Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(43): 21666-21672, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31597740

RESUMO

Regulatory T (Treg) cells expressing the transcription factor Foxp3 play an important role in maintaining immune homeostasis. Chronic inflammation is associated with reduced Foxp3 expression, function, and loss of phenotypic stability. Previous studies have established the importance of TNF receptor 2 (TNFR2) in the generation and/or activation of Treg cells. In this study, we assess the importance of TNFR2 in healthy mice and under inflammatory conditions. Our findings reveal that, in health, TNFR2 is important not only for the generation of Treg cells, but also for regulating their functional activity. We also show that TNFR2 maintains Foxp3 expression in Treg cells by restricting DNA methylation at the Foxp3 promoter. In inflammation, loss of TNFR2 results in increased severity and chronicity of experimental arthritis, reduced total numbers of Treg cells, reduced accumulation of Treg cells in inflamed joints, and loss of inhibitory activity. In addition, we demonstrate that, under inflammatory conditions, loss of TNFR2 causes Treg cells to adopt a proinflammatory Th17-like phenotype. It was concluded that TNFR2 signaling is required to enable Treg cells to promote resolution of inflammation and prevent them from undergoing dedifferentiation. Consequently, TNFR2-specific agonists or TNF1-specific antagonists may be useful in the treatment of autoimmune disease.


Assuntos
Doenças Autoimunes/imunologia , Metilação de DNA/genética , Fatores de Transcrição Forkhead/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Regiões Promotoras Genéticas/genética
2.
EBioMedicine ; 33: 282-288, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29983350

RESUMO

BACKGROUND: Dupuytren's disease is a common fibrotic condition of the hand that causes irreversible flexion contractures of the fingers, with no approved therapy for early stage disease. Our previous analysis of surgically-excised tissue defined tumour necrosis factor (TNF) as a potential therapeutic target. Here we assessed the efficacy of injecting nodules of Dupuytren's disease with a TNF inhibitor. METHODS: Patients were randomised to receive adalimumab on one occasion in dose cohorts of 15 mg in 0.3 ml, 35 mg in 0.7 ml, or 40 mg in 0.4 ml, or an equivalent volume of placebo in a 3:1 ratio. Two weeks later the injected tissue was surgically excised and analysed. The primary outcome measure was levels of mRNA expression for α-smooth muscle actin (ACTA2). Secondary outcomes included levels of α-SMA and collagen proteins. The trial was registered with ClinicalTrial.gov (NCT03180957) and the EudraCT (2015-001780-40). FINDINGS: We recruited 28 patients, 8 assigned to the 15 mg, 12 to the 35 mg and 8 to the 40 mg adalimumab cohorts. There was no change in mRNA levels for ACTA2, COL1A1, COL3A1 and CDH11. Levels of α-SMA protein expression in patients treated with 40 mg adalimumab (1.09 ±â€¯0.09 ng per µg of total protein) were significantly lower (p = 0.006) compared to placebo treated patients (1.51 ±â€¯0.09 ng/µg). The levels of procollagen type I protein expression were also significantly lower (p < 0.019) in the sub group treated with 40 mg adalimumab (474 ±â€¯84 pg/µg total protein) compared with placebo (817 ±â€¯78 pg/µg). There were two serious adverse events, both considered unrelated to the study drug. INTERPRETATION: In this dose-ranging study, injection of 40 mg of adalimumab in 0.4 ml resulted in down regulation of the myofibroblast phenotype as evidenced by reduction in expression of α-SMA and type I procollagen proteins at 2 weeks. These data form the basis of an ongoing phase 2b clinical trial assessing the efficacy of intranodular injection of 40 mg adalimumab in 0.4 ml compared to an equivalent volume of placebo in patients with early stage Dupuytren's disease. FUNDING: Health Innovation Challenge Fund (Wellcome Trust and Department of Health) and 180 Therapeutics LP.


Assuntos
Actinas/metabolismo , Adalimumab/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Colágeno Tipo I/metabolismo , Contratura de Dupuytren/tratamento farmacológico , Actinas/genética , Adalimumab/farmacologia , Anti-Inflamatórios/farmacologia , Colágeno Tipo I/genética , Método Duplo-Cego , Regulação para Baixo , Esquema de Medicação , Contratura de Dupuytren/genética , Contratura de Dupuytren/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções , Masculino , Resultado do Tratamento
3.
Nat Commun ; 8(1): 1595, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29150600

RESUMO

Pattern recognition underpins innate immunity; the accurate identification of danger, including infection, injury, or tumor, is key to an appropriately targeted immune response. Pathogen detection is increasingly well defined mechanistically, but the discrimination of endogenous inflammatory triggers remains unclear. Tenascin-C, a matrix protein induced upon tissue damage and expressed by tumors, activates toll-like receptor 4 (TLR4)-mediated sterile inflammation. Here we map three sites within tenascin-C that directly and cooperatively interact with TLR4. We also identify a conserved inflammatory epitope in related proteins from diverse families, and demonstrate that its presence targets molecules for TLR detection, while its absence enables escape of innate immune surveillance. These data reveal a unique molecular code that defines endogenous proteins as inflammatory stimuli by marking them for recognition by TLRs.


Assuntos
Imunidade Inata , Inflamação/metabolismo , Tenascina/metabolismo , Receptor 4 Toll-Like/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Tenascina/química , Tenascina/genética , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/genética
4.
Arthritis Rheumatol ; 66(10): 2728-38, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24965881

RESUMO

OBJECTIVE: Tumor necrosis factor (TNF) signals via 2 receptors, TNFR type I (TNFRI) and TNFRII, with distinct cellular distribution and signaling functions. In rheumatoid arthritis (RA), the net effect of TNFR signaling favors inflammatory responses while inhibiting the activity of regulatory T cells. TNFRII signaling has been shown to promote Treg cell function. To assess the relative contributions of TNFRI and TNFRII signaling to inflammatory and regulatory responses in vivo, we compared the effect of TNF blockade, hence TNFRI/II, versus TNFRI alone in collagen-induced arthritis (CIA) as a model of RA. METHODS: Mice with established arthritis were treated for 10 days with anti-mouse TNFRI domain antibody (dAb; DMS5540), an isotype control dAb (DMS5538), or murine TNFRII genetically fused with mouse IgG1 Fc domain (mTNFRII-Fc) beginning on the day of arthritis onset, and disease progression was monitored. Systemic cytokine concentrations and numbers of T cell subsets in lymph nodes and spleens were measured, and intrinsic Treg cell function was determined by ex vivo suppression assays. RESULTS: Progression of CIA was suppressed similarly by TNFRI (DMS5540) and TNFRI/II (mTNFRII-Fc) blockade. However, blockade of TNFRI/II led to increased effector T cell activity, which was not observed after selective TNFRI blockade, suggesting an immunoregulatory role of TNFRII. In support of this, TNFRI blockade, but not TNFRI/II blockade, expanded and activated Treg cells. Furthermore, a dramatic increase in expression of the Treg cell signature genes FoxP3 and TNFRII was observed in joints undergoing remission, which supports the notion that these molecules have a physiologic role in the resolution of inflammation. CONCLUSION: We propose that a therapeutic strategy that targets TNFRI while sparing TNFRII has the potential to both inhibit inflammation and promote Treg cell activity, which might be superior to TNF blockade.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Domínio Único/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Artrite Experimental/imunologia , Proliferação de Células/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Inflamação/tratamento farmacológico , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Domínio Único/farmacologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/imunologia
5.
Drug Des Devel Ther ; 7: 201-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23569359

RESUMO

Psoriasis is a chronic inflammatory skin disease, most commonly resulting in the occurrence of red and silver scaly plaques. About 30% of psoriasis sufferers develop psoriatic arthritis (PsA), a disorder that presents with additional joint inflammation and other clinical features. At present, the most effective treatment for moderate and severe psoriasis and PsA are biologics such as antitumor necrosis factor alpha therapy. Biologics are costly and typically require repeated injections; hence, the development of novel, orally available, small molecular inhibitors that are less expensive to produce is highly desirable. The phosphodiesterase 4 inhibitor apremilast is a small molecular inhibitor that acts by increasing cyclic adenosine monophosphate levels, ultimately suppressing tumor necrosis alpha production. Apremilast has been tested in a number of psoriasis and PsA pilot and Phase II trials to evaluate its efficacy and safety. More recently, three larger double-blinded, and randomized multicenter studies demonstrate that apremilast is efficacious in the treatment of psoriasis and PsA, with significantly higher numbers of apremilast-treated patients achieving endpoints of a 75% reduction compared to baseline in Psoriasis Area and Severity Index (PASI-75) or American College of Rheumatology-20 scores, relative to placebo. This encouraging data, along with a tolerable incidence of mild to moderate adverse events, has led to the initiation of several large Phase III trials that aim to further validate apremilast as a treatment for psoriasis and PsA. Here, we provide an overview of the current treatments for psoriasis and PsA, and summarize the findings from multiple Phase II clinical trials where the effects of apremilast in the treatment of psoriasis and PsA patients have been investigated.


Assuntos
Artrite Psoriásica/tratamento farmacológico , Psoríase/tratamento farmacológico , Talidomida/análogos & derivados , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Artrite Psoriásica/fisiopatologia , Ensaios Clínicos Fase II como Assunto , Humanos , Inibidores da Fosfodiesterase 4/efeitos adversos , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Psoríase/patologia , Índice de Gravidade de Doença , Talidomida/efeitos adversos , Talidomida/farmacologia , Talidomida/uso terapêutico
6.
Arthritis Res Ther ; 12(3): R107, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20525198

RESUMO

INTRODUCTION: Type 4 phosphodiesterases (PDE4) play an important role in immune cells through the hydrolysis of the second messenger, cAMP. Inhibition of PDE4 has previously been shown to suppress immune and inflammatory responses, demonstrating PDE4 to be a valid therapeutic target for immune-mediated pathologies. We assessed the anti-inflammatory effects of a novel PDE4 inhibitor, apremilast, in human synovial cells from rheumatoid arthritis (RA) patients, as well as two murine models of arthritis. METHODS: Cells liberated from tissue excised from arthritic joints of RA patients were cultured in the presence of increasing concentrations of apremilast for 48 hours and spontaneous tumour necrosis factor-alpha (TNFalpha) production was analysed in culture supernatants by ELISA. In addition, arthritis was induced in BALB/c and DBA/1 mice by passive transfer of anti-type II collagen mAb and immunisation with type II collagen, respectively. Mice with established arthritis received 5 or 25 mg/kg apremilast and disease severity was monitored relative to mice receiving vehicle alone. At the end of the study, paws were removed and processed for histopathological assessment. Behavioural effects of apremilast, relative to rolipram, were assessed in naïve DBA/1 mice using an automated activity monitor (LABORAS). RESULTS: Apremilast dose dependently inhibited spontaneous release of TNFalpha from human rheumatoid synovial membrane cultures. Furthermore, apremilast significantly reduced clinical score in both murine models of arthritis over a ten day treatment period and maintained a healthy joint architecture in a dose-dependent manner. Importantly, unlike rolipram, apremilast demonstrated no adverse behavioural effects in naïve mice. CONCLUSIONS: Apremilast is an orally available PDE4 inhibitor that reduces TNFalpha production from human synovial cells and significantly suppresses experimental arthritis. Apremilast appears to be a potential new agent for the treatment of rheumatoid arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/metabolismo , Inibidores da Fosfodiesterase 4 , Membrana Sinovial/metabolismo , Talidomida/análogos & derivados , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Reumatoide/patologia , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Inibidores de Fosfodiesterase/efeitos adversos , Inibidores de Fosfodiesterase/uso terapêutico , Rolipram/efeitos adversos , Rolipram/uso terapêutico , Índice de Gravidade de Doença , Membrana Sinovial/efeitos dos fármacos , Talidomida/farmacologia , Talidomida/uso terapêutico
7.
J Exp Med ; 205(11): 2491-7, 2008 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-18936235

RESUMO

IL-17 is implicated in the pathogenesis of rheumatoid arthritis (RA) and has previously been shown to be induced by tumor necrosis factor (TNF) in vitro. The aim of this study was to assess the impact of TNF inhibition on IL-17 production in collagen-induced arthritis, a model of RA. TNF blockade using TNFR-Fc fusion protein or anti-TNF monoclonal antibody reduced arthritis severity but, unexpectedly, expanded populations of Th1 and Th17 cells, which were shown by adoptive transfer to be pathogenic. Th1 and Th17 cell populations were also expanded in collagen-immunized TNFR p55(-/-) but not p75(-/-) mice. The expression of IL-12/IL-23 p40 was up-regulated in lymph nodes (LN) from p55(-/-) mice, and the expansion of Th1/Th17 cells was abrogated by blockade of p40. Treatment of macrophages with rTNF also inhibited p40 production in vitro. These findings indicate that at least one of the ways in which TNF regulates Th1/Th17 responses in arthritis is by down-regulating the expression of p40. Finally, although TNF blockade increased numbers of Th1 and Th17 cells in LN, it inhibited their accumulation in the joint, thereby providing an explanation for the paradox that anti-TNF therapy ameliorates arthritis despite increasing numbers of pathogenic T cells.


Assuntos
Artrite Experimental/imunologia , Imunoglobulina G/farmacologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Células Th1/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Transferência Adotiva , Animais , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Etanercepte , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Receptores do Fator de Necrose Tumoral , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/metabolismo
8.
Traffic ; 8(9): 1190-204, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17605758

RESUMO

Intercellular transfer of cell surface proteins is widespread and facilitates several recently discovered means for immune cell communication. Here, we examined the molecular mechanism for intercellular exchange of the natural killer (NK) cell receptor KIR2DL1 and HLA-C, prototypical proteins that swap between NK cells and target cells. Transfer was contact dependent and enhanced for cells expressing cognate receptor/ligand pairs but did not depend on KIR2DL1 signaling. To a lesser extent, proteins transferred independent from specific recognition. Intracellular domains of transferred proteins were not exposed to the extracellular environment and transferred proteins were removed by brief exposure to low pH. By fluorescence microscopy, transferred proteins localized to discrete regions on the recipient cell surface. Higher resolution scanning electron micrographs revealed that transferred proteins were located within specific membranous structures. Transmission electron microscopy of the immune synapse revealed that membrane protrusions from one cell interacted with the apposing cell surface within the synaptic cleft. These data, coupled with previous observations, lead us to propose that intercellular protein transfer is mediated by membrane protrusions within and surrounding the immunological synapse.


Assuntos
Membrana Celular/metabolismo , Extensões da Superfície Celular/metabolismo , Junções Intercelulares/metabolismo , Células Matadoras Naturais/metabolismo , Proteínas de Membrana/metabolismo , Ácidos/farmacologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Comunicação Celular/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Extensões da Superfície Celular/ultraestrutura , Invaginações Revestidas da Membrana Celular/metabolismo , Invaginações Revestidas da Membrana Celular/ultraestrutura , Antígenos HLA-C/genética , Antígenos HLA-C/metabolismo , Humanos , Junções Intercelulares/ultraestrutura , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Microscopia Eletrônica , Compostos Orgânicos/metabolismo , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Pirimidinas/farmacologia , Receptores KIR2DL1/genética , Receptores KIR2DL1/imunologia , Receptores KIR2DL1/metabolismo , Transfecção , Quinases da Família src/antagonistas & inibidores
9.
J Immunol ; 178(9): 5606-11, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17442943

RESUMO

NKG2D is an activating receptor expressed on all human NK cells and a subset of T cells. In cytolytic conjugates between NK cells and target cells expressing its ligand MHC class I chain-related gene A, NKG2D accumulates at the immunological synapse with GM1-rich microdomains. Furthermore, NKG2D is specifically recruited to detergent-resistant membrane fractions upon ligation. However, in the presence of a strong inhibitory stimulus, NKG2D-mediated cytotoxicity can be intercepted, and recruitment of NKG2D to the immunological synapse and detergent-resistant membrane fractions is blocked. Also, downstream phosphorylation of Vav-1 triggered by NKG2D ligation is circumvented by coengaging inhibitory receptors. Thus, we propose that one way in which inhibitory signaling can control NKG2D-mediated activation is by blocking its recruitment to GM1-rich membrane domains. The accumulation of activating NK cell receptors in GM1-rich microdomains may provide the necessary platform from which stimulatory signals can proceed.


Assuntos
Gangliosídeo G(M1)/metabolismo , Células Matadoras Naturais/imunologia , Microdomínios da Membrana/metabolismo , Receptores Imunológicos/metabolismo , Actinas/metabolismo , Células Cultivadas , Gangliosídeo G(M1)/análise , Humanos , Células Matadoras Naturais/química , Ativação Linfocitária , Microdomínios da Membrana/química , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Fosforilação , Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptores Imunológicos/análise , Receptores Imunológicos/antagonistas & inibidores , Receptores KIR2DL1 , Receptores de Células Matadoras Naturais
10.
Cell ; 122(5): 789-801, 2005 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-16143108

RESUMO

Dietary heme iron is an important nutritional source of iron in carnivores and omnivores that is more readily absorbed than non-heme iron derived from vegetables and grain. Most heme is absorbed in the proximal intestine, with absorptive capacity decreasing distally. We utilized a subtractive hybridization approach to isolate a heme transporter from duodenum by taking advantage of the intestinal gradient for heme absorption. Here we show a membrane protein named HCP 1 (heme carrier protein 1), with homology to bacterial metal-tetracycline transporters, mediates heme uptake by cells in a temperature-dependent and saturable manner. HCP 1 mRNA was highly expressed in duodenum and regulated by hypoxia. HCP 1 protein was iron regulated and localized to the brush-border membrane of duodenal enterocytes in iron deficiency. Our data indicate that HCP 1 is the long-sought intestinal heme transporter.


Assuntos
Duodeno/metabolismo , Heme/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/metabolismo , Células CHO , Proteínas de Transporte/metabolismo , Clonagem Molecular , Cricetinae , Duodeno/citologia , Células Epiteliais/metabolismo , Células HeLa , Humanos , Hipóxia/metabolismo , Absorção Intestinal/fisiologia , Ferro/metabolismo , Deficiências de Ferro , Proteínas de Membrana Transportadoras/genética , Camundongos , Dados de Sequência Molecular , Oócitos/metabolismo , Transportador de Folato Acoplado a Próton , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Ratos , Alinhamento de Sequência , Transferrina/metabolismo , Xenopus , Peixe-Zebra
11.
J Exp Med ; 199(7): 1005-10, 2004 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-15051759

RESUMO

Cell surface proteins major histocompatibility complex (MHC) class I-related chain A (MICA) and UL16-binding proteins (ULBP) 1, 2, and 3 are up-regulated upon infection or tumor transformation and can activate human natural killer (NK) cells. Patches of cross-linked raft resident ganglioside GM1 colocalized with ULBP1, 2, 3, or MICA, but not CD45. Thus, ULBPs and MICA are expressed in lipid rafts at the cell surface. Western blotting revealed that glycosylphosphatidylinositol (GPI)-anchored ULBP3 but not transmembrane MICA, MHC class I protein, or transferrin receptor, accumulated in detergent-resistant membranes containing GM1. Thus, MICA may have a weaker association with lipid rafts than ULBP3, yet both proteins accumulate at an activating human NK cell immune synapse. Target cell lipid rafts marked by green fluorescent protein-tagged GPI also accumulate with ULBP3 at some synapses. Electron microscopy reveals constitutive clusters of ULBP at the cell surface. Regarding a specific molecular basis for the organization of these proteins, ULBP1, 2, and 3 and MICA are lipid modified. ULBP1, 2, and 3 are GPI anchored, and we demonstrate here that MICA is S-acylated. Finally, expression of a truncated form of MICA that lacks the putative site for S-acylation and the cytoplasmic tail can be expressed at the cell surface, but is unable to activate NK cells.


Assuntos
Proteínas de Transporte/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Receptores Imunológicos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Sequência de Bases , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Primers do DNA/genética , Proteínas Ligadas por GPI , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Células Matadoras Naturais/ultraestrutura , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Proteínas de Membrana , Microscopia Eletrônica , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores de Células Matadoras Naturais , Linfócitos T/ultraestrutura
12.
J Immunol ; 170(6): 2862-70, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12626536

RESUMO

In this study, we report the organization of cytoskeletal and large transmembrane proteins at the inhibitory and activating NK cell immunological or immune synapse (IS). Filamentous actin accumulates at the activating, but not the inhibitory, NK cell IS. However, surprisingly, ezrin and the associated protein CD43 are excluded from the inhibitory, but not the activating, NK cell IS. This distribution of ezrin and CD43 at the inhibitory NK cell IS is similar to that previously seen at the activating T cell IS. CD45 is also excluded from the inhibitory, but not activating, NK cell IS. In addition, electron microscopy reveals wide and narrow domains across the synaptic cleft. Target cell HLA-C, located by immunogold labeling, clusters where the synaptic cleft spans the size of HLA-C bound to the inhibitory killer Ig-like receptor. These data are consistent with assembly of the NK cell IS involving a combination of cytoskeletal-driven mechanisms and thermodynamics favoring the organization of receptor/ligand pairs according to the size of their extracellular domains.


Assuntos
Actinas/metabolismo , Antígenos CD , Citotoxicidade Imunológica , Junções Intercelulares/imunologia , Células Matadoras Naturais/imunologia , Antígenos Comuns de Leucócito/metabolismo , Ativação Linfocitária/imunologia , Fosfoproteínas/metabolismo , Sialoglicoproteínas/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actinas/ultraestrutura , Comunicação Celular/imunologia , Linhagem Celular Transformada , Células Clonais , Proteínas do Citoesqueleto , Antígenos HLA-C/metabolismo , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/ultraestrutura , Antígenos Comuns de Leucócito/biossíntese , Antígenos Comuns de Leucócito/ultraestrutura , Leucossialina , Microscopia Confocal , Microscopia Imunoeletrônica , Fosfoproteínas/biossíntese , Fosfoproteínas/ultraestrutura , Receptores Imunológicos/biossíntese , Receptores KIR2DL1 , Sialoglicoproteínas/biossíntese , Sialoglicoproteínas/ultraestrutura , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA