Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 8(86): eadg0878, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37624910

RESUMO

During persistent antigen stimulation, such as in chronic infections and cancer, CD8 T cells differentiate into a hypofunctional programmed death protein 1-positive (PD-1+) exhausted state. Exhausted CD8 T cell responses are maintained by precursors (Tpex) that express the transcription factor T cell factor 1 (TCF-1) and high levels of the costimulatory molecule CD28. Here, we demonstrate that sustained CD28 costimulation is required for maintenance of antiviral T cells during chronic infection. Low-level CD28 engagement preserved mitochondrial fitness and self-renewal of Tpex, whereas stronger CD28 signaling enhanced glycolysis and promoted Tpex differentiation into TCF-1neg exhausted CD8 T cells (Tex). Furthermore, enhanced differentiation by CD28 engagement did not reduce the Tpex pool. Together, these findings demonstrate that continuous CD28 engagement is needed to sustain PD-1+ CD8 T cells and suggest that increasing CD28 signaling promotes Tpex differentiation into more functional effector-like Tex, possibly without compromising long-term responses.


Assuntos
Antígenos CD28 , Fator 1 de Transcrição de Linfócitos T , Fator 1 de Transcrição de Linfócitos T/genética , Receptor de Morte Celular Programada 1 , Linfócitos T CD8-Positivos , Diferenciação Celular , Fatores de Transcrição
2.
Stem Cell Reports ; 18(4): 829-840, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36963390

RESUMO

The thymus is critical for the establishment of a functional and self-tolerant adaptive immune system but involutes with age, resulting in reduced naive T cell output. Generation of a functional human thymus from human pluripotent stem cells (hPSCs) is an attractive regenerative strategy. Direct differentiation of thymic epithelial progenitors (TEPs) from hPSCs has been demonstrated in vitro, but functional thymic epithelial cells (TECs) only form months after transplantation of TEPs in vivo. We show the generation of TECs in vitro in isogenic stem cell-derived thymic organoids (sTOs) consisting of TEPs, hematopoietic progenitor cells, and mesenchymal cells, differentiated from the same hPSC line. sTOs support T cell development, express key markers of negative selection, including the autoimmune regulator (AIRE) protein, and facilitate regulatory T cell development. sTOs provide the basis for functional patient-specific thymic organoid models, allowing for the study of human thymus function, T cell development, and transplant immunity.


Assuntos
Células-Tronco Pluripotentes , Timo , Humanos , Linfócitos T , Células Epiteliais/metabolismo , Diferenciação Celular/fisiologia , Organoides
3.
JCI Insight ; 6(24)2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34752416

RESUMO

TNF ligation of TNF receptor 1 (TNFR1) promotes either inflammation and cell survival by (a) inhibiting RIPK1's death-signaling function and activating NF-κB or (b) causing RIPK1 to associate with the death-inducing signaling complex to initiate apoptosis or necroptosis. The cellular source of TNF that results in RIPK1-dependent cell death remains unclear. To address this, we employed in vitro systems and murine models of T cell-dependent transplant or tumor rejection in which target cell susceptibility to RIPK1-dependent cell death could be genetically altered. We show that TNF released by T cells is necessary and sufficient to activate RIPK1-dependent cell death in target cells and thereby mediate target cell cytolysis independently of T cell frequency. Activation of the RIPK1-dependent cell death program in target cells by T cell-derived TNF accelerates murine cardiac allograft rejection and synergizes with anti-PD1 administration to destroy checkpoint blockade-resistant murine melanoma. Together, the findings uncover a distinct immunological role for TNF released by cytotoxic effector T cells following cognate interactions with their antigenic targets. Manipulating T cell TNF and/or target cell susceptibility to RIPK1-dependent cell death can be exploited to either mitigate or augment T cell-dependent destruction of allografts and malignancies to improve outcomes.


Assuntos
Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Fatores de Transcrição TCF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Morte Celular , Humanos , Camundongos
4.
Nat Immunol ; 22(6): 757-768, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34031614

RESUMO

Maturation of B cells within germinal centers (GCs) generates diversified B cell pools and high-affinity B cell antigen receptors (BCRs) for pathogen clearance. Increased receptor affinity is achieved by iterative cycles of T cell-dependent, affinity-based B cell positive selection and clonal expansion by mechanisms hitherto incompletely understood. Here we found that, as part of a physiologic program, GC B cells repressed expression of decay-accelerating factor (DAF/CD55) and other complement C3 convertase regulators via BCL6, but increased the expression of C5b-9 inhibitor CD59. These changes permitted C3 cleavage on GC B cell surfaces without the formation of membrane attack complex and activated C3a- and C5a-receptor signals required for positive selection. Genetic disruption of this pathway in antigen-activated B cells by conditional transgenic DAF overexpression or deletion of C3a and C5a receptors limited the activation of mechanistic target of rapamycin (mTOR) in response to BCR-CD40 signaling, causing premature GC collapse and impaired affinity maturation. These results reveal that coordinated shifts in complement regulation within the GC provide crucial signals underlying GC B cell positive selection.


Assuntos
Linfócitos B/imunologia , Ativação do Complemento , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Centro Germinativo/imunologia , Animais , Animais Geneticamente Modificados , Linfócitos B/metabolismo , Antígenos CD55/genética , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Linhagem Celular Tumoral , Hematopoiese Clonal/imunologia , Centro Germinativo/citologia , Centro Germinativo/metabolismo , Humanos , Ativação Linfocitária , Camundongos , Tonsila Palatina/citologia , Tonsila Palatina/patologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/metabolismo
5.
Mol Ther ; 28(8): 1795-1805, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32497512

RESUMO

Dendritic cell vaccines are a promising strategy for the treatment of cancer and infectious diseases but have met with mixed success. We report on a lentiviral vector-based dendritic cell vaccine strategy that generates a cluster of differentiation 8 (CD8) T cell response that is much stronger than that achieved by standard peptide-pulsing approaches. The strategy was tested in the mouse lymphocytic choriomeningitis virus (LCMV) model. Bone marrow-derived dendritic cells from SAMHD1 knockout mice were transduced with a lentiviral vector expressing the GP33 major-histocompatibility-complex (MHC)-class-I-restricted peptide epitope and CD40 ligand (CD40L) and injected into wild-type mice. The mice were highly protected against acute and chronic variant CL-13 LCMVs, resulting in a 100-fold greater decrease than that achieved with peptide epitope-pulsed dendritic cells. Inclusion of an MHC-class-II-restricted epitope in the lentiviral vector further increased the CD8 T cell response and resulted in antigen-specific CD8 T cells that exhibited a phenotype associated with functional cytotoxic T cells. The vaccination synergized with checkpoint blockade to reduce the viral load of mice chronically infected with CL-13 to an undetectable level. The strategy improves upon current dendritic cell vaccine strategies; is applicable to the treatment of disease, including AIDS and cancer; and supports the utility of Vpx-containing vectors.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Vetores Genéticos , Inibidores de Checkpoint Imunológico/farmacologia , Lentivirus , Vacinas Virais/imunologia , Viroses/prevenção & controle , Animais , Biomarcadores , Células Dendríticas/virologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Antígenos de Histocompatibilidade Classe II , Interações Hospedeiro-Patógeno/imunologia , Humanos , Lentivirus/genética , Coriomeningite Linfocítica/prevenção & controle , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinas Virais/administração & dosagem , Viroses/etiologia , Viroses/imunologia
6.
Diabetes ; 69(8): 1692-1707, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32381645

RESUMO

A failure in self-tolerance leads to autoimmune destruction of pancreatic ß-cells and type 1 diabetes (T1D). Low-molecular-weight dextran sulfate (DS) is a sulfated semisynthetic polysaccharide with demonstrated cytoprotective and immunomodulatory properties in vitro. However, whether DS can protect pancreatic ß-cells, reduce autoimmunity, and ameliorate T1D is unknown. In this study, we report that DS, but not dextran, protects human ß-cells against cytokine-mediated cytotoxicity in vitro. DS also protects mitochondrial function and glucose-stimulated insulin secretion and reduces chemokine expression in human islets in a proinflammatory environment. Interestingly, daily treatment with DS significantly reduces diabetes incidence in prediabetic NOD mice and, most importantly, reverses diabetes in early-onset diabetic NOD mice. DS decreases ß-cell death, enhances islet heparan sulfate (HS)/HS proteoglycan expression, and preserves ß-cell mass and plasma insulin in these mice. DS administration also increases the expression of the inhibitory costimulatory molecule programmed death-1 (PD-1) in T cells, reduces interferon-γ+CD4+ and CD8+ T cells, and enhances the number of FoxP3+ cells. Collectively, these studies demonstrate that the action of one single molecule, DS, on ß-cell protection, extracellular matrix preservation, and immunomodulation can reverse diabetes in NOD mice, highlighting its therapeutic potential for the treatment of T1D.


Assuntos
Autoimunidade/efeitos dos fármacos , Sulfato de Dextrana/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Quimiocinas/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/metabolismo , Camundongos , Óxidos de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/metabolismo
7.
Cell Metab ; 29(3): 638-652.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30581122

RESUMO

Small-molecule inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) induce human beta cells to proliferate, generating a labeling index of 1.5%-3%. Here, we demonstrate that combined pharmacologic inhibition of DYRK1A and transforming growth factor beta superfamily (TGFßSF)/SMAD signaling generates remarkable further synergistic increases in human beta cell proliferation (average labeling index, 5%-8%, and as high as 15%-18%), and increases in both mouse and human beta cell numbers. This synergy reflects activation of cyclins and cdks by DYRK1A inhibition, accompanied by simultaneous reductions in key cell-cycle inhibitors (CDKN1C and CDKN1A). The latter results from interference with the basal Trithorax- and SMAD-mediated transactivation of CDKN1C and CDKN1A. Notably, combined DYRK1A and TGFß inhibition allows preservation of beta cell differentiated function. These beneficial effects extend from normal human beta cells and stem cell-derived human beta cells to those from people with type 2 diabetes, and occur both in vitro and in vivo.


Assuntos
Diabetes Mellitus Tipo 2 , Harmina/farmacologia , Células Secretoras de Insulina , Inibidores da Monoaminoxidase/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adolescente , Adulto , Idoso , Animais , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Smad/antagonistas & inibidores , Células-Tronco , Adulto Jovem , Quinases Dyrk
8.
Diabetes ; 67(8): 1561-1575, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29764859

RESUMO

Patients with both major forms of diabetes would benefit from therapies that increase ß-cell mass. Glucose, a natural mitogen, drives adaptive expansion of ß-cell mass by promoting ß-cell proliferation. We previously demonstrated that a carbohydrate response element-binding protein (ChREBPα) is required for glucose-stimulated ß-cell proliferation and that overexpression of ChREBPα amplifies the proliferative effect of glucose. Here we found that ChREBPα reprogrammed anabolic metabolism to promote proliferation. ChREBPα increased mitochondrial biogenesis, oxygen consumption rates, and ATP production. Proliferation augmentation by ChREBPα required the presence of ChREBPß. ChREBPα increased the expression and activity of Nrf2, initiating antioxidant and mitochondrial biogenic programs. The induction of Nrf2 was required for ChREBPα-mediated mitochondrial biogenesis and for glucose-stimulated and ChREBPα-augmented ß-cell proliferation. Overexpression of Nrf2 was sufficient to drive human ß-cell proliferation in vitro; this confirms the importance of this pathway. Our results reveal a novel pathway necessary for ß-cell proliferation that may be exploited for therapeutic ß-cell regeneration.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Fator 2 Relacionado a NF-E2/agonistas , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/química , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Cadáver , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dinâmica Mitocondrial , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Biogênese de Organelas , Consumo de Oxigênio , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Interferência de RNA , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Técnicas de Cultura de Tecidos , Fatores de Transcrição/química , Fatores de Transcrição/genética
9.
Sci Transl Med ; 9(408)2017 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931657

RESUMO

Intercellular transfer of microRNAs can mediate communication between critical effector cells. We hypothesized that transfer of neutrophil-derived microRNAs to pulmonary epithelial cells could alter mucosal gene expression during acute lung injury. Pulmonary-epithelial microRNA profiling during coculture of alveolar epithelial cells with polymorphonuclear neutrophils (PMNs) revealed a selective increase in lung epithelial cell expression of microRNA-223 (miR-223). Analysis of PMN-derived supernatants showed activation-dependent release of miR-223 and subsequent transfer to alveolar epithelial cells during coculture in vitro or after ventilator-induced acute lung injury in mice. Genetic studies indicated that miR-223 deficiency was associated with severe lung inflammation, whereas pulmonary overexpression of miR-223 in mice resulted in protection during acute lung injury induced by mechanical ventilation or by infection with Staphylococcus aureus Studies of putative miR-223 gene targets implicated repression of poly(adenosine diphosphate-ribose) polymerase-1 (PARP-1) in the miR-223-dependent attenuation of lung inflammation. Together, these findings suggest that intercellular transfer of miR-223 from neutrophils to pulmonary epithelial cells may dampen acute lung injury through repression of PARP-1.


Assuntos
Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Células Epiteliais/metabolismo , Pulmão/patologia , MicroRNAs/metabolismo , Neutrófilos/metabolismo , Animais , Comunicação Celular , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Nanopartículas/química , Pneumonia/genética , Pneumonia/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transporte de RNA
10.
Nat Med ; 23(5): 623-630, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28414329

RESUMO

Adaptive thermogenesis is the process of heat generation in response to cold stimulation. It is under the control of the sympathetic nervous system, whose chief effector is the catecholamine norepinephrine (NE). NE enhances thermogenesis through ß3-adrenergic receptors to activate brown adipose tissue and by 'browning' white adipose tissue. Recent studies have reported that alternative activation of macrophages in response to interleukin (IL)-4 stimulation induces the expression of tyrosine hydroxylase (TH), a key enzyme in the catecholamine synthesis pathway, and that this activation provides an alternative source of locally produced catecholamines during the thermogenic process. Here we report that the deletion of Th in hematopoietic cells of adult mice neither alters energy expenditure upon cold exposure nor reduces browning in inguinal adipose tissue. Bone marrow-derived macrophages did not release NE in response to stimulation with IL-4, and conditioned media from IL-4-stimulated macrophages failed to induce expression of thermogenic genes, such as uncoupling protein 1 (Ucp1), in adipocytes cultured with the conditioned media. Furthermore, chronic treatment with IL-4 failed to increase energy expenditure in wild-type, Ucp1-/- and interleukin-4 receptor-α double-negative (Il4ra-/-) mice. In agreement with these findings, adipose-tissue-resident macrophages did not express TH. Thus, we conclude that alternatively activated macrophages do not synthesize relevant amounts of catecholamines, and hence, are not likely to have a direct role in adipocyte metabolism or adaptive thermogenesis.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Macrófagos/imunologia , Norepinefrina/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Termogênese/imunologia , Tirosina 3-Mono-Oxigenase/genética , Adaptação Fisiológica , Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Western Blotting , Composição Corporal/imunologia , Catecolaminas/metabolismo , Diferenciação Celular , Meios de Cultivo Condicionados , Metabolismo Energético/genética , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Interleucina-4/imunologia , Interleucina-4/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Termogênese/genética , Proteína Desacopladora 1/genética
11.
J Clin Invest ; 126(10): 3942-3960, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27617858

RESUMO

Protective T cell memory is an acquired trait that is contingent upon the preservation of its constituents and therefore vulnerable to the potentially deleterious effects of organismal aging. Here, however, we have found that long-term T cell memory in a natural murine host-pathogen system can substantially improve over time. Comprehensive molecular, phenotypic, and functional profiling of aging antiviral CD8+ memory T cells (CD8+ TM) revealed a pervasive remodeling process that promotes the gradual acquisition of distinct molecular signatures, of increasingly homogeneous phenotypes, and of diversified functionalities that combine to confer a CD8+ TM-autonomous capacity for enhanced recall responses and immune protection. Notably, the process of CD8+ TM aging is characterized by a progressive harmonization of memory and naive T cell traits, is broadly amenable to experimental acceleration or retardation, and serves as a constitutional component for the "rebound model" of memory T cell maturation. By casting CD8+ TM populations within the temporal framework of their slowly evolving properties, this model establishes a simple ontogenetic perspective on the principal organization of CD8+ T cell memory that may directly inform the development of improved diagnostic, prophylactic, and therapeutic modalities.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Animais , Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/virologia , Degranulação Celular , Diferenciação Celular , Células Cultivadas , Senescência Celular , Quimiocinas/metabolismo , Citotoxicidade Imunológica , Granzimas/metabolismo , Memória Imunológica , Lectinas Tipo C/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transcriptoma
12.
Diabetes ; 61(2): 436-46, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22210319

RESUMO

More than one-half of the ~50 human chemokines have been associated with or implicated in the pathogenesis of type 1 diabetes, yet their actual expression patterns in the islet environment of type 1 diabetic patients remain, at present, poorly defined. Here, we have integrated a human islet culture system, murine models of virus-induced and spontaneous type 1 diabetes, and the histopathological examination of pancreata from diabetic organ donors with the goal of providing a foundation for the informed selection of potential therapeutic targets within the chemokine/receptor family. Chemokine (C-C motif) ligand (CCL) 5 (CCL5), CCL8, CCL22, chemokine (C-X-C motif) ligand (CXCL) 9 (CXCL9), CXCL10, and chemokine (C-X3-C motif) ligand (CX3CL) 1 (CX3CL1) were the major chemokines transcribed (in an inducible nitric oxide synthase-dependent but not nuclear factor-κB-dependent fashion) and translated by human islet cells in response to in vitro inflammatory stimuli. CXCL10 was identified as the dominant chemokine expressed in vivo in the islet environment of prediabetic animals and type 1 diabetic patients, whereas CCL5, CCL8, CXCL9, and CX3CL1 proteins were present at lower levels in the islets of both species. Of importance, additional expression of the same chemokines in human acinar tissues emphasizes an underappreciated involvement of the exocrine pancreas in the natural course of type 1 diabetes that will require consideration for additional type 1 diabetes pathogenesis and immune intervention studies.


Assuntos
Quimiocinas/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Anti-Inflamatórios/farmacologia , Quimiocinas/genética , Diabetes Mellitus Tipo 1/etiologia , Humanos , Interleucina-1beta/farmacologia , Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , NF-kappa B/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
13.
Antioxid Redox Signal ; 15(8): 2221-34, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21126189

RESUMO

Traditionally, the single most unique feature of the immune system has been attributed to its capability to discriminate between self (e.g., host proteins) and nonself (e.g., pathogens). More recently, an emerging immunologic concept involves the notion that the immune system responds via a complex system for sensing signals of danger, such as pathogens or host-derived signals of cellular distress (e.g., ischemia), while remaining unresponsive to nondangerous motifs. Experimental studies have provided strong evidence that the production and signaling effects of extracellular adenosine are dramatically enhanced during conditions of limited oxygen availability as occurs during ischemia. As such, adenosine would fit the bill of signaling molecules that are enhanced during situations of cellular distress. In contrast to a danger signal, we propose here that extracellular adenosine operates as a countermeasure, in fact as a safety signal, to both restrain potentially harmful immune responses and to maintain and promote general tissue integrity during conditions of limited oxygen availability.


Assuntos
Adenosina/metabolismo , Hipóxia/fisiopatologia , Inflamação/metabolismo , Isquemia/metabolismo , Animais , Humanos , Isquemia/fisiopatologia , Transdução de Sinais/fisiologia
14.
J Clin Invest ; 120(3): 907-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20197626

RESUMO

The chemokines are a large family of mainly secreted molecules involved in the regulation of numerous physiological and pathophysiological processes. Despite many years of investigation, the precise cellular sources of most chemokines have remained incompletely defined as a consequence of the limited availability of suitable reagents to visualize the expression of chemokine proteins at the single-cell level. Here, we developed a simple flow cytometry-based assay using commercially available chemokine-specific antibodies for efficient cell-associated detection of 37 of 39 murine chemokines. To demonstrate the utility of this methodology, we used it to reevaluate the nature of homeostatic chemokines in the hematopoietic compartment, to delineate the complete chemokine profiles of NK cells and B cells in response to major polyclonal stimuli, and to assess the chemokine response of DCs to bacterial infection. The versatility of this analytical methodology was further demonstrated by its application to selected human chemokines and should greatly facilitate any future investigation into chemokine biology at large.


Assuntos
Quimiocinas/biossíntese , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/fisiologia , Animais , Anticorpos/química , Anticorpos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células COS , Linhagem Celular , Quimiocinas/imunologia , Chlorocebus aethiops , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
15.
J Virol ; 80(1): 270-80, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16352552

RESUMO

CTLA-4 is considered one of the most potent negative regulators of T-cell activation. To circumvent experimental limitations due to fatal lymphoproliferative disease associated with genetic ablation of CTLA-4, we have used radiation chimeras reconstituted with a mixture of CTLA-4+/+ and CTLA-4-/- bone marrow that retain a normal phenotype and allow the evaluation of long-term T-cell immunity under conditions of intrinsic CTLA-4 deficiency. Following virus infection, we profiled primary, memory, and secondary CD8+ and CD4+ T-cell responses directed against eight different viral epitopes. Our data demonstrate unaltered antigen-driven proliferation, acquisition of effector functions, distribution of epitope hierarchies, T-cell receptor repertoire selection, functional avidities, and long-term memory maintenance in the absence of CTLA-4. Moreover, regulation of memory T-cell survival and homeostatic proliferation, as well as secondary responses, was equivalent in virus-specific CTLA4+/+ and CTL-A-4-/- T-cell populations. Thus, lack of CTLA-4 expression by antigen-specific T cells can be compensated for by extrinsic factors in the presence of CTLA-4 expression by other cells. These findings have implications for the physiologic, pathological, and therapeutic regulation of costimulation.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Antígeno CTLA-4 , Regulação da Expressão Gênica/imunologia , Inativação Gênica , Ativação Linfocitária/imunologia , Camundongos , Linfócitos T Reguladores/imunologia
16.
Proc Natl Acad Sci U S A ; 101(25): 9357-62, 2004 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-15197277

RESUMO

Heightened protection from infectious disease as conferred by vaccination or pathogen exposure relies on the effective generation and preservation of specific immunological memory. T cells are irreducibly required for the control of most viral infections, and maintenance of CD8(+)T cell memory is regulated by at least two cytokines, IL-7 and IL-15, which support survival (IL-7, IL-15) and basal homeostatic proliferation (IL-15) of specific CD8(+) memory T cells (T(M)). In contrast, the factors governing the homeostasis of pathogen-specific CD4(+)T(M) remain at present unknown. Here, we used a physiologic in vivo model system for viral infection to delineate homeostatic features and mechanisms of antiviral CD4(+)T(M) preservation in direct juxtaposition to CD8(+)T cell memory. Basal homeostatic proliferation is comparable between specific CD4(+) and CD8(+)T(M) and independent of immunodominant determinants and functional avidities but regulated in a tissue-specific fashion. IL-7, identified as the dominant cytokine, and IL-15, an accessory cytokine, regulate basal homeostatic proliferation and survival of antiviral CD4(+)T(M). Interestingly, a role for these cytokines in regulation of CD4(+)T cell memory is not readily discernible in the generic "memory-phenotype" population, apparently a consequence of its heterogeneous composition. We also describe a prominent, nonredundant role for IL-7 in supporting basal homeostatic proliferation of CD8(+)T(M). We propose that homeostatic control of antiviral CD4(+) and CD8(+) T cell memory is fundamentally similar and characterized by quantitative, rather than qualitative, differences.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Interleucina-7/imunologia , Ativação Linfocitária/imunologia , Vírus/imunologia , Animais , Afinidade de Anticorpos/imunologia , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Divisão Celular , Imunofenotipagem , Linfonodos/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia
17.
Trends Immunol ; 24(10): 540-5, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14552838

RESUMO

The concept of therapeutic immune regulation aiming to treat autoimmune diseases has been validated in multiple animal models, yet, the development of strategies for treatment of human autoimmune diseases remains problematic. Main obstacles are the contradictory findings in different model systems, as well as the contrasting functions of regulatory lymphocytes and cytokines. By drawing examples primarily from experimental type 1 diabetes, we propose that regulatory cells and cytokines can be classified according to the baseline at which they operate in healthy individuals and disease states that are not accompanied by severe systemic immune deficiency or skewing. Consequently, deletion or neutralization of regulatory cells or cytokines operative at high levels to maintain systemic homeostasis should constitute a therapeutic strategy for immune enhancement (e.g. tumor- and pathogen-specific immunity), whereas boosting these factors will have limited effects if the therapeutic goal is a downmodulation of immune responses (e.g. autoimmunity). Conversely, regulatory cells and cytokines operative at low homeostatic levels should unfold therapeutic capacities by further embellishment but not additional reduction.


Assuntos
Doenças Autoimunes/terapia , Autoimunidade , Citocinas/uso terapêutico , Imunoterapia , Linfócitos T/imunologia , Animais , Doenças Autoimunes/imunologia , Humanos
18.
J Virol ; 77(6): 3799-808, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610154

RESUMO

CD4 and CD8 T lymphocytes infiltrate the parenchyma of mouse brains several weeks after intracerebral, intraperitoneal, or oral inoculation with the Chandler strain of mouse scrapie, a pattern not seen with inoculation of prion protein knockout (PrP(-/-)) mice. Associated with this cellular infiltration are expression of MHC class I and II molecules and elevation in levels of the T-cell chemokines, especially macrophage inflammatory protein 1beta, IFN-gamma-inducible protein 10, and RANTES. T cells were also found in the central nervous system (CNS) in five of six patients with Creutzfeldt-Jakob disease. T cells harvested from brains and spleens of scrapie-infected mice were analyzed using a newly identified mouse PrP (mPrP) peptide bearing the canonical binding motifs to major histocompatibility complex (MHC) class I H-2(b) or H-2(d) molecules, appropriate MHC class I tetramers made to include these peptides, and CD4 and CD8 T cells stimulated with 15-mer overlapping peptides covering the whole mPrP. Minimal to modest K(b) tetramer binding of mPrP amino acids (aa) 2 to 9, aa 152 to 160, and aa 232 to 241 was observed, but such tetramer-binding lymphocytes as well as CD4 and CD8 lymphocytes incubated with the full repertoire of mPrP peptides failed to synthesize intracellular gamma interferon (IFN-gamma) or tumor necrosis factor alpha (TNF-alpha) cytokines and were unable to lyse PrP(-/-) embryo fibroblasts or macrophages coated with (51)Cr-labeled mPrP peptide. These results suggest that the expression of PrP(sc) in the CNS is associated with release of chemokines and, as shown previously, cytokines that attract and retain PrP-activated T cells and, quite likely, bystander activated T cells that have migrated from the periphery into the CNS. However, these CD4 and CD8 T cells are defective in such an effector function(s) as IFN-gamma and TNF-alpha expression or release or lytic activity.


Assuntos
Encéfalo/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Síndrome de Creutzfeldt-Jakob/imunologia , Scrapie/imunologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Antígenos H-2/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo
19.
J Virol ; 76(9): 4357-63, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11932402

RESUMO

Transmissible spongiform encephalopathy or prion diseases are fatal neurodegenerative disorders of humans and animals often initiated by oral intake of an infectious agent. Current evidence suggests that infection occurs initially in the lymphoid tissues and subsequently in the central nervous system (CNS). The identity of infected lymphoid cells remains controversial, but recent studies point to the involvement of both follicular dendritic cells (FDC) and CD11c(+) lymphoid dendritic cells. FDC generation and maintenance in germinal centers is dependent on lymphotoxin alpha (LT-alpha) and LT-beta signaling components. We report here that by the oral route, LT-alpha -/- mice developed scrapie while LT-beta -/- mice did not. Furthermore, LT-alpha -/- mice had a higher incidence and shorter incubation period for developing disease following inoculation than did LT-beta -/- mice. Transplantation of lymphoid tissues from LT-beta -/- mice, which have cervical and mesenteric lymph nodes, into LT-alpha -/- mice, which do not, did not alter the incidence of CNS scrapie. In other studies, a virus that is tropic for and alters functions of CD11c(+) cells did not alter the kinetics of neuroinvasion of scrapie. Our results suggest that neither FDC nor CD11c(+) cells are essential for neuroinvasion after high doses of RML scrapie. Further, it is possible that an as yet unidentified cell found more abundantly in LT-alpha -/- than in LT-beta -/- mice may assist in the amplification of scrapie infection in the periphery and favor susceptibility to CNS disease following peripheral routes of infection.


Assuntos
Predisposição Genética para Doença , Linfotoxina-alfa/genética , Proteínas de Membrana/genética , Proteínas PrPSc/patogenicidade , Scrapie/fisiopatologia , Animais , Encéfalo/metabolismo , Células Dendríticas/imunologia , Células Dendríticas Foliculares/imunologia , Integrina alfaXbeta2/metabolismo , Tecido Linfoide/citologia , Linfotoxina-beta , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas PrPSc/metabolismo , Baço/citologia
20.
Immunity ; 16(3): 403-15, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11911825

RESUMO

Systemic treatment with antibody to CD40 ligand (aCD40L) can prevent autoimmunity and transplant rejection in several animal models and is currently under evaluation in clinical trials. While it is known that aCD40L administration inhibits expansion and effector functions of aggressive T cells, it is still unclear whether additional regulatory mechanisms are operative. Here we demonstrate that a single episode of CD40L blockade during development of the autoaggressive immune response completely prevented autoimmune disease in the RIP-LCMV mouse model for virally induced type 1 diabetes. Interestingly, protection could be transferred by a highly potent, bitypic cell population sharing phenotypic and functional properties of both natural killer (NK) and dendritic cells (DC). Furthermore, protection of prediabetic recipients was autoantigen specific and did not result in generalized immunosuppression. The origin, function, and therapeutic potential of these bitypic NK/DC regulatory cells is discussed.


Assuntos
Ligante de CD40/imunologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/prevenção & controle , Insulina/genética , Células Matadoras Naturais/imunologia , Vírus da Coriomeningite Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA