Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 298
Filtrar
1.
medRxiv ; 2023 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-36993739

RESUMO

In autoimmune Type 1 diabetes (T1D), immune cells progressively infiltrate and destroy the islets of Langerhans - islands of endocrine tissue dispersed throughout the pancreas. However, it is unclear how this process, called 'insulitis', develops and progresses within this organ. Here, using highly multiplexed CO-Detection by indEXing (CODEX) tissue imaging and cadaveric pancreas samples from pre-T1D, T1D, and non-T1D donors, we examine pseudotemporal-spatial patterns of insulitis and exocrine inflammation within large pancreatic tissue sections. We identify four sub-states of insulitis characterized by CD8 + T cells at different stages of activation. We further find that exocrine compartments of pancreatic lobules affected by insulitis have distinct cellularity, suggesting that extra-islet factors may make particular lobules permissive to disease. Finally, we identify "staging areas" - immature tertiary lymphoid structures away from islets where CD8 + T cells appear to assemble before they navigate to islets. Together, these data implicate the extra-islet pancreas in autoimmune insulitis, greatly expanding the boundaries of T1D pathogenesis.

2.
Am J Transplant ; 17(11): 2945-2954, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28675676

RESUMO

Early subclinical inflammation in kidney transplants is associated with later graft fibrosis and dysfunction. Regulatory T cells (Tregs) can reverse established inflammation in animal models. We conducted a pilot safety and feasibility trial of autologous Treg cell therapy in three kidney transplant recipients with subclinical inflammation noted on 6-month surveillance biopsies. Tregs were purified from peripheral blood and polyclonally expanded ex vivo using medium containing deuterated glucose to label the cells. All patients received a single infusion of ~320 × 106 (319, 321, and 363.8 × 106 ) expanded Tregs. Persistence of the infused Tregs was tracked. Graft inflammation was monitored with follow-up biopsies and urinary biomarkers. Nearly 1 × 109 (0.932, 0.956, 1.565 × 109 ) Tregs were successfully manufactured for each patient. There were no infusion reactions or serious therapy-related adverse events. The infused cells demonstrated patterns of persistence and stability similar to those observed in non-immunosuppressed subjects receiving the same dose of Tregs. Isolation and expansion of Tregs is feasible in kidney transplant patients on immunosuppression. Infusion of these cells was safe and well tolerated. Future trials will test the efficacy of polyclonal and donor alloantigen-reactive Tregs for the treatment of inflammation in kidney transplants.


Assuntos
Rejeição de Enxerto/terapia , Inflamação/terapia , Falência Renal Crônica/cirurgia , Transplante de Rim/efeitos adversos , Linfócitos T Reguladores/imunologia , Adolescente , Adulto , Idoso , Feminino , Seguimentos , Taxa de Filtração Glomerular , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto , Humanos , Inflamação/etiologia , Inflamação/patologia , Isoantígenos , Testes de Função Renal , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Complicações Pós-Operatórias , Prognóstico , Fatores de Risco , Doadores de Tecidos , Adulto Jovem
3.
Am J Transplant ; 13(11): 3010-20, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24102808

RESUMO

Regulatory T cell (Treg) therapy has the potential to induce transplantation tolerance so that immunosuppression and associated morbidity can be minimized. Alloantigen-reactive Tregs (arTregs) are more effective at preventing graft rejection than polyclonally expanded Tregs (PolyTregs) in murine models. We have developed a manufacturing process to expand human arTregs in short-term cultures using good manufacturing practice-compliant reagents. This process uses CD40L-activated allogeneic B cells to selectively expand arTregs followed by polyclonal restimulation to increase yield. Tregs expanded 100- to 1600-fold were highly alloantigen reactive and expressed the phenotype of stable Tregs. The alloantigen-expanded Tregs had a diverse TCR repertoire. They were more potent than PolyTregs in vitro and more effective at controlling allograft injuries in vivo in a humanized mouse model.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica/imunologia , Isoantígenos/imunologia , Transplante de Pele , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Animais , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Tolerância ao Transplante
4.
Diabetologia ; 56(2): 391-400, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23086558

RESUMO

AIMS/HYPOTHESIS: Type 1 diabetes results from a chronic autoimmune process continuing for years after presentation. We tested whether treatment with teplizumab (a Fc receptor non-binding anti-CD3 monoclonal antibody), after the new-onset period, affects the decline in C-peptide production in individuals with type 1 diabetes. METHODS: In a randomised placebo-controlled trial we treated 58 participants with type 1 diabetes for 4-12 months with teplizumab or placebo at four academic centres in the USA. A central randomisation centre used computer generated tables to allocate treatments. Investigators, patients, and caregivers were blinded to group assignment. The primary outcome was a comparison of C-peptide responses to a mixed meal after 1 year. We explored modification of treatment effects in subgroups of patients. RESULTS: Thirty-four and 29 subjects were randomized to the drug and placebo treated groups, respectively. Thirty-one and 27, respectively, were analysed. Although the primary outcome analysis showed a 21.7% higher C-peptide response in the teplizumab-treated group (0.45 vs 0.371; difference, 0.059 [95% CI 0.006, 0.115] nmol/l) (p = 0.03), when corrected for baseline imbalances in HbA(1c) levels, the C-peptide levels in the teplizumab-treated group were 17.7% higher (0.44 vs 0.378; difference, 0.049 [95% CI 0, 0.108] nmol/l, p = 0.09). A greater proportion of placebo-treated participants lost detectable C-peptide responses at 12 months (p = 0.03). The teplizumab group required less exogenous insulin (p < 0.001) but treatment differences in HbA(1c) levels were not observed. Teplizumab was well tolerated. A subgroup analysis showed that treatment benefits were larger in younger individuals and those with HbA(1c) <6.5% at entry. Clinical responders to teplizumab had an increase in circulating CD8 central memory cells 2 months after enrolment compared with non-responders. CONCLUSIONS/INTERPRETATIONS: This study suggests that deterioration in insulin secretion may be affected by immune therapy with teplizumab after the new-onset period but the magnitude of the effect is less than during the new-onset period. Our studies identify characteristics of patients most likely to respond to this immune therapy. TRIAL REGISTRATION: ClinicalTrials.gov NCT00378508 FUNDING: This work was supported by grants 2007-502, 2007-1059 and 2006-351 from the JDRF and grants R01 DK057846, P30 DK20495, UL1 RR024139, UL1RR025780, UL1 RR024131 and UL1 RR024134 from the NIH.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Peptídeo C/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Adolescente , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Insulina/metabolismo , Masculino
5.
Am J Transplant ; 10(8): 1870-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20659093

RESUMO

The applicability of islet transplantation as treatment for type 1 diabetes is limited by renal and islet toxicities of currently available immunosuppressants. We describe a novel immunosuppressive regimen using the antileukocyte functional antigen-1 antibody efalizumab which permits long-term islet allograft survival while reducing the need for corticosteroids and calcineurin inhibitors (CNI). Eight patients with type 1 diabetes and hypoglycemic unawareness received intraportal allogeneic islet transplants. Immunosuppression consisted of antithymocyte globulin induction followed by maintenance with efalizumab and sirolimus or mycophenolate. When efalizumab was withdrawn from the market in mid 2009, all patients were transitioned to regimens consisting of mycophenolate and sirolimus or mycophenolate and tacrolimus. All patients achieved insulin independence and four out of eight patients became independent after single-islet transplants. Insulin independent patients had no further hypoglycemic events, hemoglobin A1c levels decreased and renal function remained stable. Efalizumab was well tolerated and no serious adverse events were encountered. Although long-term follow-up is limited by discontinuation of efalizumab and transition to conventional imunnosuppression (including CNI in four cases), these results demonstrate that insulin independence after islet transplantation can be achieved with a CNI and steroid-free regimen. Such an approach may minimize renal and islet toxicity and thus further improve long-term islet allograft survival.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Antígeno-1 Associado à Função Linfocitária/administração & dosagem , Adolescente , Adulto , Anticorpos Monoclonais Humanizados , Soro Antilinfocitário/uso terapêutico , Glicemia/metabolismo , Feminino , Humanos , Terapia de Imunossupressão/métodos , Masculino , Pessoa de Meia-Idade , Ácido Micofenólico/uso terapêutico , Sirolimo/uso terapêutico , Tacrolimo/administração & dosagem
6.
Clin Exp Immunol ; 161(2): 250-67, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20491795

RESUMO

Type 1 diabetes is an autoimmune disease whose clinical onset signifies a lifelong requirement for insulin therapy and increased risk of medical complications. To increase the efficiency and confidence with which drug candidates advance to human type 1 diabetes clinical trials, we have generated and validated a mathematical model of type 1 diabetes pathophysiology in a well-characterized animal model of spontaneous type 1 diabetes, the non-obese diabetic (NOD) mouse. The model is based on an extensive survey of the public literature and input from an independent scientific advisory board. It reproduces key disease features including activation and expansion of autoreactive lymphocytes in the pancreatic lymph nodes (PLNs), islet infiltration and beta cell loss leading to hyperglycaemia. The model uses ordinary differential and algebraic equations to represent the pancreas and PLN as well as dynamic interactions of multiple cell types (e.g. dendritic cells, macrophages, CD4+ T lymphocytes, CD8+ T lymphocytes, regulatory T cells, beta cells). The simulated features of untreated pathogenesis and disease outcomes for multiple interventions compare favourably with published experimental data. Thus, a mathematical model reproducing type 1 diabetes pathophysiology in the NOD mouse, validated based on accurate reproduction of results from multiple published interventions, is available for in silico hypothesis testing. Predictive biosimulation research evaluating therapeutic strategies and underlying biological mechanisms is intended to deprioritize hypotheses that impact disease outcome weakly and focus experimental research on hypotheses likely to provide insight into the disease and its treatment.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/fisiopatologia , Modelos Biológicos , Algoritmos , Animais , Simulação por Computador , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Modelos Imunológicos , Pâncreas/imunologia , Pâncreas/fisiopatologia
7.
Gut ; 58(10): 1363-73, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19505881

RESUMO

BACKGROUND AND AIMS: Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) has been shown to act as a negative regulator of T cell function and has been implicated in the regulation of T helper 1 (Th1)/Th2 development and the function of regulatory T cells. Tests were carried out to determine whether anti-CTLA-4 treatment would alter the polarisation of naive T cells in vivo. METHODS: Mice were treated with anti-CTLA-4 monoclonal antibody (mAb) (UC10-4F10) at the time of immunisation or colonic instillation of trinitrobenzene sulfonic acid (TNBS). The cytokines produced by lymph node cells after in vitro antigenic stimulation and the role of indoleamine 2,3 dioxygenase (IDO) and of interleukin-10 (IL-10) were tested, and the survival of mice was monitored. RESULTS: Injection of anti-CTLA-4 mAb in mice during priming induced the development of adaptive CD4(+) regulatory T cells which expressed high levels of ICOS (inducible co-stimulator), secreted IL-4 and IL-10. This treatment inhibited Th1 memory responses in vivo and repressed experimental intestinal inflammation. The anti-CTLA-4-induced amelioration of disease correlated with IDO expression and infiltration of ICOS(high) Foxp3(+) T cells in the intestine, suggesting that anti-CTLA-4 acted indirectly through the development of regulatory T cells producing IL-10 and inducing IDO. CONCLUSIONS: These observations emphasise the synergy between IL-10 and IDO as anti-inflammatory agents and highlight anti-CTLA-4 treatment as a potential novel immunotherapeutic approach for inducing adaptive regulatory T cells.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD/imunologia , Colite/tratamento farmacológico , Interleucina-10/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Antígenos CD/metabolismo , Antígeno CTLA-4 , Colite/induzido quimicamente , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/efeitos dos fármacos , Ácido Trinitrobenzenossulfônico
8.
Am J Transplant ; 8(10): 2086-96, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18828769

RESUMO

Regulatory T cells (Treg) are critical regulators of immune tolerance. Both IL-2 and CD28-CD80/CD86 signaling are critical for CD4(+)CD25(+)FOXP3(+) Treg survival in mice. Yet, both belatacept (a second-generation CTLA-4Ig) and basiliximab (an anti-CD25 monoclonal antibody) are among the arsenal of current immunotherapies being used in kidney transplant patients. In this study, we explored the direct effect of basiliximab and belatacept on the Tregs in peripheral blood both in the short term and long term and in kidney biopsies of patients with acute rejection. We report that the combined belatacept/basiliximab therapy has no long-term effect on circulating Tregs when compared to a calcineurin inhibitor (CNI)-treated group. Moreover, belatacept-treated patients had a significantly greater number of FOXP3(+) T cells in graft biopsies during acute rejection as compared to CNI-treated patients. Finally, it appears that the basiliximab caused a transient loss of both FOXP3(+) and FOXP3(-) CD25(+) T cells in the circulation in both treatment groups raising important questions about the use of this therapy in tolerance promoting therapeutic protocols.


Assuntos
Transplante de Rim/métodos , Receptores de Interleucina-2/antagonistas & inibidores , Linfócitos T Reguladores/metabolismo , Abatacepte , Adulto , Anticorpos Monoclonais/administração & dosagem , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Basiliximab , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Inibidores de Calcineurina , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunoconjugados/administração & dosagem , Imunossupressores/administração & dosagem , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Masculino , Pessoa de Meia-Idade , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem
9.
Am J Transplant ; 7(6): 1457-63, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17511675

RESUMO

The involvement of CD4(+)CD25(+) regulatory T cells (Treg) in general immune homeostasis and protection from autoimmune syndromes is now well established. Similarly, there has been increasing evidence for Treg involvement in allograft rejection and current immunotherapies. However, despite significant advances in understanding the development, function, and therapeutic efficacy of Treg in certain well-defined rodent models, the relevance of Treg to clinical transplantation remains unclear. In this review, we summarize our current understanding of the role of Treg in immunity and organ transplantation in experimental and clinical settings. In addition, we review advances in using Treg as a form of immune therapy. The goal is to highlight the complexities and opportunities in the field and to provide evidence to support the use of antigen-specific Tregs in the context of transplantation to facilitate a robust and selective state of immune tolerance.


Assuntos
Antígenos CD4/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Linfócitos T Reguladores/imunologia , Imunologia de Transplantes , Antígenos CD/imunologia , Autoantígenos/imunologia , Fatores de Transcrição Forkhead/imunologia , Humanos
10.
Clin Immunol ; 115(1): 3-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15870014

RESUMO

Regulatory T cells (Tregs) have been shown to be critical in the balance between autoimmunity and tolerance and have been implicated in several human autoimmune diseases. However, the small number of Tregs in peripheral blood limits their therapeutic potential. Therefore, we developed a protocol that would allow for the expansion of Tregs while retaining their suppressive activity. We isolated CD4+CD25 hi cells from human peripheral blood and expanded them in vitro in the presence of anti-CD3 and anti-CD28 magnetic Xcyte Dynabeads and high concentrations of exogenous Interleukin (IL)-2. Tregs were effectively expanded up to 200-fold while maintaining surface expression of CD25 and other markers of Tregs: CD62L, HLA-DR, CCR6, and FOXP3. The expanded Tregs suppressed proliferation and cytokine secretion of responder PBMCs in co-cultures stimulated with anti-CD3 or alloantigen. Treg expansion is a critical first step before consideration of Tregs as a therapeutic intervention in patients with autoimmune or graft-versus-host disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Receptores de Interleucina-2/imunologia , Doenças Autoimunes/terapia , Linfócitos T CD4-Positivos/citologia , Técnicas de Cultura de Células , Citometria de Fluxo , Fatores de Transcrição Forkhead , Antígenos HLA-DR/imunologia , Humanos , Separação Imunomagnética , Imunofenotipagem , Imunoterapia/métodos , Interleucina-2/imunologia , Selectina L/imunologia , Ativação Linfocitária/imunologia , Receptores CCR6 , Receptores de Quimiocinas/imunologia , Proteínas Repressoras
11.
Immunol Rev ; 182: 149-63, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11722631

RESUMO

Regulation of the immune response to self-antigens is a complex process that depends on maintaining self-tolerance while retaining the capacity to mount a robust immune response to foreign antigens. Autoreactive T cells specific for these autoantigens are present in most normal individuals but are kept under control by multiple diverse peripheral tolerance mechanisms. In the last few years, there has been a re-emergence of suppressor cells as among the most central of these regulatory mechanisms. These cells, which express CD4, CD25, and CD62L, develop in the thymus and survive in a CD28-dependent manner in the periphery to maintain the homeostatic equilibrium of immunity and tolerance. In this review, we will summarize studies of these regulatory cells as they relate to autoimmune diseases and more specifically to type 1 diabetes and attempt to address some of the many outstanding questions. Finally, evidence is provided to support the ability of anti-CD3 mAbs to stimulate the regulatory T cells and reset the rheostat of immune tolerance in an animal model of autoimmune diabetes, the NOD mouse.


Assuntos
Autoimunidade/imunologia , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Linfócitos T Reguladores/imunologia , Animais , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/terapia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Células Th1/metabolismo , Células Th2/metabolismo
12.
J Exp Med ; 194(5): 677-84, 2001 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-11535635

RESUMO

An increasing number of studies have documented the central role of T cell costimulation in autoimmunity. Here we show that the autoimmune diabetes-prone nonobese diabetic (NOD) mouse strain, deficient in B7-2 costimulation, is protected from diabetes but develops a spontaneous autoimmune peripheral polyneuropathy. All the female and one third of the male mice exhibited limb paralysis with histologic and electrophysiologic evidence of severe demyelination in the peripheral nerves beginning at 20 wk of age. No central nervous system lesions were apparent. The peripheral nerve tissue was infiltrated with dendritic cells, CD4(+), and CD8(+) T cells. Finally, CD4(+) T cells isolated from affected animals induced the disease in NOD.SCID mice. Thus, the B7-2-deficient NOD mouse constitutes the first model of a spontaneous autoimmune disease of the peripheral nervous system, which has many similarities to the human disease, chronic inflammatory demyelinating polyneuropathy (CIDP). This model demonstrates that NOD mice have "cryptic" autoimmune defects that can polarize toward the nervous tissue after the selective disruption of CD28/B7-2 costimulatory pathway.


Assuntos
Antígenos CD/imunologia , Glicoproteínas de Membrana/imunologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doenças do Sistema Nervoso Periférico/imunologia , Linfócitos T/imunologia , Envelhecimento , Animais , Antígenos CD/genética , Antígeno B7-2 , Encéfalo/imunologia , Encéfalo/patologia , Cruzamentos Genéticos , Gânglios Espinais/imunologia , Gânglios Espinais/patologia , Inflamação , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Doença Autoimune do Sistema Nervoso Experimental/genética , Doença Autoimune do Sistema Nervoso Experimental/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/patologia , Nós Neurofibrosos/imunologia , Nós Neurofibrosos/patologia , Nervo Isquiático/imunologia , Nervo Isquiático/patologia
13.
J Clin Invest ; 108(6): 895-903, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11560959

RESUMO

Primary T cell proliferative responses to TCR ligation plus CD28 costimulation are surprisingly heterogeneous. Many cells that enter G1 fail to progress further through the cell cycle, and some of these cells subsequently fail to divide upon restimulation, even in the presence of IL-2. Such IL-2-refractory anergy is distinct from the IL-2-reversible anergy induced by TCR occupancy in the absence of CD28 costimulation. Here, we focus on the contributions of cell cycle progression and costimulatory (CD28/CTLA-4) signals in the regulation of anergy. We show that CD28 costimulation is not sufficient for anergy avoidance and that activated T cells must progress through the cell cycle in order to escape anergy. Induction of this "division-arrest" form of anergy requires CTLA-4 signaling during the primary response. Also, cell division per se is not sufficient for anergy avoidance: the few T cells that undergo multiple rounds of cell division during overt CD28 costimulatory blockade do not escape the ultimate induction of clonal anergy. Anergy avoidance by primary T cells is thus a multistep process: in order to participate in a productive immune response, an individual T cell activated through its antigen receptor must receive CD28 costimulation and progress through the cell cycle. Anergy may be induced either through a combination of CTLA-4 signaling and the failure of cell cycle progression, or through a proliferation-independent mechanism in which TCR ligation occurs in the absence of CD28.


Assuntos
Antígenos de Diferenciação/imunologia , Antígenos CD28/imunologia , Anergia Clonal , Imunoconjugados , Linfócitos T/imunologia , Proteínas Supressoras de Tumor , Abatacepte , Animais , Antígenos CD , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Antígeno CTLA-4 , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Feminino , Humanos , Técnicas In Vitro , Interleucina-2/farmacologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais , Linfócitos T/citologia
14.
Arthritis Rheum ; 44(8): 1730-5, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11508422

RESUMO

The development of effective, new, biologically based therapies for RA has created real excitement and justifiable optimism in recent years among rheumatologists and among patients with rheumatic diseases. Recent advances in our understanding of the mechanisms of immune activation and immune tolerance provide further cause for optimism. Against this background, the establishment of the ITN is an important step. However, significant hurdles remain to be cleared. First, despite dramatic scientific progress, restoration of immune tolerance in the face of an established autoimmune response is still an elusive goal, even in the laboratory. Not only does the ITN face this fundamental scientific challenge, but it also faces daunting practical and political challenges. For example, can the ITN influence the research agenda of the pharmaceutical and biotechnology industries? This question and other important questions will only be answered as the ITN matures. Autoimmune disease, although individually uncommon, affects more than 2% of Americans. The rheumatologist is especially aware of the devastating potential of autoimmune diseases. If the ITN succeeds in linking basic research into the mechanisms of autoimmunity with clinical trials of promising new therapies, it can be expected to play a critical role in advancing the practice of clinical rheumatology.


Assuntos
Doenças Autoimunes/imunologia , Doenças Reumáticas/imunologia , Tolerância a Antígenos Próprios , Animais , Doenças Autoimunes/terapia , Autoimunidade , Humanos , Terapia de Imunossupressão , Ativação Linfocitária , Modelos Biológicos , Doenças Reumáticas/terapia
15.
J Immunol ; 167(4): 1996-2003, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11489981

RESUMO

A novel costimulatory molecule expressed on activated T cells, inducible costimulator (ICOS), and its ligand, B7-related protein-1 (B7RP-1), were recently identified. ICOS costimulation leads to the induction of Th2 cytokines without augmentation of IL-2 production, suggesting a role for ICOS in Th2 cell differentiation and expansion. In the present study, a soluble form of murine ICOS, ICOS-Ig, was used to block ICOS/B7RP-1 interactions in a Th2 model of allergic airway disease. In this model, mice are sensitized with inactivated Schistosoma mansoni eggs and are subsequently challenged with soluble S. mansoni egg Ag directly in the airways. Treatment of C57BL/6 mice with ICOS-Ig during sensitization and challenge attenuated airway inflammation, as demonstrated by a decrease in cellular infiltration into the lung tissue and airways, as well as by a decrease in local IL-5 production. These inhibitory effects were not due to a lack of T cell priming nor to a defect in Th2 differentiation. In addition, blockade of ICOS/B7RP-1 interactions during ex vivo restimulation of lung Th2 effector cells prevented cytokine production. Thus, blockade of ICOS signaling can significantly reduce airway inflammation without affecting Th2 differentiation in this model of allergic airway disease.


Assuntos
Antígenos de Diferenciação de Linfócitos T/fisiologia , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Células Th2/citologia , Células Th2/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Helmintos/administração & dosagem , Antígeno B7-1/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/biossíntese , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Imunoglobulina E/sangue , Imunossupressores/administração & dosagem , Ligante Coestimulador de Linfócitos T Induzíveis , Proteína Coestimuladora de Linfócitos T Induzíveis , Inflamação/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Schistosoma mansoni/imunologia , Células Th2/metabolismo
16.
J Immunol ; 167(5): 2555-60, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11509595

RESUMO

Ag-specific immune tolerance in clinical organ transplantation is currently an unrealized but critical goal of transplant biology. The specificity and avidity of multimerized MHC-peptide complexes suggests their potential ability to modulate T cell sensitization and effector functions. In this study, we examined the ability of MHC-peptide dimers to modulate T cell function both in vitro and in vivo. Soluble MHC dimers induced modulation of surface TCR expression and inhibited T cell cytolytic activity at nanomolar concentrations in vitro. Furthermore, engagement of TCR by soluble dimers resulted in phosphorylation of the TCR zeta-chain and recruitment and phosphorylation of zeta-associated protein-70 to the signaling complex, the latter of which increased upon dimer cross-linking. Significantly, Ag-specific inhibition of an alloreactive TCR-transgenic T cell population in vivo resulted in consequent outgrowth of an allogeneic tumor. The prolonged Ag-specific suppression of expansion and/or effector function of cognate T cells in vivo suggests that soluble MHC dimers may be a means of inducing sustained Ag-specific T cell unresponsiveness in vivo.


Assuntos
Antígenos H-2/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dimerização , Antígenos H-2/química , Humanos , Tolerância Imunológica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Proteínas Nucleares , Proteínas Tirosina Quinases/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/metabolismo , Imunologia de Transplantes , Proteína-Tirosina Quinase ZAP-70
18.
Arterioscler Thromb Vasc Biol ; 21(6): 1011-6, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11397712

RESUMO

To determine whether T cells and B cells influence lipid metabolism and atherosclerosis, we crossed apolipoprotein E-deficient (apoE degrees ) mice with recombination activating gene 2-deficient (RAG2 degrees ) mice. Total plasma cholesterol levels were approximately 20% higher in male apoE degrees mice compared with the apoE degrees RAG2 degrees mice at 8 weeks of age, and plasma triglyceride levels were 2.5-fold higher in the apoE degrees mice even when plasma cholesterol levels were similar. Male mice with plasma cholesterol levels between 400 and 600 mg/dL at 8 weeks of age were euthanized at 27 and 40 weeks of age. The aortic root lesion area in the apoE degrees RAG2 degrees mice, compared with that in the immune-competent apoE degrees mice, was 81% and 57% smaller at 27 and 40 weeks of age, respectively. In contrast, there was no difference in the size of the brachiocephalic trunk lesions. Similar results were obtained with mice euthanized at 40 weeks of age that had 8-week cholesterol levels between 300 and 399 mg/dL. In apoE degrees RAG2 degrees mice, aortic root atherosclerosis was more profoundly suppressed at lower cholesterol levels. Thus, T and B cells and their products differentially influence the development of atherosclerosis at different sites. We also demonstrate a profound effect of the immune system on plasma lipid homeostasis.


Assuntos
Apolipoproteínas E/genética , Arteriosclerose/imunologia , Hospedeiro Imunocomprometido , Lipoproteínas/sangue , Animais , Aorta/patologia , Arteriosclerose/sangue , Arteriosclerose/patologia , Tronco Braquiocefálico/patologia , Proteínas de Ligação a DNA/genética , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
J Exp Med ; 193(11): 1327-32, 2001 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-11390440

RESUMO

One striking feature of spontaneous autoimmune diabetes is the prototypic formation of lymphoid follicular structures within the pancreas. Lymphotoxin (LT) has been shown to play an important role in the formation of lymphoid follicles in the spleen. To explore the potential role of LT-mediated microenvironment in the pathogenesis of insulin-dependent diabetes mellitus (IDDM), an LTbeta receptor-immunoglobulin fusion protein (LTbetaR-Ig) was administered to nonobese diabetic mice. Early treatment with LTbetaR-Ig prevented insulitis and IDDM, suggesting that LT plays a critical role in the insulitis development. LTbetaR-Ig treatment at a late stage of the disease also dramatically reversed insulitis and prevented diabetes. Moreover, LTbetaR-Ig treatment prevented the development of IDDM by diabetogenic T cells in an adoptive transfer model. Thus, LTbetaR-Ig can disassemble the well established lymphoid microenvironment in the islets, which is required for the development and progression of IDDM.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/patologia , Receptores do Fator de Necrose Tumoral/fisiologia , Animais , Moléculas de Adesão Celular , Feminino , Imunoglobulinas/fisiologia , Receptor beta de Linfotoxina , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Mucoproteínas/fisiologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia
20.
J Immunol ; 166(11): 6491-9, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11359799

RESUMO

We have previously shown that exposure of P815 tumor cells to melphalan (L-phenylalanine mustard; L-PAM) leads to up-regulation of B7-1 surface expression, and this L-PAM-induced up-regulation requires de novo RNA synthesis and is associated with accumulation of B7-1 mRNA. Here we show that the effect of L-PAM on B7-1 surface expression can be mimicked by exposing P815 tumor cells to oxidative stress but not to heat shock. Moreover, the antioxidant N-acetyl-L-cysteine prevented the L-PAM-induced accumulation of B7-1 mRNA in P815 tumor cells, suggesting that reactive oxygen species are involved in the transcriptional regulation of L-PAM-induced B7-1 gene expression. Although AP-1 and NF-kappaB are regarded as redox-sensitive transcription factors and the promoter/enhancer region of the B7-1 gene contains an AP-1 and an NF-kappaB binding site, exposure of P815 tumor cells to L-PAM led to rapid and transient activation only of NF-kappaB, but not AP-1, that bound specifically to a probe containing the respective binding site in the murine or human B7-1 gene. Moreover, exposure of P815 tumor cells to a cell-permeable peptide that selectively inhibits NF-kappaB activation by blocking the activation of the IkappaB-kinase complex was found to inhibit the L-PAM-induced B7-1 mRNA accumulation, indicating that NF-kappaB activation is essential for the L-PAM-induced B7-1 gene expression. Taken together, these results indicate that L-PAM leads to activation of B7-1 gene expression by activating NF-kappaB via a pathway that involves reactive oxygen species.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Sarcoma de Mastócitos/imunologia , Melfalan/farmacologia , Acetilcisteína/farmacologia , Sequência de Aminoácidos , Animais , Antígenos CD/biossíntese , Antineoplásicos Alquilantes/antagonistas & inibidores , Antioxidantes/farmacologia , Antígeno B7-2 , Ligação Competitiva , Membrana Celular/imunologia , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Núcleo Celular/química , Elementos Facilitadores Genéticos/efeitos dos fármacos , Elementos Facilitadores Genéticos/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Temperatura Alta , Humanos , Peróxido de Hidrogênio/farmacologia , Quinase I-kappa B , Substâncias Macromoleculares , Sarcoma de Mastócitos/química , Sarcoma de Mastócitos/genética , Sarcoma de Mastócitos/metabolismo , Melfalan/antagonistas & inibidores , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Família Multigênica/imunologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Sondas de Oligonucleotídeos/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/farmacologia , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...