Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Front Immunol ; 12: 687582, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456907

RESUMO

High levels of inflammation play an important role in chronic heart failure (CHF). Patients with CHF have elevated levels of pro-inflammatory cytokines circulating systemically, mainly TNF and IL-6. However, there are almost no studies that relate these levels to the functional status of patients in CHF, much less to their CMV serostatus. In this study, patients with CHF (n=40; age=54.9 ± 6.3; New York Heart Association functional classification (NYHA, I-III) and healthy controls (n=40; age=53.5 ± 7.1) were analyzed. The serum concentrations of nine pro- and anti-inflammatory cytokines were measured by Luminex® xMap Technology and the basal level of mRNA expression of some immune molecules was quantified by TaqMan™ Array in CD4+ T-lymphocytes. The concentration of these cytokines in culture supernatants in response to anti-CD3 and LPS was also measured. The percentage of CD28null T-cells was determined, as well as the antibody titer against CMV. We found a higher concentration of all cytokines studied in CHF serum compared to healthy controls, as well as a direct correlation between functional status in CHF patients and levels of inflammatory cytokines. Moreover, the highest cytokine concentrations were found in patients with higher concentrations of lymphocytes lacking CD28 molecule. The cytokine production was much higher in CMV+ patients, and the production of these cytokines was found mainly in the T-lymphocytes of CMV+ patients in response to anti-CD3. Anti-CMV antibody levels were positively correlated with cytokine levels. The baseline expression of specific mRNA of the main molecules involved in the Th1 response, as well as molecules related to the CD4+CD28 null subset was higher in CMV+ patients. The cytokine concentrations are higher in CHF CMV+ patients and these concentrations are related to the production of antibodies against CMV. These high levels of cytokines are also associated with the more differentiated CD28null lymphocyte populations. All this, together with the dynamics of the pathology itself, makes CMV+ patients present a worse functional status and possibly a worse evolution of the pathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citocinas/sangue , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Insuficiência Cardíaca/imunologia , Mediadores da Inflamação/sangue , Inflamação/imunologia , Anticorpos Antivirais/sangue , Biomarcadores/sangue , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Estudos de Casos e Controles , Doença Crônica , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/diagnóstico , Infecções por Citomegalovirus/virologia , Feminino , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico , Interações Hospedeiro-Patógeno , Humanos , Inflamação/sangue , Inflamação/diagnóstico , Masculino , Pessoa de Meia-Idade , Prognóstico
2.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34214472

RESUMO

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Assuntos
Antígenos CD28/deficiência , Padrões de Herança/genética , Papillomaviridae/fisiologia , Pele/virologia , Linfócitos T/imunologia , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/imunologia , Criança , Endopeptidases/metabolismo , Feminino , Genes Recessivos , Células HEK293 , Homozigoto , Humanos , Imunidade Humoral , Memória Imunológica , Células Jurkat , Queratinócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Oncogenes , Papiloma/patologia , Papiloma/virologia , Linhagem , Sinais Direcionadores de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Cardiovasc Res ; 117(8): 1935-1948, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32647892

RESUMO

AIMS: Inflammation has important roles in atherosclerosis. CD4+CD28null (CD28null) T cells are a specialized T lymphocyte subset that produce inflammatory cytokines and cytotoxic molecules. CD28null T cells expand preferentially in patients with acute coronary syndrome (ACS) rather than stable angina and are barely detectable in healthy subjects. Importantly, ACS patients with CD28null T-cell expansion have increased risk for recurrent acute coronary events and poor prognosis, compared to ACS patients in whom this cell subset does not expand. The mechanisms regulating CD28null T-cell expansion in ACS remain elusive. We therefore investigated the role of cytokines in CD28null T-cell expansion in ACS. METHODS AND RESULTS: High-purity sorted CD4+ T cells from ACS patients were treated with a panel of cytokines (TNF-α, IL-1ß, IL-6, IL-7, and IL-15), and effects on the number, phenotype, and function of CD28null T cells were analysed and compared to the control counterpart CD28+ T-cell subset. IL-7- and IL-15-induced expansion of CD28null T cells from ACS patients, while inflammatory cytokines TNF-α, IL-1ß, and IL-6 did not. The mechanisms underlying CD28null T-cell expansion by IL-7/IL-15 were preferential activation and proliferation of CD28null T cells compared to control CD28+ T cells. Additionally, IL-7/IL-15 markedly augmented CD28null T-cell cytotoxic function and interferon-γ production. Further mechanistic analyses revealed differences in baseline expression of component chains of IL-7/IL-15 receptors (CD127 and CD122) and increased baseline STAT5 phosphorylation in CD28null T cells from ACS patients compared to the control CD28+ T-cell subset. Notably, we demonstrate that CD28null T-cell expansion was significantly inhibited by Tofacitinib, a selective JAK1/JAK3 inhibitor that blocks IL-7/IL-15 signalling. CONCLUSION: Our novel data show that IL-7 and IL-15 drive the expansion and function of CD28null T cells from ACS patients suggesting that IL-7/IL-15 blockade may prevent expansion of these cells and improve patient outcomes.


Assuntos
Síndrome Coronariana Aguda/imunologia , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inflamação/imunologia , Interleucina-15/farmacologia , Interleucina-7/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Síndrome Coronariana Aguda/metabolismo , Idoso , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citotoxicidade Imunológica/efeitos dos fármacos , Feminino , Humanos , Inflamação/metabolismo , Interferon gama/metabolismo , Janus Quinase 1/metabolismo , Janus Quinase 3/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Fosforilação , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
4.
Oxid Med Cell Longev ; 2019: 4848560, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31565151

RESUMO

Although CD28 is associated with the expression of inflammatory mediators, apoptosis-related protein, immunosuppression, and tumorigenesis, the effects of CD28 deficiency on blast exposure-induced lung injury have not been investigated. In this study, we have explored the effects of CD28 on blast exposure-induced lung injury and studied its potential molecular mechanisms. A mouse model of blast exposure-induced acute lung injury was established. Sixty C57BL/6 wild-type (WT) and CD28 knockout (CD28-/-) mice were randomly divided into control or model groups. Lung tissue samples were collected 24 h and 48 h after blast injury. Histopathological changes and the expressions of inflammatory-related proteins were detected by hematoxylin-eosin, immunohistochemistry, and immunofluorescence staining. Apoptosis and oxidative stress were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and reactive oxygen species (ROS). Inflammation, apoptosis, oxidative stress, and related pathway protein expression were studied by western blotting. In addition, the levels of CD3 and CD28 proteins were measured by flow cytometry. In the current study, we found that CD28 deficiency significantly inhibited blast exposure-induced increases in the lung weight/body weight ratio and wet weight/dry weight ratio; decreased the infiltration of CD44+ leukocytes, CD163+ macrophages, and CD3+ T cells into the lungs; reduced the expressions of proinflammatory cytokines including IL-1ß, TNF-α, and IL-6; and markedly increased IL-10 expression. CD28 deficiency also significantly attenuated blast exposure-induced ROS, MDA5, and IREα expressions; increased SOD-1 expression; lowered the number of apoptotic cells and Bax, Caspase-3, and active Caspase-8 expressions; and increased Bcl-2 expression. Additionally, CD28 deficiency significantly ameliorated blast exposure-induced increases of p-PI3K and p-Akt and ameliorated the decrease in the p-FoxO1 expression. Our results suggest that CD28 deficiency has a protective effect on blast exposure-induced lung injury, which might be associated with the PI3K/Akt/FoxO1 signaling pathway.


Assuntos
Traumatismos por Explosões/imunologia , Antígenos CD28/deficiência , Proteína Forkhead Box O1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pneumonia/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T/imunologia , Animais , Apoptose/fisiologia , Traumatismos por Explosões/metabolismo , Traumatismos por Explosões/patologia , Antígenos CD28/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/fisiologia , Pneumonia/metabolismo , Distribuição Aleatória , Transdução de Sinais , Linfócitos T/patologia
5.
PLoS One ; 13(8): e0202522, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30148845

RESUMO

Protective immunity to blood-stage malaria is attributed to Plasmodium-specific IgG and effector-memory T helper 1 (Th1) cells. However, mice lacking the costimulatory receptor CD28 (CD28KO) maintain chronic parasitemia at low levels and do not succumb to infection, suggesting that other immune responses contribute to parasite control. We report here that CD28KO mice develop long-lasting non-sterile immunity and survive lethal parasite challenge. This protection correlated with a progressive increase of anti-parasite IgM serum levels during chronic infection. Serum IgM from chronically infected CD28KO mice recognize erythrocytes infected with mature parasites, and effectively control Plasmodium infection by promoting parasite lysis and uptake. These antibodies also recognize autoantigens and antigens from other pathogens. Chronically infected CD28KO mice have high numbers of IgM+ plasmocytes and experienced B cells, exhibiting a germinal-center independent Fas+GL7-CD38+CD73- phenotype. These cells are also present in chronically infected C57BL/6 mice although in lower numbers. Finally, IgM+ experienced B cells from cured C57BL/6 and CD28KO mice proliferate and produce anti-parasite IgM in response to infected erythrocytes. This study demonstrates that CD28 deficiency results in the generation of germinal-center independent IgM+ experienced B cells and the production of protective IgM during experimental malaria, providing evidence for an additional mechanism by which the immune system controls Plasmodium infection.


Assuntos
Antígenos CD28/genética , Imunoglobulina M/imunologia , Malária/genética , Plasmodium chabaudi/imunologia , 5'-Nucleotidase/genética , ADP-Ribosil Ciclase 1/genética , Animais , Anticorpos Antiprotozoários/genética , Anticorpos Antiprotozoários/imunologia , Antígenos de Diferenciação/genética , Linfócitos B/imunologia , Linfócitos B/parasitologia , Antígenos CD28/deficiência , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Eritrócitos/parasitologia , Centro Germinativo/imunologia , Centro Germinativo/parasitologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Malária/sangue , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Knockout , Plasmodium chabaudi/patogenicidade , Receptor fas/genética
6.
Clin Exp Rheumatol ; 36(6): 1008-1013, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29745892

RESUMO

OBJECTIVES: To determine whether the CD4+CD28null T-cells subpopulation predicts the occurrence of damage in SLE. METHODS: This longitudinal study was conducted in consecutive SLE patients seen every six months in our Rheumatology Department since 2012. Patients in whom CD4+CD28null T-cells had been measured and who had at least one subsequent visit were included in the study. Survival analyses (univariable and multivariable Cox-regression models) were performed to determine the risk of overall and domain damage (as per the SLICC Damage Index - SDI) as a function of the frequency of this T-cell subpopulation. The multivariable model was adjusted for pertinent confounders. All analyses were performed using SPSS 21.0. RESULTS: One hundred and nineteen patients were evaluated; their mean (SD) age was 43.5 (11.9) years, 113 (95.0%) were female. Disease duration was 7.8 (7.0) years, the SLEDAI 5.3 (4.1) and the SDI 1.0 (1.4). The percentage of CD4+CD28null T-cells was 17.4 (14.0). The mean follow-up was 2.1 (0.8) years, and the mean number of visits per patient 3.5 (1.1). Forty-six (38.7%) patients increase at least one SDI point. In the univariable and multivariable analyses, the percentage of CD4+CD28null predicted the occurrence of lung damage [HR: 1.042 (CI95%: 1.001-1.085); p=0.047 and HR: 1.099 (CI95%1.020-1.184); p=0.013, respectively] but neither the total SDI score nor all other SDI domain scores were predicted by the percentage of CD4+CD28null cells. CONCLUSIONS: In SLE patients, CD4+CD28null T-cells predict the occurrence of new lung damage, independently of other risk factors but not of overall damage or damage on other domains.


Assuntos
Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Pulmão/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Adulto , Antimaláricos/uso terapêutico , Antígenos CD28/sangue , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Progressão da Doença , Feminino , Humanos , Imunossupressores/uso terapêutico , Estudos Longitudinais , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Peru , Prognóstico , Fatores de Risco , Fatores de Tempo
7.
J Immunol ; 199(12): 3972-3980, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29093061

RESUMO

Follicular regulatory T (TFR) cells are a newly defined regulatory T cell (Treg) subset that suppresses follicular helper T cell-mediated B cell responses in the germinal center reaction. The precise costimulatory signal requirements for proper TFR cell differentiation and function are still not known. Using conditional knockout strategies of CD28, we previously demonstrated that loss of CD28 signaling in Tregs caused autoimmunity in mice (termed CD28-ΔTreg mice), characterized by lymphadenopathy, accumulation of activated T cells, and cell-mediated inflammation of the skin and lung. In this study, we show that CD28 signaling is required for TFR cell differentiation. Treg-specific deletion of CD28 caused a reduction in TFR cell numbers and function, which resulted in increased germinal center B cells and Ab production. Moreover, residual CD28-deficient TFR cells showed a diminished suppressive capacity as assessed by their ability to inhibit Ab responses in vitro. Surprisingly, genetic deletion of B cells in CD28-ΔTreg mice prevented the development of lymphadenopathy and CD4+ T cell activation, and autoimmunity that mainly targeted skin and lung tissues. Thus, autoimmunity occurring in mice with CD28-deficient Tregs appears to be driven primarily by loss of TFR cell differentiation and function with resulting B cell-driven inflammation.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Antígenos CD28/deficiência , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Formação de Anticorpos , Doenças Autoimunes/etiologia , Doenças Autoimunes/patologia , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Feminino , Centro Germinativo/imunologia , Tolerância Imunológica/imunologia , Pulmão/imunologia , Pulmão/patologia , Linfadenopatia/etiologia , Linfadenopatia/imunologia , Linfadenopatia/prevenção & controle , Depleção Linfocítica , Linfopoese , Camundongos , Especificidade de Órgãos , Pele/imunologia , Pele/patologia , Organismos Livres de Patógenos Específicos , Linfócitos T Reguladores/classificação
8.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27396441

RESUMO

BACKGROUND: Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS: C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1ß were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS: Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).


Assuntos
Antígenos CD28/deficiência , Deleção de Genes , Ruptura Cardíaca Pós-Infarto/metabolismo , Infarto do Miocárdio/metabolismo , Transdução de Sinais , Remodelação Ventricular , Animais , Antígenos CD28/metabolismo , Regulação da Expressão Gênica , Ruptura Cardíaca Pós-Infarto/genética , Ruptura Cardíaca Pós-Infarto/patologia , Ruptura Cardíaca Pós-Infarto/fisiopatologia , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miofibroblastos/metabolismo , Miofibroblastos/ultraestrutura
9.
PLoS One ; 11(3): e0150826, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26950734

RESUMO

BACKGROUND: End-stage renal disease patients have a dysfunctional, prematurely aged peripheral T-cell system. Here we hypothesized that the degree of premature T-cell ageing before kidney transplantation predicts the risk for early acute allograft rejection (EAR). METHODS: 222 living donor kidney transplant recipients were prospectively analyzed. EAR was defined as biopsy proven acute allograft rejection within 3 months after kidney transplantation. The differentiation status of circulating T cells, the relative telomere length and the number of CD31+ naive T cells were determined as T-cell ageing parameters. RESULTS: Of the 222 patients analyzed, 30 (14%) developed an EAR. The donor age and the historical panel reactive antibody score were significantly higher (p = 0.024 and p = 0.039 respectively) and the number of related donor kidney transplantation was significantly lower (p = 0.018) in the EAR group. EAR-patients showed lower CD4+CD28null T-cell numbers (p<0.01) and the same trend was observed for CD8+CD28null T-cell numbers (p = 0.08). No differences regarding the other ageing parameters were found. A multivariate Cox regression analysis showed that higher CD4+CD28null T-cell numbers was associated with a lower risk for EAR (HR: 0.65, p = 0.028). In vitro, a significant lower percentage of alloreactive T cells was observed within CD28null T cells (p<0.001). CONCLUSION: Immunological ageing-related expansion of highly differentiated CD28null T cells is associated with a lower risk for EAR.


Assuntos
Antígenos CD28/deficiência , Rejeição de Enxerto/imunologia , Transplante de Rim/efeitos adversos , Linfócitos T/citologia , Linfócitos T/metabolismo , Adulto , Fatores Etários , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Contagem de Células , Diferenciação Celular , Senescência Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Risco , Telômero/genética , Timo/imunologia
10.
Cell Rep ; 12(2): 163-71, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26146074

RESUMO

Defective antibody production in aging is broadly attributed to immunosenescence. However, the precise immunological mechanisms remain unclear. Here, we demonstrate an increase in the ratio of inhibitory T follicular regulatory (TFR) cells to stimulatory T follicular helper (TFH) cells in aged mice. Aged TFH and TFR cells are phenotypically distinct from those in young mice, exhibiting increased programmed cell death protein-1 expression but decreased ICOS expression. Aged TFH cells exhibit defective antigen-specific responses, and programmed cell death protein-ligand 1 blockade can partially rescue TFH cell function. In contrast, young and aged TFR cells have similar suppressive capacity on a per-cell basis in vitro and in vivo. Together, these studies reveal mechanisms contributing to defective humoral immunity in aging: an increase in suppressive TFR cells combined with impaired function of aged TFH cells results in reduced T-cell-dependent antibody responses in aged mice.


Assuntos
Envelhecimento , Formação de Anticorpos/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Antígenos/imunologia , Antígenos CD28/deficiência , Antígenos CD28/genética , Imunidade Humoral , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/imunologia , Nódulos Linfáticos Agregados/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia
11.
Immunology ; 146(2): 185-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190355

RESUMO

Inflammation contributes to the development and perpetuation of several disorders and T lymphocytes orchestrate the inflammatory immune response. Although the role of T cells in inflammation is widely recognized, specific therapies that tackle inflammatory networks in disease are yet to be developed. CD4(+) CD28(null) T cells are a unique subset of helper T lymphocytes that recently shot back into the limelight as potential catalysts of inflammation in several inflammatory disorders such as autoimmunity, atherosclerosis and chronic viral infections. In contrast to conventional helper T cells, CD4(+) CD28(null) T cells have an inbuilt ability to release inflammatory cytokines and cytotoxic molecules that can damage tissues and amplify inflammatory pathways. It comes as no surprise that patients who have high numbers of these cells have more severe disease and poor prognosis. In this review, I provide an overview on the latest advances in the biology of CD4(+) CD28(null) T cells. Understanding the complex functions and dynamics of CD4(+) CD28(null) T cells may open new avenues for therapeutic intervention to prevent progression of inflammatory diseases.


Assuntos
Antígenos CD28/imunologia , Citotoxicidade Imunológica , Inflamação/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anti-Inflamatórios/uso terapêutico , Antígenos CD28/deficiência , Morte Celular , Proliferação de Células , Citocinas/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Fenótipo , Prognóstico , Índice de Gravidade de Doença , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia
12.
Cell Cycle ; 14(14): 2340-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26131730

RESUMO

Maintenance of normal core body temperature is vigorously defended by long conserved, neurovascular homeostatic mechanisms that assist in heat dissipation during prolonged, heat generating exercise or exposure to warm environments. Moreover, during febrile episodes, body temperature can be significantly elevated for at least several hours at a time. Thus, as blood cells circulate throughout the body, physiologically relevant variations in surrounding tissue temperature can occur; moreover, shifts in core temperature occur during daily circadian cycles. This study has addressed the fundamental question of whether the threshold of stimulation needed to activate lymphocytes is influenced by temperature increases associated with physiologically relevant increases in temperature. We report that the need for co-stimulation of CD4+ T cells via CD28 ligation for the production of IL-2 is significantly reduced when cells are exposed to fever-range temperature. Moreover, even in the presence of sufficient CD28 ligation, provision of extra heat further increases IL-2 production. Additional in vivo and in vitro data (using both thermal and chemical modulation of membrane fluidity) support the hypothesis that the mechanism by which temperature modulates co-stimulation is linked to increases in membrane fluidity and membrane macromolecular clustering in the plasma membrane. Thermally-regulated changes in plasma membrane organization in response to physiological increases in temperature may assist in the geographical control of lymphocyte activation, i.e., stimulating activation in lymph nodes rather than in cooler surface regions, and further, may temporarily and reversibly enable CD4+ T cells to become more quickly and easily activated during times of infection during fever.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Animais , Anquirinas/metabolismo , Antígenos CD28/deficiência , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Ensaio de Imunoadsorção Enzimática , Polarização de Fluorescência , Humanos , Interleucina-2/análise , Interleucina-2/genética , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Fluidez de Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Espectrina/metabolismo , Temperatura , Acetato de Tetradecanoilforbol/farmacologia , beta-Ciclodextrinas/farmacologia
13.
J Immunol ; 195(2): 477-87, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26041540

RESUMO

A substantial proportion of CD8(+) T cells in adults lack the expression of the CD28 molecule, and the aging of the immune system is associated with a steady expansion of this T cell subset. CD28(-)CD8(+) T cells are characterized by potent effector functions but impaired responses to antigenic challenge. CD28 acts as the primary T cell costimulatory receptor, but there are numerous additional receptors that can costimulate the activation of T cells. In this study, we have examined such alternative costimulatory pathways regarding their functional role in CD28(-)CD8(+) T cells. Our study showed that most costimulatory molecules have a low capacity to activate CD28-deficient T cells, whereas the engagement of the CD2 molecule by its ligand CD58 clearly costimulated proliferation, cytokine production, and effector function in this T cell subset. CD58 is broadly expressed on APCs including dendritic cells. Blocking CD58 mAb greatly reduced the response of human CD28(-)CD8(+) T cells to allogeneic dendritic cells, as well as to viral Ags. Our results clearly identify the CD58/CD2 axis as the primary costimulatory pathway for CD8 T cells that lack CD28. Moreover, we show that engagement of CD2 amplifies TCR signals in CD28(-)CD8(+) T cells, demonstrating that the CD2-CD58 interaction has a genuine costimulatory effect on this T cell subset. CD2 signals might promote the control of viral infection by CD28(-)CD8(+) T cells, but they might also contribute to the continuous expansion of CD28(-)CD8(+) T cells during chronic stimulation by persistent Ag.


Assuntos
Antígenos CD2/imunologia , Antígenos CD28/imunologia , Antígenos CD58/imunologia , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/imunologia , Transdução de Sinais/imunologia , Idoso , Sequência de Aminoácidos , Antígenos Virais/química , Antígenos Virais/imunologia , Antígenos Virais/farmacologia , Antígenos CD2/genética , Antígenos CD28/deficiência , Antígenos CD28/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Antígenos CD58/genética , Antígenos CD8/genética , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Regulação da Expressão Gênica , Humanos , Imunofenotipagem , Ativação Linfocitária/efeitos dos fármacos , Dados de Sequência Molecular , Orthomyxoviridae/imunologia , Peptídeos/química , Peptídeos/imunologia , Peptídeos/farmacologia , Cultura Primária de Células , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
14.
J Immunol ; 194(9): 4162-74, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25825447

RESUMO

In patients receiving allogeneic hematopoietic cell transplantation, chronic graft-versus-host disease (cGVHD) remains a frequent complication and resembles autoimmune diseases such as systemic lupus erythematosus and systemic sclerosis. Our previous work demonstrated the critical role of CD28 costimulation of donor T cells for GVHD induction. In this study, we investigate the role of CD28 costimulation of host T cells in cGVHD. CD28-intact mice as hosts showed systemic lupus erythematosus-type cGVHD, whereas CD28-deficient mice developed a distinct phenotype of cGVHD, with fibrotic damage in skin and internal organs, resembling systemic sclerosis. This phenotype was due to a lack of signaling through the C-terminal proline-rich motif within host CD28's cytoplasmic tail, a motif previously shown to be required for development of regulatory T cells (Tregs) and function of conventional T cells. Adoptive transfer experiments demonstrated that a defect in host CD4(+)CD25(+) Tregs, but not in conventional T cells, was responsible for disease phenotype. Host Treg deficiency altered the cytokine pattern of donor CD4(+) T cells and the Ag specificity of autoantibodies, and these might lead to phenotypic change. Thus, host CD28 signaling controlled the pathogenesis of cGVHD through effects on host Tregs, whose status impacts qualitatively on the allogeneic immune responses.


Assuntos
Antígenos CD28/deficiência , Doença Enxerto-Hospedeiro/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD28/genética , Antígenos CD28/imunologia , Doença Crônica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Reguladores/metabolismo , Transplante Homólogo
15.
J Infect Dis ; 211(5): 835-45, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25240171

RESUMO

We postulated that the activation of proinflammatory signaling by methicillin-resistant Staphylococcus aureus (MRSA) strain USA300 is a major factor in the pathogenesis of severe pneumonia and a target for immunomodulation. Local activation of T cells in the lung was a conserved feature of multiple strains of S. aureus, in addition to USA300. The pattern of Vß chain activation was consistent with known superantigens, but deletion of SelX or SEK and SEQ was not sufficient to prevent T-cell activation, indicating the participation of multiple genes. Using Rag2(-/-), Cd4(-/-), and Cd28(-/-) mice, we observed significantly improved clearance of MRSA from the airways and decreased lung pathology, compared with findings for wild-type controls. The improved outcome correlated with decreased production of proinflammatory cytokines (tumor necrosis factor, KC, interleukin 6, and interleukin 1ß). Our data suggest that T-cell-mediated hypercytokinemia induced by infection with MRSA strain USA300 contributes to pathogenesis and may be a therapeutic target for improving outcomes of this common infection in a clinical setting.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Citocinas/metabolismo , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/patologia , Animais , Antígenos CD28/deficiência , Antígenos CD4/genética , Citocinas/sangue , Proteínas de Ligação a DNA/deficiência , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Superantígenos/genética , Superantígenos/imunologia
16.
Immunology ; 144(4): 598-610, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25318356

RESUMO

NOD.H-2h4 mice develop spontaneous autoimmune thyroiditis (SAT) with chronic inflammation of thyroids by T and B cells. B-cell deficient (B(-/-) ) mice are resistant to SAT but develop SAT if regulatory T (Treg) cells are transiently depleted. We established a transfer model using splenocytes from CD28(-/-)  B(-/-) mice (effector cells and antigen-presenting cells) cultured with or without sorted Treg cells from Foxp3-GFP wild-type (WT) or B(-/-) mice. After transfer to mice lacking T cells, mice given Treg cells from B(-/-) mice had significantly lower SAT severity scores than mice given Treg cells from WT mice, indicating that Treg cells in B(-/-) mice are more effective suppressors of SAT than Treg cells in WT mice. Treg cells from B(-/-) mice differ from WT Treg cells in expression of CD27, tumour necrosis factor receptor (TNFR) II p75, and glucocorticoid-induced TNFR-related protein (GITR). After transient depletion using anti-CD25 or diphtheria toxin, the repopulating Treg cells in B(-/-) mice lack suppressor function, and expression of CD27, GITR and p75 is like that of WT Treg cells. If B(-/-) Treg cells develop with B cells in bone marrow chimeras, their phenotype is like that of WT Treg cells. Addition of B cells to cultures of B(-/-) Treg and T effector cells abrogates their suppressive function and their phenotype is like that of WT Treg cells. These results establish for the first time that Treg cells in WT and B(-/-) mice differ both functionally and in expression of particular cell surface markers. Both properties are altered after transient depletion and repopulation of B(-/-) Treg cells, and by the presence of B cells during Treg cell development or during interaction with effector T cells.


Assuntos
Linfócitos B/imunologia , Linfócitos T Reguladores/imunologia , Tireoidite Autoimune/imunologia , Transferência Adotiva , Animais , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Transplante de Medula Óssea , Antígenos CD28/deficiência , Antígenos CD28/genética , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Predisposição Genética para Doença , Antígenos Comuns de Leucócito/genética , Camundongos Endogâmicos NOD , Camundongos Knockout , Fenótipo , Índice de Gravidade de Doença , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Tireoidite Autoimune/genética , Tireoidite Autoimune/metabolismo , Tireoidite Autoimune/prevenção & controle , Quimeras de Transplante
17.
Proc Natl Acad Sci U S A ; 112(2): 524-9, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548162

RESUMO

Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) is an essential regulator of T-cell responses, and its absence precipitates lethal T-cell hyperactivity. However, whether CTLA-4 acts simply to veto the activation of certain clones or plays a more nuanced role in shaping the quality of T-cell responses is not clear. Here we report that T cells in CTLA-4-deficient mice show spontaneous T-follicular helper (T(FH)) differentiation in vivo, and this is accompanied by the appearance of large germinal centers (GCs). Remarkably, short-term blockade with anti-CTLA-4 antibody in wild-type mice is sufficient to elicit T(FH) generation and GC development. The latter occurs in a CD28-dependent manner, consistent with the known role of CTLA-4 in regulating the CD28 pathway. CTLA-4 can act by down-regulating CD80 and CD86 on antigen presenting cells (APCs), thereby altering the level of CD28 engagement. To mimic reduced CD28 ligation, we used mice heterozygous for CD28, revealing that the magnitude of CD28 engagement is tightly linked to the propensity for T(FH) differentiation. In contrast, other parameters of T-cell activation, including CD62L down-regulation and Ki67 expression, were relatively insensitive to altered CD28 level. Altered T(FH) generation as a result of graded reduction in CD28 was associated with decreased numbers of GC B cells and a reduction in overall GC size. These data support a model in which CTLA-4 control of immunity goes beyond vetoing T-cell priming and encompasses the regulation of T(FH) differentiation by graded control of CD28 engagement.


Assuntos
Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Imunidade Adaptativa , Animais , Autoanticorpos/biossíntese , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígenos CD28/deficiência , Antígenos CD28/genética , Antígeno CTLA-4/deficiência , Antígeno CTLA-4/genética , Diferenciação Celular/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Heterozigoto , Ligantes , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Imunológicos
18.
Circulation ; 131(8): 709-20, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25527700

RESUMO

BACKGROUND: The number of CD4(+)CD28(null) (CD28(null)) T cells, a unique subset of T lymphocytes with proinflammatory and cell-lytic phenotype, increases markedly in patients with acute coronary syndrome (ACS). ACS patients harboring high numbers of CD28(null) T cells have increased risk of recurrent severe acute coronary events and unfavorable prognosis. The mechanisms that govern the increase in CD28(null) T cells in ACS remain elusive. We investigated whether apoptosis pathways regulating T-cell homeostasis are perturbed in CD28(null) T cells in ACS. METHODS AND RESULTS: We found that CD28(null) T cells in ACS were resistant to apoptosis induction via Fas-ligation or ceramide. This was attributable to a dramatic reduction in proapoptotic molecules Bim, Bax, and Fas in CD28(null) T cells, whereas antiapoptotic molecules Bcl-2 and Bcl-xL were similar in CD28(null) and CD28(+) T cells. We also show that Bim is phosphorylated in CD28(null) T cells and degraded by the proteasome. Moreover, we demonstrate for the first time that proteasomal inhibition restores the apoptosis sensitivity of CD28(null) T cells in ACS. CONCLUSIONS: We show that CD28(null) T cells in ACS harbor marked defects in molecules that regulate T-cell apoptosis, which tips the balance in favor of antiapoptotic signals and endows these cells with resistance to apoptosis. We demonstrate that the inhibition of proteasomal activity allows CD28(null) T cells to regain sensitivity to apoptosis. A better understanding of the molecular switches that control the apoptosis sensitivity of CD28(null) T cells may reveal novel strategies for targeted elimination of these T cells in ACS patients.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Síndrome Coronariana Aguda/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/patologia , Proteínas de Membrana/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Síndrome Coronariana Aguda/epidemiologia , Anticorpos Anti-Idiotípicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas/metabolismo , Homeostase/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fenótipo , Recidiva , Fatores de Risco , Proteína X Associada a bcl-2/metabolismo
19.
J Immunol ; 193(3): 1223-32, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24951814

RESUMO

Adenoviral vectors have long been forerunners in the development of effective CD8 T cell-based vaccines; therefore, it is imperative that we understand the factors controlling the induction of robust and long-lasting transgene-specific immune responses by these vectors. In this study, we investigated the organ sites, molecules, and cell subsets that play a critical role in the priming of transgene-specific CD8 T cells after vaccination with a replication-deficient adenoviral vector. Using a human adenovirus serotype 5 (Ad5) vector and genetically engineered mice, we found that CD8(+) and/or CD103(+) dendritic cells in the draining lymph node played a critical role in the priming of Ad5-induced CD8 T cell responses. Moreover, we found that CD80/86, but not CD28, was essential for efficient generation of both primary effectors and memory CD8 T cells. Interestingly, the lack of CD28 expression resulted in a delayed primary response, whereas memory CD8 T cells generated in CD28-deficient mice appeared almost normal in terms of both phenotype and effector cytokine profile, but they exhibited a significantly reduced proliferative capacity upon secondary challenge while retaining immediate in vivo effector capabilities: in vivo cytotoxicity and short-term in vivo protective capacity. Overall, our data point to an absolute requirement for professional APCs and the expression of the costimulatory molecules CD80/86 for efficient CD8 T cell priming by adenoviral vectors. Additionally, our results suggest the existence of an alternative receptor for CD80/86, which may substitute, in part, for CD28.


Assuntos
Infecções por Adenovirus Humanos/prevenção & controle , Adenovírus Humanos/imunologia , Antígeno B7-1/deficiência , Antígeno B7-2/deficiência , Antígenos CD28/deficiência , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Engenharia Genética , Infecções por Adenovirus Humanos/genética , Adenovírus Humanos/genética , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/virologia , Antígeno B7-1/genética , Antígeno B7-2/genética , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Ligantes , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinação
20.
Cancer Immunol Res ; 2(7): 610-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24819296

RESUMO

Tumor cells use various methods of immunosuppression to overcome antitumor immunity. One such method is that of programmed death ligand-1 (PD-L1 or B7-H1), which upon binding its receptor PD-1 on T cells triggers apoptotic death of the activated T cells. Overexpression of the costimulatory molecule CD80 on PD-L1(+) tumor cells, or inclusion of a soluble form of CD80 (CD80-Fc), maintains the activation of PD-1(+)-activated T cells. Using T cells from CD28-deficient mice and antibodies to block CD28 on human T cells, we now report that a soluble form of CD80 mediates this effect by simultaneously neutralizing PD-1-PD-L1-mediated immunosuppression and by providing CD80-CD28 costimulation, and is more effective than antibodies to PD-L1 or PD-1 in maintaining IFNγ production by PD-1(+) activated T cells. Therefore, soluble CD80 may be a more effective therapeutic than these checkpoint antibodies for facilitating the development and maintenance of antitumor immunity because it has the dual functions of preventing PD-L1-mediated immunosuppression and simultaneously delivering the second signal for T-cell activation.


Assuntos
Antígeno B7-1/imunologia , Antígeno B7-H1/imunologia , Melanoma/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD28/deficiência , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas , Evasão Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...