Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Regul Integr Comp Physiol ; 321(5): R639-R654, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34431382

RESUMO

After an ischemic event, there is activation of fibroblasts leading to scar formation. It is critical to limit the profibrotic remodeling and activate the reparative remodeling phase to limit cardiac diastolic dysfunction. Mesenchymal stem cell (MSC) exosomes offer significant protection against ischemia-related systolic dysfunction. Here, we studied if MSC exosomes would offer protection against profibrotic events in mouse hearts subjected to acute ischemia [1 h left coronary artery (LCA) occlusion] or chronic ischemia (7 days LCA occlusion). After acute ischemia, there was activation of inflammatory signals, more in the peri-infarct than in the infarct area, in the saline (vehicle)-treated mice. At the same time, there was expression of cardiac remodeling signals (vimentin, collagens-1 and -3, and fibronectin), more in the infarct area. Treatment with MSC exosomes before LCA ligation suppressed inflammatory signals during acute and chronic ischemia. Furthermore, exosome treatment promoted pro-reparative cardiac extracellular matrix (ECM) remodeling in both infarct and peri-infarct areas by suppressing fibronectin secretion and by modulating collagen secretion to reduce fibrotic scar formation through altered cellular signaling pathways. Proteomics study revealed intense expression of IL-1ß and activation of profibrotic signals in the saline-treated hearts and their suppression in MSC exosome-treated hearts. To our knowledge, this is the first report on the infarct and peri-infarct area proteomics of ischemic mice hearts to explain MSC exosome-mediated suppression of scar formation in the ischemic mouse hearts.


Assuntos
Exossomos/transplante , Fibroblastos/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Isquemia Miocárdica/cirurgia , Miocárdio/metabolismo , Proteoma , Proteômica , Remodelação Ventricular , Animais , Western Blotting , Linhagem Celular , Movimento Celular , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Exossomos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/patologia , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Masculino , Espectrometria de Massas , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia
2.
Mol Cell Biochem ; 476(4): 1691-1704, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33423165

RESUMO

Mesenchymal stem cell (MSC) exosomes may limit cardiac injury, and even reverse cardiac damage in animal models of ischemia. To understand exosome-mediated improvement in cardiac function we examined the proteomic alternations in the MSC exosome-treated mice hearts subjected to left coronary artery (LCA) ligation, with particular emphasis on peri-infarct areas. At 7 days after LCA ligation, left ventricular end systolic thickness, infarct size and survival of mice were studied. Mass spectrometric analysis of infarct and peri-infarct areas was carried out. Expression of inflammatory markers (LOX-1 and NLRP3) and cell death markers (Bax, Bcl-2, Caspases 1 and 3 and GSDMD) were investigated by Western blots and immunofluorescence. Proteomic analysis of the infarct and peri-infarct areas in saline-treated hearts revealed differentially expressed proteins involved in inflammation and apoptotic cell death, while showing depletion of processes governing cell death. Exosome treatment significantly improved the proteomic profile in both infarct and peri-infarct areas, more so in the peri-infarct areas. The infarct size was smaller (9 ± 1%), and cardiac contractile function (fractional shortening) was preserved in the exosome-treated mice (28 ± 2%). Survival of exosome-treated mice was also better. White blood cell accumulation in and around the infarct area, expression of LOX-1 and NLRP3 inflammasome, and markers of cell death (cleaved Caspase-3, Caspase-1, GSDMD, Bcl-2 and Bax) were dramatically reduced by MSC exosome treatment (all p < 0.01). In cultured primary mouse cardiomyocytes, treatment with MSC exosomes essentially reversed inflammation-induced pro-apoptotic and inflammatory signals (p < 0.01). MSC exosomes exert their cardioprotective effects by suppressing inflammation and pro-apoptotic processes, particularly in the peri-infarct areas, resulting in preservation of cardiac function after LCA ligation.


Assuntos
Exossomos , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio , Animais , Linhagem Celular Transformada , Exossomos/metabolismo , Exossomos/patologia , Exossomos/transplante , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle
3.
Basic Res Cardiol ; 115(6): 66, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33180196

RESUMO

Proprotein convertase subtilisin/Kexin type 9 (PCSK9) and pyroptosis both play important roles in myocardial infarction. This study was designed to test the hypothesis that PCSK9 regulates pyroptosis in cardiomyocytes during chronic myocardial ischemia. Primary cardiomyocytes were isolated from WT and PCSK9-/- mice. HL-1 cardiomyocytes were used to set up PCSK9-deficient (PCSK9-/-) and PCSK9-upregulated (PCSK9CRISPRa) cardiomyocyte cell line with CRISPR/Cas9 knockout or activation plasmid. Additional studies were performed with chronic myocardial ischemia in WT and PCSK9-/- mice. We observed that PCSK9 initiates mitochondrial DNA (mtDNA) damage, activates NLRP3 inflammasome signaling (NLRP3, ASC, Caspase-1, IL-1ß, and IL-18), and subsequently induces Caspase-1-dependent pyroptosis. There was an intense expression of PCSK9 and pyroptosis marker, GSDMD-NT, in the zone bordering the infarct area. PCSK9-/- significantly suppressed expression of NLRP3 inflammasome signaling, GSDMD-NT, and LDH release. Furthermore, serum levels of PCSK9, NLPR3 inflammasome signaling, and pyroptosis (GSDMD and LDH release) were significantly elevated in patients with chronic myocardial ischemia as compared to those in age-matched healthy subjects. Human hearts with recent infarcts also showed high expression of PCSK9 and GSDMD-NT in the border zone similar to that in the infarcted mouse heart. These observations provide compelling evidence for the role of PCSK9 in regulating Caspase-1-dependent pyroptosis via mtDNA damage and may qualify pro-inflammatory cytokines and pyroptosis as potential targets to treat PCSK9-related cardiovascular diseases.


Assuntos
Dano ao DNA , DNA Mitocondrial/metabolismo , Mitocôndrias Cardíacas/enzimologia , Isquemia Miocárdica/enzimologia , Miócitos Cardíacos/enzimologia , Pró-Proteína Convertase 9/metabolismo , Piroptose , Idoso , Animais , Estudos de Casos e Controles , Caspase 1/metabolismo , Linhagem Celular , Doença Crônica , DNA Mitocondrial/genética , Modelos Animais de Doenças , Feminino , Humanos , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Isquemia Miocárdica/genética , Isquemia Miocárdica/patologia , Miócitos Cardíacos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Pró-Proteína Convertase 9/genética , Transdução de Sinais
4.
Cardiovasc Drugs Ther ; 31(5-6): 593-608, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28956198

RESUMO

PURPOSE: Heart failure (HF) remains a major cause of morbidity and mortality worldwide. Although various therapies developed over the last two decades have shown improved long term outcomes in patients with established HF, there has been little progress in preventing the adverse cardiac remodeling that initiates HF. To fill the gap in treatment, current research efforts are focused on understanding novel mechanisms and signaling pathways. Immune activation, inflammation, oxidative stress, alterations in mitochondrial bioenergetics, and autophagy have been postulated as important pathophysiological events in this process. An improved understanding of these complex processes could facilitate a therapeutic shift toward molecular targets that can potentially alter the course of HF. METHODS: In this review, we address the role of immunity, inflammation, and oxidative stress as well as other novel emerging concepts in the pathophysiology of HF that may have therapeutic implications. CONCLUSION: Based on the experimental and clinical studies presented here, we anticipate that a better understanding of the pathophysiology of HF will open the door for new therapeutic targets. A one-size-fits-all approach may not be appropriate for all patients with HF, and further clinical trials utilizing molecular targeting in HF may result in improved outcomes.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cardiotônicos/uso terapêutico , Fibrose , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Fatores Imunológicos/imunologia , Inflamação , Terapia de Alvo Molecular
6.
Clin Sci (Lond) ; 130(15): 1353-62, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27129181

RESUMO

Studies have indicated that dipeptidyl peptidase-4 (DPP-4) inhibitors and glucagon-like peptide-1 (GLP-1) agonists reduce infarct size after myocardial ischaemia. Whether these agents modify cardiac remodelling after ischaemia is unclear. Furthermore, it is not known if combination of the two types of drugs is superior to either agent alone. We investigated the modulatory effect of the DPP-4 inhibitor linagliptin alone, the GLP-1 activator liraglutide alone, or the two agents together on myocardial infarct size, left ventricular contractile function and cardiac remodelling signals after a brief period of left coronary artery (LCA) occlusion. C57BL/6 mice were treated with vehicle, the DPP-4 inhibitor linagliptin, the GLP-1 activator liraglutide, or both agents together for 5 days, and then subjected to LCA occlusion (1 h) and reperfusion (3 h). Ischaemia-reperfusion increased reactive oxygen species (ROS) generation and expression of NADPH oxidase (p47(phox), p22(phox) and gp91(phox) subtypes), collagens, fibronectin and proinflammatory cytokines (interleukin 6, tumour necrosis factor α and monocyte chemoattractant protein-1) in the LCA-supplied regions. Pre-treatment with linagliptin or liraglutide reduced infarct size, protected cardiomyocytes from injury and preserved cardiac contractile function in a similar fashion. It is interesting that profibrotic (collagen deposition) signals were expressed soon after ischaemia-reperfusion. Both linagliptin and liraglutide suppressed ROS generation, NADPH oxidase and proinflammatory signals, and reduced collagen deposition. Addition of linagliptin or liraglutide had no significant additive effect above and beyond that of liraglutide and linagliptin given alone. In conclusion, linagliptin and liraglutide can improve cardiac contractile function and indices of cardiac remodelling, which may be related to their role in inhibition of ROS production and proinflammatory cytokines after ischaemia.


Assuntos
Cardiotônicos/farmacologia , Colágeno/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Incretinas/farmacologia , Linagliptina/farmacologia , Liraglutida/farmacologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Citocinas/metabolismo , Citoproteção , Modelos Animais de Doenças , Quimioterapia Combinada , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
7.
Am J Ther ; 23(1): e298-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-24368608

RESUMO

Eptifibatide is a commonly and widely used drug for management of acute coronary syndrome and during percutaneous coronary intervention. It is usually well tolerated with no major adverse effects. We report a rare case of life-threatening thrombocytopenia secondary to eptifibatide along with a literature review of available evidence.


Assuntos
Peptídeos/efeitos adversos , Inibidores da Agregação Plaquetária/efeitos adversos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Trombocitopenia/induzido quimicamente , Idoso , Eptifibatida , Humanos , Masculino
8.
Acta Pharmacol Sin ; 37(10): 1349-1358, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27498780

RESUMO

AIM: Glucagon-like peptide-1 (GLP-1) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors can not only lower blood glucose levels, but also alleviate cardiac remodeling after myocardial ischemia and hypertension. In the present study, we investigated the effects of a DPP-4 inhibitor (linagliptin) and a GLP-1 activator (liraglutide) on glucose- and angiotensin II (Ang II)-induced collagen formation and cytoskeleton reorganization in cardiac fibroblasts in vitro, and elucidated the related mechanisms. METHODS: Cardiac fibroblasts were isolated from the hearts of 6-week-old C57BL/6 mice, and then exposed to different concentrations of glucose or Ang II for 24 h. The expression of fibrotic signals (fibronectin, collagen-1, -3 and -4), as well as ERK1/2 and NF-κB-p65 in the fibroblasts was examined using Western blotting assays. F-actin degradation was detected under inverted laser confocal microscope in fibroblasts stained with Rhodamine phalloidin. RESULTS: Glucose (1-40 mmol/L) and Ang II (10-8-10-5 mol/L) dose-dependently increased the expression of fibronectin, collagens, phospho-ERK1/2 and phospho-NF-κB-p65 in cardiac fibroblasts. High concentrations of glucose (≥40 mmol/L) and Ang II (≥10-6 mol/L) caused a significant degradation of F-actin (less assembly F-actin fibers and more disassembly fibers). ERK1/2 inhibitor U0126 (10 µmol/L) and NF-κB inhibitor JSH-23 (10 µmol/L) both markedly suppressed glucose- and angiotensin II-induced fibronectin and collagen expressions in cardiac fibroblasts. Furthermore, pretreatment with liraglutide (10-100 nmol/L) or linagliptin (3 and 30 nmol/L) significantly decreased glucose- and Ang II-induced expression of fibrotic signals, phospho-ERK1/2 and phospho-NF-κB-p65 in cardiac fibroblasts. Moreover, pretreatment with liraglutide (30 nmol/L) or liraglutide (100 nmol/L) markedly inhibited glucose-induced F-actin degradation, however, only liraglutide inhibited Ang II-induced F-actin degradation. CONCLUSION: Linagliptin and liraglutide inhibit glucose- and Ang II-induced collagen formation in cardiac fibroblasts via activation of the ERK/NF-κB/pathway. Linagliptin and liraglutide also markedly inhibit glucose-induced F-actin degradation in cardiac fibroblasts, but only liraglutide inhibits Ang II-induced F-actin degradation.


Assuntos
Colágeno/biossíntese , Citoesqueleto/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Fibroblastos/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/agonistas , Linagliptina/farmacologia , Liraglutida/farmacologia , Miocárdio/metabolismo , Actinas/metabolismo , Angiotensina II/farmacologia , Animais , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Glucose/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia
9.
J Mol Cell Cardiol ; 80: 101-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25584774

RESUMO

Endothelin-1 (ET-1) plays a major role in regulating myocardial fibrosis in several pathological conditions, such as hypertension and diabetes. Aging is an independent risk factor for myocardial fibrosis. We hypothesized that ET-1 upregulation may be a basis of enhanced collagen synthesis in the senescent fibroblasts resulting in cardiac fibrosis with aging. To examine this hypothesis, we cultured mouse cardiac fibroblasts to passage-30 (P30). ß-Galactosidase activity and several other aging markers were markedly increased in P30 (vs. P3) fibroblasts, indicating that these cells were indeed undergoing senescence. Importantly, ET-1 expression was markedly upregulated in P30 (vs. P3) fibroblasts. Of note, estrogen receptor-α (ER-α), an important negative regulator of ET-1, was downregulated in P30 fibroblasts. We also studied aged (130-weeks old, female) mice hearts, and observed that ET-1 was upregulated and ER-α was downregulated in these hearts (vs. 6-week old mice hearts, female). Similar observations were made in the fibroblasts isolated from aged mice hearts. ET-1 upregulation with aging was also seen in ≈70-year old (vs. ≈30-year old) human heart sections. In concert with ET-1 upregulation, the expression of fibronectin and collagens was found to be markedly increased in P30 cardiac fibroblasts in culture, fibroblasts isolated from the aged mice hearts, and in aged human hearts. Interestingly, inhibition of ET-1 in the senescent P30 fibroblasts by 2 different strategies (the use of siRNA and the use of endothelin converting enzyme inhibitors) markedly suppressed expression of fibrosis signals. Further, treatment with synthetic ET-1 enhanced fibronectin and collagen expression in P3 cardiac fibroblasts. These observations in mice and human hearts suggest that aging-related cardiac fibrosis is, at least partially, dependent on the upregulation of ET-1.


Assuntos
Envelhecimento/genética , Endotelina-1/genética , Regulação da Expressão Gênica , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Senescência Celular/genética , Endotelina-1/metabolismo , Enzimas Conversoras de Endotelina , Ativação Enzimática , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Técnicas de Silenciamento de Genes , Humanos , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/metabolismo , Camundongos , Interferência de RNA , Transdução de Sinais , Regulação para Cima , beta-Galactosidase/metabolismo
10.
Biochem Biophys Res Commun ; 467(1): 135-9, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26393906

RESUMO

Previous studies have shown that oxidized low-density lipoprotein (ox-LDL) inhibits macrophage migration, but the precise mechanisms remain unclear. Lectin-like ox-LDL receptor-1 (LOX-1) is a scavenger receptor that is expressed in macrophages and binds ox-LDL. Calpains, a family of calcium-dependent proteases, influence several aspects of cell migration. In this study, we investigated the role of LOX-1 in macrophage migration in response to ox-LDL and the involvement of calpains in this process. Peritoneal macrophages from wild type C57BL/6 mice were exposed to different concentrations of ox-LDL (1-20 µg/mL), and expression of LOX-1 and calpain-1 and -2, cell migration and intracellular calcium (Ca(2+)in) were measured. Our results showed that ox-LDL stimulated LOX-1 and calpain-2 expression, and inhibited calpain-1 expression in a dose- and time-dependent manner. Further, ox-LDL inhibited macrophage migration and increased Ca(2+)in concentration in macrophages. To further elucidate the role of LOX-1 in ox-LDL-impaired macrophage migration, we isolated peritoneal macrophages from LOX-1 knockout mice, and treated them with ox-LDL. Interestingly, calpain-1 expression was much higher, and calpain-2 expression was lower in LOX-1 knockout macrophages than in wild-type macrophages following exposure to ox-LDL. LOX-1 deletion significantly improved macrophage migration and decreased Ca(2+)in concentration. These data indicate that LOX-1 is, at least in part, responsible for the inhibitory effect of ox-LDL on macrophage migration and this process involves calpain-1 and -2.


Assuntos
Calpaína/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneais/metabolismo , Receptores Depuradores Classe E/metabolismo , Animais , Cálcio/metabolismo , Carbocianinas , Movimento Celular/efeitos dos fármacos , Corantes Fluorescentes , Técnicas In Vitro , Lipoproteínas LDL/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Depuradores Classe E/deficiência , Receptores Depuradores Classe E/genética
11.
J Cardiovasc Pharmacol ; 65(2): 123-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24853489

RESUMO

Growth of atherosclerotic plaque requires neovascularization (angiogenesis). To elucidate the involvement of angiotensin II (Ang II) in angiogenesis, we performed gene microarray and microRNA (miRNA) polymerase chain reaction array analyses on human coronary artery endothelial cells exposed to moderate concentration of Ang II for 2 and 12 hours. At 12, but not 2, hours, cultures treated with Ang II exhibited shifts in transcriptional activity involving 267 genes (>1.5-fold difference; P < 0.05). Resulting transcriptome was most significantly enriched for genes associated with blood vessel development, angiogenesis, and regulation of proliferation. Majority of upregulated genes implicated in angiogenesis shared a commonality of being either regulators (HES1, IL-18, and CXCR4) or targets (ADM, ANPEP, HES1, KIT, NOTCH4, PGF, and SOX18) of STAT3. In line with these findings, STAT3 inhibition attenuated Ang II-dependent stimulation of tube formation in Matrigel assay. Expression analysis of miRNAs transcripts revealed that the pattern of differential expression for miRNAs was largely consistent with proangiogenic response with a prominent theme of upregulation of miRs targeting PTEN (miR-19b-3p, miR-21-5p, 23b-3p, and 24-3p), many of which are directly or indirectly STAT3 dependent. We conclude that STAT3 signaling may be an intrinsic part of Ang II-mediated proangiogenic response in human endothelial cells.


Assuntos
Angiotensina II , Células Endoteliais/metabolismo , Neovascularização Patológica/genética , Placa Aterosclerótica , Angiotensina II/genética , Angiotensina II/metabolismo , Técnicas de Cultura de Células , Proliferação de Células/genética , Vasos Coronários/metabolismo , Vasos Coronários/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Humanos , MicroRNAs/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Receptor Notch4 , Receptores CXCR4/genética , Receptores Notch/genética , Fatores de Transcrição SOXF/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética
13.
Am J Physiol Heart Circ Physiol ; 306(12): H1700-7, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24778168

RESUMO

Myocardial infarct size is determined by the death of nonischemic border zone cardiomyocytes caused by export of injury signals from the infarct zone. The countermeasures to limit infarct size, therefore, should be aimed at nonselective blockade of most, if not all, injury signals from entering nonischemic cells. To test whether inhibition of endocytosis might limit infarct size, HL-1 cardiomyocytes were subjected to anoxia (6 h) and reoxygenation (1 h). Anoxic and reoxygenated cells showed a multifold increase in mitochondrial ROS production accompanied with upregulation of scavenger receptors lectin-like oxidized low-density lipoprotein receptor-1 and CD36 and stimulation of stress signals, including NADPH oxidase subunit p22(phox), SOD2, and beclin-1. Incubation of healthy cardiomyocytes in media from anoxic and reoxygenated cells (conditioned media) resulted in qualitatively similar responses, including increase in the generation of mitochondrial ROS, p22(phox), SOD2, and beclin-1. Anoxia and reoxygenation caused collapse of clathrin-mediated endocytosis and stimulation of macropinocytosis, whereas in cultures exposed to conditioned media, the activity of endocytosis was uniformly higher. Conditioned media also significantly aggravated cytotoxic effects of TNF-α and angiotensin II, and suppression of endocytosis reversed these trends, resulting in an overall increase of metabolic activity. Moreover, inhibition of endocytosis prevented binding of oxidized cellular fragments with greater efficiency than targeted neutralization of the scavenger receptor lectin-like oxidized low-density lipoprotein receptor-1. Many of the observations in HL-1 cardiomyocytes were confirmed in primary cardiomyocyte cultures. Our data suggest that endocytosis is upregulated in border zone cardiomyocytes, and inhibition of endocytosis may be an effective approach to prevent export of injury signals from the infarct zone.


Assuntos
Endocitose/fisiologia , Hipóxia/complicações , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/fisiologia , Oxigênio/farmacologia , Angiotensina II/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Antígenos CD36/metabolismo , Células Cultivadas , Endocitose/efeitos dos fármacos , Camundongos , Modelos Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
14.
Biochem Biophys Res Commun ; 451(4): 637-43, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25130466

RESUMO

BACKGROUND: Lectin-like oxidized low-density lipoprotein scavenger receptor-1 (LOX-1) is known to be involved in many pathophysiological events, such as inflammation. METHODS: To clarify the role of LOX-1 in mtDNA damage and NLRP3 inflammasome activation, we studied wild-type (WT) and LOX-1 knockout (KO) mice given thioglycollate, an inflammatory stimulus. RESULTS: We observed intense inflammatory response (CD45 and CD68 expression) and mtDNA damage in spleen and kidneys of WT mice given thioglycollate. The abrogation of LOX-1 (use of LOX-1 knockout mice) reduced the inflammatory response as well as mtDNA damage (P<0.05 vs. WT mice). We also observed that mice with LOX-1 deletion had markedly reduced expression of caspase-1 (P10 and P20 subunits) as well as cleaved IL-1ß and IL-18. These mice also had much less mtDNA damage and only limited NLRP3 inflammasome expression. CONCLUSIONS: These in vivo observations indicate that LOX-1 plays a key role in mtDNA damage which then leads to NLRP3 inflammasome activation during inflammation.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA/efeitos dos fármacos , DNA Mitocondrial/metabolismo , Inflamassomos/metabolismo , Inflamação/induzido quimicamente , Receptores Depuradores Classe E/fisiologia , Animais , Caspase 1/biossíntese , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores Depuradores Classe E/genética , Tioglicolatos
15.
J Cardiovasc Pharmacol ; 64(1): 47-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24705175

RESUMO

Oxidized low-density lipoprotein (ox-LDL) uptake by monocytes/macrophages plays a pivotal role in atherogenesis. This study was designed to examine the effect of glucagon-like peptide-1 (GLP-1) agonists on ox-LDL uptake in macrophages. Human primary monocytes/macrophages were incubated with native GLP-1 (nGLP-1) or GLP-1 agonist liraglutide to evaluate their effect on ox-LDL uptake and the expression of scavenger receptors (SRs), such as SR-A, CD36, and lectin-like ox-LDL SR-1, in this process. Our study showed a decrease in ox-LDL uptake and CD36 expression in macrophages treated with nGLP-1 or liraglutide. However, nGLP-1 and liraglutide did not affect the expression of other SRs SR-A and lectin-like ox-LDL SR-1. Simultaneously, there was an increase in the expression of activated protein kinase A (PKA). To examine the role of PKA in the effects of nGLP-1 or liraglutide, we treated macrophages with PK inhibitor (6-22) amide, a PKA inhibitor, followed by treatment with nGLP-1 or liraglutide. Inhibition of PKA activation markedly reversed the effect of nGLP-1 or liraglutide on ox-LDL uptake and enhanced the expression of CD36. Our results suggest that GLP-1 agonism inhibits ox-LDL uptake through PKA/CD36 pathway in macrophages. This study provides a novel insight in the mechanism of foam cell formation and the role by GLP-1 agonists therein.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/agonistas , Lipoproteínas LDL/metabolismo , Aterosclerose/patologia , Antígenos CD36/genética , Células Cultivadas , Células Espumosas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Liraglutida , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Monócitos/metabolismo
16.
J Cardiovasc Pharmacol ; 63(2): 158-66, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24220312

RESUMO

BACKGROUND: Dyslipidemia, particularly increased LDL-cholesterol level in serum, is associated with atherosclerosis and fibrosis in different organs. This study was designed to investigate the effects of increase in LDL-cholesterol on renal fibrosis. METHODS: Wild-type (WT) and LDLr knockout (KO) mice were fed standard or high fat diet (HFD), and their kidneys were collected after 26 weeks of dietary intervention for identification of fibrosis and study of potential mechanisms. Additional studies were performed in cultured renal fibroblasts. RESULTS: We observed extensive and diffuse fibrosis in the kidneys of mice given HFD (P < 0.05 vs. standard chow). Fibrosis was associated with enhanced expression of fibronectin, nicotinamide adenine dinucleotide phosphate oxidases and activated p38 and p44/42 mitogen-activated protein kinases (MAPKs) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). There was evidence for accumulation of 4-hydroxynonenal, a lipid peroxidation product, in the kidneys and of ox-LDL in the arteries of LDLr KO mice given HFD. The expression of ox-LDL receptor LOX-1 and of transforming growth factor beta 1 (TGFß1) was increased in these kidneys. All these changes were more pronounced in LDLr KO mice than in the WT mice. In in vitro studies, treatment of fibroblasts from kidneys of LDLr KO mice with ox-LDL showed intense proliferation and collagen formation (all P < 0.05, fibroblasts from WT mice kidneys). Blockade of p38 MAPK, p44/42 MAPK, or NF-κB significantly attenuated expression of profibrotic signals, collagen formation, and proliferation of fibroblasts. CONCLUSIONS: HFD induces renal fibrosis in LDLr-null mice primarily through activation of the nicotinamide adenine dinucleotide phosphate oxidase MAPK-NF-κB pathway by ox-LDL.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Nefropatias/etiologia , Lipoproteínas LDL/metabolismo , Receptores de LDL/genética , Aldeídos/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Fibroblastos/patologia , Fibrose , Nefropatias/patologia , Peroxidação de Lipídeos , Masculino , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
17.
Cardiovasc Drugs Ther ; 28(5): 425-32, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25022544

RESUMO

BACKGROUND: Anti-atherosclerotic effects of dipeptidyl peptidase-4 (DPP-4) inhibitors have been shown in many studies. Since inflammation and immune response play a key role in atherogenesis, we examined the effect of DPP-4 inhibitors on the expression of nod-like receptor family, pyrin domain containing 3 (NLRP3) Inflammasome and Interleukin-1beta (IL-1ß) in human macrophages. METHODS AND RESULTS: THP-1 macrophages were incubated with oxidized low density lipoprotein (ox-LDL) with or without DPP-4 inhibitors (sitagliptin and NVPDPP728). The effects of DPP-4 inhibitors on the expression of NLRP3, toll-like receptor 4 (TLR4) and pro-inflammatory cytokine IL-1ß were studied. Both DPP-4 inhibitors induced a significant reduction in NLRP3, TLR4 and IL-1ß expression; concurrently, there was an increase in glucagon like peptide 1 receptor (GLP-1R) expression. Simultaneously, DPP-4 inhibitors reduced phosphorylated-PKC, but not PKA, levels. To determine the role of PKC activation in the effects of DPP-4 inhibitors, cells were treated with PMA- which blocked the effect of DPP-4 inhibitors on NLRP3 and IL-1ß as well as TLR4 and GLP-1R. Over-expression of GLP-1R in macrophages with its agonist liraglutide also blocked the effects of PMA. CONCLUSION: DPP-4 inhibitors suppress NLRP3, TLR4 and IL-1ß in human macrophages through inhibition of PKC activity. This study provides novel insights into the mechanism of inhibition of inflammatory state and immune response in atherosclerosis by DPP-4 inhibitors.


Assuntos
Proteínas de Transporte/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Inflamassomos/biossíntese , Interleucina-1beta/biossíntese , Macrófagos/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores de Glucagon/metabolismo , Proteínas de Transporte/biossíntese , Técnicas de Cultura de Células , Proteínas Quinases Dependentes de AMP Cíclico/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Liraglutida , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nitrilas/antagonistas & inibidores , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Pirazinas/antagonistas & inibidores , Pirazinas/farmacologia , Pirrolidinas/antagonistas & inibidores , Pirrolidinas/farmacologia , Receptores de Glucagon/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fosfato de Sitagliptina , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Receptor 4 Toll-Like/biossíntese , Triazóis/antagonistas & inibidores , Triazóis/farmacologia
18.
Cardiovasc Drugs Ther ; 28(5): 441-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25005756

RESUMO

Lectin-like oxidized low-density lipoprotein (ox-LDL) receptor-1 (LOX-1) is a major receptor for ox-LDL in endothelial cells. Its activation regulates endothelial proliferation, differentiation, migration and apoptosis. Recent in vitro studies show that LOX-1 activation by ox-LDL and angiotensin II (Ang II) induces angiogenesis via activation of NADPH oxidase and subsequent increase in ROS production. In this study, we investigated the effect of LOX-1 gene deletion (LOX-1 knockout or KO mice) on angiogenesis in response to prolonged Ang II infusion in vivo. Our studies showed that Ang II (vs. saline) infusion enhanced capillary formation in subcutaneously injected Matrigel® plugs. Ang II infusion also resulted in marked angiogenesis in the hearts as determined by CD31 immunopositivity. There was an increased expression (RT-PCR and Western blotting) of CD31 and VEGF in the hearts of mice infused with Ang II, indicating pro-angiogenic miliue. More importantly, LOX-1 KO mice reveled markedly limited angiogenesis in the Matrigel® plugs as well as in the hearts despite similar infusion with Ang II (all P < 0.05 vs. wild-type mice). In addition, the hearts of LOX-1 KO mice had attenuated expression of pro-inflammatory and angiogenic signals MCP-1 and IL-1ß following Ang II Infusion. Lastly, the rise in blood pressure in response to Ang II was less in the LOX-1 KO mice (P < 0.05 vs. wild-type mice). Our findings suggest that LOX-1 participates in angiogenesis in hypertension, which may be related to a state of inflammation.


Assuntos
Angiotensina II/farmacologia , Coração/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Depuradores Classe E/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Quimiocina CCL2/biossíntese , Colágeno/fisiologia , Combinação de Medicamentos , Coração/efeitos dos fármacos , Interleucina-1beta/biossíntese , Laminina/fisiologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteoglicanas/fisiologia , Receptores Depuradores Classe E/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese
19.
Can J Physiol Pharmacol ; 92(7): 524-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24959993

RESUMO

As a major receptor for oxidized low density lipoprotein (ox-LDL), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is upregulated in many pathophysiological events, including endothelial cell dysfunction and smooth muscle cell growth, as well as monocyte migration and transformation into foam cells, which are present in atherosclerosis and myocardial ischemia. Excessive production of reactive oxygen species (ROS) increases LOX-1 expression, induces mitochondrial DNA damage, and activates autophagy. Damaged mitochondrial DNA that escapes from autophagy induces an inflammatory response. This paper reviews the potential link between LOX-1, mitochondrial DNA damage, autophagy, and immune response in atherosclerosis.


Assuntos
Aterosclerose/imunologia , Aterosclerose/metabolismo , Autofagia , Dano ao DNA , DNA Mitocondrial/metabolismo , Estresse Oxidativo , Receptores Depuradores Classe E/metabolismo , Aterosclerose/patologia , Humanos
20.
Theranostics ; 14(6): 2427-2441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646649

RESUMO

Background: MER proto-oncogene tyrosine kinase (MerTK) is a key receptor for efferocytosis, a process for the clearance of apoptotic cells. MerTK is mainly expressed in macrophages and immature dendritic cells. There are very limited reports focused on MerTK biology in aortic endothelial cells (ECs). It remains unclear for the role of blood flow patterns in regulating MerTK-mediated efferocytosis in aortic ECs. This study was designed to investigate whether endothelial MerTK and EC efferocytosis respond to blood flow patterns during atherosclerosis. Methods: Big data analytics, RNA-seq and proteomics combined with our in vitro and in vivo studies were applied to reveal the potential molecular mechanisms. Partial carotid artery ligation combined with AAV-PCSK9 and high fat diet were used to set up acute atherosclerosis in 4 weeks. Results: Our data showed that MerTK is sensitive to blood flow patterns and is inhibited by disturbed flow and oscillatory shear stress in primary human aortic ECs (HAECs). The RNA-seq data in HAECs incubated with apoptotic cells showed that d-flow promotes pro-inflammatory pathway and senescence pathway. Our in vivo data of proteomics and immunostaining showed that, compared with WT group, MerTK-/- aggravates atherosclerosis in d-flow areas through upregulation of endothelial dysfunction markers (e.g. IL-1ß, NF-κB, TLR4, MAPK signaling, vWF, VCAM-1 and p22phox) and mitochondrial dysfunction. Interestingly, MerTK-/-induces obvious abnormal endothelial thickening accompanied with decreased endothelial efferocytosis, promoting the development of atherosclerosis. Conclusions: Our data suggests that blood flow patterns play an important role in regulating MerTK-mediated efferocytosis in aortic ECs, revealing a new promising therapeutic strategy with EC efferocytosis restoration to against atherosclerosis.


Assuntos
Aorta , Aterosclerose , Células Endoteliais , Fagocitose , c-Mer Tirosina Quinase , c-Mer Tirosina Quinase/metabolismo , c-Mer Tirosina Quinase/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Humanos , Células Endoteliais/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Camundongos , Apoptose , Proto-Oncogene Mas , Masculino , Camundongos Endogâmicos C57BL , Dieta Hiperlipídica , Células Cultivadas , Eferocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA