Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Cardiovasc Drugs ; 24(1): 5-18, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37815758

RESUMO

Acute myocardial infarction (AMI) is associated with high morbidity and mortality worldwide. Although early reperfusion is the most effective strategy to salvage ischemic myocardium, reperfusion injury can develop with the restoration of blood flow. Therefore, it is important to identify protection mechanisms and strategies for the heart after myocardial infarction. Recent studies have shown that multiple intracellular molecules and signaling pathways are involved in cardioprotection. Meanwhile, device-based cardioprotective modalities such as cardiac left ventricular unloading, hypothermia, coronary sinus intervention, supersaturated oxygen (SSO2), and remote ischemic conditioning (RIC) have become important areas of research. Herein, we review the molecular mechanisms of cardioprotection and cardioprotective modalities after ischemia-reperfusion injury (IRI) to identify potential approaches to reduce mortality and improve prognosis in patients with AMI.


Assuntos
Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Humanos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Infarto do Miocárdio/metabolismo , Coração , Transdução de Sinais/fisiologia
2.
J Cardiovasc Pharmacol ; 81(3): 175-182, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36607700

RESUMO

ABSTRACT: Atherosclerotic coronary heart disease is a common cardiovascular disease with high morbidity and mortality. In recent years, the incidence of coronary heart disease has gradually become younger, and biomarkers for predicting coronary heart disease have demonstrated valuable clinical prospects. Several studies have established an association between coronary heart disease and intestinal flora metabolites, including trimethylamine oxide (TMAO), which has attracted widespread attention from researchers. Investigations have also shown that plasma levels of TMAO and its precursors can predict cardiovascular risk in humans; however, TMAO's mechanism of action in causing coronary heart disease is not fully understood. This review examines TMAO's generation, the mechanism through which it causes coronary heart disease, and the approaches used to treat TMAO-caused coronary heart disease to possible avenues for future research on coronary heart disease and find new concepts for the treatment of the condition.


Assuntos
Doença das Coronárias , Microbioma Gastrointestinal , Humanos , Doença das Coronárias/diagnóstico , Doença das Coronárias/epidemiologia , Metilaminas/metabolismo , Biomarcadores
3.
Front Cardiovasc Med ; 9: 864600, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35770223

RESUMO

Atherosclerosis (AS) is one of the most common cardiovascular diseases (CVDs), and there is currently no effective drug to reverse its pathogenesis. Trimethylamine N-oxide (TMAO) is a metabolite of the gut flora with the potential to act as a new risk factor for CVD. Many studies have shown that TMAO is involved in the occurrence and development of atherosclerotic diseases through various mechanisms; however, the targeted therapy for TMAO remains controversial. This article summarizes the vital progress made in relation to evaluations on TMAO and AS in recent years and highlights novel probable approaches for the prevention and treatment of AS.

4.
Front Cardiovasc Med ; 8: 710963, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34368260

RESUMO

Ferroptosis is an iron-dependent cell death, which is characterized by iron overload and lipid peroxidation. Ferroptosis is distinct from apoptosis, necroptosis, autophagy, and other types of cell death in morphology and function. Ferroptosis is regulated by a variety of factors and controlled by several mechanisms, including mitochondrial activity and metabolism of iron, lipid, and amino acids. Accumulating evidence shows that ferroptosis is closely related to a majority of cardiovascular diseases (CVDs), including cardiomyopathy, myocardial infarction, ischemia/reperfusion injury, heart failure, and atherosclerosis. This review summarizes the current status of ferroptosis and discusses ferroptosis as a potential therapeutic target for CVDs.

5.
Front Med (Lausanne) ; 7: 583943, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33240906

RESUMO

Non-alcoholic fatty liver disease (NAFLD) has become a common chronic disease in the world. NAFLD is not only a simple intrahepatic lesion, but also affects the occurrence of a variety of extrahepatic complications. In particular, cardiovascular complications are particularly serious, which is the main cause of death in patients with NAFLD. To study the relationship between NAFLD and AS may be a new way to improve the quality of life in patients with NAFLD. As we all known, inflammatory response plays an important role in the occurrence and development of NAFLD and AS. In this study, we found that the accumulation of Nε-carboxymethyllysine (CML) in the liver leads to hepatic steatosis. CML can induce the expression of interleukin (IL-1ß), interleukin (IL-6), tumor necrosis factor (TNF-α), C-reactionprotein (CRP) by binding with advanced glycosylation end-product receptor (RAGE) and accelerate the development of AS. After silencing RAGE expression, the expression of pro-inflammatory cytokines was inhibited and liver and aorta pathological changes were relieved. In conclusion, CML/RAGE signal promotes the progression of non-alcoholic fatty liver disease and atherosclerosis. We hope to provide new ideas for the study of liver vascular dialogue in multi organ communication.

6.
Diabetes Metab Syndr Obes ; 13: 4141-4151, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33177854

RESUMO

BACKGROUND AND AIMS: To investigate the role of Sortilin and matrix vesicles (MVs) in Nε-Carboxymethyl-lysine (CML)-induced diabetic atherosclerotic calcification (AC). METHODS: At human level, the correlation between Sortilin and CD9 (marker proteins of MVs) in serum MVs and CML in serum was explored by enzyme-linked immunosorbent assay (ELISA) detection and Pearson correlation analysis. After a diabetic apoE-/- mouse model was constructed, the calcification of aorta and the expressions of related proteins under CML and MVs injection were observed by calcification staining, immunofluorescence staining, and Western blot. MVs levels released by smooth muscle cells (SMCs) under different treatments was detected by nanometer tracking analysis (NTA). After treating SMCs with MVs and Anti-Sortilin, cell calcification was observed by Alizarin red staining. RESULTS: Serological analysis of patients showed that the concentrations of Sortilin and CD9 in serum MVs were positively correlated with the concentration of CML in serum. Animal experiments showed that CML could promote the progression of diabetic AC and the high expression of Sortilin in plaques. Diabetic apoE-/- mouse tail vein injection of CML-induced SMCs-derived MVs obviously aggravated AC. Cell experiment results showed that a high concentration of CML significantly promoted the release of MVs from SMCs. MVs from this source could markedly worsen cell calcification, while the administration of GW4869 (a widely used extracellular vesicles biogenesis inhibitor) significantly reduced cell calcification. Finally, treatment of high concentrations of CML could also promote the recruitment of Sortilin to MVs, and administration of Anti-Sortilin could markedly reduce cell calcification caused by MVs. CONCLUSION: We proved that CML not only affects the release of MVs from SMCs but also affects the recruitment of Sortilin to MVs, thereby promoting diabetic AC. This discovery may provide a new strategy for targeted prevention of vascular calcification in diabetes.

7.
Mediators Inflamm ; 2020: 6850187, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32831637

RESUMO

BACKGROUND: Advanced glycation end products play an important role in diabetic atherosclerosis. The effects of advanced glycation end products (AGEs) on vascular smooth muscle cell- (VSMC-) derived foam cell formation and phenotypic transformation are unknown. METHODS: Serological and histological samples were obtained from diabetic amputation patients and accident amputation patients from the Affiliated Hospital of Jiangsu University. CD68/Actin Alpha 2 (ACTA2) coimmunofluorescence sections were used to quantify the number of VSMCs with macrophage-like phenotypes. Western blotting was used to detect the expression of the receptor of advanced glycation end products in vascular samples. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate the level of serum Nε-carboxymethyl-lysine (CML). In vitro oil red O staining was used to examine lipid accumulation in VSMCs stimulated by CML. The expression of VSMCs and macrophage markers was measured by western blotting and quantitative real-time PCR. Furthermore, changes in VSMC migration and secretion were detected by the Transwell assay and ELISA. RESULTS: In the arterial plaque sections of diabetic patients, VSMCs transformed to a macrophage-like phenotype. The serum CML and RAGE levels in the plaques were significantly higher in the diabetes group than those in the healthy control group and were significantly related to the number of macrophage-like VSMCs. CML stimulation promoted intracellular lipid accumulation. However, CML stimulation decreased the expression of VSMC markers and increased the expression of macrophage phenotype markers. Finally, CML promoted smooth muscle cell migration and the secretion of proinflammatory-related factors. CONCLUSIONS: CML induces VSMC-derived foam cell formation, and VSMCs transdifferentiate to a macrophage-like state, which may be mediated by the activation of RAGE.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Animais , Western Blotting , Transdiferenciação Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Células Espumosas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Interferência de RNA
8.
Front Pharmacol ; 11: 661, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32457633

RESUMO

Vascular calcification is an abnormal cell-mediated process in which bone-specific hydroxyapatite crystals are actively deposited on the blood vessel wall and is a significant pathological basis for the increased incidence and mortality of adverse cardiovascular events. Macrophages play an important regulatory role in the occurrence, development, and regression of vascular calcification. After the tissue microenvironment changes, macrophages subsequently change their polarity and phenotype or secrete functional substances as an adaptive response. As research on macrophages continue to move into this field, we gain a new understanding of the mechanism of the formation and regression of vascular calcification, which might offer valuable new intervention targets for the prevention and inhibition of vascular calcification. This review summarizes a wealth of research in this field and explores the roles of macrophages in the development process of vascular calcification.

9.
Am J Physiol Heart Circ Physiol ; 318(5): H1068-H1079, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32216615

RESUMO

The clinical risks and prognosis of diabetic vascular intimal calcification (VIC) and medial calcification (VMC) are different. This study aims to investigate the mechanism of VIC/VMC translocation. Anterior tibial arteries were collected from patients with diabetic foot amputation. The patients were then divided into VIC and VMC groups. There were plaques in all anterior tibial arteries, while the enrichment of galectin-3 in arterial plaques in the VIC group was significantly higher than that in the VMC group. Furthermore, a macrophage/vascular smooth muscle cell (VSMC) coculture system was constructed. VSMC-derived extracellular vesicles (EVs) was labeled with fluorescent probe. After macrophages were pretreated with recombinant galectin-3 protein, the migration of VSMC-derived EVs and VSMC-derived calcification was more pronounced. And anti-galectin-3 antibody can inhibit this process of EVs and calcification translocation. Then, lentivirus (LV)-treated bone marrow cells (BMCs) were transplanted into apolipoprotein E-deficient (ApoE-/-) mice, and a diabetic atherosclerosis mouse model was constructed. After 15 wk of high-fat diet, ApoE-/- mice transplanted with LV-shgalectin-3 BMCs exhibited medial calcification and a concentrated distribution of EVs in the media. In conclusion, upregulation of galectin-3 in macrophages promotes the migration of VSMC-derived EVs to the intima and induces diabetic vascular intimal calcification.NEW & NOTEWORTHY The clinical risk and prognosis of vascular intimal and medial calcification are different. Macrophage galectin-3 regulates the migration of vascular smooth muscle cell-derived extracellular vesicles and mediates diabetic vascular intimal/medial calcification translocation. This study may provide insights into the early intervention in diabetic vascular calcification.


Assuntos
Angiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Galectina 3/metabolismo , Macrófagos/metabolismo , Túnica Íntima/metabolismo , Calcificação Vascular/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Apolipoproteínas E/genética , Células Cultivadas , Angiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/patologia , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Artérias da Tíbia/metabolismo , Artérias da Tíbia/patologia , Túnica Íntima/patologia , Calcificação Vascular/patologia
10.
J Immunol Res ; 2020: 1906204, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32190703

RESUMO

BACKGROUND: Macrophage-derived foam cells play a central role in atherosclerosis, and their ultimate fate includes apoptosis, promotion of vascular inflammation, or migration to other tissues. Nε-Carboxymethyl-lysine (CML), the key active component of advanced glycation end products, induced foam cell formation and apoptosis. Previous studies have shown that the Vav1/Rac1 pathway affects the macrophage cytoskeleton and cell migration, but its role in the pathogenesis of diabetic atherosclerosis is unknown. METHODS AND RESULTS: In this study, we used anterior tibiofibular vascular samples from diabetic foot amputation patients and accident amputation patients, and histological and cytological tests were performed using a diabetic ApoE-/- mouse model and primary peritoneal macrophages, respectively. The results showed that the atherosclerotic plaques of diabetic foot amputation patients and diabetic ApoE-/- mice were larger than those of the control group. Inhibition of the Vav1/Rac1 pathway reduced vascular plaques and promoted the migration of macrophages to lymph nodes. Transwell and wound healing assays showed that the migratory ability of macrophage-derived foam cells was inhibited by CML. Cytoskeletal staining showed that advanced glycation end products inhibited the formation of lamellipodia in foam cells, and inhibition of the Vav1/Rac1 pathway restored the formation of lamellipodia. CONCLUSION: CML inhibits the migration of foam cells from blood vessels via the Vav1/Rac1 pathway, and this process affects the formation of lamellipodia.


Assuntos
Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Pé Diabético/metabolismo , Células Espumosas/fisiologia , Lisina/análogos & derivados , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Amputação Cirúrgica , Animais , Apolipoproteínas E/genética , Aterosclerose/patologia , Movimento Celular , Células Cultivadas , Pé Diabético/patologia , Humanos , Lisina/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais
11.
Heart Lung Circ ; 29(1): 112-117, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31230870

RESUMO

Vascular calcification is a key character of advanced plaque in diabetic atherosclerosis. Microcalcification induces plaque rupture, whereas macrocalcification contributes to plaque stability. However, there is still no clear explanation for the formation and transition of these two types of calcification. Based on existing work and the latest international progress, this article provides a brief review of four aspects: calcification transition in plaque; matrix vesicle-mediated calcification transition in plaque; regulation mechanism of matrix vesicle-mediated calcification transition in diabetic plaque; and proposal of a new hypothesis, which may offer a new perspective on the study of the mechanism of calcification transition in plaque.


Assuntos
Aterosclerose/metabolismo , Angiopatias Diabéticas/metabolismo , Matriz Extracelular/metabolismo , Placa Aterosclerótica/metabolismo , Calcificação Vascular/metabolismo , Animais , Aterosclerose/patologia , Angiopatias Diabéticas/patologia , Matriz Extracelular/patologia , Humanos , Placa Aterosclerótica/patologia , Calcificação Vascular/patologia
12.
J Cardiovasc Pharmacol ; 75(3): 200-207, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31868826

RESUMO

Nuclear factor of activated T cell cytoplasmic 1 (NFATc1), a crucial member of the transcription factor NFAT family, is indispensable in the immune system and the morphogenesis of cardiac valves and septa and is also vital in osteoclasts and atherosclerotic calcification. Currently, osteoporosis and vascular diseases are severely hazardous to health and quality of life, and the 2 conditions always coincide with each other. The bone-vascular axis calcification paradox serves as a bridge between bone and vascular diseases, linking these 2 seemingly separate diseases, and the receptor activator of NF-κB (RANK)/receptor activator of nuclear factor kappa-B ligand (RANKL)/osteoprotegerin (OPG) system may be the common mechanism of the bone-vascular axis calcification paradox. NFATc1 provides a new therapeutic target for bone and vascular diseases. However, the specific mechanism by which NFATc1 acts on the bone-vascular axis calcification paradox, whether NFATc1 is related to the RANK/RANKL/OPG system, and how to use NFATc1 as a therapeutic target to avoid its side effects in other systems requires further study.


Assuntos
Artérias/metabolismo , Aterosclerose/metabolismo , Remodelação Óssea , Fatores de Transcrição NFATC/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Calcificação Vascular/metabolismo , Animais , Artérias/patologia , Aterosclerose/patologia , Humanos , Osteoblastos/patologia , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Calcificação Vascular/patologia
13.
J Cardiovasc Pharmacol ; 74(5): 372-378, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31361703

RESUMO

Matrix mineralization can be divided into physiological mineralization and pathological mineralization. There is a consensus among existing studies that matrix vesicles (MVs) are the starting sites of bone mineralization, and each component of MVs serves a certain function in mineralization. In addition, ectopic MVs pathologically promote undesired calcification, the primary focus of which is the promotion of vascular calcification. However, the specific mechanisms of the actions of MVs in bone-vascular axis cross-talk have not been fully elucidated. This review summarizes the latest research in this field and explores the roles of MVs in the bone-vascular axis with the aim of generating new ideas for the prevention and treatment of vascular calcification and bone metabolic disease.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Matriz Óssea/metabolismo , Calcificação Fisiológica , Micropartículas Derivadas de Células/metabolismo , Calcificação Vascular/metabolismo , Animais , Doenças Ósseas Metabólicas/patologia , Matriz Óssea/patologia , Micropartículas Derivadas de Células/patologia , Humanos , Transdução de Sinais , Calcificação Vascular/patologia
14.
Acta Diabetol ; 56(4): 457-472, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30603868

RESUMO

AIMS: Macrocalcification and microcalcification present different clinical risks, but the regulatory of their formation was unclear. Therefore, this study explored the underlying mechanisms of macrocalcification and microcalcification in diabetes mellitus. METHODS: Anterior tibial arteries of amputated diabetic feet were collected. According to the calcium content, patients were divided into less-calcification group and more-calcification group. And calcification morphology in plaques was observed. For further study, an in vivo mouse diabetic atherosclerosis model and an in vitro primary mouse aortic smooth muscle cell model were established. After the receptors for AGEs (RAGE) or galectin-3 were silenced, calcified nodule sizes and sortilin expression were determined. Scanning electron microscopy (SEM) was performed to detect the aggregation of matrix vesicles with the inhibition or promotion of sortilin. RESULTS: Both macro- and microcalcification were found in human anterior tibial artery plaques. Macrocalcification formed after the silencing of RAGE, and microcalcification formed after the silencing of galectin-3. In the process of RAGE- or galcetin-3-induced calcification, sortilin played an important role downstream. SEM showed that sortilin promoted the aggregation of MVs in the early stage of calcification and formed larger calcified nodules. CONCLUSION: RAGE downregulated sortilin and then transmitted microcalcification signals, whereas galectin-3 upregulated sortilin, which accelerated the aggregation of MVs in the early stage of calcification and mediated the formation of macrocalcifications, These data illustrate the progression of two calcification types and suggest sortilin as a potential target for early intervention of calcification and as an effective biomarker for the assessment of long-term clinical risk and prognosis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Galectina 3/fisiologia , Placa Aterosclerótica/genética , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Calcificação Vascular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Amputação Cirúrgica , Animais , Aorta/metabolismo , Aorta/patologia , Proteínas Sanguíneas , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/cirurgia , Pé Diabético/patologia , Pé Diabético/cirurgia , Galectinas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/metabolismo , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estreptozocina , Artérias da Tíbia/metabolismo , Artérias da Tíbia/patologia , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia
15.
Glycoconj J ; 35(5): 443-450, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29987432

RESUMO

The formation of advanced glycation end-products(AGEs) is an important cause of metabolic memory in diabetic patients and a key factor in the formation of atherosclerosis(AS) plaques in patients with diabetes mellitus. Related studies showed that AGEs could disrupt hemodynamic steady-state and destroy vascular wall integrity through the endothelial barrier damage, foam cell(FC) formation, apoptosis, calcium deposition and other aspects. At the same time, AGEs could initiate oxidative stress and inflammatory response cascade via receptor-depended and non-receptor-dependent pathways, promoting plaques to develop from a steady state to a vulnerable state and eventually tend to rupture and thrombosis. Numerous studies have confirmed that these pathological processes mentioned above could lead to acute coronary heart disease(CHD) and other acute cardiovascular and cerebrovascular events. However, the specific role of AGEs in the progression and regression of AS plaques has not yet been fully elucidated. In this paper, the formation, source, metabolism, physical and chemical properties of AGEs and their role in the migration of FCs and plaque calcification are briefly described, we hope to provide new ideas for the researchers that struggling in this field.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Placa Aterosclerótica/metabolismo , Animais , Apoptose , Células Espumosas/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/patologia
16.
Biomed Pharmacother ; 97: 1020-1031, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29136780

RESUMO

Among the various complications of type 2 diabetes mellitus, atherosclerosis causes the highest disability and morbidity. A multitude of macrophage-derived foam cells are retained in atherosclerotic plaques resulting not only from recruitment of monocytes into lesions but also from a reduced rate of macrophage migration from lesions. Nε-carboxymethyl-Lysine (CML), an advanced glycation end product, is responsible for most complications of diabetes. This study was designed to investigate the mechanism of CML/CD36 accelerating atherosclerotic progression via inhibiting foam cell migration. In vivo study and in vitro study were performed. For the in vivo investigation, CML/CD36 accelerated atherosclerotic progression via promoting the accumulation of macrophage-derived foam cells in aorta and inhibited macrophage-derived foam cells in aorta migrating to the para-aorta lymph node of diabetic apoE-/- mice. For the in vitro investigation, CML/CD36 inhibited RAW264.7-derived foam cell migration through NOX-derived ROS, FAK phosphorylation, Arp2/3 complex activation and F-actin polymerization. Thus, we concluded that CML/CD36 inhibited foam cells of plaque migrating to para-aorta lymph nodes, accelerating atherosclerotic progression. The corresponding mechanism may be via free cholesterol, ROS generation, p-FAK, Arp2/3, F-actin polymerization.


Assuntos
Aterosclerose/patologia , Antígenos CD36/metabolismo , Lisina/análogos & derivados , Placa Aterosclerótica/patologia , Animais , Aorta/metabolismo , Apolipoproteínas E/genética , Inibição de Migração Celular/fisiologia , Movimento Celular , Colesterol/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 2/complicações , Modelos Animais de Doenças , Progressão da Doença , Células Espumosas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Lisina/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células RAW 264.7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA