Ncf1(m1J) splenocytes was significantly delayed compared with NOD splenocytes, suggesting macrophage ROS and modulation of effector responses are critical for diabetes. The adaptive immune response was also altered by the absence of NOX activity, as purified T cells from NOD-Ncf1(m1J) mice exhibited delayed transfer kinetics. Cotransfer experiments demonstrated the defect was intrinsic to NOX-deficient CD8(+) T cells. After stimulation, cytotoxic T cells exhibited decreased effector function in the absence of superoxide production. CONCLUSIONS: These data demonstrate that the impaired autoreactive response of NOX-deficient NOD-Ncf1(m1J) immune system results from an alteration in the antigen-presenting cell-T-cell axis rather than failure of neutrophils to act as effector cells and that ROS signaling is important for the initiation of ß-cell-directed autoimmunity by T cells.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/etiologia , Macrófagos/imunologia , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Transferência Adotiva , Animais , Diabetes Mellitus Tipo 1/imunologia , Feminino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , NADH NADPH Oxirredutases/deficiência , NADPH Oxidase 1 , Neutrófilos/imunologia
13.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21690321

RESUMO

Neutrophils are essential for successful host eradication of bacterial pathogens and for survival to polymicrobial sepsis. During inflammation, the bone marrow provides a large reserve of neutrophils that are released into the peripheral circulation where they traverse to sites of infection. Although neutrophils are essential for survival, few studies have investigated the mechanisms responsible for neutrophil mobilization from the bone marrow during polymicrobial sepsis. Using a cecal ligation and puncture model of polymicrobial sepsis, we demonstrated that neutrophil mobilization from the bone marrow is not dependent on TLR4, MyD88, TRIF, IFNARα/ß, or CXCR2 pathway signaling during sepsis. In contrast, we observed that bone marrow CXCL12 mRNA abundance and specific CXCL12 levels are sharply reduced, whereas splenic CXCR4 mRNA and cell surface expression are increased during sepsis. Blocking CXCL12 activity significantly reduced blood neutrophilia by inhibiting bone marrow release of granulocytes during sepsis. However, CXCL12 inhibition had no impact on the expansion of bone marrow neutrophil precursors and hematopoietic progenitors. Bone marrow neutrophil retention by CXCL12 blockade prevented blood neutrophilia, inhibited peritoneal neutrophil accumulation, allowed significant peritoneal bacterial invasion, and increased polymicrobial sepsis mortality. We concluded that changes in the pattern of CXCL12 signaling during sepsis are essential for neutrophil bone marrow mobilization and host survival but have little impact on bone marrow granulopoiesis.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Quimiocina CXCL12/fisiologia , Infiltração de Neutrófilos/imunologia , Sepse/imunologia , Sepse/microbiologia , Transdução de Sinais/imunologia , Doença Aguda , Animais , Células da Medula Óssea/microbiologia , Quimiocina CXCL12/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mielopoese/genética , Mielopoese/imunologia , Infiltração de Neutrófilos/genética , Sepse/mortalidade , Transdução de Sinais/genética , Análise de Sobrevida
14.
J Immunol ; 186(1): 195-202, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21106855

RESUMO

Sepsis, the systemic inflammatory response to microbial infection, induces changes in both innate and adaptive immunity that presumably lead to increased susceptibility to secondary infections, multiorgan failure, and death. Using a model of murine polymicrobial sepsis whose severity approximates human sepsis, we examined outcomes and defined requirements for survival after secondary Pseudomonas aeruginosa pneumonia or disseminated Listeria monocytogenes infection. We demonstrate that early after sepsis neutrophil numbers and function are decreased, whereas monocyte recruitment through the CCR2/MCP-1 pathway and function are enhanced. Consequently, lethality to Pseudomonas pneumonia is increased early but not late after induction of sepsis. In contrast, lethality to listeriosis, whose eradication is dependent upon monocyte/macrophage phagocytosis, is actually decreased both early and late after sepsis. Adaptive immunity plays little role in these secondary infectious responses. This study demonstrates that sepsis promotes selective early, impaired innate immune responses, primarily in neutrophils, that lead to a pathogen-specific, increased susceptibility to secondary infections.


Assuntos
Bacteriemia/imunologia , Bacteriemia/mortalidade , Imunidade Inata , Sepse/imunologia , Sepse/mortalidade , Animais , Bacteriemia/patologia , Ceco , Modelos Animais de Doenças , Predisposição Genética para Doença , Imunidade Inata/genética , Ligadura , Listeriose/imunologia , Listeriose/mortalidade , Listeriose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/mortalidade , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/mortalidade , Infecções por Pseudomonas/patologia , Punções , Sepse/patologia , Fatores de Tempo
15.
J Immunol ; 185(9): 5247-58, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20881184

RESUMO

Reactive oxygen species are used by the immune system to eliminate infections; however, they may also serve as signaling intermediates to coordinate the efforts of the innate and adaptive immune systems. In this study, we show that by eliminating macrophage and T cell superoxide production through the NADPH oxidase (NOX), T cell polarization was altered. After stimulation with immobilized anti-CD3 and anti-CD28 or priming recall, T cells from NOX-deficient mice exhibited a skewed Th17 phenotype, whereas NOX-intact cells produced cytokines indicative of a Th1 response. These findings were corroborated in vivo by studying two different autoimmune diseases mediated by Th17 or Th1 pathogenic T cell responses. NOX-deficient NOD mice were Th17 prone with a concomitant susceptibility to experimental allergic encephalomyelitis and significant protection against type 1 diabetes. These data validate the role of superoxide in shaping Th responses and as a signaling intermediate to modulate Th17 and Th1 T cell responses.


Assuntos
Autoimunidade/imunologia , Linhagem da Célula/imunologia , NADPH Oxidases/deficiência , Superóxidos/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Animais , Western Blotting , Separação Celular , Citocinas/biossíntese , Citocinas/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , NADPH Oxidases/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/enzimologia , Linfócitos T Auxiliares-Indutores/imunologia
16.
Endocrinol Metab Clin North Am ; 39(3): 541-61, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20723819

RESUMO

In 1922, Leonard Thompson received the first injections of insulin prepared from the pancreas of canine test subjects. From pancreatectomized dogs to the more recent development of animal models that spontaneously develop autoimmune syndromes, animal models have played a meaningful role in furthering diabetes research. Of these animals, the nonobese diabetic (NOD) mouse is the most widely used for research in type 1 diabetes (T1D) because the NOD shares several genetic and immunologic traits with the human form of the disease. In this article, the authors discuss the similarities and differences in NOD and human T1D and the potential role of NOD mice in future preclinical studies, aiming to provide a better understanding of the genetic and immune defects that lead to T1D.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Camundongos Endogâmicos NOD , Animais , Compreensão , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Cães , Estudos de Avaliação como Assunto , Humanos , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos Endogâmicos NOD/imunologia , Camundongos Endogâmicos NOD/fisiologia
17.
Ann N Y Acad Sci ; 1150: 157-66, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19120287

RESUMO

Protection of pancreatic beta cells is an approach to prevent autoimmune type 1 diabetes (T1D) and to protect transplanted islets. Reactive oxygen species (ROS) are important mediators of beta cell death during the development of T1D. We have examined the role of elevated ROS dissipation in the prevention of T1D using the ALR mouse strain. The selection of ALR, for resistance against alloxan-induced free radical-mediated diabetes, led to a strain of mice with an elevated systemic as well as pancreatic ROS dissipation. Independent genetic mapping studies have identified ALR-derived diabetes protective loci. Conplastic and congenic mouse as well as cell line studies have confirmed the genetic mapping and demonstrated that the elevated ROS dissipation protects ALR beta cells from autoimmune destruction. Our data support the hypothesis that elevated ROS dissipation protects beta cells against autoimmune destruction and prevents T1D development.


Assuntos
Citoproteção , Diabetes Mellitus Tipo 1/etiologia , Espécies Reativas de Oxigênio/metabolismo , Aloxano , Animais , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Resistência a Medicamentos/genética , Resistência a Medicamentos/fisiologia , Predisposição Genética para Doença , Genoma Mitocondrial/fisiologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/fisiologia , Camundongos , Camundongos Endogâmicos , Locos de Características Quantitativas , Espécies Reativas de Oxigênio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...