Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.119
Filtrar
1.
Discov Med ; 36(182): 571-580, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38531797

RESUMO

BACKGROUND: The apoptosis of vascular smooth muscle cells (VSMCs) contributes to the progression of atherosclerosis (AS). Long intergenic non-protein coding RNA 1128 (LINC01128) has been implicated in AS, and this study aims to uncover the role and mechanism of LINC01128 in regulating oxidized low-density lipoprotein (oxLDL)-induced VSMCs. METHODS: The position of LINC01128 in cells and its target genes were predicted using bioinformatics. The localization of LINC01128 in human VSMCs was determined through fluorescence in situ hybridization. VSMCs were transfected, and the interaction between LINC01128 and fucosyltransferase 8 (FUT8) was validated by chromatin immunoprecipitation assay. The apoptotic VSMC model was established using oxLDL. LINC01128 expression in VSMCs was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR), and FUT8 expression was detected by qRT-PCR and western blot. VSMC viability, migration, invasion abilities, and apoptosis were assessed using cell counting kit-8, transwell assay, and flow cytometry, respectively. RESULTS: OxLDL (200 µg/mL) upregulated the expression of LINC01128 and FUT8 mRNA, as well as FUT8 protein, in VSMCs. LINC01128 was expressed in the nucleus of VSMCs and bound to FUT8. Knockdown of LINC01128 alleviated the inhibitory effects of oxLDL (200 µg/mL) on viability, migration, and invasion, and mitigated the promotion of apoptosis and FUT8 expression in VSMCs. On the other hand, FUT8 overexpression enhanced the suppressive effects of oxLDL (200 µg/mL) on viability, migration, and invasion activities, and amplified the facilitating effect of oxLDL on apoptosis in VSMCs. Moreover, FUT8 overexpression reversed the impact of LINC01128 silencing on viability, migration, invasion, and apoptosis in oxLDL-stimulated VSMCs. CONCLUSION: The knockdown of LINC01128 downregulates FUT8, inhibiting the progression of VSMCs in AS.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Músculo Liso Vascular/metabolismo , Hibridização in Situ Fluorescente , Aterosclerose/metabolismo , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Apoptose , Proliferação de Células , MicroRNAs/metabolismo , Movimento Celular , Células Cultivadas
2.
Clinics (Sao Paulo) ; 79: 100343, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38554490

RESUMO

OBJECTIVE: Atherosclerosis (AS) is a chronic inflammatory disease of the arterial wall, in which Human Vascular Smooth Muscle Cells (HVSMCs) are involved. Nevertheless, the functions and mechanisms of circRNAs in oxidized Low-Density Lipoprotein (ox-LDL)-induced vascular smooth muscle cells remain unclear. METHODS: Circ-ABCA1 expression was measured in the models of AS. Then, in the vitro model, oligonucleotide transfection was performed, followed by an analysis of VSMC proliferation, migration, inflammation, and phenotypic switch. Also, in the in vivo model, mice were injected with shRNA lentivirus, followed by histological examination of aortic tissues. Finally, the interaction of circ-ABCA1, miR-885-5p, and ROCK2 was identified. RESULTS: Circ-ABCA1, was confirmed to be overexpressed in ox-LDL-induced VSMCs and mouse models of AS. Functionally, silencing circ-ABCA1 via oligonucleotide transfection suppressed VSMC proliferation, migration, inflammation, and phenotypic switch in vitro and prevented AS development in mice in vivo. Mechanistically, circ-ABCA1 absorbed miR-885-5p, which targeted ROCK2. CONCLUSION: Taken together, the data from this study suggest that circ-ABCA1 mediates cellular inflammation and phenotype switching through the miR-885-5p/ROCK2 axis in ox-LDL-induced VSMCs, and the circ-ABCA1/miR-885-5p/ROCK2 axis is a new potential biomarker for the treatment of AS.


Assuntos
MicroRNAs , Músculo Liso Vascular , Humanos , Animais , Camundongos , Fenótipo , Inflamação , Lipoproteínas LDL/farmacologia , Miócitos de Músculo Liso , Oligonucleotídeos , MicroRNAs/genética , Proliferação de Células , Apoptose , Movimento Celular , Transportador 1 de Cassete de Ligação de ATP
3.
J Anim Sci ; 1022024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38477705

RESUMO

This study investigated the effects of feather meal (FM) processing methods on production parameters, blood biochemical indices, intestinal morphology, digestive and hepatic enzyme activities, and gastrointestinal tract pH and microflora of broilers. A total of 480-d-old male broilers were used for 42 d in a completely randomized design with eight treatments and five replicates (12 chicks/replicate). Treatments were 1) a control diet (without FM), 2) a diet containing 4% raw FM (RFM), 3) a diet containing 4% processed FM (PFM) by autoclave (Au-PFM), 4) a diet containing 4% fermented FM (FFM) by Bacillus licheniformis (Bl-FFM), 5) a diet containing 4% FFM by Bacillus subtilis (Bs-FFM), 6) a diet containing 4% FFM by Aspergillus niger (An-FFM), 7) a diet containing 4% FFM by B. licheniformis + B. subtilis + A. niger (Co-FFM), and 8) a diet containing 4% PFM by an enzyme (En-PFM). Results showed that in the FFMs the contents of ash, ether extract, total volatile nitrogen, and amino acids including Lys, Met, Thr, Trp, His, Leu, Gly, Ile, Phe, and Tyr increased (P < 0.05), while crude fiber, crude protein, and dry matter content decreased (P < 0.05). Compared with the control, the Co-FFM diet had no significant differences (P > 0.05) in total body weight gain (2,827 vs. 2,791 g/chick), total feed intake (5,018 vs. 4,991 g/chick), European production efficiency factor (375 vs. 377), European Broiler Index (371 vs. 371), and feed conversion ratio (1.77 vs. 1.78 g/g). Feeding FFM decreased (P < 0.05) serum total cholesterol (1.46-fold), triglyceride (1.61-fold), very low-density lipoprotein cholesterol (1.61-fold), and low-density lipoprotein cholesterol (2.27-fold) compared to the control. Also, FFM increased (P < 0.05) villus height (1,045 to 1,351, 661 to 854, and 523 to 620 µm), and villus height to crypt depth ratio (6.15 to 8.45, 4.55 to 7.04, and 4.27 to 5.45), in the duodenum, jejunum, and ileum, respectively, compared to the control. Compared to the control, the Co-FFM diet increased (P < 0.05) protease (34, 39, and 45 %) in the pancreas, duodenum, and jejunum, as well as amylase (73, and 97 %) activities in the duodenum, and jejunum, respectively. Diets containing FFM reduced (P < 0.05) pH in the crop, gizzard, and ileum, and decreased (P < 0.05) Escherichia coli (6.12 to 5.70) count in ileum compared to the control. The Co-FFM diet increased (P < 0.05) lactic acid bacteria count in crop (6.77 to 7.50) and ileum (6.94 to 7.73), also decreased (P < 0.05) coliforms (6.31 to 5.75) count in ileum compared to the control. In conclusion, FM fermentation, particularly Co-FFM, improves the nutritional value of FM, converting it into a decent source of dietary protein for broilers.


Fermentation represents an attractive alternative method for feather meal (FM) efficient bioconversion and its nutritional value enhancement. This study investigated the effects of FM processing methods on broilers. Experimental diets were 1) a control diet (without FM), 2) a diet containing 4% raw FM (RFM), 3) a diet containing 4% processed FM (PFM) by autoclave (Au-PFM), 4) a diet containing 4% fermented FM (FFM) by Bacillus licheniformis (Bl-FFM), 5) a diet containing 4% FFM by Bacillus subtilis (Bs-FFM), 6) a diet containing 4% FFM by Aspergillus niger (An-FFM), 7) a diet containing 4% FFM by B. licheniformis + B. subtilis + A. niger (Co-FFM), and 8) a diet containing 4% PFM by an enzyme (En-PFM). Results showed that FFMs increased the contents of ash, ether extract, total volatile nitrogen, and amino acids including Lys, Met, Thr, Trp, His, Leu, Gly, Ile, Phe, and Tyr, while decreased crude fiber, crude protein, and dry matter content. The production parameters of birds fed Co-FFM were similar to the control group. In addition, FFMs decreased serum total cholesterol (1.46-fold), triglyceride (1.61-fold), very low-density lipoprotein cholesterol (1.61-fold), and low-density lipoprotein cholesterol (2.27-fold). Furthermore, Co-FFM improved intestinal morphology, enzyme activities, and beneficial bacterial populations. In conclusion, Co-FFM, improves the nutritional value of FM, converting it into a decent source of dietary protein for broilers.


Assuntos
Galinhas , Plumas , Animais , Masculino , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Colesterol , Dieta/veterinária , Suplementos Nutricionais , Lipoproteínas LDL/farmacologia
4.
Lipids Health Dis ; 23(1): 76, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38468335

RESUMO

BACKGROUND: Atherosclerosis (AS) is a persistent inflammatory condition triggered and exacerbated by several factors including lipid accumulation, endothelial dysfunction and macrophages infiltration. Nobiletin (NOB) has been reported to alleviate atherosclerosis; however, the underlying mechanism remains incompletely understood. METHODS: This study involved comprehensive bioinformatic analysis, including multidatabase target prediction; GO and KEGG enrichment analyses for function and pathway exploration; DeepSite and AutoDock for drug binding site prediction; and CIBERSORT for immune cell involvement. In addition, target intervention was verified via cell scratch assays, oil red O staining, ELISA, flow cytometry, qRT‒PCR and Western blotting. In addition, by establishing a mouse model of AS, it was demonstrated that NOB attenuated lipid accumulation and the extent of atherosclerotic lesions. RESULTS: (1) Altogether, 141 potentially targetable genes were identified through which NOB could intervene in atherosclerosis. (2) Lipid and atherosclerosis, fluid shear stress and atherosclerosis may be the dominant pathways and potential mechanisms. (3) ALB, AKT1, CASP3 and 7 other genes were identified as the top 10 target genes. (4) Six genes, including PPARG, MMP9, SRC and 3 other genes, were related to the M0 fraction. (5) CD36 and PPARG were upregulated in atherosclerosis samples compared to the normal control. (6) By inhibiting lipid uptake in RAW264.7 cells, NOB prevents the formation of foam cell. (7) In RAW264.7 cells, the inhibitory effect of oxidized low-density lipoprotein on foam cells formation and lipid accumulation was closely associated with the PPARG signaling pathway. (8) In vivo validation showed that NOB significantly attenuated intra-arterial lipid accumulation and macrophage infiltration and reduced CD36 expression. CONCLUSIONS: Nobiletin alleviates atherosclerosis by inhibiting lipid uptake via the PPARG/CD36 pathway.


Assuntos
Aterosclerose , Flavonas , PPAR gama , Animais , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Macrófagos , Células Espumosas , Lipoproteínas LDL/farmacologia , Antígenos CD36/genética , Antígenos CD36/metabolismo
5.
Cell Signal ; 117: 111092, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38331013

RESUMO

SUMO-specific protease 3 (SENP3) participates in the removal of SUMOylation and maintains the balance of the SUMO system, which ensures normal functioning of substrates and cellular activities. In the present study, we found that SENP3 expression was significantly reduced in ox-LDL-stimulated macrophages. SENP3 overexpression suppressed and SENP3 knockdown promoted macrophage foam cell formation. Moreover, SENP3 inhibited cholesterol uptake, CD36 expression, and NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome activation in ox-LDL-stimulated macrophages. Ox-LDL-stimulated NLRP3 SUMOylation was reduced by SENP3. Blocking NLRP3 SUMOylation inhibited foam cell formation and NLRP3 inflammasome activation. Thus, this study revealed that SENP3 inhibits macrophage foam cell formation by deSUMOylating NLRP3 and regulating NLRP3 inflammasome activation, which may provide a potentially innovative approach to treatment of atherosclerosis.


Assuntos
Células Espumosas , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Células Espumosas/metabolismo , Inflamassomos/metabolismo , Peptídeo Hidrolases/metabolismo , Macrófagos/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Endopeptidases/metabolismo
6.
Hypertension ; 81(4): 861-875, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38361240

RESUMO

BACKGROUND: Chemerin, an inflammatory adipokine, is upregulated in preeclampsia, and its placental overexpression results in preeclampsia-like symptoms in mice. Statins may lower chemerin. METHODS: Chemerin was determined in a prospective cohort study in women suspected of preeclampsia and evaluated as a predictor versus the sFlt-1 (soluble fms-like tyrosine kinase-1)/PlGF (placental growth factor) ratio. Chemerin release was studied in perfused placentas and placental explants with or without the statins pravastatin and fluvastatin. We also addressed statin placental passage and the effects of chemerin in chorionic plate arteries. RESULTS: Serum chemerin was elevated in women with preeclampsia, and its addition to a predictive model yielded significant effects on top of the sFlt-1/PlGF ratio to predict preeclampsia and its fetal complications. Perfused placentas and explants of preeclamptic women released more chemerin and sFlt-1 and less PlGF than those of healthy pregnant women. Statins reversed this. Both statins entered the fetal compartment, and the fetal/maternal concentration ratio of pravastatin was twice that of fluvastatin. Chemerin constricted plate arteries, and this was blocked by a chemerin receptor antagonist and pravastatin. Chemerin did not potentiate endothelin-1 in chorionic plate arteries. In explants, statins upregulated low-density lipoprotein receptor expression, which relies on the same transcription factor as chemerin, and NO release. CONCLUSIONS: Chemerin is a biomarker for preeclampsia, and statins both prevent its placental upregulation and effects, in an NO and low-density lipoprotein receptor-dependent manner. Combined with their capacity to improve the sFlt-1/PlGF ratio, this offers an attractive mechanism by which statins may prevent or treat preeclampsia.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Pré-Eclâmpsia , Humanos , Gravidez , Feminino , Animais , Camundongos , Placenta/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Fator de Crescimento Placentário , Pravastatina/farmacologia , Regulação para Cima , Estudos Prospectivos , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/prevenção & controle , Fluvastatina/metabolismo , Fluvastatina/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Biomarcadores , Quimiocinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo
7.
Cell Signal ; 117: 111114, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38387686

RESUMO

Obesity has long been thought to be a main cause of hyperlipidemia. As a systemic disease, the impact of obesity on organs, tissues and cells is almost entirely negative. However, the relationship between obesity and bone loss is highly controversial. On the one hand, obesity has long been thought to have a positive effect on bone due to increased mechanical loading on the skeleton, conducive to increasing bone mass to accommodate the extra weight. On the other hand, obesity-related metabolic oxidative modification of low-density lipoprotein (LDL) in vivo causes a gradual increase of oxidized LDL (ox-LDL) in the bone marrow microenvironment. We have reported that low-density lipoprotein receptor-related protein 6 (LRP6) acts as a receptor of ox-LDL and mediates the bone marrow stromal cells (BMSCs) uptake of ox-LDL. We detected elevated serum ox-LDL in obese mice. We found that ox-LDL uptake by LRP6 led to an increase of intracellular reactive oxygen species (ROS) in BMSCs, and N-acetyl-L-cysteine (NAC) alleviated the cellular senescence and impairment of osteogenesis induced by ox-LDL. Moreover, LRP6 is a co-receptor of Wnt signaling. We found that LRP6 preferentially binds to ox-LDL rather than dickkopf-related protein 1 (DKK1), both inhibiting Wnt signaling and promoting BMSCs senescence. Mesoderm development LRP chaperone (MESD) overexpression inhibits ox-LDL binding to LRP6, attenuating oxidative stress and BMSCs senescence, eventually rescuing bone phenotype.


Assuntos
Medula Óssea , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Animais , Camundongos , Medula Óssea/metabolismo , Proteínas de Transporte/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Obesidade/complicações , Estresse Oxidativo
8.
Mol Biol Rep ; 51(1): 365, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409611

RESUMO

A low-frequency variant of sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SVEP1) is associated with the risk of coronary artery disease, as determined by a genome-wide association study. SVEP1 induces vascular smooth muscle cell proliferation and an inflammatory phenotype to promote atherosclerosis. In the present study, qRT‒PCR demonstrated that the mRNA expression of SVEP1 was significantly increased in atherosclerotic plaques compared to normal tissues. Bioinformatics revealed that EGR1 was a transcription factor for SVEP1. The results of the luciferase reporter assay, siRNA interference or overexpression assay, mutational analysis and ChIP confirmed that EGR1 positively regulated the transcriptional activity of SVEP1 by directly binding to its promoter. EGR1 promoted human coronary artery smooth muscle cell (HCASMC) proliferation and migration via SVEP1 in response to oxidized low-density lipoprotein (ox-LDL) treatment. Moreover, the expression level of EGR1 was increased in atherosclerotic plaques and showed a strong linear correlation with the expression of SVEP1. Our findings indicated that EGR1 binding to the promoter region drive SVEP1 transcription to promote HCASMC proliferation and migration.


Assuntos
MicroRNAs , Placa Aterosclerótica , Humanos , Placa Aterosclerótica/metabolismo , Vasos Coronários/metabolismo , Estudo de Associação Genômica Ampla , Movimento Celular , Lipoproteínas LDL/farmacologia , Células Cultivadas , Proliferação de Células/genética , Miócitos de Músculo Liso/metabolismo , MicroRNAs/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Moléculas de Adesão Celular/genética
9.
Discov Med ; 36(181): 343-354, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38409839

RESUMO

BACKGROUND: Oxidative stress, propelled by reactive oxygen species (ROS), serves as a significant catalyst for atherosclerosis (AS), a primary contributor to vascular diseases on a global scale. Antioxidant therapy via nanomedicine has emerged as a pivotal approach in AS treatment. Nonetheless, challenges such as inadequate targeting, subpar biocompatibility, and limited antioxidant effectiveness have restrained the widespread utilization of nanomedicines in AS treatment. This study aimed to synthesize a specialized peptide-modified liposome capable of encapsulating two antioxidant enzymes, intending to enhance targeted antioxidant therapy for AS. METHODS: The film dispersion method was employed for liposome preparation. Fluorescence quantification was conducted to assess the drug encapsulation rate. Characterization of liposome particle size was performed using dynamic light scattering (DLS) and transmission electron microscopy (TEM). Laser confocal microscopy and flow cytometry were utilized to analyze liposome cell uptake and target foam cells. Antioxidant analysis was conducted using 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) staining, while pro-lipid efflux analysis utilized Oil Red O (ORO) staining. Safety evaluation was performed using Hematoxylin and Eosin (H&E) staining. The level of inflammatory factors was determined through enzyme-linked immunosorbent assay (ELISA). The degree of lipid oxidation at the cellular level was assessed using the malonaldehyde (MDA) assay. In vivo targeting analysis was conducted using small animal live imaging. RESULTS: Our in vitro and in vivo findings substantiated that the modification of Lyp-1 led to increased delivery of antioxidant enzymes into foam cells (p < 0.05), the primary pathological cells within AS plaques. Upon accumulation in foam cells, liposomes loaded with superoxide dismutase (SOD) and catalase (CAT) (LyP-lip@SOD/CAT) effectively mitigated excess ROS and shielded macrophages from ROS-induced damage (p < 0.01). Furthermore, the reduction in ROS levels notably hindered the endocytosis of oxidized low-density lipoprotein (Ox-LDL) by activated macrophages, subsequently alleviating lipid accumulation at atherosclerotic lesion sites, evident from both in vitro and in vivo ORO staining results (p < 0.01). LyP-lip@SOD/CAT significantly curbed the secretion of inflammatory factors at the plaque site (p < 0.001). Additionally, LyP-lip@SOD/CAT demonstrated commendable biological safety. CONCLUSIONS: In this study, we effectively synthesized LyP-lip@SOD/CAT and established its efficacy as a straightforward and promising nano-agent for antioxidant therapy targeting atherosclerosis.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Células Espumosas , Lipossomos/farmacologia , Superóxido Dismutase/farmacologia , Catalase/farmacologia , Antioxidantes/uso terapêutico , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio , Estresse Oxidativo , Aterosclerose/tratamento farmacológico , Lipoproteínas LDL/farmacologia
10.
Biomed Pharmacother ; 172: 116268, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38359489

RESUMO

Atherosclerosis is a lipid-driven inflammatory arterial disease, with one crucial factor is oxidized low-density lipoprotein (ox-LDL), which can induce endothelial dysfunction through endoplasmic reticulum stress (ERS). Interleukin-37 (IL-37) exerts vascular protective functions. This study aims to investigates whether IL-37 can alleviate ERS and autophagy induced by ox-LDL, therely potentialy treating atherosclerosis. We found that ox-LDL enhances the wound healing rate in Rat Coronary Artery Endothelial Cells (RCAECs) and IL-37 reduce the ox-LDL-induced pro-osteogenic response, ERS, and autophagy by binding to Smad3. In RCAECs treated with ox-LDL and recombinant human IL-37, the wound healing rate was mitigated. The expression of osteogenic transcription factors and proteins involved in the ERS pathway was reduced in the group pretreated with IL-37 and ox-LDL. However, these responses were not alleviated when Smads silenced. Electron microscopy revealed that the IL-37/Smad3 complex could suppress endoplasmic reticulum autophagy under ox-LDL stimulation. Thus, IL-37 might treat atherosclerosis through its multi-protective effect by binding Smad3.


Assuntos
Aterosclerose , Células Endoteliais , Interleucina-1 , Animais , Humanos , Ratos , Aterosclerose/tratamento farmacológico , Aterosclerose/prevenção & controle , Aterosclerose/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Fatores de Transcrição/metabolismo , Interleucina-1/uso terapêutico
11.
Mol Cell Endocrinol ; 584: 112161, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38280475

RESUMO

BACKGROUND: Atherosclerosis (AS) is commonly regarded as a key driver accounted for the leading causes of morbidity and mortality among cardiovascular and cerebrovascular diseases. A growing body of evidence indicates that autophagy in macrophages involved in AS might be a potential therapeutic target. C1q/TNF-related protein 9 (CTRP9) has been proven to delay the progression of cardiovascular diseases. However, the relations between CTRP9 and Sirt1, as well as their effects on macrophages autophagy have not been fully explored. METHODS: Macrophages were differentiated from mononuclear cells collected from peripheral blood samples of healthy donors. The in vitro AS models were constructed by ox-LDL treatment. Cell viability was determined by CCK-8 assay. Immunofluorescence assay of LC3 was implemented for evaluating autophagy activity. Oil Red O staining was performed for lipid accumulation detection. ELISA, cholesterol concentration assay and cholesterol efflux analysis were conducted using commercial kits. Cycloheximide assay was implemented for revealing protein stability. RT-qPCR was used for mRNA expression detection, and western blotting was performed for protein level monitoring. RESULTS: CTRP9 attenuated impaired cell viability, autophagy inhibition and increased lipid accumulation induced by ox-LDL. Moreover, CTRP9 maintained Sirt1 protein level through enhancing its stability through de-ubiquitination, which was mediated by upregulated USP22 level. CRTP9 exerted its protective role in promoting autophagy and reducing lipid accumulation through the USP22/Sirt1 axis. CONCLUSION: Collectively, CTRP9 alleviates lipid accumulation and facilitated the macrophages autophagy by upregulating USP22 level and maintaining Sirt1 protein expression, thereby exerting a protective role in AS progression in vitro.


Assuntos
Aterosclerose , Sirtuína 1 , Humanos , Sirtuína 1/genética , Sirtuína 1/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C1q/farmacologia , Macrófagos/metabolismo , Lipoproteínas LDL/farmacologia , Colesterol/metabolismo , Aterosclerose/metabolismo , Autofagia , Ubiquitinação
12.
Int Heart J ; 65(1): 135-145, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38296567

RESUMO

Atherosclerosis may be caused or developed by an immune response and antioxidation imbalance. MicroRNA-375 (miR-375) or G-protein-coupled receptor 39 (GPR39) is involved in vascular endothelial cell injury, but their role in atherosclerosis is unknown. This experiment aimed to determine the action of the miR-375/GPR39 axis in atherosclerosis.Human aortic endothelial cells (HAECs) were treated with 10 ng/mL of oxidised low-density lipoprotein (ox-LDL) for 24 hours to induce HAEC injury, which was treated by the miR-375 inhibitor, GPR39 inhibitor, or agonist. High-fat diet (HFD) -induced ApoE-/- mice were made as an atherosclerosis model for miR-375 inhibitor treatment. Cell Counting Kit-8 was applied to detect HAEC viability. HAEC apoptosis and ROS levels were measured using flow cytometry. Vascular histopathology and the GPR39 expression were detected using hematoxylin-eosin and immunohistochemistry. The expressions of interleukin (IL) -6, IL-1ß, and tumour necrosis factor-α (TNF-α) were assessed using an enzyme-linked immunosorbent assay. The miR-375, GPR39, NOX-4, and p-IκBα/IκBα levels were measured using quantitative reverse transcription polymerase chain reaction or western blot.MiR-375 and GPR39 levels increased and decreased in ox-LDL-treated HAECs, respectively. The miR-375 inhibitor or GPR39 agonist promoted cell viability and inhibited apoptosis in ox-LDL-induced HAEC injury. The miR-375 inhibitor also significantly downregulated the IL-6, IL-1ß, TNF-α, p-IκBα/IκBα, ROS, and NOX-4 expressions to alleviate oxidative stress and inflammation, which were reversed by the GPR39 inhibitor. An in vivo experiment proved that the miR-375 inhibitor upregulated the GPR39 expression and improved inflammation, oxidative stress, and endothelial cell damage associated with atherosclerosis.The miR-375 inhibitor improved inflammation, oxidative stress, and cell damage in ox-LDL-induced HAECs and HFD-induced ApoE-/- mice by promoting the GPR39 expression, which provided a new theoretical basis for the clinical treatment of atherosclerosis.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Animais , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Células Endoteliais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Aterosclerose/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Estresse Oxidativo , Inflamação/metabolismo , Apolipoproteínas E , Apoptose
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(1): 52-59, 2024 Jan 20.
Artigo em Chinês | MEDLINE | ID: mdl-38293976

RESUMO

OBJECTIVE: To investigate the effects of galangin on angiogenic activity of oxidized low-density lipoprotein (ox-LDL)-induced human aortic endothelial cells (HAECs) and explore the underlying mechanisms. METHODS: HAECs incubated with 10, 20, 40, and 80 µmol/L galangin for 24 h were assessed for cell viability changes using MTT assay to determine the cytotoxicity of galangin. HAECs treated with 5 mg/mL ox-LDL and incubated with 20 and 40 µmol/L galangin for 24 h, and the cells overexpressing lncRNA H19 and incubated with 40 µmol/L galangin for 24 h were examined for lncRNA H19 level with qRT-PCR. The migration and tube formation capacity of the cells were observed using scratch assay and angiogenesis assay, and ROS levels in the cells were detected with flow cytometry. The protein expression levels of VEGFA, MMP-2 and MMP-9 in the treated cells were detected with Western blotting. RESULTS: Galangin at 10, 20, or 40 µmol/L produced no obvious toxicity (P>0.05), whereas 80 µmol/L galangin significantly inhibited the viability of HAECs (P<0.01). Treatment with ox-LDL significantly increased the expression of lncRNA H19 in HAECs. Galangin significantly lowered lncRNA H19 expression in ox-LDL-induced HAECs, suppressed cell migration, angiogenesis and ROS production level, and reduced the protein levels of VEGFA, MMP-2 and MMP-9 (P<0.01). The effects of galangin were blocked by overexpression of lncRNA H19 in the cardiomyocytes. CONCLUSION: The therapeutic effect of galangin for atherosclerosis is mediated by inhibiting lncRNA H19 expression to reduce ox-LDL-induced migration, oxidative stress, and angiogenesis of HAECs.


Assuntos
Flavonoides , MicroRNAs , RNA Longo não Codificante , Humanos , Células Endoteliais , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Lipoproteínas LDL/farmacologia , Apoptose
14.
Med Sci Monit ; 30: e942733, 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38273650

RESUMO

BACKGROUND C1q/tumor necrosis factor-related protein 13 (CTRP13) preserves endothelial function and possesses anti-oxidation activity. However, its effects on ferroptosis of human umbilical vein endothelial cells (HUVECs) remain unclear. We investigated the effects of CTRP13 on HUVEC ferroptosis induced by oxidized low-density lipoprotein (ox-LDL) and explored the underlying mechanisms of CTRP13 against ferroptosis via the AMPK/KLF4 pathway. MATERIAL AND METHODS Cell Counting Kit-8 assay was used to evaluate cell viability. Lactate dehydrogenase activity and malondialdehyde content analysis were performed to evaluate the cell membrane integrity and lipid peroxidation. Mito-Tracker, JC-1, and 2',7'-dichlorofluorescein di-acetate were used to evaluate the biological activity of mitochondria, mitochondrial membrane potential, and reactive oxygen species (ROS) in endothelial cells. The ferroptosis indicator expressions, recombinant solute carrier family 7, member 11, glutathione peroxidase 4 (GPX4), and acyl-CoA synthetase long-chain family member 4 were examined using real-time reverse transcription-polymerase chain reaction and Western blot. Immunofluorescence staining detected GPX4 location in endothelial cells. RESULTS The results demonstrate that CTRP13 (450 ng/mL) prevented HUVEC ferroptosis by inhibiting ROS overproduction and mitochondrial dysfunction, and CTRP13 accelerated antioxidant enzyme expression levels, such as heme oxygenase 1, superoxide dismutase 1, and superoxide dismutase 2, compared with the ox-LDL (100 µg/mL) group for 48 h. Additionally, CTRP13 treatment increased p-AMPK/AMPK expression by 47.65% (P<0.05) while decreasing Krüppel-like factor 4 expression by 37.43% (P<0.05) in ox-LDL-induced HUVECs and elucidated the protective effect on endothelial dysfunction from ferroptosis. CONCLUSIONS These findings provide new insights for understanding the effects and mechanism of CTRP13 on preventing endothelial cell ferroptosis.


Assuntos
Aterosclerose , Ferroptose , Humanos , Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Aterosclerose/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Espécies Reativas de Oxigênio/metabolismo
15.
J Mol Histol ; 55(1): 109-120, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38165567

RESUMO

Endothelial cells are a crucial component of the vessel-tissue wall and exert an important role in atherosclerosis (AS). To explore the role of Orientin in AS, human vascular endothelial cells (HUVECs) were induced by oxidized low-density lipoprotein (ox-LDL) to simulate the vascular endothelial injury during AS. Cell viability was detected by CCK-8 assay. Oxidative stress and inflammation related markers were measured using kits, RT-qPCR or western blot. Besides, cell apoptosis was assessed with TUNEL staining and cell autophagy was evaluated by LC3 immunofluorescent staining. Additionally, western blot was utilized to evaluate the expression of Sestrin 1 (SESN1) and proteins in AMPK/mTOR signaling. Afterwards, SESN1 was silenced to determine the expression of autophagy-related proteins. The further application of autophagy inhibitor 3-methyladenine (3-MA) was used to clarify the regulatory mechanism of Orientin on autophagy. Results showed that the decreased viability of HUVECs caused by ox-LDL induction was elevated by Orientin. Oxidative stress and inflammation were also attenuated after Orientin addition in HUVECs under ox-LDL condition. Moreover, Orientin suppressed apoptosis and induced autophagy of HUVECs stimulated by ox-LDL, accompanied by enhanced level of phospho (p)-AMPK and declined level of p-mTOR. Interestingly, SESN1 level was elevated by Orientin, and SESN1 depletion alleviated autophagy and reduced p-AMPK expression but enhanced p-mTOR expression. The further experiments indicated that SESN1 silencing or 3-MA addition reversed the inhibitory effects of Orientin on the oxidative stress, inflammation and apoptosis of HUVECs. Collectively, Orientin could induce autophagy by activating SESN1 expression, thereby regulating AMPK/mTOR signaling in ox-LDL-induced HUVECs.


Assuntos
Proteínas Quinases Ativadas por AMP , Flavonoides , Glucosídeos , Sestrinas , Humanos , Sestrinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Proteínas Quinases Ativadas por AMP/metabolismo , Estresse Oxidativo , Lipoproteínas LDL/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Apoptose , Autofagia , Inflamação/metabolismo
16.
Cell Mol Life Sci ; 81(1): 62, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38280036

RESUMO

Endothelial injury and dysfunction in the artery wall fuel the process of atherosclerosis. As a key epigenetic regulator, Ash2l (Absent, small, or homeotic-Like 2) is involved in regulating vascular injury and its complications. However, the role of Ash2l in atherosclerosis has not yet been fully elucidated. Here, we found increased Ash2l expression in high-cholesterol diet-fed ApoE-/- mice and oxidized LDL (oxLDL) treated endothelial cells (ECs). Furthermore, Ash2l promoted the scavenger receptors transcription by catalyzing histone H3 lysine 4 (H3K4) trimethylation at the promoter region of transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) and triggered the activation of the pro-inflammatory nuclear factor-kappa B (NF-κB) by enhancing interaction between CD36 and toll-like receptor 4 (TLR4). Meanwhile, enhanced expression of scavenger receptors drove more oxLDL uptake by ECs. In vivo studies revealed that ECs-specific Ash2l knockdown reduced atherosclerotic lesion formation and promoted fibrous cap stability in the aorta of ApoE-/- mice, which was partly associated with a reduced endothelial activation by suppressing scavenger receptors and the uptake of lipids by ECs. Collectively, our findings identify Ash2l as a novel regulator that mediates endothelial injury and atherosclerosis. Targeting Ash2l may provide valuable insights for developing novel therapeutic candidates for atherosclerosis.


Assuntos
Aterosclerose , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Aterosclerose/metabolismo , NF-kappa B/metabolismo , Receptores Depuradores/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo
17.
Mol Biotechnol ; 66(1): 112-122, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37043109

RESUMO

Kruppel-like factor 15 (KLF15) is involved in many cardiovascular diseases and is abnormally expressed in atherosclerosis (AS), but the regulatory mechanism of KLF15 in AS has not been reported so far. RT-qPCR was used to detect the expression of KLF15 and ATG14 in AS patients. Subsequently, human aortic endothelial cells (HAECs) were induced by oxidized low densitylipoprotein (ox-LDL), and the expression of KLF15 in model cells was detected. KLF15 was overexpressed in cells by lipofection transfection, and then CCK8, flow cytometry, Western blot, ELISA, and related assay kits were used to detect cell viability, apoptosis, inflammatory response as well as oxidative stress, respectively. The targeted regulatory relationship between KLF15 and autophagy-related 14 (ATG14) was detected by ChIP and luciferase reporter assays. Following ATG14 silencing in KLF15-overexpressing cells, immunofluorescence and Western blot were used to detect the autophagy. Finally, after the addition of 3-Methyladenine (3-MA), an autophagy inhibitor, the aforementioned experiments were conducted again to further explore the mechanism. The expression of KLF15 and ATG14 were decreased in AS patients and ox-LDL-induced HAECs. Overexpression of KLF15 protected ox-LDL-induced HAECs from damage, which might be achieved through transcriptional regulation of ATG14. In addition, KLF15 could promote autophagy through transcriptional activation of ATG14. KLF15 transcriptionally activated ATG14 to promote autophagy and attenuate damage of ox-LDL-induced HAECs.


Assuntos
Células Endoteliais , MicroRNAs , Humanos , Células Endoteliais/metabolismo , Transdução de Sinais , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Autofagia/genética , Apoptose , MicroRNAs/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo
18.
Inflammation ; 47(1): 421-437, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37898994

RESUMO

Despite the substantial progress in deciphering the pathogenesis of atherosclerosis (AS), cardiovascular mortality is still increasing. Therefore, atherosclerotic cardiovascular disease remains a sweeping epidemic that jeopardizes human health. Disentangling the molecular underpinnings of AS is imperative in the molecular cardiology field. Overwhelming evidence has indicated that the recognition of a fascinating class of players, known as long non-coding RNAs (lncRNAs), provides causality for coordinating AS. However, the function and mechanism of HOTAIRM1 are still poorly understood in human umbilical vein endothelial cells (HUVECs) and AS. Herein, we primarily underscored that lncRNA HOTAIRM1 is potentially responsible for AS; as such, it was dramatically up-regulated in HUVECs upon ox-LDL stimulation. Functionally, HOTAIRM1 knockdown attenuated HUVEC proliferation and potentiated apoptosis in the absence and presence of ox-LDL. Furthermore, HOTAIRM1 was preferentially located in the nuclei of HUVECs. Mechanistically, HOXA4 is directly bound to the HOTAIRM1 promoter and activated its transcription. Of note, a positive feedback signaling between HOXA4 and HOTAIRM1 was determined. Intriguingly, the interplay between HOTAIRM1 and HSPA5 occurred in an RNA-binding protein pattern and a transcription-dependent regulatory manner. In addition, HSPA5 overexpression partially antagonized HUVEC proliferation inhibition of HOTAIRM1 depletion. Taken together, our findings delineate a pivotal functional interaction among HOXA4, HOTAIRM1, and HSPA5 as a novel regulatory circuit for modulating HUVEC proliferation. An in-depth investigation of the HOXA4-HOTAIRM1-HSPA5 axis promises to yield significant breakthroughs in identifying the molecular mechanisms governing AS and developing therapeutic avenues for AS.


Assuntos
MicroRNAs , RNA Longo não Codificante , Humanos , Apoptose , Proliferação de Células , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lipoproteínas LDL/farmacologia , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo
19.
Pflugers Arch ; 476(1): 59-74, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37910205

RESUMO

The formation of foam cells, lipid-loaded macrophages, is the hallmark event of atherosclerosis. Since cigarette smoking is a risk factor for developing atherosclerosis, the current study investigated the effects of cigarette smoke extract (CSE) on different events like expressions of genes involved in lipid influx and efflux, lipophagy, etc., that play vital roles in foam cell formation. The accumulation of lipids after CSE treatment U937 macrophage cells was examined by staining lipids with specific dyes: Oil red O and BODIPY493/503. Results showed an accumulation of lipids in CSE-treated cells, confirming foam cell formation by CSE treatment. To decipher the mechanism, the levels of CD36, an ox-LDL receptor, and ABCA1, an exporter of lipids, were examined in CSE-treated and -untreated U937 cells by real-time PCR and immunofluorescence analysis. Consistent with lipid accumulation, an increased level of CD36 and a reduction in ABCA1 were observed in CSE-treated cells. Moreover, CSE treatment caused inhibition of lipophagy-mediated lipid degradation by blocking lipid droplets (LDs)-lysosome fusion and increasing the lysosomal pH. CSE also impaired mitochondrial lipid oxidation. Thus, the present study demonstrates that CSE treatment affects lipid homeostasis by altering its influx and efflux, lysosomal degradation, and mitochondrial utilization, leading to the formation of lipid-loaded foam cells. Moreover, the current study also showed that the leucine supplement caused a significant reduction of CSE-induced foam cell formation in vitro. Thus, the current study provides insight into CS-induced atherosclerosis and an agent to combat the disease.


Assuntos
Aterosclerose , Fumar Cigarros , Humanos , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Gotículas Lipídicas/metabolismo , Células U937 , Aterosclerose/metabolismo
20.
F S Sci ; 5(1): 80-91, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38043603

RESUMO

OBJECTIVES: To assess the effect of simvastatin on uterine leiomyoma growth and extracellular matrix (ECM) deposition. DESIGN: Laboratory analysis of human leiomyoma cell culture, xenograft in a mouse model, and patient tissue from a clinical trial. SETTING: Academic research center. PATIENT(S): Tissue culture from human leiomyoma tissue and surgical leiomyoma tissue sections from a placebo-controlled randomized clinical trial. INTERVENTION(S): Simvastatin treatment. MAIN OUTCOME MEASURE(S): Serum concentrations, xenograft volumes, and protein expression. RESULTS: Mice xenografted with 3-dimensional human leiomyoma cultures were divided as follows: 7 untreated controls; 12 treated with activated simvastatin at 10 mg/kg body weight; and 15 at 20 mg/kg body weight. Simvastatin was detected in the serum of mice injected at the highest dose. Xenograft volumes were significantly smaller (mean 53% smaller at the highest concentration). There was dissolution of compact ECM, decreased ECM formation, and lower collagen protein expression in xenografts. Membrane type 1 matrix metalloproteinase was increased in vitro and in vivo. Matrix metalloproteinase 2 and low-density lipoprotein receptor-related protein 1 were increased in vitro. CONCLUSIONS: Simvastatin exhibited antitumoral activity with ECM degradation and decreased leiomyoma tumor volume in vivo. Activation of the matrix metalloproteinase 2, membrane type 1 matrix metalloproteinase, and low-density lipoprotein receptor-related protein 1 pathway may explain these findings.


Assuntos
Leiomioma , Neoplasias Uterinas , Feminino , Humanos , Camundongos , Animais , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/farmacologia , Sinvastatina/farmacologia , Sinvastatina/metabolismo , Sinvastatina/uso terapêutico , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 14 da Matriz/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , Leiomioma/tratamento farmacológico , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Peso Corporal , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...