Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.403
Filtrar
1.
J Clin Invest ; 134(19)2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39352379

RESUMO

Clonal hematopoiesis of indeterminate potential (CHIP) has emerged as a previously unrecognized, potent, age-related, and common risk factor for atherosclerosis. Somatic mutations in certain known leukemia driver genes give rise to clones of mutant cells in peripheral blood. The increased risk of developing hematologic malignancy does not, on its own, explain excess mortality in individuals with CHIP. Cardiovascular disease accounts for much of this gap. Experimental evidence supports the causality of certain CHIP mutations in accelerated atherosclerosis. CHIP due to mutations in different driver genes varies in their promotion of atherosclerotic events and in the region of augmented atherosclerotic involvement. For example, CHIP due to mutations in DNMT3a appears less atherogenic than CHIP that arises from TET2 or JAK2, forms of CHIP that incite inflammation. The recognition of certain CHIP mutations as promoters of atherosclerotic risk has opened new insights into understanding of the pathophysiology of this disease. The accentuated cardiovascular risk and involvement of distinct pathways of various forms of CHIP also inform novel approaches to allocation of targeted therapies, affording a step toward personalized medicine.


Assuntos
Aterosclerose , Hematopoiese Clonal , DNA Metiltransferase 3A , Dioxigenases , Mutação , Humanos , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Hematopoiese Clonal/genética , DNA Metiltransferase 3A/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Animais , Janus Quinase 2/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Risco
2.
Sci Signal ; 17(855): eadt2301, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39316664

RESUMO

Early exposure to cholesterol primes later atherosclerosis through phenotypic changes to resident-like arterial macrophages.


Assuntos
Aterosclerose , Macrófagos , Placa Aterosclerótica , Humanos , Placa Aterosclerótica/patologia , Placa Aterosclerótica/metabolismo , Macrófagos/metabolismo , Macrófagos/citologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Animais , Colesterol/metabolismo
3.
Atherosclerosis ; 397: 118570, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39276419

RESUMO

BACKGROUND AND AIMS: CCN4/WISP-1 regulates various cell behaviours that contribute to atherosclerosis progression, including cell adhesion, migration, proliferation and survival. We therefore hypothesised that CCN4 regulates the development and progression of atherosclerotic plaques. METHODS: We used a high fat fed ApoE-/- mouse model to study atherosclerotic plaque progression in the brachiocephalic artery and aortic root. In protocol 1, male ApoE-/- mice with established plaques were given a CCN4 helper-dependent adenovirus to see the effect of treatment with CCN4, while in protocol 2 male CCN4-/-ApoE-/- were compared to CCN4+/+ApoE-/- mice to assess the effect of CCN4 deletion on plaque progression. RESULTS: CCN4 overexpression resulted in reduced occlusion of the brachiocephalic artery with less apoptosis, fewer macrophages, and attenuated lipid core size. The amount of plaque found on the aortic root was also reduced. CCN4 deficiency resulted in increased apoptosis and occlusion of the brachiocephalic artery as well as increased plaque in the aortic root. Additionally, in vitro cells from CCN4-/-ApoE-/- mice had higher apoptotic levels. CCN4 deficiency did not significantly affect blood cholesterol levels or circulating myeloid cell populations. CONCLUSIONS: We conclude that in an atherosclerosis model the most important action of CCN4 is the effect on cell apoptosis. CCN4 provides pro-survival signals and leads to reduced cell death, lower macrophage number, smaller lipid core size and reduced atherosclerotic plaque burden. As such, the pro-survival effect of CCN4 is worthy of further investigation, in a bid to find a therapeutic for atherosclerosis.


Assuntos
Apoptose , Aterosclerose , Placa Aterosclerótica , Animais , Masculino , Camundongos , Aorta/patologia , Aorta/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/patologia , Aterosclerose/metabolismo , Aterosclerose/genética , Tronco Braquiocefálico/patologia , Tronco Braquiocefálico/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Proteínas de Sinalização Intercelular CCN/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE
4.
Cell Death Dis ; 15(9): 689, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39304654

RESUMO

Recent research has explored the potential of the demethylating drug 5-azacytidine (Aza) as therapy for a range of diseases. However, the therapeutic efficacy of Aza for patients of atherosclerosis remains unclear. This study investigates the therapeutic application of Aza to atherosclerosis in order to elucidate the underlying mechanisms. We generated induced Tregs (iTregs) from CD4+ T cells by using Aza in vitro, and this was followed by the intravenous infusion of iTregs for the treatment of atherosclerosis. The adoptive transfer of Aza-iTreg significantly increased peripheral blood Treg cells, suppressed inflammation, and attenuated atherosclerosis in ApoE-/- mice. Furthermore, we observed a notable demethylation of the Forkhead box P3 (Foxp3)-regulatory T cell-specific demethylated region (TSDR) and an upregulation of Foxp3 expression in the CD4+ T cells in the spleen of the ApoE-/- mice following the transfer of Aza- iTregs. We also demonstrated that Aza converted naive CD4+ T cells into Tregs by DNA methyltransferase 1 (Dnmt1)-mediated Foxp3-TSDR demethylation and the upregulation of Foxp3 expression. Conversely, the overexpression of Dnmt1 in the CD4+ T cells attenuated the Aza-induced Foxp3-TSDR demethylation and upregulation of Foxp3 expression. Our results reveal that Aza converts naive CD4+ T cells into functional Tregs by inhibiting Dnmt1, and the transfer of Aza-iTregs suppresses atherosclerosis in mice.


Assuntos
Aterosclerose , Azacitidina , Linfócitos T CD4-Positivos , Epigênese Genética , Fatores de Transcrição Forkhead , Linfócitos T Reguladores , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Azacitidina/farmacologia , Camundongos , Fatores de Transcrição Forkhead/metabolismo , Epigênese Genética/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Camundongos Endogâmicos C57BL , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Masculino , Humanos
5.
Nat Commun ; 15(1): 8273, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39333556

RESUMO

Disruption of ciliary homeostasis in vascular endothelial cells has been implicated in the development of atherosclerosis. However, the molecular basis for the regulation of endothelial cilia during atherosclerosis remains poorly understood. Herein, we provide evidence in male mice that the accumulation of lipid droplets in vascular endothelial cells induces ciliary loss and contributes to atherosclerosis. Triglyceride accumulation in vascular endothelial cells differentially affects the abundance of free fatty acid species in the cytosol, leading to stimulated lipid droplet formation and suppressed protein S-palmitoylation. Reduced S-palmitoylation of ciliary proteins, including ADP ribosylation factor like GTPase 13B, results in the loss of cilia. Restoring palmitic acid availability, either through pharmacological inhibition of stearoyl-CoA desaturase 1 or a palmitic acid-enriched diet, significantly restores endothelial cilia and mitigates the progression of atherosclerosis. These findings thus uncover a previously unrecognized role of lipid droplets in regulating ciliary homeostasis and provide a feasible intervention strategy for preventing and treating atherosclerosis.


Assuntos
Aterosclerose , Cílios , Células Endoteliais , Gotículas Lipídicas , Ácido Palmítico , Animais , Masculino , Ácido Palmítico/farmacologia , Ácido Palmítico/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/efeitos dos fármacos , Camundongos , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Cílios/metabolismo , Cílios/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Humanos , Triglicerídeos/metabolismo
6.
Circ Res ; 135(8): 856-872, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39224974

RESUMO

BACKGROUND: Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis. METHODS: Systems analyses integrating single-cell RNA sequencing and complementary immunologic approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming were assessed by integrated biochemical and genetic approaches. The intercellular propagation of homeostasis resolution was evaluated by coculture assays with donor monocytes trained by 4-PBA and recipient naive monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high-fat diet-fed ApoE-/- mouse model with IP 4-PBA administration. Furthermore, the selective efficacy of 4-PBA-trained monocytes was examined by IV transfusion of ex vivo trained monocytes by 4-PBA into recipient high-fat diet-fed ApoE-/- mice. RESULTS: In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 (intercellular adhesion molecule 1) via reducing peroxisome stress and attenuating SYK (spleen tyrosine kinase)-mTOR (mammalian target of rapamycin) signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ (peroxisome proliferator-activated receptor γ) neddylation mediated by TOLLIP (toll-interacting protein). 4-PBA-trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA-trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo. CONCLUSIONS: Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.


Assuntos
Aterosclerose , Inflamação , Monócitos , Fenilbutiratos , Animais , Aterosclerose/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Monócitos/metabolismo , Monócitos/efeitos dos fármacos , Camundongos , Inflamação/metabolismo , Fenilbutiratos/farmacologia , Camundongos Endogâmicos C57BL , Humanos , Masculino , Molécula 1 de Adesão Intercelular/metabolismo , Molécula 1 de Adesão Intercelular/genética , Camundongos Knockout para ApoE , PPAR gama/metabolismo , Reprogramação Celular/efeitos dos fármacos , Células Cultivadas , Anti-Inflamatórios/farmacologia
7.
Ann Med ; 56(1): 2407525, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39316662

RESUMO

INTRODUCTION: Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid accumulation, inflammation and apoptosis of the arterial wall. This study evaluated the effects of lysophosphatidylinositol (LPI) on endothelial cells activation and autophagy in AS. METHODS: qRT-PCR and Western blotting were done to verify the expression of ICAM1, GPR55 and SOD2. RNA-Seq was performed and screened for the different expressions of long noncoding RNAs (lncRNAs), combining bioinformatics analysis to elucidate the mechanism by which lncRNA functions. RESULTS: qRT-PCR and Western blotting results showed that LPI increased GPR55 and ICAM1 expression. RNA-Seq analysis and qRT-PCR results showed that LPI increased the expression of LINC01235, LINC00520 and LINC01963; LINC01235 was the most obvious. Mechanistically, bioinformatic analysis demonstrated that LINC01235 inhibited autophagy through sponging miR-224-3p. And miRNA-224-3p targeted RABEP1. CONCLUSIONS: LPI promoted endothelial cell activation. LPI induced the expression of LINC01235 and LINC01235 inhibited autophagy through miR-224-3p/RABEP1. Collectively, this study first reveals the function of LINC01235, which may serve as a potential therapeutic target in AS.


Assuntos
Aterosclerose , Autofagia , Lisofosfolipídeos , MicroRNAs , RNA Longo não Codificante , Receptores de Canabinoides , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Humanos , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Receptores de Canabinoides/metabolismo , Receptores de Canabinoides/genética , MicroRNAs/metabolismo , MicroRNAs/genética , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Molécula 1 de Adesão Intercelular/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo
8.
ACS Appl Mater Interfaces ; 16(39): 51944-51956, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39287614

RESUMO

Atherosclerosis is a primary cause of cardiovascular and cerebrovascular diseases, with the unpredictable rupture of vulnerable atherosclerotic plaques enriched with lipid-laden macrophages being able to lead to heart attacks and strokes. Activating macrophage autophagy presents itself as a promising strategy for preventing vulnerable plaque formation and reducing the risk of rupture. In this study, we have developed a novel metal-free nanozyme (HCN@DS) that integrates the functions of multimodal imaging-guided therapy for atherosclerosis. HCN@DS has demonstrated high macrophage-targeting abilities due to its affinity toward scavenger receptor A (SR-A), along with excellent photoacoustic and photothermal imaging capabilities for guiding the precise treatment. It combines mild photothermal effects with moderate reactive oxygen species (ROS) generation to treat atherosclerosis. This controlled approach activates autophagy in atherosclerotic macrophages, inhibiting foam cell formation by reducing the uptake of oxidized low-density lipoproteins (oxLDL) and promoting efferocytosis and cholesterol efflux in macrophages. Additionally, it prevents plaque rupture by inhibiting apoptosis and inflammation within the plaque. Therefore, this metal-free nanozyme holds great potential for reducing the risk of atherosclerosis due to its high biosafety, excellent targeting ability, dual-modality imaging capability, and appropriate modulation of autophagy.


Assuntos
Aterosclerose , Autofagia , Macrófagos , Placa Aterosclerótica , Animais , Camundongos , Autofagia/efeitos dos fármacos , Aterosclerose/diagnóstico por imagem , Aterosclerose/metabolismo , Aterosclerose/patologia , Placa Aterosclerótica/diagnóstico por imagem , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Células RAW 264.7 , Lipoproteínas LDL/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Humanos , Nanopartículas/química , Receptores Depuradores Classe A/metabolismo
9.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39337246

RESUMO

The role of ferroptosis and iron metabolism dysregulation in the pathophysiology of cardiovascular diseases is increasingly recognized. Conditions such as hypertension, cardiomyopathy, atherosclerosis, myocardial ischemia/reperfusion injury, heart failure, and cardiovascular complications associated with COVID-19 have been linked to these processes. Inflammation is central to these conditions, prompting exploration into the inflammatory and immunoregulatory molecular pathways that mediate ferroptosis and its contribution to cardiovascular disease progression. Notably, emerging evidence highlights interleukin-37 as a protective cytokine with the ability to activate the nuclear factor erythroid 2-related factor 2 pathway, inhibit macrophage ferroptosis, and attenuate atherosclerosis progression in murine models. However, a comprehensive review focusing on interleukin-37 and its protective role against ferroptosis in CVD is currently lacking. This review aims to fill this gap by summarizing existing knowledge on interleukin-37, including its regulatory functions and impact on ferroptosis in conditions such as atherosclerosis and myocardial infarction. We also explore experimental strategies and propose that targeting interleukin-37 to modulate ferroptosis presents a promising therapeutic approach for the prevention and treatment of cardiovascular diseases.


Assuntos
Doenças Cardiovasculares , Ferroptose , Interleucina-1 , Humanos , Interleucina-1/metabolismo , Doenças Cardiovasculares/metabolismo , Animais , COVID-19/metabolismo , COVID-19/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , SARS-CoV-2/metabolismo
10.
Ecotoxicol Environ Saf ; 284: 116983, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39232293

RESUMO

Microplastics and nanoplastics (MNPs) originating from plastic pollution pose potential threats to cardiovascular health, with prior studies linking MNPs to atherosclerosis. Our earlier research elucidated how nanoplastics enhance macrophages' phagocytic activity, leading to the formation of foam cells and an elevated risk of atherosclerosis. However, the specific influence of MNPs on smooth muscle cells (SMCs) in the context of MNP-induced atherosclerosis remains poorly understood. In this study, ApoE knockout (ApoE-/-) male mice with a high-fat diet were orally exposed to environmentally realistic concentrations of 2.5-250 mg/kg polystyrene nanoplastics (PS-NPs, 50 nm) for consecutive 19 weeks. Cardiovascular toxicity was comprehensively assessed through histopathological, transcriptomic, and proteomic analyses, while mechanisms underlying this toxicity were explored through in vitro studies. Herein, hematoxylin and eosin staining revealed accelerated atherosclerotic plaque development in ApoE-/- mice exposed to PS-NPs. Multi-omics analysis identified kinesin family member 15 (KIF15) as a pivotal target molecule. Both in vitro and in vivo experiments affirmed the specific upregulation of KIF15 in mouse aortic SMCs exposed to PS-NPs. Furthermore, in vitro experiments demonstrated that PS-NPs can promote the migration ability of MOVAS cells. Knockdown of Kif15 revealed its role in reducing MOVAS cell migration, with subsequent exposure to PS-NPs reversing the increased migration ability. This suggests that PS-NPs promote SMC migration by upregulating KIF15, and the migration of SMCs is closely associated with atherosclerosis outcomes. This study significantly advances our understanding of MNP-induced cardiovascular toxicity, providing valuable insights for risk assessment of human MNP exposure.


Assuntos
Aterosclerose , Microplásticos , Miócitos de Músculo Liso , Poliestirenos , Animais , Masculino , Camundongos , Apolipoproteínas E/genética , Aterosclerose/induzido quimicamente , Aterosclerose/patologia , Movimento Celular/efeitos dos fármacos , Cinesinas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Knockout para ApoE , Microplásticos/toxicidade , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Nanopartículas/toxicidade , Poliestirenos/toxicidade
11.
Exp Mol Med ; 56(9): 1967-1979, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39223261

RESUMO

Telomere dysfunction is a well-known molecular trigger of senescence and has been associated with various age-related diseases, including atherosclerosis. However, the mechanisms involved have not yet been elucidated, and the extent to which telomeres contribute to atherosclerosis is unknown. Therefore, we investigated the mechanism of metformin-induced telomere stabilization and the ability of metformin to inhibit vascular smooth muscle cell (VSMC) senescence caused by advanced atherosclerosis. The present study revealed that metformin inhibited the phenotypes of atherosclerosis and senescence in VSMCs. Metformin increased the phosphorylation of AMPK-dependent PGC-1α and thus increased telomerase activity and the protein level of TERT in OA-treated VSMCs. Mechanistically, the phosphorylation of AMPK and PGC-1α by metformin not only enhanced telomere function but also increased the protein level of TERT, whereas TERT knockdown accelerated the development of atherosclerosis and senescent phenotypes in OA-treated VSMCs regardless of metformin treatment. Furthermore, the in vivo results showed that metformin attenuated the formation of atherosclerotic plaque markers in the aortas of HFD-fed ApoE KO mice. Although metformin did not reduce plaque size, it inhibited the phosphorylation of the AMPK/PGC-1α/TERT signaling cascade, which is associated with the maintenance and progression of plaque formation, in HFD-fed ApoE KO mice. Accordingly, metformin inhibited atherosclerosis-associated phenotypes in vitro and in vivo. These observations show that the enhancement of telomere function by metformin is involved in specific signaling pathways during the progression of atherosclerosis. These findings suggest that telomere stabilization by metformin via the AMPK/p-PGC-1α pathway might provide a strategy for developing therapeutics against vascular diseases such as atherosclerosis.


Assuntos
Proteínas Quinases Ativadas por AMP , Aterosclerose , Metformina , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transdução de Sinais , Telômero , Metformina/farmacologia , Metformina/uso terapêutico , Animais , Aterosclerose/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Aterosclerose/etiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Transdução de Sinais/efeitos dos fármacos , Telômero/metabolismo , Telômero/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/efeitos dos fármacos , Masculino , Telomerase/metabolismo , Telomerase/genética , Fosforilação/efeitos dos fármacos , Progressão da Doença , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Camundongos Knockout , Homeostase do Telômero/efeitos dos fármacos , Modelos Animais de Doenças
12.
Int J Mol Sci ; 25(18)2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39337364

RESUMO

Atherosclerosis involves an inflammatory response due to plaque formation within the arteries, which can lead to ischemic stroke and heart disease. It is one of the leading causes of death worldwide, with various contributing factors such as hyperlipidemia, hypertension, obesity, diabetes, and smoking. Wall shear stress (WSS) is also known as a contributing factor of the formation of atherosclerotic plaques. Since the causes of atherosclerosis cannot be attributed to a single factor, clearly understanding the mechanisms and causes of its occurrence is crucial for preventing the disease and developing effective treatment strategies. To better understand atherosclerosis and define the correlation between various contributing factors, computational fluid dynamics (CFD) analysis is primarily used. CFD simulates WSS, the frictional force caused by blood flow on the vessel wall with various hemodynamic changes. Using apolipoprotein E knockout (ApoE-KO) mice subjected to partial ligation and a high-fat diet at 1-week, 2-week, and 4-week intervals as an atherosclerosis model, CFD analysis was conducted along with the reconstruction of carotid artery blood flow via magnetic resonance imaging (MRI) and compared to the inflammatory factors and pathological staining. In this experiment, a comparative analysis of the effects of high WSS and low WSS was conducted by comparing the standard deviation of time-averaged wall shear stress (TAWSS) at each point within the vessel wall. As a novel approach, the standard deviation of TAWSS within the vessel was analyzed with the staining results and pathological features. Since the onset of atherosclerosis cannot be explained by a single factor, the aim was to find the correlation between the thickness of atherosclerotic plaques and inflammatory factors through standard deviation analysis. As a result, the gap between low WSS and high WSS widened as the interval between weeks in the atherosclerosis mouse model increased. This finding not only linked the occurrence of atherosclerosis to WSS differences but also provided a connection to the causes of vulnerable plaques.


Assuntos
Apolipoproteínas E , Aterosclerose , Hidrodinâmica , Estresse Mecânico , Animais , Masculino , Camundongos , Apolipoproteínas E/genética , Aterosclerose/patologia , Aterosclerose/etiologia , Aterosclerose/metabolismo , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Hemodinâmica , Camundongos Knockout , Camundongos Knockout para ApoE , Modelos Cardiovasculares , Placa Aterosclerótica/patologia , Resistência ao Cisalhamento
13.
Cardiovasc Diabetol ; 23(1): 331, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39252021

RESUMO

BACKGROUND: Visceral adipose tissue in individuals with obesity is an independent cardiovascular risk indicator. However, it remains unclear whether adipose tissue influences common cardiovascular diseases, such as atherosclerosis, through its secreted exosomes. METHODS: The exosomes secreted by adipose tissue from diet-induced obesity mice were isolated to examine their impact on the progression of atherosclerosis and the associated mechanism. Endothelial apoptosis and the proliferation and migration of vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque were evaluated. Statistical significance was analyzed using GraphPad Prism 9.0 with appropriate statistical tests. RESULTS: We demonstrate that adipose tissue-derived exosomes (AT-EX) exacerbate atherosclerosis progression by promoting endothelial apoptosis, proliferation, and migration of VSMCs within the plaque in vivo. MicroRNA-132/212 (miR-132/212) was detected within AT-EX cargo. Mechanistically, miR-132/212-enriched AT-EX exacerbates palmitate acid-induced endothelial apoptosis via targeting G protein subunit alpha 12 and enhances platelet-derived growth factor type BB-induced VSMC proliferation and migration by targeting phosphatase and tensin homolog in vitro. Importantly, melatonin decreases exosomal miR-132/212 levels, thereby mitigating the pro-atherosclerotic impact of AT-EX. CONCLUSION: These data uncover the pathological mechanism by which adipose tissue-derived exosomes regulate the progression of atherosclerosis and identify miR-132/212 as potential diagnostic and therapeutic targets for atherosclerosis.


Assuntos
Apoptose , Aterosclerose , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Exossomos , Camundongos Endogâmicos C57BL , MicroRNAs , Músculo Liso Vascular , Miócitos de Músculo Liso , Placa Aterosclerótica , Animais , MicroRNAs/metabolismo , MicroRNAs/genética , Exossomos/metabolismo , Exossomos/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/genética , Proliferação de Células/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Masculino , Transdução de Sinais , Células Cultivadas , Obesidade/metabolismo , Obesidade/patologia , Camundongos Knockout para ApoE , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/efeitos dos fármacos , Doenças da Aorta/patologia , Doenças da Aorta/metabolismo , Doenças da Aorta/genética , Becaplermina/farmacologia , Becaplermina/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Camundongos , Humanos
14.
J Cell Mol Med ; 28(17): e70064, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39248527

RESUMO

Cathepsin B (CTSB) is a member of the cysteine protease family, primarily responsible for degrading unnecessary organelles and proteins within the acidic milieu of lysosomes to facilitate recycling. Recent research has revealed that CTSB plays a multifaceted role beyond its function as a proteolytic enzyme in lysosomes. Importantly, recent data suggest that CTSB has significant impacts on different cardiac pathological conditions, such as atherosclerosis (AS), myocardial infarction, hypertension, heart failure and cardiomyopathy. Especially in the context of AS, preclinical models and clinical sample imaging data indicate that the cathepsin activity-based probe can reliably image CTSB activity in foam cells and atherosclerotic plaques; concurrently, it allows synchronous diagnostic and therapeutic interventions. However, our knowledge of CTSB in cardiovascular disease is still in the early stage. This paper aims to provide a comprehensive review of the significance of CTSB in cardiovascular physiology and pathology, with the objective of laying a theoretical groundwork for the development of drugs targeting CTSB.


Assuntos
Doenças Cardiovasculares , Catepsina B , Humanos , Catepsina B/metabolismo , Doenças Cardiovasculares/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia
15.
Adv Exp Med Biol ; 1460: 575-594, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39287865

RESUMO

Lipotoxicity can mediate endothelial dysfunction in obesity. Altered endothelial cell phenotype during the pathobiological course of the lipotoxicity may lead to hemostatic abnormalities, which is a hallmark of several hematological disorders. Impaired hemostasis could also be directly related to numerous metabolic diseases such as hypertension, diabetes, and atherosclerosis. On the other hand, the local hematopoietic bone marrow (BM) renin-angiotensin system (RAS) contributes to the development of atherosclerosis via acting on the lipotoxicity processes. Local BM RAS, principally an autocrine/paracrine/intracrine hematological system, is located at the crossroads of cellular regulation, molecular interactions, and lipotoxicity-mediated vascular endothelial dysfunction. The positive regulatory role of plasma LDL on AT1 receptor-mediated hematopoietic stem cell (HSC) differentiation and the production of pro-atherogenic monocytes have been described. LDL-regulated HSC function may explain in part hypercholesterolemia-induced inflammation as well as the anti-inflammatory and anti-atherosclerotic effects of AT1 receptor blockers. The role of local adipose tissue RAS is directly related to the pathogenesis of metabolic derangements in obesity. There may be a crosstalk between local BM RAS and local adipose tissue RAS at the genomics and transcriptomics levels. This chapter aims to review hematological alterations propagating the pathological influences of lipotoxicity on the vascular endothelium.


Assuntos
Doenças Hematológicas , Obesidade , Sistema Renina-Angiotensina , Humanos , Obesidade/metabolismo , Obesidade/complicações , Doenças Hematológicas/metabolismo , Doenças Hematológicas/patologia , Doenças Hematológicas/etiologia , Sistema Renina-Angiotensina/fisiologia , Animais , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/etiologia
16.
Sci Rep ; 14(1): 21371, 2024 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-39266573

RESUMO

Atherosclerosis (AS) is a major cause of cardiovascular diseases that may lead to mortality. This study aimed to evaluate the therapeutic potential of tetrandrine in high cholesterol diet (HCD)-induced atherosclerosis, in rats, via modulation of miR-34a, as well as, Wnt5a/Ror2/ABCA1/NF-κB pathway and to compare its efficacy with atorvastatin. Induction of AS, in male rats, was done via IP administration of vitamin D3 (70 U/Kg for 3 days) together with HCD. At the end of the 9th week, rats were treated with atorvastatin at a dose of 20 mg/kg, and tetrandrine at different doses of (18.75, and 31.25 mg/kg) for 22 days. Serum inflammatory cytokines and lipid profile, liver oxidative stress parameters, and aortic tissue Wnt5a, Ror2, ABCA1, NF-κB, miR-34a levels were assessed in all experimental groups. Histopathological and Immunohistochemical assessments of aortic tissue sections were done. Results showed that tetrandrine treatment reverted the inflammatory and oxidative stress state together with reducing the serum lipids via modulating miR-34a, and Wnt5a/Ror2/ABCA1/NF-κB pathway. Moreover, it reverted the histopathological abnormalities observed in AS rats. Tetrandrine beneficial effects, in both doses, were comparable to that of atorvastatin, in most of the discussed parameters. These findings praise tetrandrine as a promising agent for management of atherosclerosis.


Assuntos
Transportador 1 de Cassete de Ligação de ATP , Aterosclerose , Benzilisoquinolinas , MicroRNAs , NF-kappa B , Proteína Wnt-5a , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Aterosclerose/patologia , Proteína Wnt-5a/metabolismo , Ratos , NF-kappa B/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Masculino , Benzilisoquinolinas/farmacologia , Benzilisoquinolinas/uso terapêutico , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Estresse Oxidativo/efeitos dos fármacos , Colecalciferol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ratos Wistar , Dieta Hiperlipídica/efeitos adversos , Colesterol na Dieta/efeitos adversos
17.
Nat Commun ; 15(1): 8034, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39271657

RESUMO

Atherosclerosis is an inflammatory disorder responsible for cardiovascular disease. Reactivation of efferocytosis, the phagocytic removal of cells by macrophages, has emerged as a translational target for atherosclerosis. Systemic blockade of the key 'don't-eat-me' molecule, CD47, triggers the engulfment of apoptotic vascular tissue and potently reduces plaque burden. However, it also induces red blood cell clearance, leading to anemia. To overcome this, we previously developed a macrophage-specific nanotherapy loaded with a chemical inhibitor that promotes efferocytosis. Because it was found to be safe and effective in murine studies, we aimed to advance our nanoparticle into a porcine model of atherosclerosis. Here, we demonstrate that production can be scaled without impairing nanoparticle function. At an early stage of disease, we find our nanotherapy reduces apoptotic cell accumulation and inflammation in the atherosclerotic lesion. Notably, this therapy does not induce anemia, highlighting the translational potential of targeted macrophage checkpoint inhibitors.


Assuntos
Anemia , Aterosclerose , Antígeno CD47 , Modelos Animais de Doenças , Inflamação , Macrófagos , Nanopartículas , Fagocitose , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanopartículas/química , Antígeno CD47/metabolismo , Antígeno CD47/antagonistas & inibidores , Suínos , Inflamação/patologia , Fagocitose/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Humanos , Placa Aterosclerótica/patologia , Camundongos , Masculino
18.
Sci Rep ; 14(1): 20684, 2024 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237581

RESUMO

Heme released from damaged and senescent red blood cells (RBCs) may contribute to oxidant-mediated cell injury. One of the recently investigated physiological processes, essential in preventing the inflammatory impact of labile heme, is its uptake from the bloodstream by endothelial cells (ECs). In this study, we investigated heme uptake by ECs starting from the model studies on the in vitro cellular level, through the endothelium layer on the ex vivo murine aortic tissues. As the cellular model, Human Aortic Endothelial Cells (HAECs) were chosen, and the concentration of labile heme was adjusted so to avoid the excessive toxic effect of the labile heme. We utilized label-free Raman imaging with two different excitation wavelengths to capture the uptake process in situ and characterize the oxidation state of the iron ion in the intercalated heme. The phenomenon of heme uptake was demonstrated in both, the healthy control C57Bl/6J and FVB animals, as well as in mice with developed atherosclerosis (ApoE/LDLR-/- mice). In the presented work, we presented for the first time Raman-based evidence on the heme uptake process by endothelial cells in both, in vitro and ex vivo systems.


Assuntos
Células Endoteliais , Heme , Análise Espectral Raman , Animais , Heme/metabolismo , Análise Espectral Raman/métodos , Células Endoteliais/metabolismo , Camundongos , Humanos , Camundongos Endogâmicos C57BL , Aterosclerose/metabolismo , Aterosclerose/patologia
19.
Lipids Health Dis ; 23(1): 279, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227809

RESUMO

BACKGROUND: NOD-like receptor protein 3 (NLRP3) inflammasome activation is indispensable for atherogenesis. Mitophagy has emerged as a potential strategy to counteract NLRP3 inflammasome activation triggered by impaired mitochondria. Our previous research has indicated that dihydromyricetin, a natural flavonoid, can mitigate NLRP3-mediated endothelial inflammation, suggesting its potential to treat atherosclerosis. However, the precise underlying mechanisms remain elusive. This study sought to investigate whether dihydromyricetin modulates endothelial mitophagy and inhibits NLRP3 inflammasome activation to alleviate atherogenesis, along with the specific mechanisms involved. METHODS: Apolipoprotein E-deficient mice on a high-fat diet were administered daily oral gavages of dihydromyricetin for 14 weeks. Blood samples were procured to determine the serum lipid profiles and quantify proinflammatory cytokine concentrations. Aortas were harvested to evaluate atherosclerotic plaque formation and NLRP3 inflammasome activation. Concurrently, in human umbilical vein endothelial cells, Western blotting, flow cytometry, and quantitative real-time PCR were employed to elucidate the mechanistic role of mitophagy in the modulation of NLRP3 inflammasome activation by dihydromyricetin. RESULTS: Dihydromyricetin administration significantly attenuated NLRP3 inflammasome activation and vascular inflammation in mice on a high-fat diet, thereby exerting a pronounced inhibitory effect on atherogenesis. Both in vivo and in vitro, dihydromyricetin treatment markedly enhanced mitophagy. This enhancement in mitophagy ameliorated the mitochondrial damage instigated by saturated fatty acids, thereby inhibiting the activation and nuclear translocation of NF-κB. Consequently, concomitant reductions in the transcript levels of NLRP3 and interleukin-1ß (IL-1ß), alongside decreased activation of NLRP3 inflammasome and IL-1ß secretion, were discerned. Notably, the inhibitory effects of dihydromyricetin on the activation of NF-κB and subsequently the NLRP3 inflammasome were determined to be, at least in part, contingent upon its capacity to promote mitophagy. CONCLUSION: This study suggested that dihydromyricetin may function as a modulator to promote mitophagy, which in turn mitigates NF-κB activity and subsequent NLRP3 inflammasome activation, thereby conferring protection against atherosclerosis.


Assuntos
Aterosclerose , Dieta Hiperlipídica , Flavonóis , Células Endoteliais da Veia Umbilical Humana , Inflamassomos , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Mitofagia/efeitos dos fármacos , Animais , Flavonóis/farmacologia , Aterosclerose/tratamento farmacológico , Aterosclerose/prevenção & controle , Aterosclerose/patologia , Aterosclerose/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Camundongos , Humanos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Dieta Hiperlipídica/efeitos adversos , Masculino , Camundongos Endogâmicos C57BL , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo
20.
Mol Med ; 30(1): 137, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227813

RESUMO

N6-methyladenosine (m6A) modification stands out among various RNA modifications as the predominant form within eukaryotic cells, influencing numerous cellular processes implicated in disease development. m6A modification has gained increasing attention in the development of atherosclerosis and has become a research hotspot in recent years. Programmed cell death (PCD), encompassing apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, plays a pivotal role in atherosclerosis pathogenesis. In this review, we delve into the intricate interplay between m6A modification and diverse PCD pathways, shedding light on their complex association during the onset and progression of atherosclerosis. Clarifying the relationship between m6A and PCD in atherosclerosis is of great significance to provide novel strategies for cardiovascular disease treatment.


Assuntos
Adenosina , Apoptose , Aterosclerose , Humanos , Adenosina/análogos & derivados , Adenosina/metabolismo , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Animais , Apoptose/genética , Autofagia/genética , RNA/genética , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA