Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 618(7966): 827-833, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37258670

RESUMO

The immune phenotype of a tumour is a key predictor of its response to immunotherapy1-4. Patients who respond to checkpoint blockade generally present with immune-inflamed5-7 tumours that are highly infiltrated by T cells. However, not all inflamed tumours respond to therapy, and even lower response rates occur among tumours that lack T cells (immune desert) or that spatially exclude T cells to the periphery of the tumour lesion (immune excluded)8. Despite the importance of these tumour immune phenotypes in patients, little is known about their development, heterogeneity or dynamics owing to the technical difficulty of tracking these features in situ. Here we introduce skin tumour array by microporation (STAMP)-a preclinical approach that combines high-throughput time-lapse imaging with next-generation sequencing of tumour arrays. Using STAMP, we followed the development of thousands of arrayed tumours in vivo to show that tumour immune phenotypes and outcomes vary between adjacent tumours and are controlled by local factors within the tumour microenvironment. Particularly, the recruitment of T cells by fibroblasts and monocytes into the tumour core was supportive of T cell cytotoxic activity and tumour rejection. Tumour immune phenotypes were dynamic over time and an early conversion to an immune-inflamed phenotype was predictive of spontaneous or therapy-induced tumour rejection. Thus, STAMP captures the dynamic relationships of the spatial, cellular and molecular components of tumour rejection and has the potential to translate therapeutic concepts into successful clinical strategies.


Assuntos
Neoplasias , Linfócitos T , Microambiente Tumoral , Humanos , Imunoterapia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Linfócitos T/imunologia , Fenótipo , Fibroblastos , Monócitos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico
2.
Nature ; 544(7648): 105-109, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28329764

RESUMO

Platelets are critical for haemostasis, thrombosis, and inflammatory responses, but the events that lead to mature platelet production remain incompletely understood. The bone marrow has been proposed to be a major site of platelet production, although there is indirect evidence that the lungs might also contribute to platelet biogenesis. Here, by directly imaging the lung microcirculation in mice, we show that a large number of megakaryocytes circulate through the lungs, where they dynamically release platelets. Megakaryocytes that release platelets in the lungs originate from extrapulmonary sites such as the bone marrow; we observed large megakaryocytes migrating out of the bone marrow space. The contribution of the lungs to platelet biogenesis is substantial, accounting for approximately 50% of total platelet production or 10 million platelets per hour. Furthermore, we identified populations of mature and immature megakaryocytes along with haematopoietic progenitors in the extravascular spaces of the lungs. Under conditions of thrombocytopenia and relative stem cell deficiency in the bone marrow, these progenitors can migrate out of the lungs, repopulate the bone marrow, completely reconstitute blood platelet counts, and contribute to multiple haematopoietic lineages. These results identify the lungs as a primary site of terminal platelet production and an organ with considerable haematopoietic potential.


Assuntos
Plaquetas/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Pulmão/irrigação sanguínea , Pulmão/citologia , Animais , Medula Óssea , Linhagem da Célula , Feminino , Pulmão/anatomia & histologia , Masculino , Megacariócitos/citologia , Camundongos , Microcirculação , Contagem de Plaquetas , Trombocitopenia/patologia
3.
Blood ; 124(17): 2625-34, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25143486

RESUMO

Evidence is emerging that platelets are major contributors to innate immune responses in conditions such as acute lung injury (ALI). Platelets form heterotypic aggregates with neutrophils, and we hypothesized that lipoxin mediators regulate formation of neutrophil-platelet aggregates (NPA) and that NPA significantly contribute to ALI. Lipopolysaccharide (LPS)-induced lung injury was accompanied by platelet sequestration, activation, intra-alveolar accumulation, and NPA formation within both blood and alveolar compartments. Using lung intravital microscopy, we observed the dynamic formation of NPA during physiologic conditions, which sharply increased with ALI. Aspirin (ASA) treatment significantly reduced lung platelet sequestration and activation, NPA formation, and lung injury. ASA treatment increased levels of ASA-triggered lipoxin (ATL; 15-epi-lipoxin A4), and blocking the lipoxin A4 receptor (ALX) with a peptide antagonist (Boc2) or using ALX knockouts (Fpr2/3(-/-)) reversed this protection. LPS increased NPA formation in vitro, which was reduced by ATL, and engagement of ALX by ATL on both neutrophils and platelets was necessary to prevent aggregation. In a model of transfusion-related acute lung injury (TRALI), Boc2 also reversed ASA protection, and treatment with ATL in both LPS and TRALI models protected from ALI. We conclude that ATL regulates neutrophil-platelet aggregation and that platelet-neutrophil interactions are a therapeutic target in lung injury.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Aspirina/farmacologia , Lipoxinas/metabolismo , Neutrófilos/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/genética , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Comunicação Celular/efeitos dos fármacos , Contagem de Leucócitos , Lipopolissacarídeos/farmacologia , Lipoxinas/sangue , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Análise de Sobrevida , Reação Transfusional
4.
Am J Respir Crit Care Med ; 191(4): 455-63, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25485813

RESUMO

RATIONALE: Primary graft dysfunction (PGD) causes early mortality after lung transplantation and may contribute to late graft failure. No effective treatments exist. The pathogenesis of PGD is unclear, although both neutrophils and activated platelets have been implicated. We hypothesized that neutrophil extracellular traps (NETs) contribute to lung injury in PGD in a platelet-dependent manner. OBJECTIVES: To study NETs in experimental models of PGD and in lung transplant patients. METHODS: Two experimental murine PGD models were studied: hilar clamp and orthotopic lung transplantation after prolonged cold ischemia (OLT-PCI). NETs were assessed by immunofluorescence microscopy and ELISA. Platelet activation was inhibited with aspirin, and NETs were disrupted with DNaseI. NETs were also measured in bronchoalveolar lavage fluid and plasma from lung transplant patients with and without PGD. MEASUREMENTS AND MAIN RESULTS: NETs were increased after either hilar clamp or OLT-PCI compared with surgical control subjects. Activation and intrapulmonary accumulation of platelets were increased in OLT-PCI, and platelet inhibition reduced NETs and lung injury, and improved oxygenation. Disruption of NETs by intrabronchial administration of DNaseI also reduced lung injury and improved oxygenation. In bronchoalveolar lavage fluid from human lung transplant recipients, NETs were more abundant in patients with PGD. CONCLUSIONS: NETs accumulate in the lung in both experimental and clinical PGD. In experimental PGD, NET formation is platelet-dependent, and disruption of NETs with DNaseI reduces lung injury. These data are the first description of a pathogenic role for NETs in solid organ transplantation and suggest that NETs are a promising therapeutic target in PGD.


Assuntos
Armadilhas Extracelulares/metabolismo , Transplante de Pulmão , Neutrófilos/metabolismo , Disfunção Primária do Enxerto/imunologia , Animais , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Ativação Plaquetária , Disfunção Primária do Enxerto/sangue , Disfunção Primária do Enxerto/patologia
5.
Basic Res Cardiol ; 110(2): 8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25604439

RESUMO

Chronic activation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway contributes to vascular inflammation and atherosclerosis by inducing expression of genes involved in cell proliferation, differentiation and migration. We aimed to investigate whether enforced expression of negative regulators, the suppressors of cytokine signaling (SOCS1 and SOCS3), inhibits harmful JAK/STAT-mediated responses and affects atherosclerosis in apolipoprotein E knockout mice. Adenovirus-mediated SOCS1 transgene expression impaired the onset and progression of atherosclerosis without impact on lipid profile, whereas SOCS3 was only effective on early atherosclerosis. Mechanistically, SOCS gene delivery, primarily SOCS1, attenuated STAT1 and STAT3 activation and reduced the expression of STAT-dependent genes (chemokine/chemokine receptors, adhesion molecules, pro-inflammatory cytokines and scavenger receptors) in aortic tissue. Furthermore, atherosclerotic plaques exhibit a more stable phenotype characterized by lower lipids, T cells and M1 macrophages and higher M2 macrophages and collagen. Atheroprotection was accompanied by a systemic alteration of T helper- and T regulatory-related genes and a reduced activation state of circulating monocytes. In vascular smooth muscle cells and macrophages, SOCS gene delivery inhibited cytokine-induced STAT activation, pro-inflammatory gene expression, cell migration and proliferation. In conclusion, targeting SOCS proteins, predominantly SOCS1, to suppress pathological mechanisms involved in atheroma plaque progression and destabilization could be an interesting anti-atherosclerotic strategy.


Assuntos
Aterosclerose/patologia , Inflamação/patologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Inflamação/genética , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Supressoras da Sinalização de Citocina/genética , Transdução Genética
6.
Am J Pathol ; 182(5): 1910-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23597852

RESUMO

Atherosclerosis is a chronic inflammatory disease of the arterial wall. NF-κB is a major regulator of inflammation that controls the expression of many genes involved in atherogenesis. Activated NF-κB was detected in human atherosclerotic plaques, and modulation of NF-κB inflammatory activity limits disease progression in mice. Herein, we investigate the anti-inflammatory and atheroprotective effects of a cell-permeable peptide containing the NF-κB nuclear localization sequence (NLS). In vascular smooth muscle cells and macrophages, NLS peptide specifically blocked the importin α-mediated nuclear import of NF-κB and prevented lipopolysaccharide-induced pro-inflammatory gene expression, cell migration, and oxidative stress. In experimental atherosclerosis (apolipoprotein E-knockout mice fed a high-fat diet), i.p., 0.13 µmol/day NLS peptide administration for 5 weeks attenuated NF-κB activation in atherosclerotic plaques. NLS peptide significantly inhibited lesion development at both early (age 10 weeks) and advanced (age 28 weeks) stages of atherosclerosis in mice, without affecting serum lipid levels. Plaques from NLS-treated mice contained fewer macrophages of pro-inflammatory M1 subtype than those from respective untreated controls. By contrast, the relative smooth muscle cell and collagen content was increased, indicating a more stable plaque phenotype. NLS peptide also attenuated pro-inflammatory gene expression and oxidative stress in aortic lesions. Our study demonstrates that targeting NF-κB nuclear translocation hampers inflammation and atherosclerosis development and identifies cell-permeable NLS peptide as a potential anti-atherosclerotic agent.


Assuntos
Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/induzido quimicamente , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Carioferinas/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Sinais de Localização Nuclear/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Placa Aterosclerótica , Transporte Proteico/efeitos dos fármacos
7.
Stroke ; 44(3): 699-707, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23422087

RESUMO

BACKGROUND AND PURPOSE: We have previously reported that intravenous injection of high-density lipoproteins (HDLs) was neuroprotective in an embolic stroke model. We hypothesized that HDL vasculoprotective actions on the blood-brain barrier (BBB) may decrease hemorrhagic transformation-associated with tissue plasminogen activator (tPA) administration in acute stroke. METHODS: We used tPA alone or in combination with HDLs in vivo in 2 models of focal middle cerebral artery occlusion (MCAO) (embolic and 4-hour monofilament MCAO) and in vitro in a model of BBB. Sprague-Dawley rats were submitted to MCAO, n=12 per group. The rats were then randomly injected with tPA (10 mg/kg) or saline with or without human plasma purified-HDL (10 mg/kg). The therapeutic effects of HDL and BBB integrity were assessed blindly 24 hours later. The integrity of the BBB was also tested using an in vitro model of human cerebral endothelial cells under oxygen-glucose deprivation. RESULTS: tPA-treated groups had significantly higher mortality and rate of hemorrhagic transformation at 24 hours in both MCAO models. Cotreatment with HDL significantly reduced stroke-induced mortality versus tPA alone (by 42% in filament MCAO, P=0.009; by 73% in embolic MCAO, P=0.05) and tPA-induced intracerebral parenchymal hematoma (by 92% in filament MCAO, by 100% in embolic MCAO; P<0.0001). This was consistent with an improved BBB integrity. In vitro, HDLs decreased oxygen-glucose deprivation-induced BBB permeability (P<0.05) and vascular endothelial cadherin disorganization. CONCLUSIONS: HDL injection decreased tPA-induced hemorrhagic transformation in rat models of MCAO. Both in vivo and in vitro results support the vasculoprotective action of HDLs on BBB under ischemic conditions.


Assuntos
Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/prevenção & controle , Lipoproteínas HDL/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual/efeitos adversos , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Linhagem Celular , Hemorragia Cerebral/epidemiologia , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fibrinolíticos/efeitos adversos , Fibrinolíticos/uso terapêutico , Humanos , Técnicas In Vitro , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Injeções Intravenosas , Lipoproteínas HDL/administração & dosagem , Lipoproteínas HDL/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Risco , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/mortalidade
8.
Brain ; 135(Pt 9): 2826-37, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22961553

RESUMO

Alzheimer's disease is a severely debilitating disease of high and growing proportions. Hypercholesterolaemia is a key risk factor in sporadic Alzheimer's disease that links metabolic disorders (diabetes, obesity and atherosclerosis) with this pathology. Hypercholesterolaemia is associated with increased levels of immunoglobulin G against oxidized lipoproteins. Patients with Alzheimer's disease produce autoantibodies against non-brain antigens and specific receptors for the constant Fc region of immunoglobulin G have been found in vulnerable neuronal subpopulations. Here, we focused on the potential role of Fc receptors as pathological players driving hypercholesterolaemia to Alzheimer's disease. In a well-established model of hypercholesterolaemia, the apolipoprotein E knockout mouse, we report increased brain levels of immunoglobulin G and upregulation of activating Fc receptors, predominantly of type IV, in neurons susceptible to amyloid ß accumulation. In these mice, gene deletion of γ-chain, the common subunit of activating Fc receptors, prevents learning and memory impairments without influencing cholesterolaemia and brain and serum immunoglobulin G levels. These cognition-protective effects were associated with a reduction in synapse loss, tau hyperphosphorylation and intracellular amyloid ß accumulation both in cortical and hippocampal pyramidal neurons. In vitro, activating Fc receptor engagement caused synapse loss, tau hyperphosphorylation and amyloid ß deposition in primary neurons by a mechanism involving mitogen-activated protein kinases and ß-site amyloid precursor protein cleaving enzyme 1. Our results represent the first demonstration that immunoglobulin G Fc receptors contribute to the development of hypercholesterolaemia-associated features of Alzheimer's disease and suggest a new potential target for slowing or preventing Alzheimer's disease in hypercholesterolaemic patients.


Assuntos
Doença de Alzheimer/genética , Transtornos Cognitivos/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de IgG/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Comportamento Animal/fisiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Hipocampo/patologia , Camundongos , Camundongos Knockout , Neurônios/patologia , Receptores de IgG/metabolismo
9.
J Am Soc Nephrol ; 23(9): 1518-27, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22859852

RESUMO

Among patients with diabetes, increased production of immunoglobulins against proteins modified by diabetes is associated with proteinuria and cardiovascular risk, suggesting that immune mechanisms may contribute to the development of diabetes complications, such as nephropathy. We investigated the contribution of IgG Fcγ receptors to diabetic renal injury in hyperglycemic, hypercholesterolemic mice. We used streptozotocin to induce diabetes in apolipoprotein E-deficient mice and in mice deficient in both apolipoprotein E and γ-chain, the common subunit of activating Fcγ receptors. After 15 weeks, the mice lacking Fcγ receptors had significantly less albuminuria and renal hypertrophy, despite similar degrees of hyperglycemia and hypercholesterolemia, immunoglobulin production, and glomerular immune deposits. Moreover, diabetic Fcγ receptor-deficient mice had less mesangial matrix expansion, inflammatory cell infiltration, and collagen and α-smooth muscle actin content in their kidneys. Accordingly, expression of genes involved in leukocyte infiltration, fibrosis, and oxidative stress was significantly reduced in diabetic kidneys and in mesangial cells cultured from Fcγ receptor-deficient mice. In summary, preventing the activation of Fcγ receptors alleviates renal hypertrophy, inflammation, and fibrosis in hypercholesterolemic mice with diabetes, suggesting that modulating Fcγ receptor signaling may be renoprotective in diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/prevenção & controle , Receptores de IgG/deficiência , Estreptozocina/efeitos adversos , Actinas/metabolismo , Albuminúria/epidemiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Colágeno/metabolismo , Nefropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Hipertrofia/epidemiologia , Incidência , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/genética , Transdução de Sinais/fisiologia
10.
J Am Soc Nephrol ; 21(5): 763-72, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20185635

RESUMO

Activation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) is an important mechanism by which hyperglycemia contributes to renal damage, suggesting that modulation of this pathway may prevent renal and vascular complications of diabetes. Here, we investigated the involvement of suppressors of cytokine signaling (SOCS) as intracellular negative regulators of JAK/STAT activation in diabetic nephropathy. In a rat model, inducing diabetes resulted in JAK/STAT activation and increased expression of SOCS1 and SOCS3. In humans, we observed increased expression of glomerular and tubulointerstitial SOCS proteins in biopsies of patients with diabetic nephropathy. In vitro, high concentrations of glucose activated JAK/STAT/SOCS in human mesangial and tubular cells. Overexpression of SOCS reversed the glucose-induced activation of the JAK/STAT pathway, expression of STAT-dependent genes (chemokines, growth factors, and extracellular matrix proteins), and cell proliferation. In vivo, intrarenal delivery of adenovirus expressing SOCS1 and SOCS3 to diabetic rats significantly improved renal function and reduced renal lesions associated with diabetes, such as mesangial expansion, fibrosis, and influx of macrophages. SOCS gene delivery also decreased the activation of STAT1 and STAT3 and the expression of proinflammatory and profibrotic proteins in the diabetic kidney. In summary, these results provide direct evidence for a link between the JAK/STAT/SOCS axis and hyperglycemia-induced cell responses in the kidney. Suppression of the JAK/STAT pathway by increasing intracellular SOCS proteins may have therapeutic potential in diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/metabolismo , Hiperglicemia/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adenoviridae , Idoso , Animais , Estudos de Casos e Controles , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Nefropatias Diabéticas/terapia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Wistar
11.
FASEB J ; 23(9): 3129-39, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19417089

RESUMO

Various studies using proteomic approaches have shown that HDL can carry many proteins other than its constitutive apolipoprotein A-I (apoA-I). Using mass spectrometry and Western blotting, we showed the presence of alpha(1)-antitrypsin (AAT) (SERPINA1, serpin peptidase inhibitor, clade A, an elastase inhibitor) in HDL, isolated either by ultracentrifugation or by selected-affinity immunosorption using an anti-apoA-I column. Furthermore, we report that HDL possesses potent antielastase activity. We further showed that only HDL but not LDL is able to bind AAT. HDL-associated AAT was able to inhibit extracellular matrix degradation, cell detachment, and apoptosis induced by elastase in human vascular smooth muscle cells (VSMCs) and in mammary artery cultured ex vivo. Degradation of fibronectin by elastase used as a marker of pericellular proteolysis was prevented by addition of HDL. Elastase present in aortic abdominal aneurysm (AAA) thrombus samples was also able to induce apoptosis of VSMCs in culture. This phenomenon was prevented by addition of HDL but not of LDL. Finally, we report that the proportion of AAT in HDL isolated from patients with an AAA is decreased relative to that from matched control subjects, suggesting a reduced capacity of HDL to inhibit elastase in these patients. In conclusion, our data provide evidence of a new potential antiatherogenic property of HDL attributable to AAT and its antielastase activity.


Assuntos
Anoikis , Lipoproteínas HDL/química , Miócitos de Músculo Liso/citologia , Elastase Pancreática/antagonistas & inibidores , alfa 1-Antitripsina/fisiologia , Aterosclerose/prevenção & controle , Humanos , Lipoproteínas HDL/análise , Músculo Liso Vascular , Ligação Proteica , Proteínas/análise
12.
Arterioscler Thromb Vasc Biol ; 29(4): 525-31, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19164812

RESUMO

OBJECTIVE: Suppressors of cytokine signaling (SOCS) proteins are intracellular regulators of receptor signal transduction, mainly Janus kinase/signal transducers and activators of transcription (JAK/STAT). We investigated the effects of SOCS modulation on the JAK/STAT-dependent responses in vascular cells, and their implication in atherosclerotic plaque development. METHODS AND RESULTS: Immunohistochemistry in human plaques revealed a high expression of SOCS1 and SOCS3 by vascular smooth muscle cells (VSMCs) and macrophages in the inflammatory region of the shoulders, when compared to the fibrous area. SOCS were also increased in aortic lesions from apoE(-/-) mice. In cultured VSMCs, endothelial cells, and monocytes, SOCS1 and SOCS3 were transiently induced by proinflammatory cytokines, proatherogenic lipoproteins, and immune molecules. Furthermore, overexpression of SOCS suppressed STAT activation and reduced inflammatory gene expression and cell growth, whereas SOCS knockdown increased these cell responses. In vivo, antisense oligodeoxynucleotides targeting SOCS3 exacerbated the atherosclerotic process in apoE(-/-) mice by increasing the size, leukocyte content, and chemokine expression in the lesions. CONCLUSIONS: SOCS expressed in atherosclerotic lesions are key regulators of vascular cell responses. Activation of this endogenous antiinflammatory pathway might be of interest in the treatment of atherosclerosis.


Assuntos
Aterosclerose/enzimologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Aorta/enzimologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/patologia , Artérias Carótidas/enzimologia , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/enzimologia , Fosforilação , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Tempo , Transfecção
13.
J Clin Invest ; 130(4): 2041-2053, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31961827

RESUMO

Cystic fibrosis (CF) lung disease is characterized by an inflammatory response that can lead to terminal respiratory failure. The cystic fibrosis transmembrane conductance regulator (CFTR) is mutated in CF, and we hypothesized that dysfunctional CFTR in platelets, which are key participants in immune responses, is a central determinant of CF inflammation. We found that deletion of CFTR in platelets produced exaggerated acute lung inflammation and platelet activation after intratracheal LPS or Pseudomonas aeruginosa challenge. CFTR loss of function in mouse or human platelets resulted in agonist-induced hyperactivation and increased calcium entry into platelets. Inhibition of the transient receptor potential cation channel 6 (TRPC6) reduced platelet activation and calcium flux, and reduced lung injury in CF mice after intratracheal LPS or Pseudomonas aeruginosa challenge. CF subjects receiving CFTR modulator therapy showed partial restoration of CFTR function in platelets, which may be a convenient approach to monitoring biological responses to CFTR modulators. We conclude that CFTR dysfunction in platelets produces aberrant TRPC6-dependent platelet activation, which is a major driver of CF lung inflammation and impaired bacterial clearance. Platelets and TRPC6 are what we believe to be novel therapeutic targets in the treatment of CF lung disease.


Assuntos
Plaquetas/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Pulmão/metabolismo , Pneumonia Bacteriana/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Animais , Plaquetas/patologia , Fibrose Cística/genética , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Knockout , Ativação Plaquetária/genética , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia , Canal de Cátion TRPC6/genética , Canal de Cátion TRPC6/metabolismo
14.
Circ Res ; 99(11): 1188-96, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17053192

RESUMO

IgG Fc receptors (FcgammaRs) play a role in activating the immune system and in maintaining peripheral tolerance, but their role in atherosclerosis is unknown. We generated double-knockout (DKO) mice by crossing apolipoprotein E-deficient mice (apoE(-/-)) with FcgammaR gamma chain-deficient mice (gamma(-/-)). The size of atherosclerotic lesions along the aorta was approximately 50% lower in DKO compared with apoE(-/-) control mice, without differences in serum lipid levels. The macrophage and T-cell content of lesions in the DKO were reduced by 49+/-6% and 56+/-8%, respectively, compared with the content in apoE(-/-) lesions. Furthermore, the expression of monocyte chemoattractant protein-1 (MCP-1), RANTES (Regulated on Activated Normal T-cell Expressed and Secreted), and intercellular adhesion molecule-1 (ICAM-1) and the activation of nuclear factor-kappaB (NF-kappaB) were significantly reduced in aortic lesions from DKO mice. In vitro, vascular smooth muscle cells (VSMCs) from both gamma(-/-) and DKO mice failed to respond to immune complexes, as shown by impaired chemokine expression and NF-kappaB activation. ApoE(-/-) mice have higher levels of activating FcgammaRI and FcgammaRIIIA, and inhibitory FcgammaRIIB, compared with wild-type mice. The DKO mice express only the inhibitory FcgammaRIIB receptor. We conclude that FcgammaR deficiency limits development and progression of atherosclerosis. In addition to leukocytes, FcgammaR activation in VSMCs contributes to the inflammatory process, in part, by regulating chemokine expression and leukocyte invasion of the vessel wall. These results underscore the critical role of FcgammaRs in atherogenesis and support the use of immunotherapy in the treatment of this disease.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Receptores de IgG/deficiência , Animais , Complexo Antígeno-Anticorpo/metabolismo , Aterosclerose/etiologia , Células Cultivadas , Quimiocinas/metabolismo , Regulação para Baixo , Feminino , Expressão Gênica , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Receptores de IgG/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 26(8): 1864-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16741149

RESUMO

OBJECTIVE: Activation of transcription factor NF-kappaB is an important step in the development of vascular damage, because it controls inducible genes, including many inflammatory mediators. The pharmacological modulation of this process is the main objective in the design of new therapies for atherosclerosis. In this work we analyzed the effects of the natural compound parthenolide (PTN), an NF-kappaB inhibitor. METHODS AND RESULTS: In vascular smooth muscle cells (VSMCs) and monocytes stimulated with lipopolysaccharide (LPS), nontoxic doses of PTN reduced IkappaBalpha degradation, NF-kappaB activation, and MCP-1 expression, without inhibiting AP-1 and MAPK. In apoE mice, treatment with low (2 mg/kg, 20 weeks), medium (4 mg/kg, 10 weeks), and high (10 mg/kg, 10 weeks) dose of PTN reduced the size of aortic lesion, decreased macrophage, and increased VSMC content in the lesions. Treated mice showed reduced serum levels of MCP-1 and attenuated NF-kappaB activity, but not AP-1, in the lesions. Moreover, PTN affects neither apoptotic cell death nor oxidative stress in cultured cells and mice. CONCLUSIONS: NF-kappaB inhibition by PTN retards atherosclerotic lesions in apoE mice, by reducing lesion size and changing plaque composition. This natural compound could represent a novel therapeutic approach to inflammation during vascular damage.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Inflamação/etiologia , Inflamação/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Sesquiterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/etiologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Gorduras na Dieta/administração & dosagem , Humanos , Camundongos
16.
Atherosclerosis ; 253: 75-80, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27591364

RESUMO

BACKGROUND AND AIMS: HDL-cholesterol concentration is a reliable negative risk factor for acute cerebral infarction (ACI). Beyond quantitative aspects, our aim was to determine whether lipoprotein profiles and HDL functionality were altered at the acute phase of ischemic stroke. METHODS: Blood was taken from ACI patients within 4.5 h of symptom onset. Lipoproteins were separated by electrophoresis for determination of particle size. HDLs were isolated from plasma of patients (n = 10) and controls (n = 10) by ultracentrifugation. The relative amounts of paraoxonase 1 (PON1), α1antitrypsin (AAT) and myeloperoxidase (MPO) were determined by Western blot. HDL functional assays were performed on human-brain endothelial cells stimulated with TNFα. RESULTS: Stroke patients had higher proportion of large HDL particles relative to controls (37.8 ± 11.8 vs. 28.4 ± 6.6, p = 0.04). HDLs from patients contained significantly less ApoA1 (1.63 ± 0.42 vs. 2.54 ± 0.71 mg/mL, p = 0.0026) and PON1 (4598 ± 1921 vs. 6598 ± 1127 AU, p = 0.01) than those from controls, whereas MPO and AAT were more abundant in HDLs isolated from ACI patients (respectively 3657 ± 1457 vs. 2012 ± 1234 and 3347 ± 917 vs. 2472 ± 470 AU, p = 0.014 and p = 0.015). HDLs reduced the expression of VCAM1, MCP1 and MMP3 mRNA induced by TNFα in blood-brain barrier endothelial cells. HDLs from patients were less effective in inhibiting TNFα-induced transcription of these genes (respectively 38.6 vs. 55.6% for VCAM1, p = 0.047, 44 vs. 48.1% for MCP1, p = 0.015 and 70 vs. 74% for MMP3, p = 0.024). CONCLUSIONS: ACI may be associated with a modified distribution of HDL particles (increased proportion of large particles) and HDL-binding proteins, resulting in an inappropriate protection of endothelial cells under ischemic conditions.


Assuntos
Lipoproteínas HDL/sangue , Acidente Vascular Cerebral/sangue , Fator de Necrose Tumoral alfa/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Apolipoproteína A-I/sangue , Arildialquilfosfatase/sangue , Infarto Encefálico/sangue , Isquemia Encefálica/sangue , Estudos de Casos e Controles , Eletroforese , Endotélio Vascular/patologia , Feminino , Humanos , Inflamação , Masculino , Pessoa de Meia-Idade , Tamanho da Partícula , Peroxidase/sangue , RNA Mensageiro/metabolismo , alfa 1-Antitripsina/sangue
17.
Curr Drug Targets Inflamm Allergy ; 4(3): 341-51, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16101544

RESUMO

The mesangium occupies a central anatomical position in the glomerulus, and also plays an important regulatory role in immune-mediated glomerular diseases, with an active participation in the response to local inflammation. In general, the mesangial cell responses to the pathological stimuli are associated with the main events of glomerular injury: leukocyte infiltration, cell proliferation and fibrosis. Leukocyte migration and infiltration into the glomerulus is responsible for the initiation and amplification of glomerular injury, and is mediated by adhesion molecules and chemokines, which can be locally synthesized by mesangial cells. The increase in mesangial cell number is also due to proliferation of intrinsic mesangial cell population. Regulatory mechanisms of mesangial cell replication include a complex array of factors which control cell proliferation, survival and apoptosis. Mesangial matrix accumulation leading to glomerulosclerosis, is a consequence of an imbalance between matrix production and degradation, and is controlled by growth factors and pro-inflammatory cytokines. The initial phase of immune-mediated glomerular inflammation depends on the interaction of immune complexes with specific Fc receptors in infiltrating leukocytes and resident mesangial cells, the ability of immune complexes to activate complement system, and on local inflammatory processes. Activated mesangial cells then produce many inflammatory mediators leading to amplification of the injury. This review will focus on the biological functions of mesangial cells that contribute to glomerular injury, with special attention to immune-mediated glomerulonephritis. Furthermore, new therapies based on the pathophysiology of the mesangial cell that are being developed in experimental models are also proposed.


Assuntos
Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/patologia , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/patologia , Animais , Proteínas do Sistema Complemento/fisiologia , Fibrose , Mesângio Glomerular/imunologia , Mesângio Glomerular/fisiopatologia , Glomerulonefrite/imunologia , Glomerulonefrite/fisiopatologia , Humanos , Mediadores da Inflamação/fisiologia , Receptores Fc/efeitos dos fármacos
18.
Bio Protoc ; 5(12)2015.
Artigo em Inglês | MEDLINE | ID: mdl-27390765

RESUMO

The intratracheal instillation technique is used to deliver a variety of agents to the lungs ranging from pathogens (bacteria, viruses), toxins, to therapeutic agents. To model lung inflammation and injury, LPS can be administrated via intranasal, intratracheal, or aerosol approaches. Each technique has its limitations. The intratracheal technique can involve the non-invasive instillation method (via the oro-tracheal route) or a direct injection into the trachea. Here, we describe an optimized method for direct visual instillation of LPS via the non-invasive oro-tracheal route.

19.
Bio Protoc ; 5(12)2015 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-29104887

RESUMO

Transfusion-related acute lung injury (TRALI) is defined as acute lung injury that occurs within 6 hours of a blood product transfusion. TRALI continues to be a leading cause of transfusion-related mortality and we have developed a mouse model of TRALI to better understand the mechanisms by which injury occurs and to test therapeutic approaches. Our model is a two-event model based on immune priming and the challenge of BALB/c wild-type mice with cognate MHC Class I monoclonal antibody (MHC I mAb). Immune priming with LPS mimics the primed state of recipients (first event) that is important for the development of TRALI. Donor HLA antibodies are frequently implicated in TRALI reactions, and cognate MHC Class I antibody (second event) produces acute lung injury in primed animals. Here, we describe a detailed protocol with high reproducibility within animals.

20.
PLoS One ; 8(6): e66754, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23805273

RESUMO

Immunity contributes to arterial inflammation during atherosclerosis. Oxidized low-density lipoproteins induce an autoimmune response characterized by specific antibodies and immune complexes in atherosclerotic patients. We hypothesize that specific Fcγ receptors for IgG constant region participate in atherogenesis by regulating the inflammatory state of lesional macrophages. In vivo we examined the role of activating Fcγ receptors in atherosclerosis progression using bone marrow transplantation from mice deficient in γ-chain (the common signaling subunit of activating Fcγ receptors) to hyperlipidemic mice. Hematopoietic deficiency of Fcγ receptors significantly reduced atherosclerotic lesion size, which was associated with decreased number of macrophages and T lymphocytes, and increased T regulatory cell function. Lesions of Fcγ receptor deficient mice exhibited increased plaque stability, as evidenced by higher collagen and smooth muscle cell content and decreased apoptosis. These effects were independent of changes in serum lipids and antibody response to oxidized low-density lipoproteins. Activating Fcγ receptor deficiency reduced pro-inflammatory gene expression, nuclear factor-κB activity, and M1 macrophages at the lesion site, while increasing anti-inflammatory genes and M2 macrophages. The decreased inflammation in the lesions was mirrored by a reduced number of classical inflammatory monocytes in blood. In vitro, lack of activating Fcγ receptors attenuated foam cell formation, oxidative stress and pro-inflammatory gene expression, and increased M2-associated genes in murine macrophages. Our study demonstrates that activating Fcγ receptors influence the macrophage phenotypic balance in the artery wall of atherosclerotic mice and suggests that modulation of Fcγ receptor-mediated inflammatory responses could effectively suppress atherosclerosis.


Assuntos
Aterosclerose/fisiopatologia , Macrófagos/metabolismo , Receptores de IgG/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/mortalidade , Transplante de Medula Óssea , Colágeno/metabolismo , Modelos Animais de Doenças , Hiperlipidemias/patologia , Lipídeos/sangue , Lipoproteínas LDL/sangue , Lipoproteínas LDL/imunologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Placa Aterosclerótica/patologia , Receptores de IgG/deficiência , Linfócitos T/citologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa