Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(11)2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35682871

RESUMO

The endothelium has a fundamental role in the cardiovascular complications of coronavirus disease 2019 (COVID-19). Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) particularly affects endothelial cells. The virus binds to the angiotensin-converting enzyme 2 (ACE-2) receptor (present on type 2 alveolar cells, bronchial epithelial cells, and endothelial cells), and induces a cytokine storm. The cytokines tumor necrosis factor alpha, interleukin-1 beta, and interleukin-6 have particular effects on endothelial cells-leading to endothelial dysfunction, endothelial cell death, changes in tight junctions, and vascular hyperpermeability. Under normal conditions, apoptotic endothelial cells are removed into the bloodstream. During COVID-19, however, endothelial cells are detached more rapidly, and do not regenerate as effectively as usual. The loss of the endothelium on the luminal surface abolishes all of the vascular responses mediated by the endothelium and nitric oxide production in particular, which results in greater contractility. Moreover, circulating endothelial cells infected with SARS-CoV-2 act as vectors for viral dissemination by forming clusters that migrate into the circulation and reach distant organs. The cell clusters and the endothelial dysfunction might contribute to the various thromboembolic pathologies observed in COVID-19 by inducing the formation of intravascular microthrombi, as well as by triggering disseminated intravascular coagulation. Here, we review the contributions of endotheliopathy and endothelial-cell-derived extracellular vesicles to the pathogenesis of COVID-19, and discuss therapeutic strategies that target the endothelium in patients with COVID-19.


Assuntos
COVID-19 , Doenças Vasculares , COVID-19/complicações , Citocinas/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , SARS-CoV-2 , Doenças Vasculares/metabolismo
2.
Sci Rep ; 11(1): 7464, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33811249

RESUMO

Chronic kidney disease (CKD) worsens ischemic stroke severity in both patients and animals. In mice, these poorer functional outcomes are associated with decreased brain activity of AMP-activated protein kinase (AMPK), a molecule that recently emerged as a potential therapeutic target for ischemic stroke. The antidiabetic drug metformin, a well-known activator of AMPK, has improved stroke outcomes in diabetic patients with normal renal function. We investigated whether chronic metformin pre-conditioning can rescue AMPK activity and prevent stroke damage in non-diabetic mice with CKD. Eight-week-old female C57BL/6J mice were assigned to CKD or SHAM groups. CKD was induced through right kidney cortical electrocautery, followed by left total nephrectomy. Mice were then allocated to receive metformin (200 mg/kg/day) or vehicle for 5 weeks until stroke induction by transient middle cerebral artery occlusion (tMCAO). The infarct volumes were lower in CKD mice exposed to metformin than in vehicle-treated CKD mice 24 h after tMCAO. Metformin pre-conditioning of CKD mice improved their neurological score, grip strength, and prehensile abilities. It also enhanced AMPK activation, reduced apoptosis, increased neuron survival and decreased microglia/macrophage M1 signature gene expression as well as CKD-induced activation of the canonical NF-κB pathway in the ischemic lesions of CKD mice.


Assuntos
Metformina/uso terapêutico , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/prevenção & controle , Adenilato Quinase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Peso Corporal , Infarto Encefálico/sangue , Infarto Encefálico/complicações , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/genética , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Gliose/sangue , Gliose/complicações , Gliose/tratamento farmacológico , Infarto da Artéria Cerebral Média/sangue , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/genética , Precondicionamento Isquêmico , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Metformina/sangue , Metformina/farmacologia , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Modelos Biológicos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/genética , Acidente Vascular Cerebral/genética
3.
Sci Rep ; 9(1): 6432, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015533

RESUMO

Ischemic stroke is highly prevalent in chronic kidney disease (CKD) patients and has been associated with a higher risk of neurological deterioration and in-hospital mortality. To date, little is known about the processes by which CKD worsens ischemic stroke. This work aimed to investigate the cellular and molecular mechanism associated with ischemic stroke severity in an in vivo model of CKD. CKD was induced through right kidney cortical electrocautery in 8-week-old female C57BL/6 J mice followed by left total nephrectomy. Transient middle cerebral artery occlusion (tMCAO) was performed 6 weeks after left nephrectomy. Twenty-four hours after tMCAO, the infarct volumes were significantly wider in CKD than in SHAM mice. CKD mice displayed decreased neuroscore, impaired ability to remain on rotarod device, weaker muscular strength and decreased prehensile score. Apoptosis, neuronal loss, glial cells recruitment and microglia/macrophages M1 signature genes CD32, CD86, IL-1ß, IL-6, MCP1 and iNOS were significantly increased within ischemic lesions of CKD mice. This effect was associated with decreased AMP kinase phosphorylation and increased activation of the NFΚB pathway. Pharmacological targeting of AMP kinase activity, which is known to block microglia/macrophages M1 polarization, appears promising to improve stroke recovery in CKD.


Assuntos
Isquemia Encefálica/fisiopatologia , Córtex Renal/metabolismo , Debilidade Muscular/fisiopatologia , Neurônios/metabolismo , Insuficiência Renal Crônica/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Adenilato Quinase/genética , Adenilato Quinase/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose/genética , Isquemia Encefálica/complicações , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Eletrocoagulação , Feminino , Regulação da Expressão Gênica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Córtex Renal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Debilidade Muscular/complicações , Debilidade Muscular/genética , Debilidade Muscular/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/patologia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Teste de Desempenho do Rota-Rod , Índice de Gravidade de Doença , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo
4.
Cancer Lett ; 444: 147-161, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30583074

RESUMO

Overall survival of patients with metastatic non-small cell lung cancer (NSCLC) has significantly improved with platinum-based salt treatments and recently with targeted therapies and immunotherapies. However, treatment failure occurs due to acquired or emerging tumor resistance. We developed a monoclonal antibody against the proform of neurotensin (LF-NTS mAb) that alters the homeostasis of tumors overexpressing NTSR1. Neurotensin is frequently overexpressed along with its high affinity receptor (NTSR1) in tumors from epithelial origins. This ligand/receptor complex contributes to the progression of many tumor types by activation of the cellular effects involved in tumor progression (proliferation, survival, migration, and invasion). We demonstrate that LF-NTS mAb operates on the plasticity of tumor cells overexpressing NTSR1 and lowers their aggressiveness. The mAb enables the restoration of platinum-based therapies responsiveness, while also decreasing metastatic processes. Efficacy dosage with long-term treatment showed no obvious adverse events, while demonstrating improvement in the performance status. Our data suggests that LF-NTS mAb is an ideal candidate to be safely added to the conventional standard of care in order to improve its efficacy.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neurotensina/antagonistas & inibidores , Receptores de Neurotensina/antagonistas & inibidores , Adenocarcinoma de Pulmão/imunologia , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/patologia , Animais , Antineoplásicos/farmacologia , Apoptose , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neurotensina/imunologia , Neurotensina/metabolismo , Prognóstico , Receptores de Neurotensina/imunologia , Receptores de Neurotensina/metabolismo , Estudos Retrospectivos , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Glia ; 66(8): 1709-1723, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29624734

RESUMO

Neuroinflammation and iron accumulation are hallmarks of a variety of adult neurodegenerative diseases. In Sanfilippo syndrome (mucopolysaccharidosis type III, MPSIII, a pediatric neurodegenerative disease that shares some features with adult neurodegenerative diseases), the progressive accumulation of heparan sulfate oligosaccharides (HSOs) induces microglia and astrocytes to produce pro-inflammatory cytokines leading to severe neuroinflammation. The objectives of the present study were (1) to measure the local iron concentration and to assess iron metabolism in the brain of a MPSIIIB murine model and (2) to identify the brain cells involved in this accumulation. We found that iron accumulation in MPSIIIB mice primarily affected the cerebral cortex where hepcidin levels were higher than in wild-type mice, and increased with aging. This increase was correlated with low expression of ferroportin 1 (FPN1), and thus brain iron retention. Moreover, we showed in vitro that HSOs are directly responsible for the production of hepcidin and the relative decrease in FPN1 expression when added to cultures of microglia and, to a lesser extent, to cultures of astrocytes. In contrast, no significant differences were observed in neurons. Hepcidin induction results from activation of the TLR4 pathway and STAT3 signaling, and leads to iron retention within microglia. Our results show that microglia have a key role in cerebral hepcidin overexpression and thus in the brain iron accumulation observed in the MPSIIIB model.


Assuntos
Encéfalo/metabolismo , Ferro/metabolismo , Microglia/metabolismo , Mucopolissacaridose III/metabolismo , Animais , Astrócitos/metabolismo , Camundongos Knockout , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo
6.
Eur J Med Chem ; 122: 702-722, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27451257

RESUMO

An important part of pathogenesis of Alzheimer's disease (AD) is attributed to the contribution of AGE (Advanced Glycation Endproducts) and ALE (Advanced Lipid peroxidation Endproducts). In order to attenuate the progression of AD, we designed a new type of molecules that consist of two trapping parts for reactive carbonyl species (RCS) and reactive oxygen species (ROS), precursors of AGE and ALE, respectively. These molecules also chelate transition metals, the promoters of ROS formation. In this paper, synthesis of the new AGE/ALE inhibitors and evaluation of their physicochemical and biological properties (carbonyl trapping capacity, antioxidant activity, Cu(2+)-chelating capacity, cytotoxicity and protective effect against in vitro MGO-induced apoptosis in the model AD cell-line PC12) are described. It is found that compounds 40b and 51e possess promising therapeutic potentials for treating AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Diaminas/química , Diaminas/farmacologia , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Peroxidação de Lipídeos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cobre/química , Diaminas/uso terapêutico , Malondialdeído/metabolismo , Células PC12 , Aldeído Pirúvico/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
7.
J Cell Physiol ; 230(2): 347-55, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24976589

RESUMO

Inorganic phosphate (Pi) is an essential nutrient to all living organisms. Nevertheless, hyperphosphatemia is now recognized as a risk factor for cardiovascular events and mortality in chronic kidney disease (CKD) patients. To our knowledge, the mechanisms by which elevated Pi alters smooth muscle cell proliferation have been poorly addressed. Therefore, in this study, we investigated the effects of Pi on cell cycle regulation and apoptosis in human aortic smooth muscle cells (HAoSMC). HAoSMC were treated with physiologic (1 mM) or high (2 and 3 mM) Pi concentrations. We showed that Pi not only decreased significantly cell viability (P < 0.001) but also induced apoptosis of HAoSMC. Moreover, Pi treatment blocked G1/S cell cycle progression by increasing cell number in G0/G1 phase up to 82.4 ± 3.4% for 3 mM vs 76.2 ± 3.1% for control (P < 0.01) while decreasing cell number in S phase. Accordingly, this was associated with a decrease protein expression of cyclin E and its associated CDK (CDK2), and phosphorylated retinoblastoma protein. Moreover, we observed an increase of protein expression of cell cycle inhibitors p15, p21, and p27. Interestingly, we also found that induction of cell cycle arrest was partially dependent on phosphate uptake. Our results demonstrated that Pi reduced HAoSMC proliferation by inducing apoptosis and cell cycle arrest. Indeed, we showed for the first time that Pi affected HAoSMC cell cycle by blocking G1/S progression. These findings would be useful for a better understanding of molecular mechanisms involved in vascular complications observed in CKD patients.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Fosfatos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Fosfatos/metabolismo , Proteína do Retinoblastoma/metabolismo
8.
Biochem Pharmacol ; 80(9): 1373-85, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20691164

RESUMO

Photodynamic therapy (PDT) is a poor treatment option for nodular basal cell carcinomas and squamous cell carcinomas. As a result, the search for new photosensitizers with better effectiveness is of current interest. The photocytotoxicity of conjugates (P-R) of a water-soluble tri-cationic porphyrin (P-H) having similar efficiency of production of singlet oxygen, the PDT cytotoxin, has been assessed in vitro. Links between uptake, intracellular localization, photooxidative stress, photocytotoxicity and ability to induce programmed cell death are established. Conjugates bearing methyl (P-Me), Di-O-isopropylidene-(-d-galactopyranosyl (P-OGal) or N,N'-dicyclohexylureidooxycarbonyl (P-DDC) chains are efficiently taken-up by proliferating NCTC 2544 keratinocytes. The relative order of photocytotoxicity is P-OGal >P-DDC=P-Me≫P-H. The photocytotoxic potential of P-Me, P-OGal and P-DDC equals that of endogenous protoporphyrin IX induced by δ-aminolevulinic acid or its esters, the pro-drugs currently employed for PDT of skin lesions. Microfluorometry shows that P-Me, P-OGal, and P-DDC localize in endocytotic or pinocytotic vesicles but not in mitochondria or nucleus. Absence of annexin V binding, caspase activation or chromatin condensation suggests that cell photosensitization by P-R does not induce apoptosis. On the other hand, P-OGal photocytotoxicity correlates with appearance of multiple vesicles that have hallmarks of autophagy compartments, being decorated with the marker LC3 in cells transfected with an expression vector encoding GFP-LC3. p38 and JNK phosphorylation and inhibition of ERK1/2 phosphorylation suggest close relationship between mortality of NCTC 2544 keratinocytes and MAPK pathway impairment. Given their potentially easy formulation, water-soluble P-R are promising powerful photosensitizers for PDT of skin lesions.


Assuntos
Apoptose/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Fotoquimioterapia , Porfirinas/farmacologia , Anexina A5/análise , Autofagia/efeitos dos fármacos , Caspases/metabolismo , Células Cultivadas , Humanos , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Estresse Oxidativo
9.
Arterioscler Thromb Vasc Biol ; 27(3): 614-20, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17170367

RESUMO

OBJECTIVE: Based on their role in T-cell homing into nonlymphoid tissue, we examined the role of the homeostatic chemokines CCL19 and CCL21 and their common receptor CCR7 in coronary artery disease (CAD). METHODS AND RESULTS: We performed studies in patients with stable (n=40) and unstable (n=40) angina and healthy controls (n=20), in vitro studies in T-cells and macrophages, and studies in apolipoprotein-E-deficient (ApoE-/-) mice and human atherosclerotic carotid plaques. We found increased levels of CCL19 and CCL21 within the atherosclerotic lesions of the ApoE-/- mice, in human atherosclerotic carotid plaques, and in plasma of CAD patients. Whereas strong CCR7 expression was seen in T-cells from murine and human atherosclerotic plaques, circulating T-cells from angina patients showed decreased CCR7 expression. CCL19 and CCL21 promoted an inflammatory phenotype in T-cells and macrophages and increased matrix metalloproteinase (MMP) and tissue factor levels in the latter cell type. Although aggressive statin therapy increased CCR7 and decreased CCL19/CCL21 levels in peripheral blood from CAD patients, conventional therapy did not. CONCLUSIONS: The abnormal regulation of CCL19 and CCL21 and their common receptor in atherosclerosis could contribute to disease progression by recruiting T-cells and macrophages to the atherosclerotic lesions and by promoting inflammatory responses in these cells.


Assuntos
Quimiocinas CC/metabolismo , Doença das Coronárias/sangue , Doença das Coronárias/tratamento farmacológico , Ácidos Heptanoicos/uso terapêutico , Pirróis/uso terapêutico , Sinvastatina/uso terapêutico , Angioplastia Coronária com Balão/métodos , Animais , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Atorvastatina , Biópsia por Agulha , Células Cultivadas , Quimiocina CCL19 , Quimiocina CCL21 , Quimiocinas CC/genética , Doença das Coronárias/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Leucócitos Mononucleares , Camundongos , Camundongos Transgênicos , Prognóstico , RNA Mensageiro/análise , Receptores CCR7 , Receptores de Quimiocinas/metabolismo , Valores de Referência , Fatores de Risco , Sensibilidade e Especificidade , Resultado do Tratamento
10.
Hepatology ; 43(5): 932-42, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16628670

RESUMO

Cell entry of hepatitis C virus, pseudoparticles (HCVpp) and cell culture grown virus (HCVcc), requires the interaction of viral glycoproteins with CD81 and other as yet unknown cellular factors. One of these is likely to be the scavenger receptor class B type I (SR-BI). To further understand the role of SR-BI, we examined the effect of SR-BI ligands on HCVpp and HCVcc infectivity. Oxidized low-density lipoprotein (oxLDL), but not native LDL, potently inhibited HCVpp and HCVcc cell entry. Pseudoparticles bearing unrelated viral glycoproteins or bovine viral diarrhea virus were not affected. A dose-dependent inhibition was observed for HCVpp bearing diverse viral glycoproteins with an approximate IC50 of 1.5 microg/mL apolipoprotein content, which is within the range of oxLDL reported to be present in human plasma. The ability of lipoprotein components to bind to target cells associated with their antiviral activity, suggesting a mechanism of action which targets a cell surface receptor critical for HCV infection of the host cell. However, binding of soluble E2 to SR-BI or CD81 was not affected by oxLDL, suggesting that oxLDL does not act as a simple receptor blocker. At the same time, oxLDL incubation altered the biophysical properties of HCVpp, suggesting a ternary interaction of oxLDL with both virus and target cells. In conclusion, the SR-BI ligand oxLDL is a potent cell entry inhibitor for a broad range of HCV strains in vitro. These findings suggest that SR-BI is an essential component of the cellular HCV receptor complex.


Assuntos
Carcinoma Hepatocelular/virologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/patogenicidade , Lipoproteínas LDL/farmacologia , Neoplasias Hepáticas/virologia , Antígenos CD36/fisiologia , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia , Oxirredução , Células Tumorais Cultivadas
11.
Arterioscler Thromb Vasc Biol ; 26(5): 1169-76, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16484596

RESUMO

OBJECTIVE: Lipid-loaded macrophage-derived foam cells populate atherosclerotic lesions and produce many pro-inflammatory and plaque-destabilizing factors. An excessive accumulation of extensively oxidized low-density lipoprotein (OxLDL) or free cholesterol (FC), both of which are believed to be major lipid components of macrophages in advanced lesions, rapidly induces apoptosis in macrophages. Indeed, there is evidence of macrophage death in lesions, but how the surviving macrophages avoid death induced by OxLDL, FC, and other factors is not known. METHODS AND RESULTS: Minimally oxidized LDL (mmLDL), which is an early product of progressive LDL oxidation in atherosclerotic lesions, countered OxLDL-induced or FC-induced apoptosis and stimulated macrophage survival both in cell culture and in vivo. DNA fragmentation and caspase-3 activity in OxLDL-treated peritoneal macrophages were significantly reduced by coincubation with mmLDL. In a separate set of experiments, mmLDL significantly reduced annexin V binding to macrophages in which apoptosis was induced by FC loading. In both cellular models, mmLDL activated a pro-survival PI3K/Akt signaling pathway, and PI3K inhibitors, wortmannin and LY294002, eliminated the pro-survival effect of mmLDL. Immunohistochemical examination demonstrated phospho-Akt in murine atherosclerotic lesions. CONCLUSIONS: Minimally oxidized LDL, an early form of oxidized LDL in atherosclerotic lesions, may contribute to prolonged survival of macrophage foam cells in lesions via a PI3K/Akt-dependent mechanism.


Assuntos
Apoptose/efeitos dos fármacos , Aterosclerose/patologia , Colesterol/toxicidade , Lipoproteínas LDL/metabolismo , Macrófagos/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Feminino , Lipoproteínas LDL/toxicidade , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores de LDL/fisiologia , Transdução de Sinais
12.
Arterioscler Thromb Vasc Biol ; 25(6): 1213-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15718493

RESUMO

OBJECTIVE: Innate immune responses to oxidized low-density lipoprotein LDL (LDL) regulate the development of atherosclerosis. We demonstrated previously that an early form of oxidized LDL, minimally modified LDL (mmLDL), triggers cytoskeletal rearrangements in macrophages via CD14 and Toll-like receptor 4 (TLR4)/MD-2. Because lipopolysaccharide (LPS) activation of TLR4 leads to proinflammatory gene expression, in this study, we asked whether mmLDL also induced proinflammatory signaling. METHODS AND RESULTS: We studied cytokine secretion and signaling in J774 and primary peritoneal macrophages stimulated with mmLDL, which was prepared by incubating LDL with cells expressing human 15-lipoxygenase. MmLDL stimulated robust phosphoinositide 3-kinase (PI3K) activation, and Akt and extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, which exceeded that induced by LPS. On the other hand, although mmLDL induced nuclear factor kappaB (NF-kappaB) p65 translocation to the nucleus, there was no detectable NF-kappaB activation. However, mmLDL induced early mRNA and protein expression of the cytokines MIP-2, MCP-1, tumor necrosis factor-alpha, and interleukin-6. Chemokine MIP-2 but not MCP-1 secretion depended on TLR4/MyD88, ERK1/2, and PI3K signaling. In turn, TLR4 regulated phosphorylation of ERK1/2 but not of Akt, suggesting that mmLDL-induced PI3K activation is TLR4 independent. CONCLUSIONS: In macrophages, mmLDL activates TLR4-dependent and -independent signaling pathways, resulting in secretion of proinflammatory cytokines. These results provide new insights into the inflammatory origins of atherosclerosis.


Assuntos
Aterosclerose/imunologia , Citocinas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneais/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos de Diferenciação/genética , Aterosclerose/metabolismo , Células CHO , Quimiocina CCL4 , Quimiocina CXCL2 , Quimiocinas/genética , Quimiocinas/metabolismo , Cricetinae , Citocinas/genética , Feminino , Fibroblastos/citologia , Proteínas Inflamatórias de Macrófagos/genética , Proteínas Inflamatórias de Macrófagos/metabolismo , Macrófagos Peritoneais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator 88 de Diferenciação Mieloide , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Imunológicos/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
13.
J Lipid Res ; 46(5): 969-76, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15722561

RESUMO

We have previously shown that CD36 recognizes oxidation products of phospholipids on oxidized LDL (OxLDL) such as 1-palmitoyl-2-(5'-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC). The current study was designed to examine whether the phosphocholine (PC) headgroup in POVPC constitutes an obligatory binding target for CD36. To examine the contribution of PC in the binding of POVPC to CD36, we used well-defined synthetic oxidized phospholipids (OxPLs) cross-linked to BSA or to a hexapeptide. The OxPL adducts were then tested for their ability to bind to CD36-transfected cells and for their ability to inhibit OxLDL binding to CD36. Both POVPC-BSA and POVPC-peptide adducts were high-affinity ligands for CD36 and potent inhibitors of OxLDL binding. Enzymatic removal of the entire PC moiety of the POVPC-peptide, or of the choline headgroup alone, as well as substitution of the choline headgroup by ethanolamine abrogated the inhibitory activity of POVPC. Interestingly, PC by itself or cross-linked to BSA did not show any intrinsic competition activity. In conclusion, our data demonstrate that the PC headgroup of OxPL alone is sufficient for binding to CD36, but only if presented in the correct conformation as in OxPL of OxLDL or as in POVPC-peptide adducts.


Assuntos
Antígenos CD36/metabolismo , Fosforilcolina/metabolismo , Animais , Células COS , Ligantes , Metabolismo dos Lipídeos , Lipoproteínas LDL/metabolismo , Espectrometria de Massas , Peptídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA