Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
JCI Insight ; 6(15)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34185707

RESUMO

We previously reported heightened expression of the human endogenous retroviral protein HERV-K deoxyuridine triphosphate nucleotidohydrolase (dUTPase) in circulating monocytes and pulmonary arterial (PA) adventitial macrophages of patients with PA hypertension (PAH). Furthermore, recombinant HERV-K dUTPase increased IL-6 in PA endothelial cells (PAECs) and caused pulmonary hypertension in rats. Here we show that monocytes overexpressing HERV-K dUTPase, as opposed to GFP, can release HERV-K dUTPase in extracellular vesicles (EVs) that cause pulmonary hypertension in mice in association with endothelial mesenchymal transition (EndMT) related to induction of SNAIL/SLUG and proinflammatory molecules IL-6 as well as VCAM1. In PAECs, HERV-K dUTPase requires TLR4-myeloid differentiation primary response-88 to increase IL-6 and SNAIL/SLUG, and HERV-K dUTPase interaction with melanoma cell adhesion molecule (MCAM) is necessary to upregulate VCAM1. TLR4 engagement induces p-p38 activation of NF-κB in addition to p-pSMAD3 required for SNAIL and pSTAT1 for IL-6. HERV-K dUTPase interaction with MCAM also induces p-p38 activation of NF-κB in addition to pERK1/2-activating transcription factor-2 (ATF2) to increase VCAM1. Thus in PAH, monocytes or macrophages can release HERV-K dUTPase in EVs, and HERV-K dUTPase can engage dual receptors and signaling pathways to subvert PAEC transcriptional machinery to induce EndMT and associated proinflammatory molecules.


Assuntos
Retrovirus Endógenos , Transição Epitelial-Mesenquimal/imunologia , Hipertensão Pulmonar , Macrófagos/imunologia , Monócitos/imunologia , Artéria Pulmonar , Pirofosfatases/metabolismo , Animais , Antígeno CD146/metabolismo , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/patogenicidade , Células Endoteliais/metabolismo , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/virologia , Inflamação/metabolismo , Inflamação/virologia , Camundongos , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Transdução de Sinais , Fatores de Transcrição da Família Snail/metabolismo
2.
Int J Mol Sci ; 21(14)2020 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-32708482

RESUMO

According to a widespread theory, thrombotic masses are not formed in the pulmonary artery (PA) but result from migration of blood clots from the venous system. This concept has prevailed in clinical practice for more than a century. However, a new technologic era has brought forth more diagnostic possibilities, and it has been shown that thrombotic masses in the PA could, in many cases, be found without any obvious source of emboli. Chronic obstructive pulmonary disease, asthma, sickle cell anemia, emergency and elective surgery, viral pneumonia, and other conditions could be complicated by PA thrombosis development without concomitant deep vein thrombosis (DVT). Different pathologies have different causes for local PA thrombotic process. As evidenced by experimental results and clinical observations, endothelial and platelet activation are the crucial mechanisms of this process. Endothelial dysfunction can impair antithrombotic function of the arterial wall through downregulation of endothelial nitric oxide synthase (eNOS) or via stimulation of adhesion receptor expression. Hypoxia, proinflammatory cytokines, or genetic mutations may underlie the procoagulant phenotype of the PA endothelium. Both endotheliocytes and platelets could be activated by protease mediated receptor (PAR)- and receptors for advanced glycation end (RAGE)-dependent mechanisms. Hypoxia, in particular induced by high altitudes, could play a role in thrombotic complications as a trigger of platelet activity. In this review, we discuss potential mechanisms of PA thrombosis in situ.


Assuntos
Plaquetas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Endotélio Vascular/metabolismo , Ativação Plaquetária/imunologia , Artéria Pulmonar/metabolismo , Embolia Pulmonar/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Trombose/metabolismo , Plaquetas/enzimologia , Plaquetas/imunologia , Hipóxia Celular , Micropartículas Derivadas de Células/patologia , Citocinas/metabolismo , Endotélio Vascular/enzimologia , Endotélio Vascular/imunologia , Proteína HMGB1/metabolismo , Humanos , Óxido Nítrico Sintase Tipo III/metabolismo , Artéria Pulmonar/enzimologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Embolia Pulmonar/genética , Embolia Pulmonar/fisiopatologia , Embolia Pulmonar/virologia , Receptor PAR-1/metabolismo , Fatores de Risco
3.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31996456

RESUMO

An increasing body of evidence suggests that bone marrow-derived myeloid cells play a critical role in the pathophysiology of pulmonary hypertension (PH). However, the true requirement for myeloid cells in PH development has not been demonstrated, and a specific disease-promoting myeloid cell population has not been identified. Using bone marrow chimeras, lineage labeling, and proliferation studies, we determined that, in murine hypoxia-induced PH, Ly6Clo nonclassical monocytes are recruited to small pulmonary arteries and differentiate into pulmonary interstitial macrophages. Accumulation of these nonclassical monocyte-derived pulmonary interstitial macrophages around pulmonary vasculature is associated with increased muscularization of small pulmonary arteries and disease severity. To determine if the sensing of hypoxia by nonclassical monocytes contributes to the development of PH, mice lacking expression of hypoxia-inducible factor-1α in the Ly6Clo monocyte lineage were exposed to hypoxia. In these mice, vascular remodeling and PH severity were significantly reduced. Transcriptome analyses suggest that the Ly6Clo monocyte lineage regulates PH through complement, phagocytosis, Ag presentation, and chemokine/cytokine pathways. Consistent with these murine findings, relative to controls, lungs from pulmonary arterial hypertension patients displayed a significant increase in the frequency of nonclassical monocytes. Taken together, these findings show that, in response to hypoxia, nonclassical monocytes in the lung sense hypoxia, infiltrate small pulmonary arteries, and promote vascular remodeling and development of PH. Our results demonstrate that myeloid cells, specifically cells of the nonclassical monocyte lineage, play a direct role in the pathogenesis of PH.


Assuntos
Hipertensão Pulmonar/imunologia , Hipóxia/complicações , Macrófagos Alveolares/imunologia , Monócitos/imunologia , Remodelação Vascular/imunologia , Animais , Antígenos Ly/metabolismo , Transplante de Medula Óssea , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Humanos , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/cirurgia , Hipóxia/imunologia , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Transplante de Pulmão , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Quimeras de Transplante/imunologia , Remodelação Vascular/genética
4.
Eur J Pharmacol ; 867: 172823, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31770525

RESUMO

Leukotriene B4 (LTB4) has been found to contribute to pulmonary arterial smooth muscle cells (PASMCs) proliferation and pulmonary arterial remodeling therefore the development of pulmonary arterial hypertension (PAH). Yet, the underlying molecular mechanisms remain poorly understood. The present study aims to address this issue. Our results demonstrate that LTB4 dose- and time-dependently induced proliferation of primary cultured rat PASMCs, this was accompanied with the activation of phosphatidylinositol-3-kinase/Akt (PI3K/Akt) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways, and consequent inactivation of glycogen synthase kinase-3ß (GSK-3ß), up-regulation of ß-catenin and induction of cyclin D1 expression. The presence of PI3K inhibitor (LY294002) or MEK inhibitor (U0126) or prior silencing of ß-catenin with siRNA suppressed LTB4-induced cyclin D1 up-regulation and PASMCs proliferation. In addition, inactivation or lack of GSK-3ß up-regulated ß-catenin and cyclin D1 in PASMCs. Taken together, our study indicates that activation of PI3K/Akt and ERK1/2 pathways mediates LTB4-induced PASMCs proliferation by modulating GSK-3ß/ß-catenin/cyclin D1 axis and suggests that targeting this pathway might have potential value in alleviating vascular remodeling and benefit PAH.


Assuntos
Hipertensão Pulmonar/imunologia , Leucotrieno B4/imunologia , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/patologia , Remodelação Vascular/imunologia , Animais , Butadienos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Ciclina D1/imunologia , Ciclina D1/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Hipertensão Pulmonar/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Cultura Primária de Células , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Remodelação Vascular/efeitos dos fármacos , beta Catenina/genética , beta Catenina/metabolismo
5.
Immunol Lett ; 218: 22-29, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31866401

RESUMO

OBJECTIVE: Pulmonary arterial hypertension (PAH) is an intractable complication in connective tissue diseases, but the pathological mechanisms responsible for progression remain obscure. This study aims to test whether patient IgG possesses biological activity promoting the migration of pulmonary artery smooth muscle cells (PASMCs). METHODS: Cell migration was estimated by lamellipodia formation and by utilizing a Boyden chamber method. The specificity of autoantibodies was established by western blotting, ELISA, and immunocytochemistry. The target antigen was investigated by mass spectrometry. RESULTS: IgG obtained from a patient with systemic lupus erythematosus (SLE) accompanied by PAH was found to promote lamellipodia formation and migration of PASMCs. The IgG bound to a ∼50 kDa protein expressed on the cell membrane, and in the cytoplasm and nucleus. This molecule was identified as enolase 1. Removal of enolase 1-binding antibodies from the IgG fraction, or treatment of the cells with an enolase inhibitor, significantly suppressed the migration of PASMCs. CONCLUSION: Patients with SLE may possess autoantibodies to enolase 1 which stimulate the migration of PASMCs and are likely to play a role in the progression of PAH.


Assuntos
Autoanticorpos/imunologia , Biomarcadores Tumorais/imunologia , Proteínas de Ligação a DNA/imunologia , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Miócitos de Músculo Liso/metabolismo , Fosfopiruvato Hidratase/imunologia , Hipertensão Arterial Pulmonar/complicações , Hipertensão Arterial Pulmonar/metabolismo , Proteínas Supressoras de Tumor/imunologia , Sequência de Aminoácidos , Autoanticorpos/sangue , Autoantígenos/imunologia , Biomarcadores Tumorais/química , Biomarcadores Tumorais/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Ligantes , Miócitos de Músculo Liso/imunologia , Fosfopiruvato Hidratase/química , Fosfopiruvato Hidratase/metabolismo , Ligação Proteica , Hipertensão Arterial Pulmonar/diagnóstico , Artéria Pulmonar/citologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
6.
Int J Chron Obstruct Pulmon Dis ; 14: 1323-1332, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31417249

RESUMO

Background: Previous studies have shown that the arterial wall is a potential source of inflammatory markers in COPD. Here, we sought to compare the expression of acute phase reactants (APRs) in COPD patients and controls both at the local (pulmonary arteries and lung parenchyma) and systemic (peripheral blood leukocytes and plasma) compartments. Methods: Consecutive patients undergoing elective surgery for suspected primary lung cancer were eligible for the study. Patients were categorized either as COPD or control group based on the spirometry results. Pulmonary arteries and lung parenchyma sections, peripheral blood leukocytes, and plasma samples were obtained from all participants. Gene expression levels of C-reactive protein (CRP) and serum amyloid A (SAA1, SAA2, and SAA4) were evaluated in tissue samples and peripheral blood leukocytes by reverse transciption-PCR. Plasma CRP and SAA protein levels were measured by enzyme-linked immunosorbent assays. Proteins were evaluated in paraffin-embedded lung tissues by immunohistochemistry. Results: A total of 40 patients with COPD and 62 controls were enrolled. We did not find significant differences in the gene expression between COPD and control group. Both CRP and SAA were overexpressed in the lung parenchyma compared with pulmonary arteries and peripheral blood leukocytes. The expression of SAA was significantly higher in the lung parenchyma than in the pulmonary artery (2-fold higher for SAA1 and SAA4, P=0.015 and P<0.001, respectively; 8-fold higher for SAA2, P<0.001) and peripheral blood leukocytes (16-fold higher for SAA1, 439-fold higher for SAA2, and 5-fold higher for SAA4; P<0.001). No correlation between plasma levels of inflammatory markers and their expression in the lung and peripheral blood leukocytes was observed. Conclusions: The expression of SAA in lung parenchyma is higher than in pulmonary artery and peripheral blood leukocytes. Notably, no associations were noted between lung expression of APRs and their circulating plasma levels, making the leakage of inflammatory proteins from the lung to the bloodstream unlikely. Based on these results, other potential sources of systemic inflammation in COPD (eg, the liver) need further scrutiny.


Assuntos
Reação de Fase Aguda , Pulmão , Linfócitos/imunologia , Artéria Pulmonar , Doença Pulmonar Obstrutiva Crônica , Proteína Amiloide A Sérica/análise , Proteínas de Fase Aguda/análise , Proteínas de Fase Aguda/imunologia , Reação de Fase Aguda/sangue , Reação de Fase Aguda/imunologia , Correlação de Dados , Feminino , Humanos , Pulmão/imunologia , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/patologia , Espirometria/métodos
8.
Int J Mol Sci ; 19(9)2018 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150544

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.


Assuntos
Micropartículas Derivadas de Células/imunologia , Exossomos/imunologia , Hipertensão Pulmonar/imunologia , Imunomodulação/imunologia , Macrófagos/imunologia , Células-Tronco Mesenquimais/imunologia , Animais , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/transplante , Exossomos/metabolismo , Exossomos/transplante , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipertensão Pulmonar/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Artéria Pulmonar/imunologia , Artéria Pulmonar/fisiopatologia
9.
PLoS One ; 13(7): e0199206, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990318

RESUMO

Although the accumulation of amyloidogenic proteins in neuroinflammatory conditions is generally considered pathologic, in a murine model of multiple sclerosis, amyloid-forming fibrils, comprised of hexapeptides, are anti-inflammatory. Whether these molecules modulate systemic inflammatory conditions remains unknown. We hypothesized that an amylin hexapeptide that forms fibrils can attenuate the systemic inflammatory response in a murine model of sepsis. To test this hypothesis, mice were pre-treated with either vehicle or amylin hexapeptide (20 µg) at 12 hours and 6 hours prior to intraperitoneal (i.p.) lipopolysaccharide (LPS, 20 mg/kg) administration. Illness severity and survival were monitored every 6 hours for 3 days. Levels of pro- (IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-10) cytokines were measured via ELISA at 1, 3, 6, 12, and 24 hours after LPS (i.p.). As a metric of lung injury, pulmonary artery endothelial cell (PAEC) barrier function was tested 24 hours after LPS administration by comparing lung wet-to-dry ratios, Evan's blue dye (EBD) extravasation, lung histology and caspase-3 activity. Compared to controls, pretreatment with amylin hexapeptide significantly reduced mortality (p<0.05 at 72 h), illness severity (p<0.05), and pro-inflammatory cytokine levels, while IL-10 levels were elevated (p<0.05). Amylin pretreatment attenuated LPS-induced lung injury, as demonstrated by decreased lung water and caspase-3 activity (p<0.05, versus PBS). Hence, in a murine model of systemic inflammation, pretreatment with amylin hexapeptide reduced mortality, disease severity, and preserved lung barrier function. Amylin hexapeptide may represent a novel therapeutic tool to mitigate sepsis severity and lung injury.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Proteínas Amiloidogênicas/farmacologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Pulmão/efeitos dos fármacos , Oligopeptídeos/farmacologia , Sepse/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/mortalidade , Proteínas Amiloidogênicas/síntese química , Animais , Caspase 3/genética , Caspase 3/imunologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica , Inflamação , Interferon gama/genética , Interferon gama/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Lipopolissacarídeos/administração & dosagem , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/síntese química , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Sepse/induzido quimicamente , Sepse/imunologia , Sepse/mortalidade , Análise de Sobrevida , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
10.
Physiol Rep ; 6(11): e13721, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29894584

RESUMO

The importance of myeloid cells in promoting neovascularization has been shown in a number of pathological settings in several organs. However, the specific role of macrophages in promoting systemic angiogenesis during pulmonary ischemia is not fully determined. Our past work suggested that cells of monocytic lineage contributed to systemic angiogenesis in the lung since clodronate-induced depletion of all macrophages resulted in attenuated neovascularization. Our current goals were to define the population of macrophages important for systemic vessel growth into the lung after the onset of pulmonary ischemia in mice. Interstitial macrophages (CD64+ MerTK+ CD11b+ ) increased significantly as did the percent of CD45+ Ly6G+ neutrophils 1 day after the induction of left lung ischemia, despite the fact there was limited cell recruitment due to complete obstruction of the left pulmonary artery in this ischemia model. Since both interstitial macrophages and neutrophils express CD11b, we used CD11b+ DTR mice and showed the critical role for these cells since CD11b+ depleted mice showed no systemic angiogenesis 7 days after the onset of ischemia when compared to control mice. Coculture of mouse aortic endothelial cells with macrophages showed increased proliferation relative to endothelial cells in culture without inflammatory cells, or pulmonary artery endothelial cells. We conclude that CD11b+ leukocytes, trapped within the lung at the onset of ischemia, contribute to growth factor release, and are critical for new blood vessel proliferation.


Assuntos
Isquemia/complicações , Pulmão/irrigação sanguínea , Pulmão/imunologia , Macrófagos/imunologia , Neovascularização Patológica , Animais , Antígeno CD11b/imunologia , Células Cultivadas , Citocinas/imunologia , Células Endoteliais/imunologia , Inflamação/imunologia , Leucócitos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Artéria Pulmonar/imunologia
11.
Respir Res ; 18(1): 64, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28424078

RESUMO

BACKGROUND: Interleukin 6 (IL-6) is a predictive factor of poor prognosis in patients with acute respiratory distress syndrome (ARDS). However, its acute pulmonary hemodynamic effects and role in lung injury have not been investigated in a clinically relevant murine model of ARDS. METHODS: We used adult C57Bl6 wild-type (WT) and IL-6 knock-out (IL-6KO) mice. The animals received intravenous recombinant human IL-6 (rHuIL-6) or vehicle followed by intratracheal lipopolysaccharide (LPS) or saline before undergoing low tidal volume mechanical ventilation (MV) for 5 h. Before sacrifice, right ventricular systolic pressure and cardiac output were measured and total pulmonary resistance was calculated. After sacrifice, lung inflammation, edema and injury were assessed with bronchoalveolar lavage (BAL) and histology. In other experiments, right ventricular systolic pressure was recorded during hypoxic challenges in uninjured WT mice pretreated with rHuIL-6 or rHuIL-6 + non-selective nitric oxide synthase inhibitor L-NAME or vehicle. RESULTS: IL-6KO(LPS+MV) mice showed a faster deterioration of lung elastic properties and more severe bronchoalveolar cellular inflammation as compared to WT(LPS+MV). Treatment with rHuIL-6 partially prevented this lung deterioration. Total pulmonary resistance was higher in IL-6KO(LPS+MV) mice and this increase was abolished in rHuIL-6-treated IL-6KO mice. Finally, rHuIL-6 reduced hypoxic pulmonary vasoconstriction in uninjured WT mice, an effect that was abolished by co-treatment with L-NAME. CONCLUSIONS: In a double-hit murine model of ARDS, IL-6 deficient mice experienced more severe bronchoalveolar cellular inflammation as compared to wild-type littermates. Furthermore, IL-6 deficiency caused marked acute pulmonary hypertension, which may be, at least partially, due to vasoactive mechanisms. A dysregulation of nitric oxide synthase may account for this observation, a hypothesis that will need to be investigated in future studies.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Anti-Inflamatórios/imunologia , Velocidade do Fluxo Sanguíneo/imunologia , Interleucina-6/imunologia , Artéria Pulmonar/imunologia , Circulação Pulmonar/imunologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Cardiovasc Pharmacol ; 67(5): 394-401, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26814361

RESUMO

Endothelial dysfunction and inflammation are believed to be 2 primary instigators of pulmonary arterial hypertension (PH). C1q/TNF-related protein 9 (CTRP9) plays important roles in anti-inflammation and improvement of epithelial function. However, the role of CTRP9 in the progression of PH remains still unclear. In this study, the role and mechanism of CTRP9 in the PH progression were explored. First, serum CTRP9 contents and CTRP9 mRNA expression in the pulmonary artery epithelial cells from patients with PH were detected. Our data on enzyme-linked immunosorbent assay and real-time quantitative Polymerase Chain Reaction showed that CTRP9 mRNA and protein content were markedly downregulated in the patients with PH. Then the pcDNA-CTRP9 expression vector or CTRP9 siRNA was transfected into the primary pulmonary artery epithelial cells from the patients with PH in vitro. CTRP9 overexpression significantly improved endothelial NOS protein expression and reduced the secretion of endothelin-1 (ET-1) and matrix metalloproteinase-2 (MMP-2), whereas knockdown of CTRP9 sharply reduced eNOS protein expression and promoted the secretion of ET-1 and MMP-2 in the cultured human epithelial cells. Moreover, the levels of phosphatidylinositol 3-kinase (PI3K) and pAkt were reduced in the epithelial cells and CTRP9 overexpression activated the PI3K/Akt pathway. CTRP9 could inhibit cell apoptosis and eNOS expression reduction in the cells pretreated with the PI3K/Akt inhibitor LY294002 and resist LY294002-induced ET-1 and MMP-2 secretion. Finally, to verify the role of CTRP9 in the progression of PH in vivo, the pcDNA-CTRP9 expression vector or CTRP9 siRNA was intravenously injected into rats with PH. Pulmonary arterial pressures of the rats were notably reduced by the pcDNA-CTRP9 injection and elevated by the CTRP9 siRNA injection. In conclusion, CTRP9 ameliorated PH by attenuating inflammation and improving endothelial cell survival and function.


Assuntos
Adiponectina/biossíntese , Células Epiteliais/imunologia , Glicoproteínas/biossíntese , Hipertensão Pulmonar/fisiopatologia , Artéria Pulmonar/imunologia , Adulto , Animais , Apoptose/efeitos dos fármacos , Cromonas/farmacologia , Endotelina-1/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral
13.
Transplantation ; 99(11): 2285-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26079744

RESUMO

BACKGROUND: Early growth response-1 (Egr-1) has been shown to be a trigger-switch transcription factor that is involved in lung ischemia-reperfusion injury (IRI). METHODS: Mouse lung transplants were performed in wild-type (WT) C57BL/6 and Egr1-knockout (KO) mice in the following donor → recipient combinations: WT → WT, KO → WT, WT → KO, and KO → KO to determine whether the presence of Egr-1 in the donor or recipient is the most critical factor for IRI. Pulmonary grafts were retrieved after 18 hours of ischemia after 4 hours of reperfusion. We analyzed graft function by analyzing arterial blood gas and histology in each combination and assessed the effects of Egr1 depletion on inflammatory cytokines that are regulated by Egr-1 as well on polymorphonuclear neutrophil (PMN) infiltration. RESULTS: Deletion of Egr1 improved pulmonary graft function in the following order of donor → recipient combinations: WT → WT < WT → KO < KO → WT < KO → KO. Polymerase chain reaction assays for Il1B, Il6, Mcp1, Mip2, Icam1, and Cox2 showed significantly lower expression levels in the KO → KO group than in the other groups. Immunohistochemistry demonstrated clear Egr-1 expression in the nuclei of pulmonary artery endothelial cells and PMN cytoplasm in the WT grafts. Flow cytometry analysis showed that Egr1 deletion reduced PMN infiltration and that the extent of reduction correlated with graft function. CONCLUSIONS: Both graft and recipient Egr-1 played a role in lung IRI, but the graft side contributed more to this phenomenon through regulation of PMN infiltration. Donor Egr-1 expression in pulmonary artery endothelial cells may play an important role in PMN infiltration, which results in IRI after lung transplantation.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Lesão Pulmonar/metabolismo , Transplante de Pulmão/efeitos adversos , Pulmão/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Quimiotaxia de Leucócito , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/deficiência , Proteína 1 de Resposta de Crescimento Precoce/genética , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Lesão Pulmonar/genética , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Fatores de Tempo
14.
Arch Pharm Res ; 38(11): 1983-91, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25981821

RESUMO

Poecilocoris lewisi (Korean name: "Kwangdaenolinjae") is a red-striped gold stink bug (insect) which has been used as a crude drug in traditional medicine of East Asia and Korea. In this study, ethanol extract and glycosaminoglycan from P. lewisi (Pl GAG), as an active substance among its components, were investigated for their potential anti-inflammatory actions. They were found to be a potent inducer of nitric oxide (NO) production from calf pulmonary artery endothelial (CPAE) cells and a stimulator of endothelial nitric oxide synthase in a dose-dependent manner. The anti-inflammatory activities were also evaluated by determining the level of adhesion molecules related to atherogenesis and pro-inflammatory cytokines, such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), secretory phospholipase A2, and prostaglandin E2, stimulated by tumor necrosis factor (TNF)-α in human umbilical vein endothelial cells (HUVEC). They also showed inhibitory effects on vascular endothelial growth factor (VEGF) production in HUVECs. Matrix metalloproteinases (MMP-2 and 9) were also inhibited by treatment with this extract or glycosaminoglycan. Furthermore, this GAG showed cytotoxicity against CT-26 colon cancer cells whereas having no cytotoxicity in CHO normal cells. Monosaccharide (amino, acidic, neutral monosaccharides) composition of used GAG was characterized by trimethylsilylated GC-MS analysis method.


Assuntos
Anti-Inflamatórios/farmacologia , Células Endoteliais/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , Heterópteros/metabolismo , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/isolamento & purificação , Células CHO , Bovinos , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Cricetinae , Cricetulus , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Células Endoteliais/imunologia , Cromatografia Gasosa-Espectrometria de Massas , Glicosaminoglicanos/administração & dosagem , Glicosaminoglicanos/isolamento & purificação , Células Endoteliais da Veia Umbilical Humana , Humanos , Óxido Nítrico/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Int Immunopharmacol ; 26(1): 188-93, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25601497

RESUMO

BACKGROUND: Baicalin has been shown to possess various pharmacological actions, a recent study revealed that baicalin can attenuate pulmonary hypertension and pulmonary vascular remodeling through the inhibition of pulmonary artery smooth muscle cell proliferation, however, the potential mechanism remains unexplored. In this study, we investigated the therapeutic effect of baicalin on a rat model of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) and attempted to further clarify the possible mechanisms underlying the anti-inflammatory. METHODS AND RESULTS: Our research showed that compared with MCT-induced PAH model rats, rats administered intragrastically with 100mg/kg baicalin showed the following after two weeks: the right ventricular systolic pressure (RVSP) and the right ventricle/left ventricle plus septum (RV/LV+S) ratio were lower (P<0.05); the intima thickening and luminal stenosis were improved (P<0.05); the mRNA levels of tumor necrosis factor alpha (TNF-α), interleukin-11ß (IL-1ß), IL-6, and endothelin-1 (ET-1) were obviously reduced by quantitative reverse transcription-polymerase chain reaction (qRT-PCR); the protein expression of transforming growth factor-ß1 (TGF-ß1), intercellular cell adhesion molecule-1 (ICAM-1) and nuclear factor-κB (NF-κB) were significantly decreased (P<0.05); and the expression of inhibitor of NF-κB (I-κB) was increased (P<0.05) through immunohistochemical and western blot. CONCLUSION: We studied the protective effects of baicalin against the lung and heart damage in experimental PAH rats; the therapeutic effects maybe through inhibiting vascular endothelial inflammatory response.


Assuntos
Anti-Inflamatórios/uso terapêutico , Flavonoides/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Monocrotalina/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Animais , Anti-Inflamatórios/administração & dosagem , Western Blotting , Citocinas/imunologia , Modelos Animais de Doenças , Flavonoides/administração & dosagem , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/patologia , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Ratos Wistar , Remodelação Vascular/efeitos dos fármacos
16.
Am J Physiol Lung Cell Mol Physiol ; 305(9): L651-64, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24039253

RESUMO

Endothelial cell (EC) inflammation is a central event in the pathogenesis of many pulmonary diseases such as acute lung injury and its more severe form acute respiratory distress syndrome. Alterations in actin cytoskeleton are shown to be crucial for NF-κB regulation and EC inflammation. Previously, we have described a role of actin binding protein cofilin in mediating cytoskeletal alterations essential for NF-κB activation and EC inflammation. The present study describes a dynamic mechanism in which LIM kinase 1 (LIMK1), a cofilin kinase, and slingshot-1Long (SSH-1L), a cofilin phosphatase, are engaged by procoagulant and proinflammatory mediator thrombin to regulate these responses. Our data show that knockdown of LIMK1 destabilizes whereas knockdown of SSH-1L stabilizes the actin filaments through modulation of cofilin phosphorylation; however, in either case thrombin-induced NF-κB activity and expression of its target genes (ICAM-1 and VCAM-1) is inhibited. Further mechanistic analyses reveal that knockdown of LIMK1 or SSH-1L each attenuates nuclear translocation and thereby DNA binding of RelA/p65. In addition, LIMK1 or SSH-1L depletion inhibited RelA/p65 phosphorylation at Ser(536), a critical event conferring transcriptional competency to the bound NF-κB. However, unlike SSH-1L, LIMK1 knockdown also impairs the release of RelA/p65 by blocking IKKß-dependent phosphorylation/degradation of IκBα. Interestingly, LIMK1 or SSH-1L depletion failed to inhibit TNF-α-induced RelA/p65 nuclear translocation and proinflammatory gene expression. Thus this study provides evidence for a novel role of LIMK1 and SSH-1L in selectively regulating EC inflammation associated with intravascular coagulation.


Assuntos
Células Endoteliais/metabolismo , Quinases Lim/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Pneumonia/metabolismo , Trombina/metabolismo , Fator de Transcrição RelA/metabolismo , Linhagem Celular , Cofilina 1/metabolismo , Coagulação Intravascular Disseminada/imunologia , Coagulação Intravascular Disseminada/metabolismo , Células Endoteliais/citologia , Células Endoteliais/imunologia , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/metabolismo , Quinases Lim/genética , NF-kappa B/metabolismo , Fosfoproteínas Fosfatases/genética , Fosforilação/fisiologia , Pneumonia/imunologia , Artéria Pulmonar/citologia , Artéria Pulmonar/imunologia , Vasculite/imunologia , Vasculite/metabolismo
17.
Br J Pharmacol ; 170(4): 835-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23889691

RESUMO

BACKGROUND AND PURPOSE: AVE 0991 (AVE) is a non-peptide compound, mimic of the angiotensin (Ang)-(1-7) actions in many tissues and pathophysiological states. Here, we have investigated the effect of AVE on pulmonary remodelling in a murine model of ovalbumin (OVA)-induced chronic allergic lung inflammation. EXPERIMENTAL APPROACH: We used BALB/c mice (6-8 weeks old) and induced chronic allergic lung inflammation by OVA sensitization (20 µg·mouse(-1) , i.p., four times, 14 days apart) and OVA challenge (1%, nebulised during 30 min, three times per·week, for 4 weeks). Control and AVE groups were given saline i.p and challenged with saline. AVE treatment (1 mg·kg(-1) ·per day, s.c.) or saline (100 µL·kg(-1) ·per day, s.c.) was given during the challenge period. Mice were anaesthetized 72 h after the last challenge and blood and lungs collected. In some animals, primary bronchi were isolated to test contractile responses. Cytokines were evaluated in bronchoalveolar lavage (BAL) and lung homogenates. KEY RESULTS: Treatment with AVE of OVA sensitised and challenged mice attenuated the altered contractile response to carbachol in bronchial rings and reversed the increased airway wall and pulmonary vasculature thickness and right ventricular hypertrophy. Furthermore, AVE reduced IL-5 and increased IL-10 levels in the BAL, accompanied by decreased Ang II levels in lungs. CONCLUSIONS AND IMPLICATIONS: AVE treatment prevented pulmonary remodelling, inflammation and right ventricular hypertrophy in OVA mice, suggesting that Ang-(1-7) receptor agonists are a new possibility for the treatment of pulmonary remodelling induced by chronic asthma.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Angiotensina I/farmacologia , Antiasmáticos/farmacologia , Asma/tratamento farmacológico , Imidazóis/farmacologia , Pulmão/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Veias Pulmonares/efeitos dos fármacos , Angiotensina II/metabolismo , Animais , Asma/induzido quimicamente , Asma/imunologia , Asma/metabolismo , Asma/fisiopatologia , Líquido da Lavagem Broncoalveolar/imunologia , Broncoconstrição/efeitos dos fármacos , Doença Crônica , Citocinas/metabolismo , Modelos Animais de Doenças , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/prevenção & controle , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mimetismo Molecular , Ovalbumina , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Veias Pulmonares/imunologia , Veias Pulmonares/metabolismo , Veias Pulmonares/fisiopatologia , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Tempo
18.
Panminerva Med ; 55(1): 93-7, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23474667

RESUMO

Both hyperthyroidism and hypothyroidism produce changes in cardiac contractility, myocardial oxygen consumption, cardiac output, blood pressure, and systemic or pulmonary vascular resistance. In almost all cases these cardiovascular changes are reversible when the underlying thyroid disorder is recognized and treated. Pulmonary hypertension (PAH) has been associated with thyroid dysfunction, but primarily with hyperthyroidism. The vast majority of patients with this form of PAH are usually older with toxic multinodular goitre. Data currently available suggest a direct influence of TH on pulmonary vasculature. Possible mechanisms include: 1) enhanced catecholamine sensitivity, causing pulmonary vasoconstriction, a reduction in pulmonary vascular compliance and an increase in vascular resistance; 2) increased metabolism of intrinsic pulmonary vasodilating substances (prostacyclin, nitric oxide); 3) decreased or impaired metabolism of vascontrictors (serotonin, endothelin 1 and tromboxane). In some cases (Graves's and Hashimoto's disease) and an autoimmune process inducing endothelial damage may play a key role. Future studies should focus on discovering the immunogenetic overlap between autoimmune thyroid diseases and PAH: common human leukocyte antigen alleles, susceptibility loci and so on. Such an understanding of the genetic and immune factors may ultimately lead to novel effective approaches in the treatment of PAH. At present, thyroid function tests should be considered in the investigation of all patients with PAH.


Assuntos
Hipertensão Pulmonar/etiologia , Artéria Pulmonar/fisiopatologia , Doenças da Glândula Tireoide/complicações , Glândula Tireoide/fisiopatologia , Animais , Pressão Arterial , Hipertensão Pulmonar Primária Familiar , Humanos , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipertireoidismo/complicações , Hipertireoidismo/fisiopatologia , Hipotireoidismo/complicações , Hipotireoidismo/fisiopatologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , Fatores de Risco , Doenças da Glândula Tireoide/imunologia , Doenças da Glândula Tireoide/metabolismo , Doenças da Glândula Tireoide/fisiopatologia , Glândula Tireoide/imunologia , Glândula Tireoide/metabolismo , Hormônios Tireóideos/metabolismo , Tireoidite Autoimune/complicações , Tireoidite Autoimune/fisiopatologia
19.
Am J Respir Cell Mol Biol ; 47(5): 688-97, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22842495

RESUMO

Oxidative stress is an important part of host innate immune response to foreign pathogens, such as bacterial LPS, but excessive activation of redox signaling may lead to pathologic endothelial cell (EC) activation and barrier dysfunction. Microtubules (MTs) play an important role in agonist-induced regulation of vascular endothelial permeability, but their impact in modulation of inflammation and EC barrier has not been yet investigated. This study examined the effects of LPS-induced oxidative stress on MT dynamics and the involvement of MTs in the LPS-induced mechanisms of Rho activation, EC permeability, and lung injury. LPS treatment of pulmonary vascular EC induced elevation of reactive oxygen species (ROS) and caused oxidative stress associated with EC hyperpermeability, cytoskeletal remodeling, and formation of paracellular gaps, as well as activation of Rho, p38 stress kinase, and NF-κB signaling, the hallmarks of endothelial barrier dysfunction. LPS also triggered ROS-dependent disassembly of the MT network, leading to activation of MT-dependent signaling. Stabilization of MTs with epothilone B, or inhibition of MT-associated guanine nucleotide exchange factor (GEF)-H1 activity by silencing RNA-mediated knockdown, suppressed LPS-induced EC barrier dysfunction in vitro, and attenuated vascular leak and lung inflammation in vivo. LPS disruptive effects were linked to activation of Rho signaling caused by LPS-induced MT disassembly and release of Rho-specific GEF-H1 from MTs. These studies demonstrate, for the first time, the mechanism of ROS-induced Rho activation via destabilization of MTs and GEF-H1-dependent activation of Rho signaling, leading to pulmonary EC barrier dysfunction and exacerbation of LPS-induced inflammation.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Permeabilidade Capilar , Microtúbulos/metabolismo , Estresse Oxidativo , Acetilcisteína/farmacologia , Lesão Pulmonar Aguda/imunologia , Animais , Antioxidantes/farmacologia , Células Cultivadas , Impedância Elétrica , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Epotilonas/farmacologia , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Humanos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/efeitos dos fármacos , Estabilidade Proteica , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Interferência de RNA , Fatores de Troca de Nucleotídeo Guanina Rho , Moduladores de Tubulina/farmacologia
20.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L1014-22, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22387295

RESUMO

In severe pulmonary arterial hypertension (PAH), vascular lesions are composed of phenotypically altered vascular and inflammatory cells that form clusters or tumorlets. Because macrophages are found in increased numbers in intravascular and perivascular space in human PAH, here we address the question whether macrophages play a role in pulmonary vascular remodeling and whether accumulation of macrophages in the lung vasculature could be compromised by the immune system. We used the mouse macrophage cell line RAW 264.7 because these cells are resistant to apoptosis, have high proliferative capacity, and resemble cells in the plexiform lesions that tend to pile up instead of maintaining a monolayer. Cells were characterized by immunocytochemistry with cell surface markers (Lycopersicon Esculentum Lectin, CD117, CD133, FVIII, CD31, VEGFR-2, and S100). Activated, but not quiescent, T cells were able to suppress RAW 264.7 cell proliferative and migration activity in vitro. The carboxyfluorescein diacetate-labeled RAW 264.7 cells were injected into the naïve Sprague Dawley (SD) rat and athymic nude rat. Twelve days later, cells were found in the lung vasculature of athymic nude rats that lack functional T cells, contributing to vascular remodeling. No labeled RAW 264.7 cells were detected in the lungs of immune-competent SD rats. Our data demonstrate that T cells can inhibit in vitro migration and in vivo accumulation of macrophage-like cells.


Assuntos
Comunicação Celular/imunologia , Pulmão/irrigação sanguínea , Macrófagos/citologia , Artéria Pulmonar/citologia , Linfócitos T/citologia , Animais , Biomarcadores/análise , Linhagem Celular , Movimento Celular/imunologia , Proliferação de Células , Hipertensão Pulmonar Primária Familiar , Fluoresceínas , Corantes Fluorescentes , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Imuno-Histoquímica , Pulmão/citologia , Pulmão/imunologia , Macrófagos/imunologia , Macrófagos/transplante , Masculino , Camundongos , Modelos Biológicos , Artéria Pulmonar/imunologia , Ratos , Ratos Nus , Ratos Sprague-Dawley , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA