Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(21): 5432-5447.e16, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34619077

RESUMO

Understanding vaccine-elicited protection against SARS-CoV-2 variants and other sarbecoviruses is key for guiding public health policies. We show that a clinical stage multivalent SARS-CoV-2 spike receptor-binding domain nanoparticle (RBD-NP) vaccine protects mice from SARS-CoV-2 challenge after a single immunization, indicating a potential dose-sparing strategy. We benchmarked serum neutralizing activity elicited by RBD-NPs in non-human primates against a lead prefusion-stabilized SARS-CoV-2 spike (HexaPro) using a panel of circulating mutants. Polyclonal antibodies elicited by both vaccines are similarly resilient to many RBD residue substitutions tested, although mutations at and surrounding position 484 have negative consequences for neutralization. Mosaic and cocktail nanoparticle immunogens displaying multiple sarbecovirus RBDs elicit broad neutralizing activity in mice and protect mice against SARS-CoV challenge even in the absence of SARS-CoV RBD in the vaccine. This study provides proof of principle that multivalent sarbecovirus RBD-NPs induce heterotypic protection and motivates advancing such broadly protective sarbecovirus vaccines to the clinic.

2.
FASEB J ; 33(3): 3137-3151, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30383447

RESUMO

Immunodeficient mice engrafted with human peripheral blood mononuclear cells (PBMCs) support preclinical studies of human pathogens, allograft rejection, and human T-cell function. However, a major limitation of PBMC engraftment is development of acute xenogeneic graft- versus-host disease (GVHD) due to human T-cell recognition of murine major histocompatibility complex (MHC). To address this, we created 2 NOD- scid IL-2 receptor subunit γ ( IL2rg) null (NSG) strains that lack murine MHC class I and II [NSG-ß-2-microglobulin ( B2M) null ( IA IE)null and NSG -( Kb Db) null ( IAnull)]. We observed rapid human IgG clearance in NSG- B2Mnull ( IA IE) null mice whereas clearance in NSG -( Kb Db) null ( IAnull) mice and NSG mice was comparable. Injection of human PBMCs into both strains enabled long-term engraftment of human CD4+ and CD8+ T cells without acute GVHD. Engrafted human T-cell function was documented by rejection of human islet allografts. Administration of human IL-2 to NSG -( Kb Db) null ( IAnull) mice via adeno-associated virus vector increased human CD45+ cell engraftment, including an increase in human regulatory T cells. However, high IL-2 levels also induced the development of GVHD. These data document that NSG mice deficient in murine MHC support studies of human immunity in the absence of acute GVHD and enable evaluation of human antibody therapeutics targeting human T cells.-Brehm, M. A., Kenney, L. L., Wiles, M. V., Low, B. E., Tisch, R. M., Burzenski, L., Mueller, C., Greiner, D. L., Shultz, L. D. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Linfócitos T/imunologia , Animais , Feminino , Genes MHC Classe I , Genes MHC da Classe II , Sobrevivência de Enxerto/imunologia , Xenoenxertos , Humanos , Transplante das Ilhotas Pancreáticas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Fenótipo
3.
J Immunol ; 198(1): 249-256, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27895179

RESUMO

Thymic dendritic cells (DC) delete self-antigen-specific thymocytes, and drive development of Foxp3-expressing immunoregulatory T cells. Unlike medullary thymic epithelial cells, which express and present peripheral self-antigen, DC must acquire self-antigen to mediate thymic negative selection. One such mechanism entails the transfer of surface MHC-self peptide complexes from medullary thymic epithelial cells to thymic DC. Despite the importance of thymic DC cross-dressing in negative selection, the factors that regulate the process and the capacity of different thymic DC subsets to acquire MHC and stimulate thymocytes are poorly understood. In this study intercellular MHC transfer by thymic DC subsets was investigated using an MHC-mismatch-based in vitro system. Thymic conventional DC (cDC) subsets signal regulatory protein α (SIRPα+) and CD8α+ readily acquired MHC class I and II from thymic epithelial cells but plasmacytoid DC were less efficient. Intercellular MHC transfer was donor-cell specific; thymic DC readily acquired MHC from TEC plus thymic or splenic DC, whereas thymic or splenic B cells were poor donors. Furthermore DC origin influenced cross-dressing; thymic versus splenic DC exhibited an increased capacity to capture TEC-derived MHC, which correlated with direct expression of EpCAM by DC. Despite similar capacities to acquire MHC-peptide complexes, thymic CD8α+ cDC elicited increased T cell stimulation relative to SIRPα+ cDC. DC cross-dressing was cell-contact dependent and unaffected by lipid raft disruption of donor TEC. Furthermore, blocking PI3K signaling reduced MHC acquisition by thymic CD8α+ cDC and plasmacytoid DC but not SIRPα+ cDC. These findings demonstrate that multiple parameters influence the efficiency of and distinct mechanisms drive intercellular MHC transfer by thymic DC subsets.


Assuntos
Células Dendríticas/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Tolerância a Antígenos Próprios/imunologia , Timo/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Separação Celular , Células Epiteliais/imunologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
4.
J Immunol ; 198(3): 1263-1273, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031339

RESUMO

Infection with Staphylococcus aureus does not induce long-lived protective immunity for reasons that are not completely understood. Human and murine vaccine studies support a role for Abs in protecting against recurring infections, but S. aureus modulates the B cell response through expression of staphylococcus protein A (SpA), a surface protein that drives polyclonal B cell expansion and induces cell death in the absence of costimulation. In this murine study, we show that SpA altered the fate of plasmablasts and plasma cells (PCs) by enhancing the short-lived extrafollicular response and reducing the pool of bone marrow (BM)-resident long-lived PCs. The absence of long-lived PCs was associated with a rapid decline in Ag-specific class-switched Ab. In contrast, when previously inoculated mice were challenged with an isogenic SpA-deficient S. aureus mutant, cells proliferated in the BM survival niches and sustained long-term Ab titers. The effects of SpA on PC fate were limited to the secondary response, because Ab levels and the formation of B cell memory occurred normally during the primary response in mice inoculated with wild-type or SpA-deficient S. aureus mutant. Thus, failure to establish long-term protective Ab titers against S. aureus was not a consequence of diminished formation of B cell memory; instead, SpA reduced the proliferative capacity of PCs that entered the BM, diminishing the number of cells in the long-lived pool.


Assuntos
Plasmócitos/efeitos dos fármacos , Proteína Estafilocócica A/farmacologia , Animais , Células Produtoras de Anticorpos/imunologia , Imunoglobulina G/biossíntese , Memória Imunológica , Interleucina-12/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Baço/imunologia , Staphylococcus aureus/imunologia
5.
Eur J Immunol ; 47(1): 144-154, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27859048

RESUMO

IL-35 is a recently identified cytokine exhibiting potent immunosuppressive properties. The therapeutic potential and effects of IL-35 on pathogenic T effector cells (Teff) and Foxp3+ Treg, however, are ill defined. We tested the capacity of IL-35 to suppress ongoing autoimmunity in NOD mice. For this purpose, an adeno-associated virus vector in which IL-35 transgene expression is selectively targeted to ß cells via an insulin promoter (AAV8mIP-IL35) was used. AAV8mIP-IL35 vaccination of NOD mice at a late preclinical stage of type 1 diabetes (T1D) suppressed ß-cell autoimmunity and prevented diabetes onset. Numbers of islet-resident conventional CD4+ and CD8+ T cells, and DCs were reduced within 4 weeks of AAV8mIP-IL35 treatment. The diminished islet T-cell pool correlated with suppressed proliferation, and a decreased frequency of IFN-γ-expressing Teff. Ectopic IL-35 also reduced islet Foxp3+ Treg numbers and proliferation, and protection was independent of induction/expansion of adaptive islet immunoregulatory T cells. These findings demonstrate that IL-35-mediated suppression is sufficiently robust to block established ß-cell autoimmunity, and support the use of IL-35 to treat T1D and other T-cell-mediated autoimmune diseases.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Terapia Genética , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Interleucinas/genética , Transferência Adotiva , Animais , Linhagem Celular , Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Modelos Animais de Doenças , Expressão Ectópica do Gene , Feminino , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Interleucinas/metabolismo , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transdução Genética
6.
J Immunol ; 197(9): 3504-3511, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27694496

RESUMO

The use of nondepleting Abs specific for CD4 and CD8 is an effective strategy to tolerize CD4+ and CD8+ T cells in a tissue-specific manner. We reported that coreceptor therapy reverses diabetes in new onset NOD mice. A striking feature of coreceptor-induced remission is the purging of T cells from the pancreatic lymph nodes (PLN) and islets of NOD mice. Evidence indicates that Abs binding to the coreceptors promotes T cell egress from these tissues. The present study examined how coreceptor therapy affects the migration of CD4+ T cells residing in the PLN of NOD mice. Anti-CD4 Ab treatment resulted in an increased frequency of PLN but not splenic CD4+ T cells that exhibited a polarized morphology consistent with a migratory phenotype. Furthermore, PLN CD4+ T cells isolated from anti-CD4 versus control Ab-treated animals displayed increased in vitro chemotaxis to chemoattractants such as sphingosine-1-phosphate and CXCL12. Notably, the latter was dependent on activation of the small Rho GTPases Rac1 and Rac2. Rac1 and Rac2 activation was increased in Ab-bound CD4+ T cells from the PLN but not the spleen, and knockdown of Rac expression blocked the heightened reactivity of Ab-bound PLN CD4+ T cells to CXCL12. Interestingly, Rac1 and Rac2 activation was independent of Rac guanine nucleotide exchange factors known to regulate T cell activity. Therefore, Ab binding to CD4 initiates a novel pathway that involves inflammation-dependent activation of Rac and establishment of altered T cell migratory properties.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Diabetes Mellitus/terapia , Neuropeptídeos/metabolismo , Linfócitos T/efeitos dos fármacos , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12/metabolismo , Diabetes Mellitus/imunologia , Feminino , Humanos , Tolerância Imunológica , Ativação Linfocitária , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Neuropeptídeos/genética , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfócitos T/imunologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteína RAC2 de Ligação ao GTP
7.
Eur J Immunol ; 46(10): 2352-2362, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27501268

RESUMO

Dysregulation of negative selection contributes to T-cell-mediated autoimmunity, such as type 1 diabetes. The events regulating thymic negative selection, however, are ill defined. Work by our group and others suggest that negative selection is inefficient early in ontogeny and increases with age. This study examines temporal changes in negative selection and the thymic DC compartment. Peptide-induced thymocyte deletion in vivo was reduced in newborn versus 4-week-old NOD mice, despite a similar sensitivity of the respective thymocytes to apoptosis induction. The temporal increase in negative selection corresponded with an elevated capacity of thymic antigen-presenting cells to stimulate T cells, along with altered subset composition and function of resident DC. The frequency of signal regulatory protein α+ (SIRPα+ ) and plasmacytoid DCs was increased concomitant with a decrease in CD8α+ DC in 4-week-old NOD thymi. Importantly, 4-week-old versus newborn thymic SIRPα+ DC exhibited increased antigen processing and presentation via the MHC class II but not class I pathway, coupled with an enhanced T-cell stimulatory capacity not seen in thymic plasmacytoid DC and CD8α+ DC. These findings indicate that the efficiency of thymic DC-mediated negative selection is limited early after birth, and increases with age paralleling expansion of functionally superior thymic SIRPα+ DC.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Receptores Imunológicos/metabolismo , Linfócitos T/fisiologia , Timócitos/fisiologia , Animais , Apoptose , Antígenos CD8/metabolismo , Diferenciação Celular , Células Cultivadas , Seleção Clonal Mediada por Antígeno , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD
8.
J Immunol ; 193(1): 21-5, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24890722

RESUMO

Thymic dendritic cells (DC) mediate self-tolerance by presenting self-peptides to and depleting autoreactive thymocytes. Despite a significant role in negative selection, the events regulating thymic DC maturation and function under steady-state conditions are poorly understood. We report that cross-talk with thymocytes regulates thymic conventional DC (cDC) numbers, phenotype, and function. In mice lacking TCR-expressing thymocytes, thymic cDC were reduced and exhibited a less mature phenotype. Furthermore, thymic cDC in TCR-transgenic mice lacking cognate Ag expression in the thymus were also immature; notably, however, thymic cDC maturation was re-established by an Ag-specific cognate interaction with CD4+ or CD8+ single-positive thymocytes (SP). Blockade of CD40L during Ag-specific interactions with CD4 SP, but not CD8 SP, limited the effect on cDC maturation. Together, these novel findings demonstrate that homeostatic maturation and function of thymic cDC are regulated by feedback delivered by CD4 SP and CD8 SP via distinct mechanisms during a cognate Ag-specific interaction.


Assuntos
Antígenos/imunologia , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Homeostase/imunologia , Timócitos/imunologia , Animais , Antígenos/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/genética , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/genética , Células Dendríticas/citologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Timócitos/citologia
9.
J Immunol ; 193(5): 2168-77, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25063876

RESUMO

IL-2, a cytokine with pleiotropic effects, is critical for immune cell activation and peripheral tolerance. Although the therapeutic potential of IL-2 has been previously suggested in autoimmune diseases, the mechanisms whereby IL-2 mitigates autoimmunity and prevents organ damage remain unclear. Using an inducible recombinant adeno-associated virus vector, we investigated the effect of low systemic levels of IL-2 in lupus-prone MRL/Fas(lpr/lpr) (MRL/lpr) mice. Treatment of mice after the onset of disease with IL-2-recombinant adeno-associated virus resulted in reduced mononuclear cell infiltration and pathology of various tissues, including skin, lungs, and kidneys. In parallel, we noted a significant decrease of IL-17-producing CD3(+)CD4(-)CD8(-) double-negative T cells and an increase in CD4(+)CD25(+)Foxp3(+) immunoregulatory T cells (Treg) in the periphery. We also show that IL-2 can drive double-negative (DN) T cell death through an indirect mechanism. Notably, targeted delivery of IL-2 to CD122(+) cytotoxic lymphocytes effectively reduced the number of DN T cells and lymphadenopathy, whereas selective expansion of Treg by IL-2 had no effect on DN T cells. Collectively, our data suggest that administration of IL-2 to lupus-prone mice protects against end-organ damage and suppresses inflammation by dually limiting IL-17-producing DN T cells and expanding Treg.


Assuntos
Antineoplásicos/farmacologia , Antígenos CD4 , Antígenos CD8 , Interleucina-2/farmacologia , Lúpus Eritematoso Sistêmico/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Interleucina-17/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos MRL lpr , Linfócitos T Reguladores/patologia
10.
J Immunol ; 191(12): 5858-66, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24198282

RESUMO

Inefficient thymic negative selection of self-specific T cells is associated with several autoimmune diseases, including type 1 diabetes. The factors that influence the efficacy of thymic negative selection, as well as the kinetics of thymic output of autoreactive T cells remain ill-defined. We investigated thymic production of ß cell-specific T cells using a thymus-transplantation model. Thymi from different aged NOD mice, representing distinct stages of type 1 diabetes, were implanted into NOD.scid recipients, and the diabetogenicity of the resulting T cell pool was examined. Strikingly, the development of diabetes-inducing ß cell-specific CD4(+) and CD8(+) T cells was regulated in an age-dependent manner. NOD.scid recipients of newborn NOD thymi developed diabetes. However, recipients of thymi from 7- and 10-d-old NOD donor mice remained diabetes-free and exhibited a progressive decline in islet infiltration and ß cell-specific CD4(+) and CD8(+) T cells. A similar temporal decrease in autoimmune infiltration was detected in some, but not all, tissues of recipient mice implanted with thymi from NOD mice lacking expression of the autoimmune regulator transcription factor, which develop multiorgan T cell-mediated autoimmunity. In contrast, recipients of 10 d or older thymi lacked diabetogenic T cells but developed severe colitis marked by increased effector T cells reactive to intestinal microbiota. These results demonstrate that thymic development of autoreactive T cells is limited to a narrow time window and occurs in a reciprocal manner compared with colonic microbiota-responsive T cells in NOD mice.


Assuntos
Envelhecimento/imunologia , Autoimunidade/fisiologia , Seleção Clonal Mediada por Antígeno , Diabetes Mellitus Tipo 1/imunologia , Camundongos Endogâmicos NOD/imunologia , Timo/imunologia , Transferência Adotiva , Animais , Animais Recém-Nascidos , Animais Lactentes , Autoantígenos/imunologia , Doenças Autoimunes/etiologia , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Colite/etiologia , Colite/imunologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , Citotoxicidade Imunológica , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Tecido Linfoide/patologia , Camundongos , Camundongos Knockout , Camundongos SCID , Especificidade de Órgãos , Pâncreas/imunologia , Pâncreas/patologia , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/patologia , Glândulas Salivares/imunologia , Glândulas Salivares/patologia , Organismos Livres de Patógenos Específicos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Timo/crescimento & desenvolvimento , Timo/patologia , Timo/transplante , Fatores de Transcrição/deficiência , Fatores de Transcrição/fisiologia , Proteína AIRE
11.
Clin Immunol ; 148(1): 136-47, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23688653

RESUMO

Monoclonal antibody (mAb) technology has revolutionized treatment options for T cell mediated diseases. However, a safe, clinically available anti-T cell antibody (ab) remains elusive. Experience with anti-T cell agents and their propensity for causing immune-mediated toxicities have hampered the development of anti-T cell mAb's. Furthermore, misunderstanding regarding mechanism(s) of action of particular antibodies can influence development and clinical prescription habits. For example, the anti-CD3 Ab OKT3 is consistently described as a depleting Ab even though original studies showed the mechanism to be non-lytic. Future anti-T cell mAbs are likely to be non-depletional and focused on the expansion of regulatory T cells. This review discusses how the properties of Abs can be exploited for manipulating pathological T cell responses in the clinic.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Linfócitos T/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/imunologia , Autoimunidade/imunologia , Citotoxicidade Imunológica , Humanos , Ativação Linfocitária , Linfócitos T/citologia , Linfócitos T/patologia
12.
Eur J Immunol ; 42(8): 2010-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22865049

RESUMO

IFN-γ is generally believed to be important in the autoimmune pathogenesis of type 1 diabetes (T1D). However, the development of spontaneous ß-cell autoimmunity is unaffected in NOD mice lacking expression of IFN-γ or the IFN-γ receptor (IFNγR), bringing into question the role IFN-γ has in T1D. In the current study, an adoptive transfer model was employed to define the contribution of IFN-γ in CD4(+) versus CD8(+) T cell-mediated ß-cell autoimmunity. NOD.scid mice lacking expression of the IFNγR ß chain (NOD.scid.IFNγRB(null)) developed diabetes following transfer of ß cell-specific CD8(+) T cells alone. In contrast, ß cell-specific CD4(+) T cells alone failed to induce diabetes despite significant infiltration of the islets in NOD.scid.IFNγRB(null) recipients. The lack of pathogenicity of CD4(+) T-cell effectors was due to the resistance of IFNγR-deficient ß cells to inflammatory cytokine-induced cell death. On the other hand, CD4(+) T cells indirectly promoted ß-cell destruction by providing help to CD8(+) T cells in NOD.scid.IFNγRB(null) recipients. These results demonstrate that IFN-γR may play a key role in CD4(+) T cell-mediated ß-cell destruction.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Receptores de Interferon/imunologia , Transferência Adotiva , Animais , Autoimunidade , Proliferação de Células , Células Cultivadas , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Interferon/deficiência , Receptor de Interferon gama
13.
J Immunol ; 186(6): 3779-86, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21317396

RESUMO

IL-2 and TGF-ß1 play key roles in the immunobiology of Foxp3-expressing CD25(+)CD4(+) T cells (Foxp3(+)Treg). Administration of these cytokines offers an appealing approach to manipulate the Foxp3(+)Treg pool and treat T cell-mediated autoimmunity such as type 1 diabetes. However, efficacy of cytokine treatment is dependent on the mode of application, and the potent pleiotropic effects of cytokines like IL-2 may lead to severe side effects. In the current study, we used a gene therapy-based approach to assess the efficacy of recombinant adeno-associated virus vectors expressing inducible IL-2 or TGF-ß1 transgenes to suppress ongoing ß cell autoimmunity in NOD mice. Intramuscular vaccination of recombinant adeno-associated virus to 10-wk-old NOD female mice and a subsequent 3 wk induction of IL-2 was sufficient to prevent diabetes and block the progression of insulitis. Protection correlated with an increased frequency of Foxp3(+)Treg in the periphery as well as in the draining pancreatic lymph nodes and islets. IL-2 induced a shift in the ratio favoring Foxp3(+)Treg versus IFN-γ-expressing T cells infiltrating the islets. Induction of IL-2 had no systemic effect on the frequency or activational status of T cells and NK cells. Induction of TGF-ß1 had no effect on the Foxp3(+)Treg pool or the progression of ß cell autoimmunity despite induced systemic levels of activated TGF-ß1 that were comparable to IL-2. These results demonstrate that inducible IL-2 gene therapy is an effective and safe approach to manipulate Foxp3(+)Treg and suppress T cell-mediated autoimmunity and that under the conditions employed, IL-2 is more potent than TGF-ß1.


Assuntos
Dependovirus/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Terapia Genética/métodos , Interleucina-2/biossíntese , Animais , Diabetes Mellitus Tipo 1/prevenção & controle , Feminino , Engenharia Genética/métodos , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , Células HEK293 , Humanos , Interleucina-2/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/prevenção & controle , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética , Transgenes
14.
Front Immunol ; 14: 1154552, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37081890

RESUMO

Inflammasome molecules make up a family of receptors that typically function to initiate a proinflammatory response upon infection by microbial pathogens. Dysregulation of inflammasome activity has been linked to unwanted chronic inflammation, which has also been implicated in certain autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, type 1 diabetes, systemic lupus erythematosus, and related animal models. Classical inflammasome activation-dependent events have intrinsic and extrinsic effects on both innate and adaptive immune effectors, as well as resident cells in the target tissue, which all can contribute to an autoimmune response. Recently, inflammasome molecules have also been found to regulate the differentiation and function of immune effector cells independent of classical inflammasome-activated inflammation. These alternative functions for inflammasome molecules shape the nature of the adaptive immune response, that in turn can either promote or suppress the progression of autoimmunity. In this review we will summarize the roles of inflammasome molecules in regulating self-tolerance and the development of autoimmunity.


Assuntos
Doenças Autoimunes , Inflamassomos , Animais , Autoimunidade , Inflamação , Tolerância a Antígenos Próprios
15.
Eur J Immunol ; 41(5): 1480-1490, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469091

RESUMO

IL-2 plays a critical role in the induction and maintenance of FoxP3-expressing regulatory T cells (FoxP3(+) Tregs). Reduced expression of IL-2 is linked to T-cell-mediated autoimmune diseases such as type 1 diabetes (T1D), in which an imbalance between FoxP3(+) Tregs and pathogenic T effectors exists. We investigated the contribution of IL-2 to dysregulation of FoxP3(+) Tregs by comparing wildtype NOD mice with animals congenic for a C57BL/6-derived disease-resistant Il2 allele and in which T-cell secretion of IL-2 is increased (NOD.B6Idd3). Although NOD mice exhibited a progressive decline in the frequency of CD62L(hi) FoxP3(+) Tregs due to an increase in CD62L(lo) FoxP3(+) Tregs, CD62L(hi) FoxP3(+) Tregs were maintained in the pancreatic lymph nodes and islets of NOD.B6Idd3 mice. Notably, the frequency of proliferating CD62L(hi) FoxP3(+) Tregs was elevated in the islets of NOD.B6Idd3 versus NOD mice. Increasing levels of IL-2 in vivo also resulted in larger numbers of CD62L(hi) FoxP3(+) Tregs in NOD mice. These results demonstrate that IL-2 influences the suppressor activity of the FoxP3(+) Tregs pool by regulating the balance between CD62L(lo) and CD62L(hi) FoxP3(+) Tregs. In NOD mice, reduced IL-2 expression leads to an increase in nonsuppressive CD62L(lo) FoxP3(+) Tregs, which in turn correlates with a pool of CD62L(hi) FoxP3(+) Tregs with limited proliferation.


Assuntos
Interleucina-2/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proliferação de Células , Diabetes Mellitus Tipo 1/imunologia , Feminino , Fatores de Transcrição Forkhead/análise , Expressão Gênica , Interleucina-2/genética , Interleucina-2/imunologia , Ilhotas Pancreáticas/imunologia , Selectina L/análise , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pâncreas/imunologia
16.
J Immunol ; 184(9): 4863-70, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20348424

RESUMO

High titers of autoantibodies against glutamic acid decarboxylase (GAD) 65 are commonly observed in patients suffering from type 1 diabetes as well as stiff-person syndrome (SPS), a disorder that affects the CNS, and a variant of SPS, progressive encephalomyelitis with rigidity and myoclonus. Although there is a considerable amount of data focusing on the role of GAD65-specific CD4(+) T cells in type 1 diabetes, little is known about their role in SPS. In this study, we show that mice possessing a monoclonal GAD65-specific CD4(+) T cell population (4B5, PA19.9G11, or PA17.9G7) develop a lethal encephalomyelitis-like disease in the absence of any other T cells or B cells. GAD65-reactive CD4(+) T cells were found throughout the CNS in direct concordance with GAD65 expression and activated microglia: proximal to the circumventricular organs at the interface between the brain parenchyma and the blood-brain barrier. In the presence of B cells, high titer anti-GAD65 autoantibodies were generated, but these had no effect on the incidence or severity of disease. In addition, GAD65-specific CD4(+) T cells isolated from the brain were activated and produced IFN-gamma. These findings suggest that GAD65-reactive CD4(+) T cells alone mediate a lethal encephalomyelitis-like disease that may serve as a useful model to study GAD65-mediated diseases of the CNS.


Assuntos
Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/imunologia , Encefalomielite/enzimologia , Encefalomielite/imunologia , Epitopos de Linfócito T/imunologia , Glutamato Descarboxilase/imunologia , Animais , Autoanticorpos/biossíntese , Linfócitos B/enzimologia , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos T CD4-Positivos/patologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Clonais , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Encefalomielite/genética , Encefalomielite/patologia , Feminino , Imunofenotipagem , Mediadores da Inflamação/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/fisiologia
17.
J Immunol ; 184(8): 4196-204, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20220085

RESUMO

There is compelling evidence that self-reactive CD8(+) T cells are a major factor in development and progression of type 1 diabetes in animals and humans. Hence, great effort has been expended to define the specificity of autoimmune CD8(+) T cells and to alter their responses. Much work has focused on tolerization of T cells using proteins or peptides. A weakness in this approach is that residual autoreactive T cells may be activated and exacerbate disease. In this report, we use a novel approach, toxin-coupled MHC class I tetramers. Used for some time to identify Ag-specific cells, in this study, we use that same property to delete the Ag-specific cells. We show that saporin-coupled tetramers can delete islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-reactive T cells in vitro and in vivo. Sequence analysis of TCRbeta-chains of IGRP(+) cells reveals the repertoire complexity in the islets is markedly decreased as NOD mice age and significantly altered in toxic tetramer-treated NOD mice. Further tetramer(+) T cells in the islets are almost completely deleted, and, surprisingly, loss of tetramer(+) T cells in the islets is long lasting. Finally, we show deletion at 8 wk of age of IGRP(+) CD8(+) T cells, but not dystophia myotonica kinase- or insulin B-reactive cells, significantly delays diabetes in NOD mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diabetes Mellitus Tipo 1/prevenção & controle , Antígenos H-2/administração & dosagem , Imunotoxinas/administração & dosagem , Proteínas Inativadoras de Ribossomos Tipo 1/toxicidade , Microglobulina beta-2/administração & dosagem , Animais , Autoantígenos/imunologia , Autoantígenos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Morte Celular/imunologia , Movimento Celular/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Progressão da Doença , Epitopos de Linfócito T/imunologia , Feminino , Glucose-6-Fosfatase/administração & dosagem , Glucose-6-Fosfatase/biossíntese , Glucose-6-Fosfatase/imunologia , Antígenos H-2/toxicidade , Antígeno de Histocompatibilidade H-2D , Imunotoxinas/toxicidade , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Mimetismo Molecular/imunologia , Proteínas/administração & dosagem , Proteínas/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Proteínas Inativadoras de Ribossomos Tipo 1/administração & dosagem , Saporinas , Microglobulina beta-2/toxicidade
18.
Proc Natl Acad Sci U S A ; 106(26): 10770-4, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19541644

RESUMO

The immune response has been implicated as a critical factor in determining the success or failure of clinical gene therapy trials. Generally, such a response is elicited by the desired transgene product or, in some cases, the delivery system. In the current study, we report the previously uncharacterized finding that a therapeutic cassette currently being used for human investigation displays alternative reading frames (ARFs) that generate unwanted protein products to induce a cytotoxic T lymphocyte (CTL) response. In particular, we tested the hypothesis that antigenic epitopes derived from an ARF in coagulation factor IX (F9) cDNA can induce CTL reactivity, subsequently killing F9-expressing hepatocytes. One peptide (p18) of 3 candidates from an ARF of the F9 transgene induced CD8(+) T cell reactivity in mice expressing the human MHC class I molecule B0702. Subsequently, upon systemic administration of adeno-associated virus (AAV) serotype 2 vectors packaged with the F9 transgene (AAV2/F9), a robust CD8(+) CTL response was elicited against peptide p18. Of particular importance is that the ARF epitope-specific CTLs eliminated AAV2/F9-transduced hepatocytes but not AAV2/F9 codon-optimized (AAV2/F9-opt)-transduced liver cells in which p18 epitope was deleted. These results demonstrate a previously undiscovered mechanism by which CTL responses can be elicited by cryptic epitopes generated from a therapeutic transgene and have significant implications for all gene therapy modalities. Such unforeseen epitope generation warrants careful analysis of transgene sequences for ARFs to reduce the potential for adverse events arising from immune responses during clinical gene therapy protocols.


Assuntos
Epitopos/imunologia , Fator IX/metabolismo , Terapia Genética/métodos , Imunidade Celular/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células COS , Chlorocebus aethiops , Técnicas de Cocultura , Citotoxicidade Imunológica/imunologia , Dependovirus/genética , Epitopos/genética , Fator IX/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Antígenos HLA-B/genética , Antígenos HLA-B/metabolismo , Humanos , Hibridomas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligopeptídeos/imunologia , Fases de Leitura Aberta/genética , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Transfecção
19.
Proc Natl Acad Sci U S A ; 106(12): 4810-5, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19251650

RESUMO

T cell-mediated autoimmune diseases such as type 1 diabetes (T1D) are believed to be the result in part of inefficient negative selection of self-specific thymocytes. However, the events regulating thymic negative selection are not fully understood. In the current study, we demonstrate that nonobese diabetic (NOD) mice lacking expression of the Mer tyrosine kinase (MerTK) have reduced inflammation of the pancreatic islets and fail to develop diabetes. Furthermore, NOD mice deficient in MerTK expression (Mer(-/-)) exhibit a reduced frequency of beta cell-specific T cells independent of immunoregulatory effectors. The establishment of bone marrow chimeric mice demonstrated that the block in beta cell autoimmunity required hematopoietic-derived cells lacking MerTK expression. Notably, fetal thymic organ cultures and self-peptide administration showed increased thymic negative selection in Mer(-/-) mice. Finally, thymic dendritic cells (DC) prepared from Mer(-/-) mice exhibited an increased capacity to induce thymocyte apoptosis in a peptide-specific manner in vitro. These findings provide evidence for a unique mechanism involving MerTK-mediated regulation of thymocyte negative selection and thymic DC, and suggest a role for MerTK in contributing to beta cell autoimmunity.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Seleção Genética , Linfócitos T/enzimologia , Linfócitos T/imunologia , Timo/enzimologia , Timo/imunologia , Animais , Células da Medula Óssea/citologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Imunidade , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Proteínas Proto-Oncogênicas/deficiência , Receptores Proteína Tirosina Quinases/deficiência , Timo/citologia , c-Mer Tirosina Quinase
20.
Adv Mater ; 34(5): e2107392, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34775659

RESUMO

Failure to establish immune tolerance leads to the development of autoimmune disease. The ability to regulate autoreactive T cells without inducing systemic immunosuppression represents a major challenge in the development of new strategies to treat autoimmune disease. Here, a translational method for bioengineering programmed death-ligand 1 (PD-L1)- and cluster of differentiation 86 (CD86)-functionalized mouse Schwann cells (SCs) to prevent and ameliorate multiple sclerosis (MS) in established mouse models of chronic and relapsing-remitting experimental autoimmune encephalomyelitis (EAE) is described. It is shown that the intravenous (i.v.) administration of immune checkpoint ligand functionalized mouse SCs modifies the course of disease and ameliorates EAE. Further, it is found that such bioengineered mouse SCs inhibit the differentiation of myelin-specific helper T cells into pathogenic T helper type-1 (Th 1) and type-17 (Th 17) cells, promote the development of tolerogenic myelin-specific regulatory T (Treg ) cells, and resolve inflammatory central nervous system microenvironments without inducing systemic immunosuppression.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Antígenos , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/terapia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/terapia , Células de Schwann/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA