Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Int Heart J ; 65(4): 738-747, 2024.
Article in English | MEDLINE | ID: mdl-39085113

ABSTRACT

The regulatory factor X7 (RFX7) is a vital mediator in atherosclerosis. This study aims to discuss the effect and underlying mechanism of RFX7 on the regulation of oxidized low-density lipoprotein (ox-LDL) -induced proliferation and migration of vascular smooth muscle cells (VSMCs).Ox-LDL was used to construct atherosclerosis in vitro model. The mRNA and protein levels of RFX7 and Sirtuin 4 (SIRT4) were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assays. The cellular functions were measured via 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), EdU, flow cytometry, and wound healing assay assays. The interaction between RFX7 and SIRT4 promoter was validated using chromatin immunoprecipitation and dual-luciferase reporter assays.The stimulation with ox-LDL elevated the viability of VSMCs and decreased the mRNA and protein levels of RFX7 and SIRT4 in VSMCs in a dose-dependent manner. Functionally, RFX7 overexpression restrained the VSMC viability, proliferation, and migration induced by ox-LDL, but facilitated VSMC apoptosis. RFX7 elevated SIRT4 expression via binding to its promoter. Furthermore, overexpressing either SIRT4 or RFX7 inactivated JAK2/STAT3 signaling, causing a decrease in VSMC proliferation and migration and an increase in VSMC apoptosis when exposed to ox-LDL. The impact of RFX7 overexpression on JAK2/STAT3 signaling and cellular function following ox-LDL exposure was abrogated by SIRT4 silencing.The heightened RFX7 expression restrained the proliferation and migration of ox-LDL-stimulated VSMCs via SIRT4-mediated inactivation of JAK2/STAT3 pathway.


Subject(s)
Cell Movement , Cell Proliferation , Janus Kinase 2 , Lipoproteins, LDL , Muscle, Smooth, Vascular , STAT3 Transcription Factor , Signal Transduction , Sirtuins , STAT3 Transcription Factor/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Janus Kinase 2/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Humans , Sirtuins/metabolism , Sirtuins/genetics , Atherosclerosis/metabolism , Cells, Cultured , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Mitochondrial Proteins
2.
Mol Med Rep ; 30(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38873985

ABSTRACT

Macrophage pyroptosis mediates vascular inflammation and atherosclerosis (AS). Hydrogen sulfide (H2S) exerts a protective role in preventing inflammation and AS. However, its molecular mechanisms of regulating the pyroptosis signaling pathway and inhibiting macrophage pyroptosis remain unexplored. The present study aimed to determine whether H2S mitigates macrophage pyroptosis by downregulating the pyroptosis signaling pathway and S­sulfhydrating caspase­1 under the stimulation of oxidized low­density lipoprotein (ox­LDL), a pro­atherosclerotic factor. Macrophages derived from THP­1 monocytes were pre­treated using exogenous H2S donors sodium hydrosulfide (NaHS) and D,L­propargylglycine (PAG), a pharmacological inhibitor of endogenous H2S­producing enzymes, alone or in combination. Subsequently, cells were stimulated with ox­LDL or the desulfhydration reagent dithiothreitol (DTT) in the presence or absence of NaHS and/or PAG. Following treatment, the levels of H2S in THP­1 derived macrophages were measured by a methylene blue colorimetric assay. The pyroptotic phenotype of THP­1 cells was observed and evaluated by light microscopy, Hoechst 33342/propidium iodide fluorescent staining and lactate dehydrogenase (LDH) release assay. Caspase­1 activity in THP­1 cells was assayed by caspase­1 activity assay kit. Immunofluorescence staining was used to assess the accumulation of active caspase­1. Western blotting and ELISA were performed to determine the expression of pyroptosis­specific markers (NLRP3, pro­caspase­1, caspase­1, GSDMD and GSDMD­N) in cells and the secretion of pyroptosis­related cytokines [interleukin (IL)­1ß and IL­18] in the cell­free media, respectively. The S­sulfhydration of pro­caspase­1 in cells was assessed using a biotin switch assay. ox­LDL significantly induced macrophage pyroptosis by activating the pyroptosis signaling pathway. Inhibition of endogenous H2S synthesis by PAG augmented the pro­pyroptotic effects of ox­LDL. Conversely, exogenous H2S (NaHS) ameliorated ox­LDL­and ox­LDL + PAG­induced macrophage pyroptosis by suppressing the activation of the pyroptosis signaling pathway. Mechanistically, ox­LDL and the DTT increased caspase­1 activity and downstream events (IL­1ß and IL­18 secretion) of the caspase­1­dependent pyroptosis pathway by reducing S­sulfhydration of pro­caspase­1. Conversely, NaHS increased S­sulfhydration of pro­caspase­1, reducing caspase­1 activity and caspase­1­dependent macrophage pyroptosis. The present study demonstrated the molecular mechanism by which H2S ameliorates macrophage pyroptosis by suppressing the pyroptosis signaling pathway and S­sulfhydration of pro­caspase­1, thereby suppressing the generation of active caspase-1 and activity of caspase-1.


Subject(s)
Caspase 1 , Hydrogen Sulfide , Lipoproteins, LDL , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Pyroptosis , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Pyroptosis/drug effects , Humans , Caspase 1/metabolism , Macrophages/metabolism , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Phosphate-Binding Proteins/metabolism , THP-1 Cells , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction/drug effects , Gasdermins , Alkynes , Glycine/analogs & derivatives , Sulfides
3.
J Mol Histol ; 55(4): 589-598, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38890233

ABSTRACT

OBJECTIVE: This study was conducted to characterize the antioxidant and anti-inflammatory properties of Rubber Seed Oil (RSO) against atherosclerosis (AS) through the study of the protective effects and mechanisms on human umbilical vein endothelial cells (HUVECs) injury induced by oxidized low-density lipoprotein (ox-LDL). METHODS: HUVECs were treated with RSO, ox-LDL, RSO + ox-LDL, respectively, followed by cell activity testing, levels of IL-1ß, IL-6, IL-10, TNF-α, ROS, NO, the mRNA expression of eNOS and protein expression of MCP-1, VCAM-1, eNOS, TLR4, NF-κB p65、p-NF-κB p65. RESULTS: Compared with the ox-LDL group, cell viability, NO level and the expression of eNOS mRNA significantly increased. and the levels of pro-inflammatory factors such as IL-1ß, IL-6, TNF-α, IL-10, ROS were significantly decreased, which was accompanied by decreases in TLR4 mRNA, TLR4, MCP-1, VCAM-1 protein expression, as well as the ratio of NF-κB p-p65/p65 in the group treated with 250 µg/ml ox-LDL + 50 µg/ml RSO, 250 µg/ml ox-LDL + 100 µg/ml RSO, 250 µg/ml ox-LDL + 150 µg/ml RSO. CONCLUSIONS: RSO can reduce the expression of pro-inflammatory mediators, oxidative factors involved in injured vascular endothelial cells, exhibiting anti-inflammatory and antioxidant properties HUVECs exposed to ox-LDL. In addition, it may alleviate endothelial cell damage by inhibiting the TLR4/NF-κB signaling pathway.


Subject(s)
Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , Plant Oils/pharmacology , Cell Survival/drug effects , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism , Cytokines/metabolism , Nitric Oxide/metabolism , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Anti-Inflammatory Agents/pharmacology , Seeds/chemistry , Signal Transduction/drug effects
4.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928513

ABSTRACT

Arterial macrophage cholesterol accumulation and impaired cholesterol efflux lead to foam cell formation and the development of atherosclerosis. Modified lipoproteins interact with toll-like receptors (TLR), causing an increased inflammatory response and altered cholesterol homeostasis. We aimed to determine the effects of TLR antagonists on cholesterol efflux and foam cell formation in human macrophages. Stimulated monocytes were treated with TLR antagonists (MIP2), and the cholesterol efflux transporter expression and foam cell formation were analyzed. The administration of MIP2 attenuated the foam cell formation induced by lipopolysaccharides (LPS) and oxidized low-density lipoproteins (ox-LDL) in stimulated THP-1 cells (p < 0.001). The expression of ATP-binding cassette transporters A (ABCA)-1, ABCG-1, scavenger receptor (SR)-B1, liver X receptor (LXR)-α, and peroxisome proliferator-activated receptor (PPAR)-γ mRNA and proteins were increased (p < 0.001) following MIP2 administration. A concentration-dependent decrease in the phosphorylation of p65, p38, and JNK was also observed following MIP2 administration. Moreover, an inhibition of p65 phosphorylation enhanced the expression of ABCA1, ABCG1, SR-B1, and LXR-α. TLR inhibition promoted the cholesterol efflux pathway by increasing the expression of ABCA-1, ABCG-1, and SR-B1, thereby reducing foam cell formation. Our results suggest a potential role of the p65/NF-kB/LXR-α/ABCA1 axis in TLR-mediated cholesterol homeostasis.


Subject(s)
ATP Binding Cassette Transporter 1 , Cholesterol , Foam Cells , Lipoproteins, LDL , Liver X Receptors , Toll-Like Receptors , Humans , Foam Cells/metabolism , Foam Cells/drug effects , Cholesterol/metabolism , Liver X Receptors/metabolism , Toll-Like Receptors/metabolism , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , PPAR gamma/metabolism , THP-1 Cells , Macrophages/metabolism , Macrophages/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Lipopolysaccharides/pharmacology , Scavenger Receptors, Class B/metabolism , Scavenger Receptors, Class B/genetics
5.
Biosci Biotechnol Biochem ; 88(7): 776-783, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38714325

ABSTRACT

Atherosclerosis (AS) is the major cause of multiple cardiovascular diseases. In addition, the lipid accumulation of human vascular smooth muscle cells (HVSMCs) can cause the occurrence of AS. Secreted frizzled-related protein 5 (Sfrp5) was known to be downregulated in AS; however, the detailed function of Sfrp5 in HVSMCs remains unclear. Specifically, we found that Sfrp5 expression in oxLDL-treated HVSMCs was downregulated. Sfrp5 overexpression inhibited the viability of HVSMCs induced by oxLDL. In addition, oxLDL-induced proliferation and migration in HVSMCs were abolished by Sfrp5 overexpression. Sfrp5 overexpression reduced oxLDL-caused oxidative stress, lipid accumulation, and inflammation in HVSMCs. Meanwhile, oxLDL treatment increased the expressions of Wnt5a, c-Myc, and ß-catenin in HVSMCs, while this phenomenon was rescued by Sfrp5 overexpression. Furthermore, the inhibitory effect of Sfrp5 upregulation on the viability and migration of HVSMCs was reversed by R-spondin 1. These results indicate that Sfrp5 overexpression could reverse oxLDL-induced lipid accumulation in HVSMCs through inactivating Wnt5a/ß-catenin signaling pathway.


Subject(s)
Cell Movement , Lipid Metabolism , Lipoproteins, LDL , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Wnt-5a Protein , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Cell Movement/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Oxidative Stress , beta Catenin/metabolism , beta Catenin/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Signal Transduction
6.
Aging (Albany NY) ; 16(9): 8070-8085, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38728249

ABSTRACT

BACKGROUND: Inflammation is one of the significant consequences of ox-LDL-induced endothelial cell (EC) dysfunction. The senescence-associated secretory phenotype (SASP) is a critical source of inflammation factors. However, the molecular mechanism by which the SASP is regulated in ECs under ox-LDL conditions remains unknown. RESULTS: The level of SASP was increased in ox-LDL-treated ECs, which could be augmented by KLF4 knockdown whereas restored by KLF4 knock-in. Furthermore, we found that KLF4 directly promoted PDGFRA transcription and confirmed the central role of the NAPMT/mitochondrial ROS pathway in KLF4/PDGFRA-mediated inhibition of SASP. Animal experiments showed a higher SASP HFD-fed mice, compared with normal feed (ND)-fed mice, and the endothelium of EC-specific KLF4-/- mice exhibited a higher proportion of SA-ß-gal-positive cells and lower PDGFRA/NAMPT expression. CONCLUSIONS: Our results revealed that KLF4 inhibits the SASP of endothelial cells under ox-LDL conditions through the PDGFRA/NAMPT/mitochondrial ROS. METHODS: Ox-LDL-treated ECs and HFD-fed mice were used as endothelial senescence models in vitro and in vivo. SA-ß-gal stain, detection of SAHF and the expression of inflammatory factors determined SASP and senescence of ECs. The direct interaction of KLF4 and PDGFRA promotor was analyzed by EMSA and fluorescent dual luciferase reporting analysis.


Subject(s)
Cellular Senescence , Endothelial Cells , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Lipoproteins, LDL , Mitochondria , Reactive Oxygen Species , Receptor, Platelet-Derived Growth Factor alpha , Kruppel-Like Factor 4/metabolism , Animals , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Reactive Oxygen Species/metabolism , Cellular Senescence/drug effects , Mitochondria/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Mice , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Humans , Endothelial Cells/metabolism , Cytokines/metabolism , Phenotype , Mice, Knockout , Human Umbilical Vein Endothelial Cells/metabolism , Male , Signal Transduction
7.
Int Heart J ; 65(3): 466-474, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38749754

ABSTRACT

Endothelial cell dysfunction is the main pathology of atherosclerosis (AS). Sirtuin 6 (SIRT6), a deacetylase, is involved in AS progression. This study aimed to investigate the impacts of SIRT6 on the pyroptosis of endothelial cells and its underlying mechanisms. ApoE-/- mice were fed a high-fat diet (HFD) to establish the AS mouse model, atherosclerotic lesions were evaluated using oil red O staining, and blood lipids and inflammatory factors were measured using corresponding kits. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to establish the cell model, and pyroptosis was evaluated by flow cytometry, ELISA, and western blot. Immunoprecipitation (IP), co-IP, western blot, and immunofluorescence were used to detect the molecular mechanisms. The results showed that SIRT6 expression was downregulated in the blood of HFD-induced mice and ox-LDL-induced HUVECs. Overexpression of SIRT6 reduced atherosclerotic lesions, blood lipids, and inflammation in vivo and suppressed pyroptosis of HUVECs in vitro. Moreover, SIRT6 interacted with ASC to inhibit the acetylation of ASC, thus, reducing the interaction between ASC and NLRP3. Moreover, SIRT6 inhibits endothelial cell pyroptosis in the aortic roots of mice by deacetylating ASC. In conclusion, SIRT6 deacetylated ASC to inhibit its interaction with NLRP3 and then suppressed pyroptosis of endothelial cells, thus, decelerating the progression of AS. The findings provide new insights into the function of SIRT6 in AS.


Subject(s)
Atherosclerosis , Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , Pyroptosis , Sirtuins , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Sirtuins/metabolism , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , CARD Signaling Adaptor Proteins/metabolism , Disease Models, Animal , Diet, High-Fat , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Inbred C57BL
8.
Int J Biol Macromol ; 269(Pt 2): 132257, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729492

ABSTRACT

Low-density lipoprotein (LDL) transports cholesterol to various tissues via the blood. Glycation of LDL occurs during hyperglycemic condition which is characterised by persistently high blood glucose level. Circulating erythrocytes can come in direct contact with glycated LDL (G-LDL). The objective of this study was to investigate the effect of G-LDL on human erythrocytes, specifically on hemoglobin, intracellular generation of reactive species and the antioxidant defence system. Isolated erythrocytes were incubated with G-LDL (3 and 6 mg/ml) and native LDL (6 mg/ml) at 37 °C for 24 h. Native LDL and G-LDL untreated erythrocytes were similarly incubated at 37 °C and served as control. G-LDL treatment increased hemolysis compared to control and native LDL-treated erythrocytes. Incubation of erythrocytes with G-LDL led to an increase in protein oxidation and lipid peroxidation while greatly decreasing the total sulfhydryl content. It also significantly enhanced hemoglobin oxidation, heme degradation, and the release of free iron moiety. Treatment with G-LDL led to an appreciable increase in the production of reactive oxygen and nitrogen species. The antioxidant power and activities of major antioxidant enzymes were drastically reduced, while critical membrane-bound enzymes were inhibited. The surface morphology of G-LDL-treated erythrocytes was altered leading to the formation of echinocytes. Importantly, treatment of erythrocytes with native LDL did not significantly affect the above-mentioned parameters and values were similar to the corresponding controls. Thus, G-LDL is cytotoxic to human erythrocytes and causes oxidative damage to cell components. This can reduce the oxygen-transporting ability of blood and also result in red cell senescence and anemia.


Subject(s)
Erythrocytes , Hemoglobins , Hemolysis , Lipoproteins, LDL , Oxidation-Reduction , Reactive Oxygen Species , Humans , Erythrocytes/metabolism , Erythrocytes/drug effects , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Reactive Oxygen Species/metabolism , Hemoglobins/metabolism , Hemolysis/drug effects , Oxidation-Reduction/drug effects , Antioxidants/pharmacology , Lipid Peroxidation/drug effects , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Oxidative Stress/drug effects , Heme/metabolism , Heme/pharmacology , Glycated Proteins
9.
Angiogenesis ; 27(3): 545-560, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38733496

ABSTRACT

Regenerative capabilities of the endothelium rely on vessel-resident progenitors termed endothelial colony forming cells (ECFCs). This study aimed to investigate if these progenitors are impacted by conditions (i.e., obesity or atherosclerosis) characterized by increased serum levels of oxidized low-density lipoprotein (oxLDL), a known inducer of Endothelial-to-Mesenchymal Transition (EndMT). Our investigation focused on understanding the effects of EndMT on the self-renewal capabilities of progenitors and the associated molecular alterations. In the presence of oxLDL, ECFCs displayed classical features of EndMT, through reduced endothelial gene and protein expression, function as well as increased mesenchymal genes, contractility, and motility. Additionally, ECFCs displayed a dramatic loss in self-renewal capacity in the presence of oxLDL. RNA-sequencing analysis of ECFCs exposed to oxLDL validated gene expression changes suggesting EndMT and identified SOX9 as one of the highly differentially expressed genes. ATAC sequencing analysis identified SOX9 binding sites associated with regions of dynamic chromosome accessibility resulting from oxLDL exposure, further pointing to its importance. EndMT phenotype and gene expression changes induced by oxLDL in vitro or high fat diet (HFD) in vivo were reversed by the silencing of SOX9 in ECFCs or the endothelial-specific conditional knockout of Sox9 in murine models. Overall, our findings support that EndMT affects vessel-resident endothelial progenitor's self-renewal. SOX9 activation is an early transcriptional event that drives the mesenchymal transition of endothelial progenitor cells. The identification of the molecular network driving EndMT in vessel-resident endothelial progenitors presents a new avenue in understanding and preventing a range of condition where this process is involved.


Subject(s)
Lipoproteins, LDL , SOX9 Transcription Factor , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Animals , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Mice , Humans , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Epithelial-Mesenchymal Transition , Mice, Inbred C57BL , Male , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Cell Self Renewal , Endothelial Cells/metabolism
10.
Brain Behav Immun ; 119: 416-430, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636563

ABSTRACT

The role of microglia in triggering the blood-brain barrier (BBB) impairment and white matter damage after chronic cerebral hypoperfusion is unclear. Here we demonstrated that the vessel-adjacent microglia were specifically activated by the leakage of plasma low-density lipoprotein (LDL), which led to BBB breakdown and ischemic demyelination. Interestingly, we found that LDL stimulation enhanced microglial phagocytosis, causing excessive engulfment of myelin debris and resulting in an overwhelming lipid burden in microglia. Surprisingly, these lipid-laden microglia exhibited a suppressed profile of inflammatory response and compromised pro-regenerative properties. Microglia-specific knockdown of LDLR or systematic medication lowering circulating LDL-C showed protective effects against ischemic demyelination. Overall, our findings demonstrated that LDL-stimulated vessel-adjacent microglia possess a disease-specific molecular signature, characterized by suppressed regenerative properties, which is associated with the propagation of demyelination during ischemic white matter damage.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Lipoproteins, LDL , Microglia , White Matter , Microglia/metabolism , Animals , White Matter/metabolism , White Matter/pathology , Mice , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Brain Ischemia/metabolism , Blood-Brain Barrier/metabolism , Male , Mice, Inbred C57BL , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Phagocytosis/physiology , Myelin Sheath/metabolism
11.
Lipids Health Dis ; 23(1): 76, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38468335

ABSTRACT

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.


Subject(s)
Atherosclerosis , Flavones , PPAR gamma , Animals , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Macrophages , Foam Cells , Lipoproteins, LDL/pharmacology , CD36 Antigens/genetics , CD36 Antigens/metabolism
12.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-38477705

ABSTRACT

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.


Subject(s)
Chickens , Feathers , Animals , Male , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Cholesterol , Diet/veterinary , Dietary Supplements , Lipoproteins, LDL/pharmacology
13.
Discov Med ; 36(182): 571-580, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38531797

ABSTRACT

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.


Subject(s)
Atherosclerosis , MicroRNAs , Humans , Muscle, Smooth, Vascular/metabolism , In Situ Hybridization, Fluorescence , Atherosclerosis/metabolism , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Apoptosis , Cell Proliferation , MicroRNAs/metabolism , Cell Movement , Cells, Cultured
14.
Prostaglandins Other Lipid Mediat ; 172: 106832, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460759

ABSTRACT

Atherosclerosis (AS) represents a prevalent initiating factor for cardiovascular events. Insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is an oncofetal RNA-binding protein that participates in cardiovascular diseases. This work aimed to elaborate the effects of IGF2BP3 on AS and the probable mechanism by using an oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) model. Results indicated that IGF2BP3 expression was declined in the blood of AS patients and ox-LDL-induced HUVECs. IGF2BP3 elevation alleviated ox-LDL-provoked viability loss, apoptosis, oxidative DNA damage and endothelial dysfunction in HUVECs. Moreover, IGF2BP3 bound SESN1 and stabilized SESN1 mRNA. Furthermore, SESN1 interference reversed the impacts of IGF2BP3 overexpression on the apoptosis, oxidative DNA damage and endothelial dysfunction of ox-LDL-challenged HUVECs. Additionally, the activation of Nrf2 signaling mediated by IGF2BP3 up-regulation in ox-LDL-treated HUVECs was blocked by SESN1 absence. Collectively, SESN1 stabilized by IGF2BP3 might protect against AS by activating Nrf2 signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , NF-E2-Related Factor 2 , Oxidative Stress , RNA, Messenger , RNA-Binding Proteins , Signal Transduction , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Oxidative Stress/drug effects , Signal Transduction/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Apoptosis/drug effects , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , RNA Stability/drug effects , DNA Damage , Sestrins
15.
Clinics (Sao Paulo) ; 79: 100343, 2024.
Article in English | MEDLINE | ID: mdl-38554490

ABSTRACT

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.


Subject(s)
MicroRNAs , Muscle, Smooth, Vascular , Humans , Animals , Mice , Phenotype , Inflammation , Lipoproteins, LDL/pharmacology , Myocytes, Smooth Muscle , Oligonucleotides , MicroRNAs/genetics , Cell Proliferation , Apoptosis , Cell Movement , ATP Binding Cassette Transporter 1
16.
Cell Biol Int ; 48(6): 848-860, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38444077

ABSTRACT

Oxidized low-density lipoprotein (oxLDL), a key component in atherosclerosis and hyperlipidemia, is a risk factor for atherothrombosis in dyslipidemia, yet its mechanism is poorly understood. In this study, we used oxLDL-induced human aortic endothelial cells (HAECs) and high-fat diet (HFD)-fed mice as a hyperlipidemia model. Phosphatidylserine (PS) exposure, cytosolic Ca2+, reactive oxygen species (ROS), and lipid peroxidation were measured by flow cytometer. TMEM16F expression was detected by immunofluorescence, western blot, and reverse transcription polymerase chain reaction. Procoagulant activity (PCA) was measured by coagulation time, intrinsic/extrinsic factor Xase, and thrombin generation. We found that oxLDL-induced PS exposure and the corresponding PCA of HAECs were increased significantly compared with control, which could be inhibited over 90% by lactadherin. Importantly, TMEM16F expression in oxLDL-induced HAECs was upregulated by enhanced intracellular Ca2+ concentration, ROS, and lipid peroxidation, which led to PS exposure. Meanwhile, the knockdown of TMEM16F by short hairpin RNA significantly inhibited PS exposure in oxLDL-induced HAECs. Moreover, we observed that HFD-fed mice dramatically increased the progress of thrombus formation and accompanied upregulated TMEM16F expression by thromboelastography analysis, FeCl3-induced carotid artery thrombosis model, and western blot. Collectively, these results demonstrate that TMEM16F-mediated PS exposure may contribute to prothrombotic status under hyperlipidemic conditions, which may serve as a novel therapeutic target for the prevention of thrombosis in hyperlipidemia.


Subject(s)
Anoctamins , Endothelial Cells , Lipoproteins, LDL , Phosphatidylserines , Reactive Oxygen Species , Animals , Humans , Mice , Anoctamins/metabolism , Blood Coagulation/drug effects , Calcium/metabolism , Cells, Cultured , Diet, High-Fat , Endothelial Cells/metabolism , Hyperlipidemias/metabolism , Lipid Peroxidation/drug effects , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Mice, Inbred C57BL , Phosphatidylserines/metabolism , Reactive Oxygen Species/metabolism , Thrombosis/metabolism , Phospholipid Transfer Proteins/metabolism
17.
Am J Hypertens ; 37(7): 465-476, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38536049

ABSTRACT

BACKGROUND: Atherosclerosis (AS) stands as the primary contributor to cardiovascular disease, a pervasive global health concern. Extensive research has underscored the pivotal role of circular RNAs (circRNAs) in cardiovascular disease development. However, the specific functions of numerous circRNAs in AS remain poorly understood. METHODS: Quantitative real-time PCR analysis revealed a significant upregulation of circ_0104652 in oxidized low-density lipoprotein (ox-LDL)-induced vascular smooth muscle cells (VSMCs). Loss-of-function experiments were subsequently employed to assess the impact of circ_0104652 on ox-LDL-induced VSMCs. RESULTS: Silencing circ_0104652 was found to impede the proliferation and migration while promoting the apoptosis of ox-LDL-stimulated VSMCs. Mechanistic assays unveiled that circ_0104652 stabilized ADAM metallopeptidase with thrombospondin type 1 motif 7 (ADAMTS7) and high mobility group box 1 (HMGB1) by recruiting eukaryotic translation initiation factor 4A3 (EIF4A3) protein. Rescue assays further confirmed that circ_0104652 exerted its influence on ox-LDL-induced VSMC proliferation through modulation of ADAMTS7 and HMGB1. CONCLUSIONS: This study elucidates the role of the circ_0104652/EIF4A3/ADAMTS7/HMGB1 axis in ox-LDL-stimulated VSMCs, providing valuable insights into the intricate mechanisms involved.


Subject(s)
ADAMTS7 Protein , Atherosclerosis , Cell Movement , Cell Proliferation , HMGB1 Protein , Lipoproteins, LDL , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , RNA, Circular , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Cell Proliferation/drug effects , RNA, Circular/metabolism , RNA, Circular/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Cell Movement/drug effects , Humans , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , ADAMTS7 Protein/metabolism , ADAMTS7 Protein/genetics , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/genetics , Cells, Cultured , Signal Transduction , Apoptosis/drug effects
18.
Cell Signal ; 117: 111092, 2024 05.
Article in English | MEDLINE | ID: mdl-38331013

ABSTRACT

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.


Subject(s)
Foam Cells , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Foam Cells/metabolism , Inflammasomes/metabolism , Peptide Hydrolases/metabolism , Macrophages/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Endopeptidases/metabolism
19.
Hypertension ; 81(4): 861-875, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38361240

ABSTRACT

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.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Pre-Eclampsia , Humans , Pregnancy , Female , Animals , Mice , Placenta/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Placenta Growth Factor , Pravastatin/pharmacology , Up-Regulation , Prospective Studies , Pre-Eclampsia/drug therapy , Pre-Eclampsia/prevention & control , Fluvastatin/metabolism , Fluvastatin/pharmacology , Vascular Endothelial Growth Factor Receptor-1 , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Biomarkers , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism
20.
Cell Signal ; 117: 111114, 2024 05.
Article in English | MEDLINE | ID: mdl-38387686

ABSTRACT

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.


Subject(s)
Bone Marrow , Low Density Lipoprotein Receptor-Related Protein-6 , Animals , Mice , Bone Marrow/metabolism , Carrier Proteins/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Obesity/complications , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL