Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Respir Res ; 16: 94, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26243260

RESUMO

BACKGROUND: Although T cells, especially CD8+, have been implicated in chronic obstructive pulmonary disease (COPD) pathogenesis, their role during acute exacerbations (AE-COPD) is uncertain. METHODS: We recruited subjects with COPD and a history of previous AE-COPD and studied them quarterly to collect blood and spontaneously expectorated sputum while stable. During exacerbations (defined by a change in symptoms plus physician diagnosis and altered medications), we collected blood and sputum before administering antibiotics or steroids. We used flow cytometry to identify leukocytes in peripheral blood, plus Luminex® analysis or ELISA to determine levels of inflammatory biomarkers in serum and sputum supernatants. RESULTS: Of 33 enrolled subjects, 13 participated in multiple stable visits and had ≥1 AE-COPD visit, yielding 18 events with paired data. Flow cytometric analyses of peripheral blood demonstrated decreased CD4+ and CD8+ T cells during AE-COPD (both absolute and as a percentage of all leukocytes) and significantly increased granulocytes, all of which correlated significantly with serum C-reactive protein (CRP) concentrations. No change was observed in other leukocyte populations during AE-COPD, although the percentage of BDCA-1+ dendritic cells expressing the activation markers CD40 and CD86 increased. During AE-COPD, sICAM-1, sVCAM-1, IL-10, IL-15 and GDF-15 increased in serum, while in sputum supernatants, CRP and TIMP-2 increased and TIMP-1 decreased. CONCLUSIONS: The decrease in CD4+ and CD8+ T cells (but not other lymphocyte subsets) in peripheral blood during AE-COPD may indicate T cell extravasation into inflammatory sites or organized lymphoid tissues. GDF-15, a sensitive marker of cardiopulmonary stress that in other settings independently predicts reduced long-term survival, is acutely increased in AE-COPD. These results extend the concept that AE-COPD are systemic inflammatory events to which adaptive immune mechanisms contribute. TRIAL REGISTRATION: NCT00281216 , ClinicalTrials.gov.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Progressão da Doença , Fator 15 de Diferenciação de Crescimento/sangue , Leucócitos Mononucleares/metabolismo , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Aguda , Idoso , Estudos de Coortes , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Escarro/metabolismo
2.
Respir Res ; 14: 33, 2013 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-23497334

RESUMO

BACKGROUND: Cigarette smoking is associated with increased frequency and duration of viral respiratory infections, but the underlying mechanisms are incompletely defined. We investigated whether smoking reduces expression by human lung macrophages (Mø) of receptors for viral nucleic acids and, if so, the effect on CXCL10 production. METHODS: We collected alveolar macrophages (AMø) by bronchoalveolar lavage of radiographically-normal lungs of subjects undergoing bronchoscopies for solitary nodules (n = 16) and of volunteers who were current or former smokers (n = 7) or never-smokers (n = 13). We measured expression of mRNA transcripts for viral nucleic acid receptors by real-time PCR in those AMø and in the human Mø cell line THP-1 following phorbol myristate acetate/vitamin D3 differentiation and exposure to cigarette smoke extract, and determined TLR3 protein expression using flow cytometry and immunohistochemistry. We also used flow cytometry to examine TLR3 expression in total lung Mø from subjects undergoing clinically-indicated lung resections (n = 25). Of these, seven had normal FEV1 and FEV1/FVC ratio (three former smokers, four current smokers); the remaining 18 subjects (14 former smokers; four current smokers) had COPD of GOLD stages I-IV. We measured AMø production of CXCL10 in response to stimulation with the dsRNA analogue poly(I:C) using Luminex assay. RESULTS: Relative to AMø of never-smokers, AMø of smokers demonstrated reduced protein expression of TLR3 and decreased mRNA for TLR3 but not TLR7, TLR8, TLR9, RIG-I, MDA-5 or PKR. Identical changes in TLR3 gene expression were induced in differentiated THP-1 cells exposed to cigarette smoke-extract in vitro for 4 hours. Among total lung Mø, the percentage of TLR3-positive cells correlated inversely with active smoking but not with COPD diagnosis, FEV1% predicted, sex, age or pack-years. Compared to AMø of never-smokers, poly(I:C)-stimulated production of CXCL10 was significantly reduced in AMø of smokers. CONCLUSIONS: Active smoking, independent of COPD stage or smoking duration, reduces both the percent of human lung Mø expressing TLR3, and dsRNA-induced CXCL10 production, without altering other endosomal or cytoplasmic receptors for microbial nucleic acids. This effect provides one possible mechanism for increased frequency and duration of viral lower respiratory tract infections in smokers. TRIAL REGISTRATION: ClinicalTrials.gov NCT00281190, NCT00281203 and NCT00281229.


Assuntos
Regulação para Baixo/genética , Macrófagos Alveolares/metabolismo , RNA de Cadeia Dupla/antagonistas & inibidores , Fumar/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores , Adulto , Idoso , Linhagem Celular , Células Cultivadas , Estudos de Coortes , Feminino , Humanos , Pulmão/citologia , Pulmão/metabolismo , Pulmão/virologia , Macrófagos Alveolares/virologia , Masculino , Pessoa de Meia-Idade , RNA de Cadeia Dupla/genética , Fumar/genética , Receptor 3 Toll-Like/biossíntese , Receptor 3 Toll-Like/genética , Adulto Jovem
3.
J Immunol ; 184(11): 6504-13, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20427767

RESUMO

Lung CD8(+) T cells might contribute to progression of chronic obstructive pulmonary disease (COPD) indirectly via IFN-gamma production or directly via cytolysis, but evidence for either mechanism is largely circumstantial. To gain insights into these potential mechanisms, we analyzed clinically indicated lung resections from three human cohorts, correlating findings with spirometrically defined disease severity. Expression by lung CD8(+) T cells of IL-18R and CD69 correlated with severity, as did mRNA transcripts for perforin and granzyme B, but not Fas ligand. These correlations persisted after correction for age, smoking history, presence of lung cancer, recent respiratory infection, or inhaled corticosteroid use. Analysis of transcripts for killer cell lectin-like receptor G1, IL-7R, and CD57 implied that lung CD8(+) T cells in COPD do not belong to the terminally differentiated effector populations associated with chronic infections or extreme age. In vitro stimulation of lung CD8(+) T cells with IL-18 plus IL-12 markedly increased production of IFN-gamma and TNF-alpha, whereas IL-15 stimulation induced increased intracellular perforin expression. Both IL-15 and IL-18 protein expression could be measured in whole lung tissue homogenates, but neither correlated in concentration with spirometric severity. Although lung CD8(+) T cell expression of mRNA for both T-box transcription factor expressed in T cells and GATA-binding protein 3 (but not retinoic acid receptor-related orphan receptor gamma or alpha) increased with spirometric severity, stimulation of lung CD8(+) T cells via CD3epsilon-induced secretion of IFN-gamma, TNF-alpha, and GM-CSF, but not IL-5, IL-13, and IL-17A. These findings suggest that the production of proinflammatory cytokines and cytotoxic molecules by lung-resident CD8(+) T cells contributes to COPD pathogenesis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interleucina-15/imunologia , Interleucina-18/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Subpopulações de Linfócitos T/imunologia , Idoso , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Separação Celular , Citocinas/biossíntese , Citocinas/imunologia , Citotoxicidade Imunológica , Feminino , Citometria de Fluxo , Volume Expiratório Forçado , Humanos , Lectinas Tipo C/biossíntese , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , RNA Mensageiro/análise , Testes de Função Respiratória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/metabolismo
4.
Am J Respir Crit Care Med ; 180(12): 1179-88, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19729666

RESUMO

RATIONALE: Dendritic cells (DCs) have not been well studied in chronic obstructive pulmonary disease (COPD), yet their integral role in activating and differentiating T cells makes them potential participants in COPD pathogenesis. OBJECTIVES: To determine the expression of maturation molecules by individual DC subsets in relationship to COPD stage and to expression of the acute activation marker CD69 by lung CD4(+) T cells. METHODS: We nonenzymatically released lung leukocytes from human surgical specimens (n = 42) and used flow cytometry to identify three DC subsets (mDC1, mDC2, and pDC) and to measure their expression of three costimulatory molecules (CD40, CD80 and CD86) and of CD83, the definitive marker of DC maturation. Spearman nonparametric correlation analysis was used to identify significant correlations between expression of DC maturation molecules and COPD severity. MEASUREMENTS AND MAIN RESULTS: Expression of CD40 by mDC1 and mDC2 and of CD86 by mDC2 was high regardless of GOLD stage, but CD80 and CD83 on these two DC subsets increased with disease progression. pDC also showed significant increases in expression of CD40 and CD80. Expression of all but one of the DC molecules that increased with COPD severity also correlated with CD69 expression on lung CD4(+) T cells from the same patients, with the exception of CD83 on mDC2. CONCLUSIONS: This cross-sectional study implies that COPD progression is associated with significant increases in costimulatory molecule expression by multiple lung DC subsets. Interactions with lung DCs may contribute to the immunophenotype of CD4(+) T cells in advanced COPD. Clinical trial registered with www.clinicaltrials.gov (NCT00281229).


Assuntos
Células Dendríticas/imunologia , Pulmão/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Idoso , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cultura de Células , Diferenciação Celular/imunologia , Estudos Transversais , Feminino , Citometria de Fluxo , Humanos , Lectinas Tipo C , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Fumar/imunologia
5.
Front Biosci (Landmark Ed) ; 14(7): 2631-46, 2009 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-19273223

RESUMO

Mononuclear phagocytes comprise a mobile, broadly dispersed and highly adaptable system that lies at the very epicenter of host defense against pathogens and the interplay of the innate and adaptive arms of immunity. Understanding the molecular mechanisms that control the response of mononuclear phagocytes to apoptotic cells and the anti-inflammatory consequences of that response is an important goal with implications for multiple areas of biomedical sciences. This review details current understanding of the heterogeneity of apoptotic cell uptake by different members of the mononuclear phagocyte family in humans and mice. It also recounts the unique role of the Tyro3 family of receptor tyrosine kinases, best characterized for Mertk, in the signal transduction leading both to apoptotic cell ingestion and the anti-inflammatory effects that result.


Assuntos
Apoptose , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Humanos , Camundongos , Fagócitos/enzimologia , Transdução de Sinais
6.
J Leukoc Biol ; 84(2): 510-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18511575

RESUMO

Apoptotic cells (AC) must be cleared by macrophages (Mø) to resolve inflammation effectively. Mertk and scavenger receptor A (SR-A) are two of many receptors involved in AC clearance. As SR-A lacks enzymatic activity or evident intracellular signaling motifs, yet seems to signal in some cell types, we hypothesized that SR-A signals via Mer receptor tyrosine kinase (Mertk), which contains a multisubstrate docking site. We induced apoptosis in murine thymocytes by dexamethasone and used Western blotting and immunoprecipitation to analyze the interaction of Mertk and SR-A in the J774A.1 (J774) murine Mø cell line and in peritoneal Mø of wild-type mice and SR-A-/- mice. Phagocytosis (but not adhesion) of AC by J774 was inhibited by anti-SR-A or function-blocking SR-A ligands. In resting J774, SR-A was associated minimally with unphosphorylated (monomeric) Mertk; exposure to AC induced a time-dependent increase in association of SR-A with Mertk in a direct or indirect manner. Anti-SR-A inhibited AC-induced phosphorylation of Mertk and of phospholipase Cgamma2, essential steps in AC ingestion. Relative to tissue Mø of wild-type mice, AC-induced Mertk phosphorylation was reduced and delayed in tissue Mø of SR-A-/- mice, as was in vitro AC ingestion at early time-points. Thus, during AC uptake by murine Mø, SR-A is essential for optimal phosphorylation of Mertk and subsequent signaling required for AC ingestion. These data support the Mertk/SR-A complex as a potential target to manipulate AC clearance and hence, resolution of inflammation and infections.


Assuntos
Apoptose/fisiologia , Macrófagos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores Depuradores Classe A/deficiência , Receptores Depuradores Classe A/fisiologia , Animais , Inflamação/fisiopatologia , Camundongos , Camundongos Knockout , Receptores Depuradores Classe A/genética , Linfócitos T/fisiologia , c-Mer Tirosina Quinase
7.
J Leukoc Biol ; 75(4): 705-13, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14704368

RESUMO

Apoptotic leukocytes must be cleared efficiently by macrophages (Mø). Apoptotic cell phagocytosis by Mø requires the receptor tyrosine kinase (RTK) MerTK (also known as c-Mer and Tyro12), the phosphatidylserine receptor (PS-R), and the classical protein kinase C (PKC) isoform betaII, which translocates to Mø membrane and cytoskeletal fractions in a PS-R-dependent manner. How these molecules cooperate to induce phagocytosis is unknown. As the phosphatidylinositol-specific phospholipase (PI-PLC) gamma2 is downstream of RTKs in some cell types and can activate classical PKCs, we hypothesized that MerTK signals via PLC gamma2. To test this hypothesis, we examined the interaction of MerTK and PLC gamma2 in resident, murine peritoneal (P)Mø and in the murine Mø cell line J774A.1 (J774) following exposure to apoptotic thymocytes. We found that as with PMø, J774 phagocytosis of apoptotic thymocytes was inhibited by antibody against MerTK. Western blotting and immunoprecipitation showed that exposure to apoptotic cells produced three time-dependent changes in PMø and J774: tyrosine phosphorylation of MerTK; association of PLC gamma2 with MerTK; and tyrosine phosphorylation of PLC gamma2. Cross-linking MerTK using antibody also induced phosphorylation of PLC gamma2 and its association with MerTK. A PI-PLC appears to be required for phagocytosis of apoptotic cells, as the PI-PLC inhibitor Et-18-OCH3 and the PLC inhibitor U73122, but not the inactive control U73343, blocked phagocytosis without impairing adhesion. On apoptotic cell adhesion to Mø, MerTK signals at least in part via PLC gamma2.


Assuntos
Apoptose/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases/metabolismo , Linfócitos T/imunologia , Fosfolipases Tipo C/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Comunicação Celular/imunologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose/efeitos dos fármacos , Fosfolipase C gama , Fosforilação/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/imunologia , Transdução de Sinais/imunologia , Fosfolipases Tipo C/imunologia , c-Mer Tirosina Quinase
8.
Am J Respir Cell Mol Biol ; 30(5): 687-93, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14527926

RESUMO

Apoptotic cells must be cleared efficiently by macrophages (Mø) to prevent autoimmunity, yet their ingestion impairs Mø microbicidal function. The principal murine resident lung phagocyte, the alveolar Mø (AMø), is specifically deficient at apoptotic cell ingestion, both in vitro and in vivo, compared with resident peritoneal Mø (PMø). To further characterize this deficiency, we assayed static adhesion in vitro using apoptotic thymocytes and resident AMø and PMø from normal C57BL/6 mice. Adhesion of apoptotic thymocytes by both types of Mø was rapid, specific, and cold-sensitive. Antibody against the receptor tyrosine kinase MerTK (Tyro12) blocked phagocytosis but not adhesion in both types of Mø. Surfactant protein A increased adhesion and phagocytosis by AMø, but not to the levels seen using PMø. Adhesion was largely cation-independent for PMø and calcium-dependent for AMø. Adhesion was not inhibited in either Mø type by mAbs against beta1 or beta3 integrins or scavenger receptor I/II (CD204), but AMø adhesion was inhibited by specific mAbs against CD11c/CD18. Thus, resident murine tissue Mø from different tissues depend on qualitatively disparate receptor systems to bind apoptotic cells. The decreased capacity of murine AMø to ingest apoptotic cells is only partially explained by reduced initial adhesion.


Assuntos
Apoptose/fisiologia , Adesão Celular/fisiologia , Macrófagos Alveolares/fisiologia , Macrófagos Peritoneais/fisiologia , Proteínas Proto-Oncogênicas , Linfócitos T/fisiologia , Timo/citologia , Animais , Anticorpos Monoclonais/metabolismo , Antígeno CD11c/metabolismo , Antígenos CD18/metabolismo , Cálcio/metabolismo , Cátions/metabolismo , Temperatura Baixa , Feminino , Macrófagos Alveolares/citologia , Macrófagos Peritoneais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteína A Associada a Surfactante Pulmonar/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase
9.
J Immunol ; 169(5): 2570-9, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12193727

RESUMO

We previously demonstrated induction and expression of CD62E and CD62P in the lungs of mice primed and then challenged with intratracheal (i.t.) SRBC. The current study examined accumulation of endogenous lymphocytes in the lungs of endothelial E- and P-selectin-deficient (E(-)P(-)) mice after i.t. SRBC challenge. Compared with syngeneic wild-type (wt) mice, E(-)P(-) mice showed an 85-95% decrease in CD8(+) T cells and B cells in the lungs at both early and late time points. In contrast, CD4(+) T cell accumulation was reduced by approximately 60% early, but equivalent to wt levels later. Surprisingly, many gammadelta T cells were found in lungs and blood of E(-)P(-) mice but were undetectable in the lungs and blood of wt mice. Absolute numbers of peripheral blood CD4, CD8, and B lymphocytes in E(-)P(-) mice equaled or exceeded the levels in wt mice, particularly after challenge. Trafficking studies using alphabeta T lymphoblasts confirmed that the recruitment of circulating cells after challenge was markedly reduced in E(-)P(-) mice. Furthermore, Ag priming occurred normally in both the selectin-deficient and wt mice, because primed lymphocytes from both groups transferred Ag sensitivity into naive wt mice. Lung production of mRNA for six CC and two CXC chemokines after challenge was equivalent by RT-PCR analysis in wt and E(-)P(-) mice. Therefore, reduced lung accumulation of alphabeta T cells and B cells in E(-)P(-) mice did not result from reduced delivery of circulating lymphocytes to the lungs, unsuccessful Ag priming, or defective pulmonary chemokine production. Selectin-dependent lymphocyte recruitment into the lungs following i.t.-SRBC challenge is subset specific and time dependent.


Assuntos
Antígenos de Grupos Sanguíneos/administração & dosagem , Movimento Celular/imunologia , Selectina E/genética , Pulmão/citologia , Pulmão/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Linfopenia/imunologia , Transferência Adotiva , Animais , Antígenos de Grupos Sanguíneos/imunologia , Inibição de Migração Celular , Movimento Celular/genética , Quimiocinas CC/biossíntese , Quimiocinas CXC , Selectina E/fisiologia , Feminino , Granulócitos/citologia , Granulócitos/imunologia , Intubação Intratraqueal , Pulmão/patologia , Ativação Linfocitária/genética , Contagem de Linfócitos , Subpopulações de Linfócitos/transplante , Linfocitose/sangue , Linfocitose/genética , Linfocitose/imunologia , Linfopenia/genética , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Selectina-P/genética , Selectina-P/fisiologia , Ovinos , Linfócitos T/imunologia
10.
J Biol Chem ; 277(39): 35906-14, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12114511

RESUMO

We showed previously that protein kinase C (PKC) is required for phagocytosis of apoptotic leukocytes by murine alveolar (AMø) and peritoneal macrophages (PMø) and that such phagocytosis is markedly lower in AMø compared with PMø. In this study, we examined the roles of individual PKC isoforms in phagocytosis of apoptotic thymocytes by these two Mø populations. By immunoblotting, AMø expressed equivalent PKC eta but lower amounts of other isoforms (alpha, betaI, betaII, delta, epsilon, mu, and zeta), with the greatest difference in betaII expression. A requirement for PKC betaII for phagocytosis was demonstrated collectively by phorbol 12-myristate 13-acetate-induced depletion of PKC betaII, by dose-response to PKC inhibitor Ro-32-0432, and by use of PKC betaII myristoylated peptide as a blocker. Exposure of PMø to phosphatidylserine (PS) liposomes specifically induced translocation of PKC betaII and other isoforms to membranes and cytoskeleton. Both AMø and PMø expressed functional PS receptor, blockade of which inhibited PKC betaII translocation. Our results indicate that murine tissue Mø require PKC betaII for phagocytosis of apoptotic cells, which differs from the PKC isoform requirement previously described in Mø phagocytosis of other particles, and imply that a crucial action of the PS receptor in this process is PKC betaII activation.


Assuntos
Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Receptores de Superfície Celular/metabolismo , Timo/citologia , Acetofenonas/farmacologia , Animais , Apoptose , Benzopiranos/farmacologia , Western Blotting , Adesão Celular , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Citometria de Fluxo , Immunoblotting , Indóis/farmacologia , Concentração Inibidora 50 , Lipossomos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ácido Mirístico/farmacologia , Fagocitose , Isoformas de Proteínas , Proteína Quinase C/química , Proteína Quinase C beta , Transporte Proteico , Pirróis/farmacologia , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...