1
|
Yang Y, Chen S, Shi G, Huang S, Cui N, Tan L, Yang X. Polyphenol extract ameliorates diabetes-related atherosclerosis through HIF-1 signaling pathway in APOE -/- mice: Possible synergism with atorvastatin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156572. [PMID: 40023112 DOI: 10.1016/j.phymed.2025.156572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Diabetes mellitus promotes atherosclerosis (AS), a major cause of cardiovascular disease. Statins, clinical drugs for AS treatment, may increase the risk of diabetes and induce hepatotoxicity. Polyphenols can alleviate AS by exerting anti-inflammatory and antioxidant effects, and play a role in liver protection. However, the effects of polyphenol extract (PE) on diabetic AS and whether PE can assist atorvastatin in managing diabetic AS with additional benefits remain poorly understood. PURPOSE This study aims to investigate the synergistic effects of a type of PE with atorvastatin on diabetic AS, as well as explore the anti-atherogenic mechanisms of PE. METHODS Polyphenol constituents in PE were identified using UPLC-MS/MS. Streptozocin -induced diabetic ApoE-/- mice were orally administered 350 and 700 mg/kg of PE, 10 mg/kg of atorvastatin, or 5 mg/kg of atorvastatin+700 mg/kg of PE for 10 weeks. Atherosclerotic lesions and plaque stability were evaluated using Oil Red O staining, Sirius red staining, and immunofluorescence. Inflammation and oxidative stress were determined using the corresponding markers. Hepatic pathological changes were assessed using hematoxylin and eosin staining. Network pharmacology prediction, molecular docking, microscale thermophoresis (MST) and western blotting were used to explore the anti-atherosclerotic mechanisms of PE. RESULTS The polyphenolic constituents of PE mainly consist of flavonoids and phenolic acids. PE treatment effectively mitigated atherosclerotic lesions in diabetic mice, particularly at high dose. Co-treatment with a half-dose of atorvastatin and PE did not restrain the anti-atherosclerotic effects of atorvastatin treatment, whereas the combination better relieved hyperglycemia, inflammation, oxidative stress, and liver damage in diabetic AS compared to that with atorvastatin alone. In addition, network pharmacology, molecular docking, MST and western blotting results indicated that PE ameliorated diabetic AS, possibly by targeting the AKT/mTOR/HIF-1 signaling pathway. CONCLUSION The PE demonstrated efficacy against diabetic AS and cooperated with atorvastatin for some additional benefits, especially in liver protection, indicating that PE has the potential to be developed as a supplement for the management of diabetes-related AS.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Shengxiang Chen
- Hubei YouTang Health Management Co. Ltd, Wuhan, Hubei, 430000, PR China
| | - Guanjin Shi
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Shanshan Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Ningning Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Le Tan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China.
| |
Collapse
|
2
|
Aguilar V, Le Master E, Paul A, Ahn SJ, Lazarko D, Febbraio M, Mehta D, Lee J, Levitan I. Endothelial Stiffening Induced by CD36-Mediated Lipid Uptake Leads to Endothelial Barrier Disruption and Contributes to Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2025. [PMID: 40207364 DOI: 10.1161/atvbaha.124.322244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Endothelial stiffening induced by Western diet was proposed to be an important factor in vascular dysfunction. In this study, we determine the role of endothelial CD36 (cluster of differentiation 36) in stiffening, disruption of aortic endothelial barrier, and atherosclerosis in mouse models of obesity and hypercholesterolemia. METHODS To address this goal, we generated an endothelial-specific inducible knockdown mouse model of CD36, Cdh5.CreERT2CD36fl/fl, on C57/BL6J wild-type and LDLR-/- genetic backgrounds. Endothelial stiffness is assessed by atomic force microscopy; endothelial barrier integrity is assessed by imaging VE-cadherin junctions and by penetration of Evans blue dye into the aortic wall. Atherosclerotic plaques are quantified using oil red O staining. RESULTS Endothelial-specific downregulation of CD36 abrogates stiffening of aortic endothelium induced by Western diet in Cdh5.CreERT2CD36fl/fl and in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Prevention of Western diet-induced endothelial stiffening by downregulation of CD36 is associated with a protective effect against endothelial barrier disruption in both mouse models and with a significant decrease in the areas of atherosclerotic lesions in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Mechanistically, stiffening of human aortic endothelial cells in vitro is induced by saturated fatty acids, particularly palmitic acid (PA), which results in activation of RhoA. Both PA-induced endothelial stiffening and RhoA activation are abrogated by CD36 siRNA. Furthermore, PA-induced endothelial stiffening of excised aortas ex vivo is lost in aortas isolated from mice, where endothelial CD36 is downregulated. We also demonstrate that PA-induced activation of RhoA and endothelial stiffening require expressing an RhoA-inhibitory protein, Rho-GDI1 (Rho guanosine dissociation inhibitor 1). Finally, we discover that PA disrupts the colocalization of RhoA with Rho-GDI1. CONCLUSIONS We conclude that stiffening of the aortic endothelium by CD36-mediated uptake of fatty acids contributes significantly to Western diet-induced vascular dysfunction and atherosclerosis. We further propose that fatty acids may activate RhoA by inducing its dissociation from Rho-GDI1.
Collapse
Affiliation(s)
- Victor Aguilar
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| | - Elizabeth Le Master
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Amit Paul
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Sang Joon Ahn
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Dana Lazarko
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada (M.F.)
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago. (D.M.)
| | - James Lee
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| |
Collapse
|
3
|
Peng X, Lian Z, O'Brien V, Xiao J, Litchfield BA, Perrard XYD, Xu L, Ni J, Mukherjee A, Simmons T, Dong H, Mullick AE, Crooke R, Pownall HJ, Simon SI, Ballantyne CM, Wu H. Foamy monocytes and atherogenesis in mice with combined hyperlipidemia and effects of antisense knockdown of apoCIII. J Lipid Res 2025; 66:100763. [PMID: 39988193 PMCID: PMC11981816 DOI: 10.1016/j.jlr.2025.100763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
Hypertriglyceridemia (HTG), particularly in combined hyperlipidemia, increases risk for atherosclerotic cardiovascular disease, but the underlying mechanisms remain incompletely understood. We sought to determine contributions of circulating monocytes to atherosclerosis associated with HTG in combined hyperlipidemia, created by transgenic expression of human apoCIII in Ldlr-/- mice (Ldlr-/-ApoCIIItg) fed Western high-fat diet (WD). Tissue culture with THP-1 and primary human monocytes was used to examine effects of triglyceride (TG)-rich lipoproteins on monocytes. Ldlr-/-ApoCIIItg mice were also treated with apoCIII antisense oligonucleotide (ASO) and examined for foamy monocytes and atherosclerosis. Compared to Ldlr-/- mice, Ldlr-/-ApoCIIItg mice fed WD had early and persistent increases in lipid accumulation within monocytes and enhanced atherosclerosis. Ldlr-/-ApoCIIItg mice versus Ldlr-/- mice had higher levels of CD11c, CD36, and cytokines in foamy monocytes, with increases in foamy monocyte adhesion to vascular cell adhesion molecule-1 and oxidized LDL uptake. Monocytes took up TG-rich lipoprotein in vivo and in vitro and changed phenotypes. Foamy monocytes infiltrated into atherosclerotic lesions, and specific and sustained depletion of CD11c+ (foamy) monocytes profoundly reduced atherosclerosis in Ldlr-/-ApoCIIItg mice on WD. Treatment with apoCIII ASO lowered plasma TG and cholesterol levels, improved foamy monocyte phenotypes, and reduced atherosclerosis in Ldlr-/-ApoCIIItg mice. In conclusion, HTG in combined hyperlipidemia accelerates atherosclerosis, in part, by increasing foamy monocyte formation and infiltration into atherosclerotic plaques. Treatment with apoCIII ASO is a potential new therapy for improving monocyte phenotypes and reducing atherosclerosis in combined hyperlipidemia.
Collapse
Affiliation(s)
- Xueying Peng
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Veronica O'Brien
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Xiao
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Lu Xu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Ni
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aparna Mukherjee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Simmons
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, TX, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Zhang N, Zhang T, Feng J, Shang J, Zhang B, Dong Q, Zhang Z, Sun C. Application of therapeutical nanoparticles with neutrophil membrane camouflaging for inflammatory plaques targeting against atherosclerosis. Mater Today Bio 2025; 30:101397. [PMID: 39802828 PMCID: PMC11722182 DOI: 10.1016/j.mtbio.2024.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Atherosclerosis is the leading cause of cardiovascular disease and myocardial infarction. Precise and effective plaque targeting is a major objective for therapeutic outcomes throughout various stages of atherosclerosis. Inspired by the natural recruitment of neutrophils in atherosclerotic plaques, we fabricated a simvastatin (ST)-loaded and neutrophil membrane-cloaked nanoplatform (NNPST) for enhancing localized payload delivery and atherosclerosis management. The resulting NNPST mimicked neutrophil function and significantly decreased macrophage-mediated phagocytosis to prolong its own circulation time in the blood. Compared to pristine nanoparticles (NPST) without a membrane coating, NNPST achieved better plaque targeting in ApoE-/- mice, as indicated by neutrophils actively recruited in atherosclerotic lesions. The higher plaque homing with NNPST was monitored by dynamic fluorescence/magnetic resonance (MR) dual-modality imaging. The results further showed that NNPST efficiently prevented atherosclerosis development mainly by suppressing local inflammatory macrophages, and the percentage of plaques in the entire aortic area was reduced to 4.75 ± 1.48 % following NNPST treatment. A biosafety assessment indicated that the biomimetic NNPST induced no noticeable toxicity in the body. This approach of neutrophil membrane-camouflaged nanoparticles offers new opportunities to various therapeutic agents for on-demand delivery in neutrophil-involved inflammatory diseases.
Collapse
Affiliation(s)
- Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Jintang Feng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Jian Shang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Beibei Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450002, PR China
| | - Qingyang Dong
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, PR China
| | - Zhang Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chunyang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| |
Collapse
|
5
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-Specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed a Western Diet. Arterioscler Thromb Vasc Biol 2025; 45:74-89. [PMID: 39569521 PMCID: PMC11668626 DOI: 10.1161/atvbaha.124.321366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pharmacological inhibition of megalin (also known as LRP2 [low-density lipoprotein receptor-related protein-2]) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. METHODS Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and PTC-LRP2 -/- littermates. To study atherosclerosis, all mice were bred to an LDL (low-density lipoprotein) receptor -/- background and fed a Western diet to induce atherosclerosis. RESULTS PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed a Western diet but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not on hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished ALB (albumin) accumulation in PTCs within 10 days of Western diet feeding. RNA-sequencing analyses demonstrated the upregulation of inflammation-related pathways in the kidney. CONCLUSIONS PTC-specific megalin deletion does not affect atherosclerosis but leads to tubulointerstitial nephritis in mice fed a Western diet, with severe pathologies in male mice.
Collapse
MESH Headings
- Animals
- Low Density Lipoprotein Receptor-Related Protein-2/genetics
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Low Density Lipoprotein Receptor-Related Protein-2/deficiency
- Diet, Western/adverse effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Female
- Male
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/metabolism
- Mice
- Mice, Inbred C57BL
- Hypercholesterolemia/genetics
- Hypercholesterolemia/complications
- Hypercholesterolemia/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Plaque, Atherosclerotic
- CD68 Molecule
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Michael K. Franklin
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Masayoshi Kukida
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
6
|
Gan G, Zhang R, Zeng Y, Lu B, Luo Y, Chen S, Lei H, Cai Z, Huang X. Fecal microbiota transplantation validates the importance of gut microbiota in an ApoE -/- mouse model of chronic apical periodontitis-induced atherosclerosis. BMC Oral Health 2024; 24:1455. [PMID: 39614243 DOI: 10.1186/s12903-024-05230-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Chronic apical periodontitis (CAP) has been linked to the development of atherosclerosis, although the underlying mechanisms remain unclear. This study aimed to investigate the role of gut microbiota disruption in CAP-induced atherosclerosis development, focusing on trimethylamine N-oxide (TMAO)-related metabolites. METHODS The study utilized fecal microbiota transplantation (FMT) to transfer gut microbiota from mice with CAP to healthy mice. Atherosclerosis development was assessed by analyzing lesions in the aortic arch and aortic root. Serum lipid and inflammatory factor levels were measured. Composition and diversity of gut microbiota were analyzed using targeted metabolomics, with a focus on the ratio of Firmicutes to Bacteroidetes. The expression of hepatic flavin-containing monooxygenase 3 (FMO3) and serum TMAO levels were also evaluated. RESULTS Mice receiving gut microbiota from CAP mice showed increased atherosclerotic lesions compared to controls, without significant differences in serum lipid or inflammatory factor levels. Alterations in gut microbiota composition were observed, characterized by an increase in the Firmicutes to Bacteroidetes ratio. Peptostreptococcaceae abundance positively correlated with atherosclerosis severity, while Odoribacteraceae showed a negative correlation. No significant differences were found in hepatic FMO3 expression or serum TMAO levels. CONCLUSIONS The study confirms the role of gut microbiota disruption in CAP-mediated atherosclerosis development, independent of serum lipid or TMAO levels. Alterations in gut microbiota composition, particularly increased Firmicutes to Bacteroidetes ratio and specific bacterial families, were associated with atherosclerosis severity. These findings highlight the intricate interplay between gut microbiota and cardiovascular health in the context of CAP.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
7
|
Zhao G, Zhao Y, Liang W, Lu H, Liu H, Deng Y, Zhu T, Guo Y, Chang L, Garcia-Barrio MT, Chen YE, Zhang J. Endothelial KLF11 is a novel protector against diabetic atherosclerosis. Cardiovasc Diabetol 2024; 23:381. [PMID: 39462409 PMCID: PMC11514907 DOI: 10.1186/s12933-024-02473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases remain the leading cause of mortality in diabetic patients, with endothelial cell (EC) dysfunction serving as the initiating step of atherosclerosis, which is exacerbated in diabetes. Krüppel-like factor 11 (KLF11), known for its missense mutations leading to the development of diabetes in humans, has also been identified as a novel protector of vascular homeostasis. However, its role in diabetic atherosclerosis remains unexplored. METHODS Diabetic atherosclerosis was induced in both EC-specific KLF11 transgenic and knockout mice in the Ldlr-/- background by feeding a diabetogenic diet with cholesterol (DDC). Single-cell RNA sequencing (scRNA-seq) was utilized to profile EC dysfunction in diabetic atherosclerosis. Additionally, gain- and loss-of-function experiments were conducted to investigate the role of KLF11 in hyperglycemia-induced endothelial cell dysfunction. RESULTS We found that endothelial KLF11 deficiency significantly accelerates atherogenesis under diabetic conditions, whereas KLF11 overexpression remarkably inhibits it. scRNA-seq profiling demonstrates that loss of KLF11 increases endothelial-to-mesenchymal transition (EndMT) during atherogenesis under diabetic conditions. Utilizing gain- and loss-of-function approaches, our in vitro study reveals that KLF11 significantly inhibits EC inflammatory activation and TXNIP-induced EC oxidative stress, as well as Notch1/Snail-mediated EndMT under high glucose exposure. CONCLUSION Our study demonstrates that endothelial KLF11 is an endogenous protective factor against diabetic atherosclerosis. These findings indicate that manipulating KLF11 could be a promising approach for developing novel therapies for diabetes-related cardiovascular complications.
Collapse
Affiliation(s)
- Guizhen Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Yang Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Wenying Liang
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Haocheng Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People's Republic of China
| | - Hongyu Liu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yongjie Deng
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Tianqing Zhu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yanhong Guo
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Lin Chang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Minerva T Garcia-Barrio
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Jifeng Zhang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
8
|
Xue C, Zeng P, Gong K, Li Q, Feng Z, Wang M, Chen S, Yang Y, Li J, Zhang S, Yin Z, Liang Y, Yan T, Yu M, Feng K, Zhao D, Yang X, Zhang X, Ma L, Iwakiri Y, Chen L, Tang X, Chen Y, Chen H, Duan Y. Nogo-B inhibition facilitates cholesterol metabolism to reduce hypercholesterolemia. Cell Rep 2024; 43:114691. [PMID: 39235944 DOI: 10.1016/j.celrep.2024.114691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/09/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
The strategy of lowering cholesterol levels by promoting cholesterol excretion is still lacking, and few molecular targets act on multiple cholesterol metabolic processes. In this study, we find that Nogo-B deficiency/inhibition simultaneously promotes hepatic uptake of cholesterol and cholesterol excretion. Nogo-B deficiency decreases cholesterol levels by activating ATP-binding cassette transporters (ABCs), apolipoprotein E (ApoE), and low-density lipoprotein receptor (LDLR) expression. We discover that Nogo-B interacts with liver X receptor α (LXRα), and Nogo-B deficiency inhibits ubiquitination degradation of LXRα, thereby enhancing its function on cholesterol excretion. Decreased cellular cholesterol levels further activate SREBP2 and LDLR expression, thereby promoting hepatic uptake of cholesterol. Nogo-B inhibition decreases atherosclerotic plaques and cholesterol levels in mice, and Nogo-B levels are correlated to cholesterol levels in human plasma. In this study, Nogo-B deficiency/inhibition not only promotes hepatic uptake of blood cholesterol but also facilitates cholesterol excretion. This study reports a strategy to lower cholesterol levels by inhibiting Nogo-B expression to promote hepatic cholesterol uptake and cholesterol excretion.
Collapse
Affiliation(s)
- Chao Xue
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Peng Zeng
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Qian Li
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zian Feng
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Mengyao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Shasha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yanfang Yang
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jiaqi Li
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Zequn Yin
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yingquan Liang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Tengteng Yan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Miao Yu
- Medical College of Soochow University, Suzhou 215031, China
| | - Ke Feng
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Dan Zhao
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xia Zhang
- Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, China
| | - Likun Ma
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Liang Chen
- College of Life Science, Anhui Medical University, Hefei 230032, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No. 17 People's South Road, Chengdu, Sichuan 610041, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Houzao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China.
| | - Yajun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
9
|
Echrish J, Pasca MI, Cabrera D, Yang Y, Harper AGS. Developing a Biomimetic 3D Neointimal Layer as a Prothrombotic Substrate for a Humanized In Vitro Model of Atherothrombosis. Biomimetics (Basel) 2024; 9:372. [PMID: 38921252 PMCID: PMC11201422 DOI: 10.3390/biomimetics9060372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Acute cardiovascular events result from clots caused by the rupture and erosion of atherosclerotic plaques. This paper aimed to produce a functional biomimetic hydrogel of the neointimal layer of the atherosclerotic plaque that can support thrombogenesis upon exposure to human blood. A biomimetic hydrogel of the neointima was produced by culturing THP-1-derived foam cells within 3D collagen hydrogels in the presence or absence of atorvastatin. Prothrombin time and platelet aggregation onset were measured after exposure of the neointimal models to platelet-poor plasma and washed platelet suspensions prepared from blood of healthy, medication-free volunteers. Activity of the extrinsic coagulation pathway was measured using the fluorogenic substrate SN-17. Foam cell formation was observed following preincubation of the neointimal biomimetic hydrogels with oxidized LDL, and this was inhibited by pretreatment with atorvastatin. The neointimal biomimetic hydrogel was able to trigger platelet aggregation and blood coagulation upon exposure to human blood products. Atorvastatin pretreatment of the neointimal biomimetic layer significantly reduced its pro-aggregatory and pro-coagulant properties. In the future, this 3D neointimal biomimetic hydrogel can be incorporated as an additional layer within our current thrombus-on-a-chip model to permit the study of atherosclerosis development and the screening of anti-thrombotic drugs as an alternative to current animal models.
Collapse
Affiliation(s)
| | | | - David Cabrera
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | | |
Collapse
|
10
|
Rao C, Liu B, Qin H, Du Z. Enoyl coenzyme a hydratase 1 attenuates aortic valve calcification by suppressing Runx2 via Wnt5a/Ca 2+ pathway. J Cell Commun Signal 2024; 18:e12038. [PMID: 38946717 PMCID: PMC11208118 DOI: 10.1002/ccs3.12038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
The morbidity and death rates of calcified aortic valves|calcific aortic valve (CAV) disease (CAVD) remain high for its limited therapeutic choices. Here, we investigated the function, therapeutic potential, and putative mechanisms of Enoyl coenzyme A hydratase 1 (ECH1) in CAVD by various in vitro and in vivo experiments. Single-cell sequencing revealed that ECH1 was predominantly expressed in valve interstitial cells and was significantly reduced in CAVs. Overexpression of ECH1 reduced aortic valve calcification in ApoE-/- mice treated with high cholesterol diet, while ECH1 silencing had the reverse effect. We also identified Wnt5a, a noncanonical Wnt ligand, was also altered when ECH1 expression was modulated. Mechanistically, we found that ECH1 exerted anti-calcific actions through suppressing Wnt signaling, since CHIR99021, a Wnt agonist, may significantly lessen the protective impact of ECH1 overexpression on the development of valve calcification. ChIP and luciferase assays all showed that ECH1 overexpression prevented Runx2 binding to its downstream gene promoters (osteopontin and osteocalcin), while CHIR99021 neutralized this protective effect. Collectively, our findings reveal a previously unrecognized mechanism of ECH1-Wnt5a/Ca2+ regulation in CAVD, implying that targeting ECH1 may be a potential therapeutic strategy to prevent CAVD development.
Collapse
Affiliation(s)
- Caijun Rao
- Department of GeriatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Baoqing Liu
- Department of Cardiovascular SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Haojie Qin
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Zhipeng Du
- Department of GastroenterologyInstitute of Liver and Gastrointestinal DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
11
|
Cervantes J, Koska J, Kramer F, Akilesh S, Alpers CE, Mullick AE, Reaven P, Kanter JE. Elevated apolipoprotein C3 augments diabetic kidney disease and associated atherosclerosis in type 2 diabetes. JCI Insight 2024; 9:e177268. [PMID: 38743496 PMCID: PMC11383354 DOI: 10.1172/jci.insight.177268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetes increases the risk of both cardiovascular disease and kidney disease. Notably, most of the excess cardiovascular risk in people with diabetes is in those with kidney disease. Apolipoprotein C3 (APOC3) is a key regulator of plasma triglycerides, and it has recently been suggested to play a role in both type 1 diabetes-accelerated atherosclerosis and kidney disease progression. To investigate if APOC3 plays a role in kidney disease in people with type 2 diabetes, we analyzed plasma levels of APOC3 from the Veterans Affairs Diabetes Trial. Elevated baseline APOC3 levels predicted a greater loss of renal function. To mechanistically test if APOC3 plays a role in diabetic kidney disease and associated atherosclerosis, we treated black and tan, brachyury, WT and leptin-deficient (OB; diabetic) mice, a model of type 2 diabetes, with an antisense oligonucleotide (ASO) to APOC3 or a control ASO, all in the setting of human-like dyslipidemia. Silencing APOC3 prevented diabetes-augmented albuminuria, renal glomerular hypertrophy, monocyte recruitment, and macrophage accumulation, partly driven by reduced ICAM1 expression. Furthermore, reduced levels of APOC3 suppressed atherosclerosis associated with diabetes. This suggests that targeting APOC3 might benefit both diabetes-accelerated atherosclerosis and kidney disease.
Collapse
Affiliation(s)
- Jocelyn Cervantes
- Division of Metabolism, Endocrinology, and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Juraj Koska
- VA Phoenix Health Care System, Phoenix, Arizona, USA
| | - Farah Kramer
- Division of Metabolism, Endocrinology, and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Charles E. Alpers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | | | - Peter Reaven
- VA Phoenix Health Care System, Phoenix, Arizona, USA
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology, and Nutrition, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Gan G, Lin S, Luo Y, Zeng Y, Lu B, Zhang R, Chen S, Lei H, Cai Z, Huang X. Unveiling the oral-gut connection: chronic apical periodontitis accelerates atherosclerosis via gut microbiota dysbiosis and altered metabolites in apoE -/- Mice on a high-fat diet. Int J Oral Sci 2024; 16:39. [PMID: 38740741 PMCID: PMC11091127 DOI: 10.1038/s41368-024-00301-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
The aim of this study was to explore the impact of chronic apical periodontitis (CAP) on atherosclerosis in apoE-/- mice fed high-fat diet (HFD). This investigation focused on the gut microbiota, metabolites, and intestinal barrier function to uncover potential links between oral health and cardiovascular disease (CVD). In this study, CAP was shown to exacerbate atherosclerosis in HFD-fed apoE-/- mice, as evidenced by the increase in plaque size and volume in the aortic walls observed via Oil Red O staining. 16S rRNA sequencing revealed significant alterations in the gut microbiota, with harmful bacterial species thriving while beneficial species declining. Metabolomic profiling indicated disruptions in lipid metabolism and primary bile acid synthesis, leading to elevated levels of taurochenodeoxycholic acid (TCDCA), taurocholic acid (TCA), and tauroursodeoxycholic acid (TDCA). These metabolic shifts may contribute to atherosclerosis development. Furthermore, impaired intestinal barrier function, characterized by reduced mucin expression and disrupted tight junction proteins, was observed. The increased intestinal permeability observed was positively correlated with the severity of atherosclerotic lesions, highlighting the importance of the intestinal barrier in cardiovascular health. In conclusion, this research underscores the intricate interplay among oral health, gut microbiota composition, metabolite profiles, and CVD incidence. These findings emphasize the importance of maintaining good oral hygiene as a potential preventive measure against cardiovascular issues, as well as the need for further investigations into the intricate mechanisms linking oral health, gut microbiota, and metabolic pathways in CVD development.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shihan Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
13
|
Qian G, Adeyanju O, Cai D, Tucker TA, Idell S, Chen SY, Guo X. DOCK2 Promotes Atherosclerosis by Mediating the Endothelial Cell Inflammatory Response. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:599-611. [PMID: 37838011 PMCID: PMC10988758 DOI: 10.1016/j.ajpath.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/16/2023]
Abstract
The pathology of atherosclerosis, a leading cause of mortality in patients with cardiovascular disease, involves inflammatory phenotypic changes in vascular endothelial cells. This study explored the role of the dedicator of cytokinesis (DOCK)-2 protein in atherosclerosis. Mice with deficiencies in low-density lipoprotein receptor and Dock2 (Ldlr-/-Dock2-/-) and controls (Ldlr-/-) were fed a high-fat diet (HFD) to induce atherosclerosis. In controls, Dock2 was increased in atherosclerotic lesions, with increased intercellular adhesion molecule (Icam)-1 and vascular cell adhesion molecule (Vcam)-1, after HFD for 4 weeks. Ldlr-/-Dock2-/- mice exhibited significantly decreased oil red O staining in both aortic roots and aortas compared to that in controls after HFD for 12 weeks. In control mice and in humans, Dock2 was highly expressed in the ECs of atherosclerotic lesions. Dock2 deficiency was associated with attenuation of Icam-1, Vcam-1, and monocyte chemoattractant protein (Mcp)-1 in the aortic roots of mice fed HFD. Findings in human vascular ECs in vitro suggested that DOCK2 was required in TNF-α-mediated expression of ICAM-1/VCAM-1/MCP-1. DOCK2 knockdown was associated with attenuated NF-κB phosphorylation with TNF-α, partially accounting for DOCK2-mediated vascular inflammation. With DOCK2 knockdown in human vascular ECs, TNF-α-mediated VCAM-1 promoter activity was inhibited. The findings from this study suggest the novel concept that DOCK2 promotes the pathogenesis of atherosclerosis by modulating inflammation in vascular ECs.
Collapse
Affiliation(s)
- Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Oluwaseun Adeyanju
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Dunpeng Cai
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, Missouri
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Shi-You Chen
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, Missouri; The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri; Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.
| | - Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.
| |
Collapse
|
14
|
Qi W, Zhu S, Feng L, Liang J, Guo X, Cheng F, Guo Y, Lan G, Liang J. Integrated Analysis of the Transcriptome and Microbial Diversity in the Intestine of Miniature Pig Obesity Model. Microorganisms 2024; 12:369. [PMID: 38399773 PMCID: PMC10891586 DOI: 10.3390/microorganisms12020369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Obesity, a key contributor to metabolic disorders, necessitates an in-depth understanding of its pathogenesis and prerequisites for prevention. Guangxi Bama miniature pig (GBM) offers an apt model for obesity-related studies. In this research, we used transcriptomics and 16S rRNA gene sequencing to discern the differentially expressed genes (DEGs) within intestinal (jejunum, ileum, and colon) tissues and variations in microbial communities in intestinal contents of GBM subjected to normal diets (ND) and high-fat, high-carbohydrate diets (HFHCD). After a feeding duration of 26 weeks, the HFHCD-fed experimental group demonstrated notable increases in backfat thickness, BMI, abnormal blood glucose metabolism, and blood lipid levels alongside the escalated serum expression of pro-inflammatory factors and a marked decline in intestinal health status when compared to the ND group. Transcriptomic analysis revealed a total of 1669 DEGs, of which 27 had similar differences in three intestinal segments across different groups, including five immune related genes: COL6A6, CYP1A1, EIF2AK2, NMI, and LGALS3B. Further, we found significant changes in the microbiota composition, with a significant decrease in beneficial bacterial populations within the HFHCD group. Finally, the results of integrated analysis of microbial diversity with transcriptomics show a positive link between certain microbial abundance (Solibacillus, norank_f__Saccharimonadaceae, Candidatus_Saccharimonas, and unclassified_f__Butyricicoccaceae) and changes in gene expression (COL6A6 and NMI). Overall, HFHCD appears to co-contribute to the initiation and progression of obesity in GBM by aggravating inflammatory responses, disrupting immune homeostasis, and creating imbalances in intestinal flora.
Collapse
Affiliation(s)
- Wenjing Qi
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Siran Zhu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Lingli Feng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Jinning Liang
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, China
| | - Xiaoping Guo
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, China
| | - Feng Cheng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Yafen Guo
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Ganqiu Lan
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| | - Jing Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (W.Q.); (G.L.)
| |
Collapse
|
15
|
Song J, Zhang Y, Frieler RA, Andren A, Wood S, Tyrrell DJ, Sajjakulnukit P, Deng JC, Lyssiotis CA, Mortensen RM, Salmon M, Goldstein DR. Itaconate suppresses atherosclerosis by activating a Nrf2-dependent antiinflammatory response in macrophages in mice. J Clin Invest 2023; 134:e173034. [PMID: 38085578 PMCID: PMC10849764 DOI: 10.1172/jci173034] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Itaconate has emerged as a critical immunoregulatory metabolite. Here, we examined the therapeutic potential of itaconate in atherosclerosis. We found that both itaconate and the enzyme that synthesizes it, aconitate decarboxylase 1 (Acod1, also known as immune-responsive gene 1 [IRG1]), are upregulated during atherogenesis in mice. Deletion of Acod1 in myeloid cells exacerbated inflammation and atherosclerosis in vivo and resulted in an elevated frequency of a specific subset of M1-polarized proinflammatory macrophages in the atherosclerotic aorta. Importantly, Acod1 levels were inversely correlated with clinical occlusion in atherosclerotic human aorta specimens. Treating mice with the itaconate derivative 4-octyl itaconate attenuated inflammation and atherosclerosis induced by high cholesterol. Mechanistically, we found that the antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), was required for itaconate to suppress macrophage activation induced by oxidized lipids in vitro and to decrease atherosclerotic lesion areas in vivo. Overall, our work shows that itaconate suppresses atherogenesis by inducing Nrf2-dependent inhibition of proinflammatory responses in macrophages. Activation of the itaconate pathway may represent an important approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anthony Andren
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sherri Wood
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel J. Tyrrell
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center
| | - Jane C. Deng
- Graduate Program in Immunology, and
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes
| | | | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Graduate Program in Immunology, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Qu W, Zhou X, Jiang X, Xie X, Xu K, Gu X, Na R, Piao M, Xi X, Sun N, Wang X, Peng X, Xu J, Tian J, Zhang J, Guo J, Zhang M, Zhang Y, Pan Z, Wang K, Yu B, Sun B, Li S, Tian J. Long Noncoding RNA Gpr137b-ps Promotes Advanced Atherosclerosis via the Regulation of Autophagy in Macrophages. Arterioscler Thromb Vasc Biol 2023; 43:e468-e489. [PMID: 37767704 DOI: 10.1161/atvbaha.123.319037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Current therapies cannot completely reverse advanced atherosclerosis. High levels of amino acids, induced by Western diet, stimulate mTORC1 (mammalian target of rapamycin complex 1)-autophagy defects in macrophages, accelerating atherosclerotic plaque progression. In addition, autophagy-lysosomal dysfunction contributes to plaque necrotic core enlargement and lipid accumulation. Therefore, it is essential to investigate the novel mechanism and molecules to reverse amino acid-mTORC1-autophagy signaling dysfunction in macrophages of patients with advanced atherosclerosis. METHODS We observed that Gpr137b-ps (G-protein-coupled receptor 137B, pseudogene) was upregulated in advanced atherosclerotic plaques. The effect of Gpr137b-ps on the progression of atherosclerosis was studied by generating advanced plaques in ApoE-/- mice with cardiac-specific knockout of Gpr137b-ps. Bone marrow-derived macrophages and mouse mononuclear macrophage cell line RAW264.7 cells were subjected to starvation or amino acid stimulation to study amino acid-mTORC1-autophagy signaling. Using both gain- and loss-of-function approaches, we explored the mechanism of Gpr137b-ps-regulated autophagy. RESULTS Our results demonstrated that Gpr137b-ps deficiency led to enhanced autophagy in macrophages and reduced atherosclerotic lesions, characterized by fewer necrotic cores and less lipid accumulation. Knockdown of Gpr137b-ps increased autophagy and prevented amino acid-induced mTORC1 signaling activation. As the downstream binding protein of Gpr137b-ps, HSC70 (heat shock cognate 70) rescued the impaired autophagy induced by Gpr137b-ps. Furthermore, Gpr137b-ps interfered with the HSC70 binding to G3BP (Ras GTPase-activating protein-binding protein), which tethers the TSC (tuberous sclerosis complex) complex to lysosomes and suppresses mTORC1 signaling. In addition to verifying that the NTF2 (nuclear transport factor 2) domain of G3BP binds to HSC70 by in vitro protein synthesis, we further demonstrated that HSC70 binds to the NTF2 domain of G3BP through its W90-F92 motif by using computational modeling. CONCLUSIONS These findings reveal that Gpr137b-ps plays an essential role in the regulation of macrophage autophagy, which is crucial for the progression of advanced atherosclerosis. Gpr137b-ps impairs the interaction of HSC70 with G3BP to regulate amino acid-mTORC1-autophagy signaling, and these results provide a new potential therapeutic direction for the treatment of advanced atherosclerosis.
Collapse
Affiliation(s)
- Wenbo Qu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xin Zhou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xinjian Jiang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xianwei Xie
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China (X. Xie)
| | - Kaijian Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xia Gu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, China (R.N.)
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Na Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Junyan Xu
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (J.X.)
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Jiangtian Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Jian Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology (J.Z.)
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Zhenwei Pan
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (K.W.)
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Bin Sun
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy (S.L.), Harbin Medical University, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases Harbin Medical University, China (S.L.)
- Department of Biopharmaceutical Sciences, College of Pharmacy Harbin Medical University, China (S.L.)
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| |
Collapse
|
17
|
Chen H, Teng D, Xu B, Wang C, Wang H, Jia W, Gong L, Dong H, Zhong L, Yang J. The SGLT2 Inhibitor Canagliflozin Reduces Atherosclerosis by Enhancing Macrophage Autophagy. J Cardiovasc Transl Res 2023; 16:999-1009. [PMID: 37126209 DOI: 10.1007/s12265-023-10390-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/23/2023] [Indexed: 05/02/2023]
Abstract
It has been shown that SGLT2 suppresses atherosclerosis (AS). Recent studies indicate that autophagy widely participates in atherogenesis. This study aimed to assess the effect of canagliflozin (CAN) on atherogenesis via autophagy. Macrophages and ApoE - / - mice were used in this study. In macrophages, the results showed that CAN promoted LC3II expression and autophagosome formation. Furthermore, the cholesterol efflux assay demonstrated that CAN enhanced cholesterol efflux from macrophages via autophagy, resulting in lower lipid droplet concentrations in macrophages. The western blot revealed that CAN regulated autophagy via the AMPK/ULK1/Beclin1 signaling pathway. CAN resulted in increased macrophage autophagy in atherosclerotic plaques of ApoE - / - mice, confirming that CAN could inhibit the progression of AS via promoting macrophage autophagy. The current study found that CAN reduced the production of atherosclerotic lesions, which adds to our understanding of how SGLT2 inhibitors function to delay the progression of AS.
Collapse
Affiliation(s)
- Hongping Chen
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Da Teng
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Bowen Xu
- Binzhou Medical University, Yantai, Shandong Province, China
| | - Chunxiao Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Hua Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Wenjuan Jia
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Lei Gong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Haibin Dong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Lin Zhong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China.
| | - Jun Yang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China.
| |
Collapse
|
18
|
Chen H, Zhang L, Mi S, Wang H, Wang C, Jia W, Gong L, Dong H, Xu B, Jing Y, Ge P, Pei Z, Zhong L, Yang J. FURIN suppresses the progression of atherosclerosis by promoting macrophage autophagy. FASEB J 2023; 37:e22933. [PMID: 37093709 DOI: 10.1096/fj.202201762rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 04/25/2023]
Abstract
FURIN, a member of the mammalian proprotein convertases (PCs) family, can promote the proteolytic maturation of proproteins. It has been shown that FURIN plays an important role in the progression of atherosclerosis (AS). Current evidence indicates that autophagy widely participates in atherogenesis. This study aimed to explore whether FURIN could affect atherogenesis via autophagy. The effect of FURIN on autophagy was studied using aortic tissues from aortic dissection patients who had BENTALL surgery, as well as macrophages and ApoE-/- mice. In atherosclerotic plaques of aortic tissues from patients, FURIN expression and autophagy were elevated. In macrophages, FURIN-shRNA and FURIN-overexpression lentivirus were used to intervene in FURIN expression. The results showed that FURIN overexpression accelerated LC3 formation in macrophages during the autophagosome formation phase. Furthermore, FURIN-induced autophagy resulted in lower lipid droplet concentrations in macrophages. The western blot revealed that FURIN regulated autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway. In vivo, FURIN overexpression resulted in increased macrophage LC3 formation in ApoE-/- mice atherosclerotic plaques, confirming that FURIN could inhibit the progression of AS by promoting macrophage autophagy. The present study demonstrated that FURIN suppressed the progression of AS by promoting macrophage autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway, which attenuated atherosclerotic lesion formation. Based on this data, current findings add to our understanding of the complexity of AS.
Collapse
Affiliation(s)
- Hongping Chen
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lihui Zhang
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Shaohua Mi
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Hua Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Chunxiao Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Wenjuan Jia
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lei Gong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Haibin Dong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Bowen Xu
- The 2nd Medical Colloge, Binzhou Medical University, Yantai, China
| | - Yanyan Jing
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Peipei Ge
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Zhigang Pei
- Department of Vascular Surgery, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, China
| | - Lin Zhong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Jun Yang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| |
Collapse
|
19
|
Metformin Directly Binds to MMP-9 to Improve Plaque Stability. J Cardiovasc Dev Dis 2023; 10:jcdd10020054. [PMID: 36826550 PMCID: PMC9962015 DOI: 10.3390/jcdd10020054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Vulnerable atherosclerotic plaque rupture is the principal mechanism that accounts for myocardial infarction and stroke. High matrix metalloproteinase-9 (MMP-9) expression and activity have been proven to lead to plaque instability. Metformin, a first-line treatment for type 2 diabetes, is beneficial to plaque vulnerability. However, the mechanism underlying its anti-atherogenic effect remains unclear. Molecular docking and surface plasmon resonance experiments showed that metformin directly interacts with MMP-9, and incubated MMP-9 overexpressing HEK293A cells with metformin (1 μmol·L-1) significantly attenuates MMP-9's activity using zymography and MMP activity assays. Moreover, metformin treatment drives MMP-9 degradation. Next, we constructed a carotid artery atherosclerotic plaque model and administered consecutive 14-day metformin (200 mg·kg-1·d-1) treatment by intragastric gavage. Immunofluorescence staining of the right carotid common artery and serum MMP activity assay results showed that metformin treatment decreased local plaque MMP-9 protein level and circulating MMP-9 activity, respectively. Histochemical staining revealed that after metformin treatment, the collagen content in plaque was significantly preserved, and the plaque vulnerability index decreased. These findings suggested that metformin improved atherosclerotic plaque stability by directly binding to MMP-9 and driving its degradation.
Collapse
|
20
|
Dog models of human atherosclerotic cardiovascular diseases. Mamm Genome 2022:10.1007/s00335-022-09965-w. [PMID: 36243810 DOI: 10.1007/s00335-022-09965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/06/2022] [Indexed: 10/17/2022]
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of death worldwide. Eighty-five percent of CVD-associated deaths are due to heart attacks and stroke. Atherosclerosis leads to heart attack and stroke through a slow progression of lesion formation and luminal narrowing of arteries. Dogs are similar to humans in terms of their cardiovascular physiology, size, and anatomy. Dog models have been developed to recapitulate the complex phenotype of human patients and understand the underlying mechanism of CVD. Different methods, including high-fat, high-cholesterol diet and genetic modification, have been used to generate dog models of human CVD. Remarkably, the location and severity of atherosclerotic lesions in the coronary arteries and branches of the carotid arteries of dog models closely resemble those of human CVD patients. Overt clinical manifestations such as stroke caused by plaque rupture and thrombosis were observed in dog models. Thus, dog models can help define the pathophysiological mechanisms of atherosclerosis and develop potential strategy for preventing and treating CVD. In this review, we summarize the progress in generating and characterizing canine models to investigate CVD and discuss the advantages and limitations of canine CVD models.
Collapse
|
21
|
Chen Z, Ouyang C, Zhang H, Gu Y, Deng Y, Du C, Cui C, Li S, Wang W, Kong W, Chen J, Cai J, Geng B. Vascular smooth muscle cell-derived hydrogen sulfide promotes atherosclerotic plaque stability via TFEB (transcription factor EB)-mediated autophagy. Autophagy 2022; 18:2270-2287. [PMID: 35090378 PMCID: PMC9542771 DOI: 10.1080/15548627.2022.2026097] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) contribute to plaque stability. VSMCs are also a major source of CTH (cystathionine gamma-lyase)-hydrogen sulfide (H2S), a protective gasotransmitter in atherosclerosis. However, the role of VSMC endogenous CTH-H2S in pathogenesis of plaque stability and the mechanism are unknown. In human carotid plaques, CTH expression in ACTA2+ cells was dramatically downregulated in lesion areas in comparison to non-lesion areas. Intraplaque CTH expression was positively correlated with collagen content, whereas there was a negative correlation with CD68+ and necrotic core area, resulting in a rigorous correlation with vulnerability index (r = -0.9033). Deletion of Cth in VSMCs exacerbated plaque vulnerability, and were associated with VSMC autophagy decline, all of which were rescued by H2S donor. In ox-LDL treated VSMCs, cth deletion reduced collagen and heightened apoptosis association with autophagy reduction, and vice versa. For the mechanism, CTH-H2S mediated VSMC autophagosome formation, autolysosome formation and lysosome function, in part by activation of TFEB, a master regulator for autophagy. Interference with TFEB blocked CTH-H2S effects on VSMCs collagen and apoptosis. Next, we demonstrated that CTH-H2S sulfhydrated TFEB at Cys212 site, facilitating its nuclear translocation, and then promoting transcription of its target genes such as ATG9A, LAPTM5 or LDLRAP1. Conclusively, CTH-H2S increases VSMC autophagy by sulfhydration and activation of TFEB, promotes collagen secretion and inhibits apoptosis, thereby attenuating atherogenesis and plaque vulnerability. CTH-H2S may act as a warning biomarker for vulnerable plaque.Abbreviations ATG9A: autophagy related 9A; CTH: cystathionine gamma-lyase; CQ: chloroquine; HASMCs: human aortic smooth muscle cells; H2S: hydrogen sulfide; LAMP1: lysosomal associated membrane protein 1; LAPTM5: lysosomal protein transmembrane 5; NaHS: sodium hydrosulfide hydrate; ox-LDL: oxidized-low density lipoprotein; PPG: DL- propagylglycine; TFEB: transcription factor EB; 3-MA: 3-methyladenine; VSMCs: vascular smooth muscle cells.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, Beijing, China
| | - Haizeng Zhang
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanrui Gu
- Department of Vascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, Beijing, China
| | - Yue Deng
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Changting Cui
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuangyue Li
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjie Wang
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jingzhou Chen
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,CONTACT Jingzhou Chen ; Jun Cai ; Bin Geng Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Cai
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Geng
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Bywaters BC, Pedraza G, Trache A, Rivera GM. Endothelial NCK2 promotes atherosclerosis progression in male but not female Nck1-null atheroprone mice. Front Cardiovasc Med 2022; 9:955027. [PMID: 36035930 PMCID: PMC9413153 DOI: 10.3389/fcvm.2022.955027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
A better understanding of endothelial dysfunction holds promise for more effective interventions for atherosclerosis prevention and treatment. Endothelial signaling by the non-catalytic region of the tyrosine kinase (NCK) family of adaptors, consisting of NCK1 and NCK2, has been implicated in cardiovascular development and postnatal angiogenesis but its role in vascular disease remains incompletely understood. Here, we report stage- and sex-dependent effects of endothelial NCK2 signaling on arterial wall inflammation and atherosclerosis development. Male and female Nck1-null atheroprone mice enabling inducible, endothelial-specific Nck2 inactivation were fed a high fat diet (HFD) for 8 or 16 weeks to model atherosclerosis initiation and progression, respectively. Analysis of aorta preparations en face during disease progression, but not initiation, showed a significant reduction in plaque burden in males, but not females, lacking endothelial NCK2 relative to controls. Markers of vascular inflammation were reduced by endothelial NCK2 deficiency in both males and females during atherosclerosis progression but not initiation. At advanced stages of disease, plaque size and severity of atherosclerotic lesions were reduced by abrogation of endothelial NCK2 signaling only in males. Collectively, our results demonstrate stage- and sex-dependent modulation of atherosclerosis development by endothelial NCK2 signaling.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- *Correspondence: Briana C. Bywaters
| | - Gladys Pedraza
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Andreea Trache
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX, United States
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Gonzalo M. Rivera
| |
Collapse
|
23
|
Suh J, Kim S, Lee S, Kim R, Park NH. Hyperlipidemia is necessary for the initiation and progression of atherosclerosis by severe periodontitis in mice. Mol Med Rep 2022; 26:273. [PMID: 35795972 PMCID: PMC9309540 DOI: 10.3892/mmr.2022.12789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/07/2022] [Indexed: 11/08/2022] Open
Abstract
Hyperlipidemia is a major risk of atherosclerosis; however, systemic inflammatory diseases such as rheumatoid arthritis, psoriasis, systemic lupus erythematosus and systemic sclerosis are also known risks for the development of atherosclerosis. Periodontitis, a local and systemic inflammatory condition, has also been reported as a risk for atherosclerosis, but the specific link between periodontitis and atherosclerosis remains somewhat controversial. We previously reported that ligature-induced periodontitis exacerbates atherosclerosis in hyperlipidemic Apolipoprotein E-deficient (ApoE−/−) mice. To understand whether hyperlipidemia is necessary for the development and exacerbation of atherosclerosis associated with periodontitis, the present study created ligature-induced periodontitis in both wild-type (WT) and ApoE−/− mice. Subsequently, the status of local, systemic and vascular inflammation, serum lipid contents and arterial lipid deposition were examined with histological analysis, µCT, en face analysis, serum lipid and cytokine measurements, reverse transcription-quantitative PCR and immunohistochemical analysis. Ligature placement induced severe periodontitis in both WT and ApoE−/− mice at the local level as demonstrated by gingival inflammation, alveolar bone loss, increased osteoclastic activities and inflammation in alveolar bone. Systemic inflammation was also induced by ligature placement in both WT and ApoE−/− mice, albeit more so in ApoE−/− mice. The serum cholesterol levels were not altered by the ligature in both WT and ApoE−/− mice. However, the vascular inflammation and arterial lipid deposition were induced by ligature-induced periodontitis only in ApoE−/− mice, but not in WT mice. The present study indicated that the coupling of systemic inflammation and hyperlipidemia was necessary for the development and exacerbation of atherosclerosis induced by ligature-induced periodontitis in mice.
Collapse
Affiliation(s)
- Jin Suh
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Sharon Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Sung Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - Reuben Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, Los Angeles, CA 90095, USA
| |
Collapse
|
24
|
Zhen H, Yan Q, Liu Y, Li Y, Yang S, Jiang Z. Chitin oligosaccharides alleviate atherosclerosis progress in ApoE-/- mice by regulating lipid metabolism and inhibiting inflammation. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.03.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
25
|
Gisterå A, Ketelhuth DFJ, Malin SG, Hansson GK. Animal Models of Atherosclerosis-Supportive Notes and Tricks of the Trade. Circ Res 2022; 130:1869-1887. [PMID: 35679358 DOI: 10.1161/circresaha.122.320263] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atherosclerotic cardiovascular disease is a major cause of death among humans. Animal models have shown that cholesterol and inflammation are causatively involved in the disease process. Apolipoprotein B-containing lipoproteins elicit immune reactions and instigate inflammation in the vessel wall. Still, a treatment that is specific to vascular inflammation is lacking, which motivates continued in vivo investigations of the immune-vascular interactions that drive the disease. In this review, we distill old notions with emerging concepts into a contemporary understanding of vascular disease models. Pros and cons of different models are listed and the complex integrative interplay between cholesterol homeostasis, immune activation, and adaptations of the vascular system is discussed. Key limitations with atherosclerosis models are highlighted, and we suggest improvements that could accelerate progress in the field. However, excessively rigid experimental guidelines or limiting usage to certain animal models can be counterproductive. Continued work in improved models, as well as the development of new models, should be of great value in research and could aid the development of cardiovascular disease diagnostics and therapeutics of the future.
Collapse
Affiliation(s)
- Anton Gisterå
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Daniel F J Ketelhuth
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.).,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark (SDU), Odense, Denmark (D.F.J.K)
| | - Stephen G Malin
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Göran K Hansson
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| |
Collapse
|
26
|
La Rose AM, Groenen AG, Halmos B, Bazioti V, Rutten MG, Krishnamurthy KA, Koster MH, Kloosterhuis NJ, Smit M, Havinga R, Mithieux G, Rajas F, Kuipers F, Oosterveer MH, Westerterp M. Increased atherosclerosis in a mouse model of glycogen storage disease type 1a. Mol Genet Metab Rep 2022; 31:100872. [PMID: 35782606 PMCID: PMC9248218 DOI: 10.1016/j.ymgmr.2022.100872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 12/02/2022] Open
Abstract
Glycogen storage disease type 1a (GSD Ia) is an inborn error of carbohydrate metabolism. Despite severe hyperlipidemia, GSD Ia patients show limited atherogenesis compared to age-and-gender matched controls. Employing a GSD Ia mouse model that resembles the severe hyperlipidemia in patients, we here found increased atherogenesis in GSD Ia. These data provide a rationale for investigating atherogenesis in GSD Ia in a larger patient cohort.
Collapse
Affiliation(s)
- Anouk M. La Rose
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anouk G. Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn G.S. Rutten
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kishore A. Krishnamurthy
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maaike H. Oosterveer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Corresponding author at: Department of Pediatrics, University Medical Center Groningen, ERIBA Building 3226 room 04.14, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
27
|
Nguyen MA, Hoang HD, Rasheed A, Duchez AC, Wyatt H, Lynn Cottee M, Graber TE, Susser L, Robichaud S, Berber İ, Geoffrion M, Ouimet M, Kazan H, Maegdefessel L, Mulvihill EE, Alain T, Rayner KJ. miR-223 Exerts Translational Control of Proatherogenic Genes in Macrophages. Circ Res 2022; 131:42-58. [PMID: 35611698 PMCID: PMC9213086 DOI: 10.1161/circresaha.121.319120] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A significant burden of atherosclerotic disease is driven by inflammation. Recently, microRNAs (miRNAs) have emerged as important factors driving and protecting from atherosclerosis. miR-223 regulates cholesterol metabolism and inflammation via targeting both cholesterol biosynthesis pathway and NFkB signaling pathways; however, its role in atherosclerosis has not been investigated. We hypothesize that miR-223 globally regulates core inflammatory pathways in macrophages in response to inflammatory and atherogenic stimuli thus limiting the progression of atherosclerosis.
Collapse
Affiliation(s)
- My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Huy-Dung Hoang
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Anne-Claire Duchez
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Mary Lynn Cottee
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.)
| | - Leah Susser
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Sabrina Robichaud
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - İbrahim Berber
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Turkey (I.B.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Mireille Ouimet
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Turkey (H.K.)
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.).,Department of Medicine, Karolinska Institute, Stockholm, Sweden (L.M.)
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Centre for Infection, Immunity & Inflammation, Faculty of Medicine, University of Ottawa, Canada (K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| |
Collapse
|
28
|
Rom O, Liu Y, Finney AC, Ghrayeb A, Zhao Y, Shukha Y, Wang L, Rajanayake KK, Das S, Rashdan NA, Weissman N, Delgadillo L, Wen B, Garcia-Barrio MT, Aviram M, Kevil CG, Yurdagul A, Pattillo CB, Zhang J, Sun D, Hayek T, Gottlieb E, Mor I, Chen YE. Induction of glutathione biosynthesis by glycine-based treatment mitigates atherosclerosis. Redox Biol 2022; 52:102313. [PMID: 35447412 PMCID: PMC9044008 DOI: 10.1016/j.redox.2022.102313] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
Lower circulating levels of glycine are consistently reported in association with cardiovascular disease (CVD), but the causative role and therapeutic potential of glycine in atherosclerosis, the underlying cause of most CVDs, remain to be established. Here, following the identification of reduced circulating glycine in patients with significant coronary artery disease (sCAD), we investigated a causative role of glycine in atherosclerosis by modulating glycine availability in atheroprone mice. We further evaluated the atheroprotective potential of DT-109, a recently identified glycine-based compound with dual lipid/glucose-lowering properties. Glycine deficiency enhanced, while glycine supplementation attenuated, atherosclerosis development in apolipoprotein E-deficient (Apoe−/−) mice. DT-109 treatment showed the most significant atheroprotective effects and lowered atherosclerosis in the whole aortic tree and aortic sinus concomitant with reduced superoxide. In Apoe−/− mice with established atherosclerosis, DT-109 treatment significantly reduced atherosclerosis and aortic superoxide independent of lipid-lowering effects. Targeted metabolomics and kinetics studies revealed that DT-109 induces glutathione formation in mononuclear cells. In bone marrow-derived macrophages (BMDMs), glycine and DT-109 attenuated superoxide formation induced by glycine deficiency. This was abolished in BMDMs from glutamate-cysteine ligase modifier subunit-deficient (Gclm−/-) mice in which glutathione biosynthesis is impaired. Metabolic flux and carbon tracing experiments revealed that glycine deficiency inhibits glutathione formation in BMDMs while glycine-based treatment induces de novo glutathione biosynthesis. Through a combination of studies in patients with CAD, in vivo studies using atherosclerotic mice and in vitro studies using macrophages, we demonstrated a causative role of glycine in atherosclerosis and identified glycine-based treatment as an approach to mitigate atherosclerosis through antioxidant effects mediated by induction of glutathione biosynthesis.
Collapse
Affiliation(s)
- Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Yuhao Liu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Alia Ghrayeb
- The Laboratory for Metabolism in Health and Disease, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Ying Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yousef Shukha
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa, 3109601, Israel; The Lipid Research Laboratory, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3525433, Israel
| | - Lu Wang
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Krishani K Rajanayake
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nabil A Rashdan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Natan Weissman
- The Laboratory for Metabolism in Health and Disease, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Luisa Delgadillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Bo Wen
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Minerva T Garcia-Barrio
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Aviram
- The Lipid Research Laboratory, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3525433, Israel
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Arif Yurdagul
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher B Pattillo
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Duxin Sun
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tony Hayek
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa, 3109601, Israel; The Lipid Research Laboratory, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3525433, Israel
| | - Eyal Gottlieb
- The Laboratory for Metabolism in Health and Disease, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Inbal Mor
- The Laboratory for Metabolism in Health and Disease, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Y Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
29
|
Bjørklund MM, Bernal JA, Bentzon JF. Atherosclerosis Induced by Adeno-Associated Virus Encoding Gain-of-Function PCSK9. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:461-473. [PMID: 35237981 DOI: 10.1007/978-1-0716-1924-7_27] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induction of atherosclerosis in mice with one or more genetic alterations (e.g., conditional deletion of a gene of interest) has traditionally required crossbreeding with Apoe or Ldlr deficient mice to achieve sufficient hypercholesterolemia. However, this procedure is time consuming and generates a surplus of mice with genotypes that are irrelevant for experiments. Several alternative methods exist that obviate the need to work in mice with germline-encoded hypercholesterolemia. In this chapter, we detail an efficient and increasingly used method to induce hypercholesterolemia in mice through adeno-associated virus-mediated transfer of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene.
Collapse
Affiliation(s)
- Martin Mæng Bjørklund
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
30
|
MEKK3-TGFβ crosstalk regulates inward arterial remodeling. Proc Natl Acad Sci U S A 2021; 118:2112625118. [PMID: 34911761 DOI: 10.1073/pnas.2112625118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 01/08/2023] Open
Abstract
Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFβ-signaling both in vitro and in vivo. Endothelial-specific TGFβR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFβR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.
Collapse
|
31
|
Yin F, Lin P, Yu WQ, Shen N, Li Y, Guo SD. The Cordyceps militaris-Derived Polysaccharide CM1 Alleviates Atherosclerosis in LDLR (-/-) Mice by Improving Hyperlipidemia. Front Mol Biosci 2021; 8:783807. [PMID: 34966782 PMCID: PMC8710727 DOI: 10.3389/fmolb.2021.783807] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/26/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerotic cardiovascular disease has a high mortality worldwide. Our lab previously purified a polysaccharide designated as CM1 with (1→4)-β-D-Glcp and (1→2)-α-D-Manp glycosyls as the backbone. In this study, we investigated the anti-atherosclerosis effect of CM1 and the underlying mechanisms of action in a low-density lipoprotein receptor knockout (LDLR(-/-) mouse model. It was found that CM1 significantly decreased the formation of atherosclerotic plaques. Mechanistically, CM1 enhanced plasma level of apolipoprotein A-I and decreased the plasma levels of triglyceride, apolipoprotein B, and total cholesterol. In the absence of LDLR, CM1 elevated the expression of very low-density lipoprotein receptor for liver uptake of plasma apolipoprotein B-containing particles and reduced hepatic triglyceride synthesis by inhibiting sterol regulatory element binding protein 1c. CM1 improved lipids excretion by increasing the liver X receptor α/ATP-binding cassette G5 pathway in small intestine. CM1 reduced lipogenesis and lipolysis by inhibiting peroxisome proliferator-activated receptor γ and adipose triglyceride lipase in epididymal fat. Furthermore, CM1 improved lipid profile in C57BL/6J mice. Collectively, CM1 can modulate lipid metabolism by multiple pathways, contributing to reduced plasma lipid level and formation of atherosclerotic plaques in LDLR(-/-) mice. This molecule could be explored as a potential compound for prevention and treatment of hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
32
|
Lee GL, Liao TL, Wu JY, Wu KK, Kuo CC. Restoration of 5-methoxytryptophan protects against atherosclerotic chondrogenesis and calcification in ApoE -/- mice fed high fat diet. J Biomed Sci 2021; 28:74. [PMID: 34749728 PMCID: PMC8573875 DOI: 10.1186/s12929-021-00771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/28/2021] [Indexed: 11/18/2022] Open
Abstract
Background Toll-like receptor-2 (TLR2) promotes vascular smooth muscle cell (VSMC) transdifferentiation to chondrocytes and calcification in a p38 MAPK-dependent manner. Vascular 5-methoxytryptophan (5-MTP) is a newly identified factor with anti-inflammatory actions. As 5-MTP targets p38 MAPK for its actions, we postulated that 5-MTP protects against vascular chondrogenesis and calcification. Methods High-fat diet-induced advanced atherosclerosis in mice were performed to investigate the effect of 5-MTP on atherosclerotic lesions and calcification. VSMCs were used to determine the role of 5-MTP in VSMC chondrogenic differentiation and calcification. Alizarin red S and Alcian blue staining were used to measure VSMC calcification and chondrogenic differentiation, respectively. Results 5-MTP was detected in aortic tissues of ApoE−/− mice fed control chow. It was reduced in ApoE−/− mice fed high-fat diet (HFD), but was restored in ApoE−/−Tlr2−/− mice, suggesting that HFD reduces vascular 5-MTP production via TLR2. Intraperitoneal injection of 5-MTP or its analog into ApoE−/− mice fed HFD reduced aortic atherosclerotic lesions and calcification which was accompanied by reduction of chondrogenesis and calcium deposition. Pam3CSK4 (Pam3), ligand of TLR2, induced SMC phenotypic switch to chondrocytes. Pretreatment with 5-MTP preserved SMC contractile proteins and blocked Pam3-induced chondrocyte differentiation and calcification. 5-MTP inhibited HFD-induced p38 MAPK activation in vivo and Pam3-induced p38 MAPK activation in SMCs. 5-MTP suppressed HFD-induced CREB activation in aortic tissues and Pam3-induced CREB and NF-κB activation in SMCs. Conclusions These findings suggest that 5-MTP is a vascular arsenal against atherosclerosis and calcification by inhibiting TLR2–mediated SMC phenotypic switch to chondrocytes and the consequent calcification. 5-MTP exerts these effects by blocking p38 MAPK activation and inhibiting CREB and NF-κB transactivation activity. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00771-1.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Lien Liao
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan. .,College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan. .,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
33
|
Ma J, Liu X, Qiao L, Meng L, Xu X, Xue F, Cheng C, Han Z, Lu Y, Zhang W, Bu P, Zhang M, An G, Lu H, Ni M, Zhang C, Zhang Y. Association Between Stent Implantation and Progression of Nontarget Lesions in a Rabbit Model of Atherosclerosis. Circ Cardiovasc Interv 2021; 14:e010764. [PMID: 34674554 DOI: 10.1161/circinterventions.121.010764] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Progression of nontarget lesions (NTLs) after percutaneous coronary intervention (PCI) has been reported. However, it remains unknown whether progression of NTLs was causally related to stenting. This study was undertaken to test the hypothesis that stent implantation triggers acute phase response and systemic inflammation which may be associated with progression of NTLs. METHODS Thirty New Zealand rabbits receiving endothelial denudation and atherogenic diet were randomly divided into stenting, sham, and control groups. Angiography and intravascular ultrasonography were performed in the stenting and sham groups, and stent implantation performed only in the stenting group. Histopathologic study was conducted and serum levels of APPs (acute phase proteins) measured in all rabbits. Proteomics analysis was performed to screen the potential proteins related to NTLs progression after stent implantation. The serum levels of APPs and inflammatory cytokines were measured in 147 patients undergoing coronary angiography or PCI. RESULTS Plaque burden in the NTLs was significantly increased 12 weeks after stent implantation in the stenting group versus sham group. Serum levels of APPs and their protein expression in NTLs were significantly increased and responsible for stenting-triggered inflammation. In patients receiving PCI, serum levels of SAA-1 (serum amyloid A protein 1), CRP (C-reactive protein), TNF (tumor necrosis factor)-α, and IL (interleukin)-6 were substantially elevated up to 1 month post-PCI. CONCLUSIONS In a rabbit model of atherosclerosis, stent implantation triggered acute phase response and systemic inflammation, which was associated with increased plaque burden and pathological features of unstable plaque in NTLs. The potential mechanism involved vessel injury-triggered acute phase response manifested as increased serum levels of SAA-1, CRP, and LBP (lipopolysaccharide-binding protein) and their protein expression in NTLs. These findings provided a new insight into the relation between stent implantation and progression of NTLs, and further studies are warranted to clarify the detailed mechanism and clinical significance of these preliminary results. Registration: URL: http://www.chictr.org.cn; Unique identifier: ChiCTR1900026393. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Jing Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linlin Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xingli Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Xue
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziqi Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wencheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guipeng An
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
34
|
Chen Y, Su J, Yan Y, Zhao Q, Ma J, Zhu M, He X, Zhang B, Xu H, Yang X, Duan Y, Han J. Intermittent Fasting Inhibits High-Fat Diet-Induced Atherosclerosis by Ameliorating Hypercholesterolemia and Reducing Monocyte Chemoattraction. Front Pharmacol 2021; 12:719750. [PMID: 34658858 PMCID: PMC8517704 DOI: 10.3389/fphar.2021.719750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/16/2021] [Indexed: 01/29/2023] Open
Abstract
Atherosclerosis is a major pathology for cardiovascular diseases (CVDs). Clinically, the intermittent fasting (IF) has been observed to reduce the risk of CVDs. However, the effect of IF on the development of atherosclerosis has not been fully elucidated. Herein, we determined the protection of IF against high-fat diet–induced atherosclerosis in pro-atherogenic low-density lipoprotein receptor deficient (LDLR-/-) mice and the potentially involved mechanisms. The LDLR-/- mice were scheduled intermittent fasting cycles of 3-day HFD feeding ad libitum and 1 day fasting, while the mice in the control group were continuously fed HFD. The treatment was lasted for 7 weeks (∼12 cycles) or 14 weeks (∼24 cycles). Associated with the reduced total HFD intake, IF substantially reduced lesions in the en face aorta and aortic root sinus. It also increased plaque stability by increasing the smooth muscle cell (SMC)/collagen content and fibrotic cap thickness while reducing macrophage accumulation and necrotic core areas. Mechanistically, IF reduced serum total and LDL cholesterol levels by inhibiting cholesterol synthesis in the liver. Meanwhile, HFD-induced hepatic lipid accumulation was attenuated by IF. Interestingly, circulating Ly6Chigh monocytes but not T cells and serum c-c motif chemokine ligand 2 levels were significantly reduced by IF. Functionally, adhesion of monocytes to the aortic endothelium was decreased by IF via inhibiting VCAM-1 and ICAM-1 expression. Taken together, our study indicates that IF reduces atherosclerosis in LDLR-/- mice by reducing monocyte chemoattraction/adhesion and ameliorating hypercholesterolemia and suggests its potential application for atherosclerosis treatment.
Collapse
Affiliation(s)
- Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Jiamin Su
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Yali Yan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Qian Zhao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Jialing Ma
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Mengmeng Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Xiaoyu He
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Hongmei Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| |
Collapse
|
35
|
Citrate Synthase and OGDH as Potential Biomarkers of Atherosclerosis under Chronic Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9957908. [PMID: 34539976 PMCID: PMC8445721 DOI: 10.1155/2021/9957908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/20/2021] [Accepted: 08/10/2021] [Indexed: 11/17/2022]
Abstract
Background Pathological changes of the adrenal gland and the possible underlying molecular mechanisms are currently unclear in the case of atherosclerosis (AS) combined with chronic stress (CS). Methods New Zealand white rabbits were used to construct a CS and AS animal model. Proteomics and bioinformatics were employed to identify hub proteins in the adrenal gland related to CS and AS. Hub proteins were detected using immunohistochemistry, immunofluorescence assays, and Western blotting. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to analyze the expression of genes. In addition, a neural network model was constructed. The quantitative relationships were inferred by cubic spline interpolation. Enzymatic activity of mitochondrial citrate synthase and OGDH was detected by the enzymatic assay kit. Function of citrate synthase and OGDH with knockdown experiments in the adrenal cell lines was performed. Furthermore, target genes-TF-miRNA regulatory network was constructed. Coimmunoprecipitation (IP) assay and molecular docking study were used to detect the interaction between citrate synthase and OGDH. Results Two most significant hub proteins (citrate synthase and OGDH) that were related to CS and AS were identified in the adrenal gland using numerous bioinformatic methods. The hub proteins were mainly enriched in mitochondrial proton transport ATP synthase complex, ATPase activation, and the AMPK signaling pathway. Compared with the control group, the adrenal glands were larger and more disordered, irregular, and necrotic in the AS+CS group. The expression of citrate synthase and OGDH was higher in the AS+CS group than in the control group, both at the protein and mRNA levels (P < 0.05). There were strong correlations among the cross-sectional areas of adrenal glands, citrate synthase, and OGDH (P < 0.05) via Spearman's rho analysis, receiver operating characteristic curves, a neural network model, and cubic spline interpolation. Enzymatic activity of citrate synthase and OGDH increased under the situation of atherosclerosis and chronic stress. Through the CCK8 assay, the adrenal cell viability was downregulated significantly after the knockdown experiment of citrate synthase and OGDH. Target genes-TF-miRNA regulatory network presented the close interrelations among the predicted microRNA, citrate synthase and OGDH. After Coimmunoprecipitation (IP) assay, the result manifested that the citrate synthase and OGDH were coexpressed in the adrenal gland. The molecular docking study showed that the docking score of optimal complex conformation between citrate synthase and OGDH was -6.15 kcal/mol. Conclusion AS combined with CS plays a significant role on the hypothalamic–pituitary–adrenal (HPA) axis, promotes adrenomegaly, increases the release of glucocorticoid (GC), and might enhance ATP synthesis and energy metabolism in the body through citrate synthase and OGDH gene targets, providing a potential research direction for future related explorations into this mechanism.
Collapse
|
36
|
Yu J, Li W, Xiao X, Huang Q, Yu J, Yang Y, Han T, Zhang D, Niu X. (-)-Epicatechin gallate blocks the development of atherosclerosis by regulating oxidative stress in vivo and in vitro. Food Funct 2021; 12:8715-8727. [PMID: 34365492 DOI: 10.1039/d1fo00846c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
(-)-Epicatechin gallate (ECG), as a compound in green tea extract polyphenols, has specific therapeutic effects against oxidative stress. Oxidative stress exists throughout the pathological development of atherosclerosis. In this study, two atherosclerosis models, oxidized low-density lipoprotein (ox-LDL)-induced vascular smooth muscle cells (VSMCs) and high fat diet (HFD)-induced ApoE-/- mice, were applied to explore the mechanism of ECG intervention on AS. In vivo and in vitro studies showed that ECG reduced the level of MDA and increased the activity of SOD, which are oxidative stress factors. ECG also improved HFD-induced disorder of lipid factor expression in the serum of ApoE-/- mice and alleviated oxidative stress by enhancing the antioxidant activity. The potential mechanism was supposed to be the inhibition of the phosphorylation of p65 by ECG in the NF-κB pathway in the aorta, thereby blocking the expression of inflammatory mediators. In addition, ECG increased the stability of atherosclerosis plaques by reducing the expression of MMP-2 and ICAM-1 in atherosclerosis diseased tissues. ECG reduced lipid accumulation in the aorta and its roots and also plaque neoplasia. Western blotting experiments indicated that ECG increased the nuclear transfer of Nrf2 and the expression of heme oxygenase 1 (HO-1) was increased. These results demonstrated that ECG significantly reduced the formation of aortic plaque in ApoE-/- mice which was possibly triggered by the inhibition of hyperlipidemia and oxidative stress that exhibited the anti-atherosclerotic potential.
Collapse
Affiliation(s)
- Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
37
|
Liu Y, Zhao Y, Shukha Y, Lu H, Wang L, Liu Z, Liu C, Zhao Y, Wang H, Zhao G, Liang W, Fan Y, Chang L, Yurdagul A, Pattillo CB, Orr AW, Aviram M, Wen B, Garcia-Barrio MT, Zhang J, Liu W, Sun D, Hayek T, Chen YE, Rom O. Dysregulated oxalate metabolism is a driver and therapeutic target in atherosclerosis. Cell Rep 2021; 36:109420. [PMID: 34320345 PMCID: PMC8363062 DOI: 10.1016/j.celrep.2021.109420] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/16/2021] [Accepted: 06/28/2021] [Indexed: 02/01/2023] Open
Abstract
Dysregulated glycine metabolism is emerging as a common denominator in cardiometabolic diseases, but its contribution to atherosclerosis remains unclear. In this study, we demonstrate impaired glycine-oxalate metabolism through alanine-glyoxylate aminotransferase (AGXT) in atherosclerosis. As found in patients with atherosclerosis, the glycine/oxalate ratio is decreased in atherosclerotic mice concomitant with suppression of AGXT. Agxt deletion in apolipoprotein E-deficient (Apoe-/-) mice decreases the glycine/oxalate ratio and increases atherosclerosis with induction of hepatic pro-atherogenic pathways, predominantly cytokine/chemokine signaling and dysregulated redox homeostasis. Consistently, circulating and aortic C-C motif chemokine ligand 5 (CCL5) and superoxide in lesional macrophages are increased. Similar findings are observed following dietary oxalate overload in Apoe-/- mice. In macrophages, oxalate induces mitochondrial dysfunction and superoxide accumulation, leading to increased CCL5. Conversely, AGXT overexpression in Apoe-/- mice increases the glycine/oxalate ratio and decreases aortic superoxide, CCL5, and atherosclerosis. Our findings uncover dysregulated oxalate metabolism via suppressed AGXT as a driver and therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Yuhao Liu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Ying Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yousef Shukha
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel; The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Haocheng Lu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lu Wang
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Cai Liu
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yang Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Huilun Wang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guizhen Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenying Liang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanbo Fan
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Lin Chang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Michael Aviram
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Bo Wen
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Minerva T Garcia-Barrio
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Duxin Sun
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tony Hayek
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel; The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Y Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Oren Rom
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA.
| |
Collapse
|
38
|
Di Cataldo V, Debatisse J, Piraquive J, Géloën A, Grandin C, Verset M, Taborik F, Labaronne E, Loizon E, Millon A, Mury P, Pialoux V, Serusclat A, Lamberton F, Ibarrola D, Lavenne F, Le Bars D, Troalen T, Confais J, Crola Da Silva C, Mechtouff L, Contamin H, Fayad ZA, Canet-Soulas E. Cortical inflammation and brain signs of high-risk atherosclerosis in a non-human primate model. Brain Commun 2021; 3:fcab064. [PMID: 33937770 PMCID: PMC8063585 DOI: 10.1093/braincomms/fcab064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 11/14/2022] Open
Abstract
Atherosclerosis is a chronic systemic inflammatory disease, inducing cardiovascular and cerebrovascular acute events. A role of neuroinflammation is suspected, but not yet investigated in the gyrencephalic brain and the related activity at blood−brain interfaces is unknown. A non-human primate model of advanced atherosclerosis was first established using longitudinal blood samples, multimodal imaging and gene analysis in aged animals. Non-human primate carotid lesions were compared with human carotid endarterectomy samples. During the whole-body imaging session, imaging of neuroinflammation and choroid plexus function was performed. Advanced plaques were present in multiple sites, premature deaths occurred and downstream lesions (myocardial fibrosis, lacunar stroke) were present in this model. Vascular lesions were similar to in humans: high plaque activity on PET and MRI imaging and systemic inflammation (high plasma C-reactive protein levels: 42 ± 14 µg/ml). We also found the same gene association (metabolic, inflammatory and anti-inflammatory markers) as in patients with similar histological features. Metabolic imaging localized abnormal brain glucose metabolism in the frontal cortex. It corresponded to cortical neuro-inflammation (PET imaging) that correlated with C-reactive protein level. Multimodal imaging also revealed pronounced choroid plexus function impairment in aging atherosclerotic non-human primates. In conclusion, multimodal whole-body inflammation exploration at the vascular level and blood−brain interfaces identified high-risk aging atherosclerosis. These results open the way for systemic and central inflammation targeting in atherosclerosis in the new era of immunotherapy.
Collapse
Affiliation(s)
- Vanessa Di Cataldo
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Justine Debatisse
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France.,Siemens-Healthcare SAS, Saint-Denis, France
| | | | - Alain Géloën
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | - Emmanuel Labaronne
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Emmanuelle Loizon
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Antoine Millon
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Pauline Mury
- LIBM Laboratory, Univ Lyon, Université Lyon 1, Lyon, France
| | | | - André Serusclat
- Radiology Department, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | | | | | | | | | | | | | - Claire Crola Da Silva
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Laura Mechtouff
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France.,Stroke Department, Hospices Civils de Lyon, Lyon, France
| | | | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
39
|
Wu Q, Sun L, Hu X, Wang X, Xu F, Chen B, Liang X, Xia J, Wang P, Aibara D, Zhang S, Zeng G, Yun C, Yan Y, Zhu Y, Bustin M, Zhang S, Gonzalez FJ, Jiang C. Suppressing the intestinal farnesoid X receptor/sphingomyelin phosphodiesterase 3 axis decreases atherosclerosis. J Clin Invest 2021; 131:142865. [PMID: 33938457 DOI: 10.1172/jci142865] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Intestinal farnesoid X receptor (FXR) signaling is involved in the development of obesity, fatty liver disease, and type 2 diabetes. However, the role of intestinal FXR in atherosclerosis and its potential as a target for clinical treatment have not been explored. The serum levels of fibroblast growth factor 19 (FGF19), which is encoded by an FXR target gene, were much higher in patients with hypercholesterolemia than in control subjects and were positively related to circulating ceramide levels, indicating a link between intestinal FXR, ceramide metabolism, and atherosclerosis. Among ApoE-/- mice fed a high-cholesterol diet (HCD), intestinal FXR deficiency (in FxrΔIE ApoE-/- mice) or direct FXR inhibition (via treatment with the FXR antagonist glycoursodeoxycholic acid [GUDCA]) decreased atherosclerosis and reduced the levels of circulating ceramides and cholesterol. Sphingomyelin phosphodiesterase 3 (SMPD3), which is involved in ceramide synthesis in the intestine, was identified as an FXR target gene. SMPD3 overexpression or C16:0 ceramide supplementation eliminated the improvements in atherosclerosis in FxrΔIE ApoE-/- mice. Administration of GUDCA or GW4869, an SMPD3 inhibitor, elicited therapeutic effects on established atherosclerosis in ApoE-/- mice by decreasing circulating ceramide levels. This study identified an intestinal FXR/SMPD3 axis that is a potential target for atherosclerosis therapy.
Collapse
Affiliation(s)
- Qing Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Xiaomin Hu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Feng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bo Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xianyi Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jialin Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Pengcheng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Daisuke Aibara
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Shaofei Zhang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Guangyi Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chuyu Yun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yu Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yicheng Zhu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Michael Bustin
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Shuyang Zhang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.,Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
40
|
Ngo ATP, Jordan KR, Mueller PA, Hagen MW, Reitsma SE, Puy C, Revenko AS, Lorentz CU, Tucker EI, Cheng Q, Hinds MT, Fazio S, Monia BP, Gailani D, Gruber A, Tavori H, McCarty OJT. Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice. J Thromb Haemost 2021; 19:1001-1017. [PMID: 33421301 PMCID: PMC8549080 DOI: 10.1111/jth.15236] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Human coagulation factor (F) XI deficiency, a defect of the contact activation system, protects against venous thrombosis, stroke, and heart attack, whereas FXII, plasma prekallikrein, or kininogen deficiencies are asymptomatic. FXI deficiency, inhibition of FXI production, activated FXI (FXIa) inhibitors, and antibodies to FXI that interfere with FXI/FXII interactions reduce experimental thrombosis and inflammation. FXI inhibitors are antithrombotic in patients, and FXI and FXII deficiencies are atheroprotective in apolipoprotein E-deficient mice. OBJECTIVES Investigate the effects of pharmacological targeting of FXI in experimental models of atherogenesis and established atherosclerosis. METHODS AND RESULTS Low-density lipoprotein receptor-knockout (Ldlr-/- ) mice were administered high-fat diet (HFD) for 8 weeks; concomitantly, FXI was targeted with anti-FXI antibody (14E11) or FXI antisense oligonucleotide (ASO). 14E11 and FXI-ASO reduced atherosclerotic lesion area in proximal aortas when compared with controls, and 14E11 also reduced aortic sinus lesions. In an established disease model, in which therapy was given after atherosclerosis had developed, Ldlr-/- mice were fed HFD for 8 weeks and then administered 14E11 or FXI-ASO weekly until 16 weeks on HFD. In this established disease model, 14E11 and FXI-ASO reduced atherosclerotic lesion area in proximal aortas, but not in aortic sinus. In cultures of human endothelium, FXIa exposure disrupted VE-Cadherin expression and increased endothelial lipoprotein permeability. Strikingly, we found that 14E11 prevented the disruption of VE-Cadherin expression in aortic sinus lesions observed in the atherogenesis mouse model. CONCLUSION Pharmacological targeting of FXI reduced atherogenesis in Ldlr-/- mice. Interference with the contact activation system may safely reduce development or progression of atherosclerosis.
Collapse
Affiliation(s)
- Anh T. P. Ngo
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Kelley R. Jordan
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Paul A. Mueller
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Matthew W. Hagen
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Stéphanie E. Reitsma
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | | | - Christina U. Lorentz
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Aronora Inc, Portland, OR, USA
| | - Erik I. Tucker
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Aronora Inc, Portland, OR, USA
| | - Quifang Cheng
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Monica T. Hinds
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Sergio Fazio
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | | | - David Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - András Gruber
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Aronora Inc, Portland, OR, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
41
|
Bi X, Du C, Wang X, Wang X, Han W, Wang Y, Qiao Y, Zhu Y, Ran L, Liu Y, Xiong J, Huang Y, Liu M, Liu C, Zeng C, Wang J, Yang K, Zhao J. Mitochondrial Damage-Induced Innate Immune Activation in Vascular Smooth Muscle Cells Promotes Chronic Kidney Disease-Associated Plaque Vulnerability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002738. [PMID: 33717842 PMCID: PMC7927614 DOI: 10.1002/advs.202002738] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/06/2020] [Indexed: 05/02/2023]
Abstract
Chronic kidney disease (CKD) is associated with accelerated atherosclerosis progression and high incidence of cardiovascular events, hinting that atherosclerotic plaques in CKD may be vulnerable. However, its cause and mechanism remain obscure. Here, it is shown that apolipoprotein E-deficient (ApoE-/-) mouse with CKD (CKD/ApoE-/- mouse) is a useful model for investigating the pathogenesis of plaque vulnerability, and premature senescence and phenotypic switching of vascular smooth muscle cells (VSMCs) contributes to CKD-associated plaque vulnerability. Subsequently, VSMC phenotypes in patients with CKD and CKD/ApoE-/- mice are comprehensively investigated. Using multi-omics analysis and targeted and VSMC-specific gene knockout mice, VSMCs are identified as both type-I-interferon (IFN-I)-responsive and IFN-I-productive cells. Mechanistically, mitochondrial damage resulting from CKD-induced oxidative stress primes the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway to trigger IFN-I response in VSMCs. Enhanced IFN-I response then induces VSMC premature senescence and phenotypic switching in an autocrine/paracrine manner, resulting in the loss of fibrous cap VSMCs and fibrous cap thinning. Conversely, blocking IFN-I response remarkably attenuates CKD-associated plaque vulnerability. These findings reveal that IFN-I response in VSMCs through immune sensing of mitochondrial damage is essential for the pathogenesis of CKD-associated plaque vulnerability. Mitigating IFN-I response may hold promise for the treatment of CKD-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Xianjin Bi
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Changhong Du
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xinmiao Wang
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xue‐Yue Wang
- Laboratory of Stem Cell & Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic Medical SciencesArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Wenhao Han
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yue Wang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yu Qiao
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yingguo Zhu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Li Ran
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yong Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jiachuan Xiong
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yinghui Huang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Mingying Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Chi Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Chunyu Zeng
- Department of CardiologyDaping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Junping Wang
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Ke Yang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jinghong Zhao
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| |
Collapse
|
42
|
Ikeda J, Scipione CA, Hyduk SJ, Althagafi MG, Atif J, Dick SA, Rajora M, Jang E, Emoto T, Murakami J, Ikeda N, Ibrahim HM, Polenz CK, Gao X, Tai K, Jongstra-Bilen J, Nakashima R, Epelman S, Robbins CS, Zheng G, Lee WL, MacParland SA, Cybulsky MI. Radiation Impacts Early Atherosclerosis by Suppressing Intimal LDL Accumulation. Circ Res 2021; 128:530-543. [PMID: 33397122 DOI: 10.1161/circresaha.119.316539] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
RATIONALE Bone marrow transplantation (BMT) is used frequently to study the role of hematopoietic cells in atherosclerosis, but aortic arch lesions are smaller in mice after BMT. OBJECTIVE To identify the earliest stage of atherosclerosis inhibited by BMT and elucidate potential mechanisms. METHODS AND RESULTS Ldlr-/- mice underwent total body γ-irradiation, bone marrow reconstitution, and 6-week recovery. Atherosclerosis was studied in the ascending aortic arch and compared with mice without BMT. In BMT mice, neutral lipid and myeloid cell topography were lower in lesions after feeding a cholesterol-rich diet for 3, 6, and 12 weeks. Lesion coalescence and height were suppressed dramatically in mice post-BMT, whereas lateral growth was inhibited minimally. Targeted radiation to the upper thorax alone reproduced the BMT phenotype. Classical monocyte recruitment, intimal myeloid cell proliferation, and apoptosis did not account for the post-BMT phenotype. Neutral lipid accumulation was reduced in 5-day lesions, thus we developed quantitative assays for LDL (low-density lipoprotein) accumulation and paracellular leakage using DiI-labeled human LDL and rhodamine B-labeled 70 kD dextran. LDL accumulation was dramatically higher in the intima of Ldlr-/- relative to Ldlr+/+ mice, and was inhibited by injection of HDL mimics, suggesting a regulated process. LDL, but not dextran, accumulation was lower in mice post-BMT both at baseline and in 5-day lesions. Since the transcript abundance of molecules implicated in LDL transcytosis was not significantly different in the post-BMT intima, transcriptomics from whole aortic arch intima, and at single-cell resolution, was performed to give insights into pathways modulated by BMT. CONCLUSIONS Radiation exposure inhibits LDL entry into the aortic intima at baseline and the earliest stages of atherosclerosis. Single-cell transcriptomic analysis suggests that LDL uptake by endothelial cells is diverted to lysosomal degradation and reverse cholesterol transport pathways. This reduces intimal accumulation of lipid and impacts lesion initiation and growth.
Collapse
Affiliation(s)
- Jiro Ikeda
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto
| | - Corey A Scipione
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada
| | - Marwan G Althagafi
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto
| | - Jawairia Atif
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Ajmera Family Transplant Centre, Toronto General Hospital Research Institute (J.A., S.A.M.), University Health Network, Toronto, Canada.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Sarah A Dick
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Ajmera Family Transplant Centre, Toronto General Hospital Research Institute (J.A., S.A.M.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program (S.A.D., S.E.)
| | - Maneesha Rajora
- Princess Margaret Cancer Centre (M.R., R.N., G.Z.), University Health Network, Toronto, Canada
| | - Erika Jang
- Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Keenan Research Centre, Unity Health (E.J., W.L.L.)
| | - Takuo Emoto
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada
| | - Junichi Murakami
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Latner Thoracic Surgery Research Laboratories (J.M.), University Health Network, Toronto, Canada
| | - Noriko Ikeda
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada
| | - Hisham M Ibrahim
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto
| | - Chanele K Polenz
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto
| | - Xiaotang Gao
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada
| | - Kelly Tai
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Jenny Jongstra-Bilen
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Ryota Nakashima
- Princess Margaret Cancer Centre (M.R., R.N., G.Z.), University Health Network, Toronto, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Peter Munk Cardiac Centre (S.E., C.S.R., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program (S.A.D., S.E.)
| | - Clinton S Robbins
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Peter Munk Cardiac Centre (S.E., C.S.R., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Gang Zheng
- Princess Margaret Cancer Centre (M.R., R.N., G.Z.), University Health Network, Toronto, Canada
| | - Warren L Lee
- Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Medicine (W.L.L.), University of Toronto.,Keenan Research Centre, Unity Health (E.J., W.L.L.)
| | - Sonya A MacParland
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute (J.I., C.A.S., S.J.H., M.G.A., J.A., S.A.D., T.E., J.M., N.I., H.M.I., C.K.P., X.G., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University Health Network, Toronto, Canada.,Peter Munk Cardiac Centre (S.E., C.S.R., M.I.C.), University Health Network, Toronto, Canada.,Laboratory Medicine and Pathobiology (J.I., C.A.S., M.G.A., E.J., H.M.I., C.K.P., J.J.-B., S.E., C.S.R., W.L.L., S.A.M., M.I.C.), University of Toronto.,Immunology (C.A.S., J.A., S.A.D., K.T., J.J.-B., S.E., C.S.R., S.A.M., M.I.C.), University of Toronto
| |
Collapse
|
43
|
Zeng P, Yang J, Liu L, Yang X, Yao Z, Ma C, Zhu H, Su J, Zhao Q, Feng K, Yang S, Zhu Y, Li X, Wang W, Duan Y, Han J, Chen Y. ERK1/2 inhibition reduces vascular calcification by activating miR-126-3p-DKK1/LRP6 pathway. Am J Cancer Res 2021; 11:1129-1146. [PMID: 33391525 PMCID: PMC7738895 DOI: 10.7150/thno.49771] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Vascular microcalcification increases the risk of rupture of vulnerable atherosclerotic lesions. Inhibition of ERK1/2 reduces atherosclerosis in animal models while its role in vascular calcification and the underlying mechanisms remains incompletely understood. Methods: Levels of activated ERK1/2, DKK1, LRP6 and BMP2 in human calcific aortic valves were determined. ApoE deficient mice received ERK1/2 inhibitor (U0126) treatment, followed by determination of atherosclerosis, calcification and miR-126-3p production. C57BL/6J mice were used to determine the effect of U0126 on Vitamin D3 (VD3)-induced medial arterial calcification. HUVECs, HAECs and HASMCs were used to determine the effects of ERK1/2 inhibitor or siRNA on SMC calcification and the involved mechanisms. Results: We observed the calcification in human aortic valves was positively correlated to ERK1/2 activity. At cellular and animal levels, U0126 reduced intimal calcification in atherosclerotic lesions of high-fat diet-fed apoE deficient mice, medial arterial calcification in VD3-treated C57BL/6J mice, and calcification in cultured SMCs and arterial rings. The reduction of calcification was attributed to ERK1/2 inhibition-reduced expression of ALP, BMP2 and RUNX2 by activating DKK1 and LRP6 expression, and consequently inactivating both canonical and non-canonical Wnt signaling pathways in SMCs. Furthermore, we determined ERK1/2 inhibition activated miR-126-3p production by facilitating its maturation through activation of AMPKα-mediated p53 phosphorylation, and the activated miR-126-3p from ECs and SMCs played a key role in anti-vascular calcification actions of ERK1/2 inhibition. Conclusions: Our study demonstrates that activation of miR-126-3p production in ECs/SMCs and interactions between ECs and SMCs play an important role in reduction of vascular calcification by ERK1/2 inhibition.
Collapse
|
44
|
Jurtz VI, Skovbjerg G, Salinas CG, Roostalu U, Pedersen L, Hecksher-Sørensen J, Rolin B, Nyberg M, van de Bunt M, Ingvorsen C. Deep learning reveals 3D atherosclerotic plaque distribution and composition. Sci Rep 2020; 10:21523. [PMID: 33299076 PMCID: PMC7726562 DOI: 10.1038/s41598-020-78632-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Complications of atherosclerosis are the leading cause of morbidity and mortality worldwide. Various genetically modified mouse models are used to investigate disease trajectory with classical histology, currently the preferred methodology to elucidate plaque composition. Here, we show the strength of light-sheet fluorescence microscopy combined with deep learning image analysis for characterising and quantifying plaque burden and composition in whole aorta specimens. 3D imaging is a non-destructive method that requires minimal ex vivo handling and can be up-scaled to large sample sizes. Combined with deep learning, atherosclerotic plaque in mice can be identified without any ex vivo staining due to the autofluorescent nature of the tissue. The aorta and its branches can subsequently be segmented to determine how anatomical position affects plaque composition and progression. Here, we find the highest plaque accumulation in the aortic arch and brachiocephalic artery. Simultaneously, aortas can be stained for markers of interest (for example the pan immune cell marker CD45) and quantified. In ApoE-/- mice we observe that levels of CD45 reach a plateau after which increases in plaque volume no longer correlate to immune cell infiltration. All underlying code is made publicly available to ease adaption of the method.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Aortic Diseases
- Apolipoproteins E/analysis
- Atherosclerosis/complications
- Atherosclerosis/pathology
- Deep Learning
- Disease Models, Animal
- Female
- Image Processing, Computer-Assisted/methods
- Imaging, Three-Dimensional/methods
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence/methods
- Plaque, Atherosclerotic/diagnostic imaging
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Receptors, LDL/analysis
Collapse
Affiliation(s)
| | - Grethe Skovbjerg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | | | | | - Louise Pedersen
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- University of Copenhagen, 1017, Copenhagen, Denmark
| | | | - Bidda Rolin
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | - Michael Nyberg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
| | | | | |
Collapse
|
45
|
Golforoush P, Yellon DM, Davidson SM. Mouse models of atherosclerosis and their suitability for the study of myocardial infarction. Basic Res Cardiol 2020; 115:73. [PMID: 33258000 PMCID: PMC7704510 DOI: 10.1007/s00395-020-00829-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Atherosclerotic plaques impair vascular function and can lead to arterial obstruction and tissue ischaemia. Rupture of an atherosclerotic plaque within a coronary artery can result in an acute myocardial infarction, which is responsible for significant morbidity and mortality worldwide. Prompt reperfusion can salvage some of the ischaemic territory, but ischaemia and reperfusion (IR) still causes substantial injury and is, therefore, a therapeutic target for further infarct limitation. Numerous cardioprotective strategies have been identified that can limit IR injury in animal models, but none have yet been translated effectively to patients. This disconnect prompts an urgent re-examination of the experimental models used to study IR. Since coronary atherosclerosis is the most prevalent morbidity in this patient population, and impairs coronary vessel function, it is potentially a major confounder in cardioprotective studies. Surprisingly, most studies suggest that atherosclerosis does not have a major impact on cardioprotection in mouse models. However, a major limitation of atherosclerotic animal models is that the plaques usually manifest in the aorta and proximal great vessels, and rarely in the coronary vessels. In this review, we examine the commonly used mouse models of atherosclerosis and their effect on coronary artery function and infarct size. We conclude that none of the commonly used strains of mice are ideal for this purpose; however, more recently developed mouse models of atherosclerosis fulfil the requirement for coronary artery lesions, plaque rupture and lipoprotein patterns resembling the human profile, and may enable the identification of therapeutic interventions more applicable in the clinical setting.
Collapse
MESH Headings
- Animals
- Aortic Diseases/complications
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/complications
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Coronary Artery Disease/complications
- Coronary Artery Disease/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Diet, High-Fat
- Disease Models, Animal
- Genetic Predisposition to Disease
- Mice, Knockout, ApoE
- Myocardial Infarction/etiology
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardium/pathology
- Phenotype
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Rupture, Spontaneous
- Scavenger Receptors, Class B/deficiency
- Scavenger Receptors, Class B/genetics
- Species Specificity
Collapse
Affiliation(s)
- Pelin Golforoush
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
46
|
Rakipovski G, Rolin B, Barascuk N, Lund HE, Bjørn Bonde MF, Djordjevic D, Wulff-Larsen PG, Petersen M, Kirk RK, Hultman K, Manfe V, Blume N, Zahn S, Lengquist M, Maegdefessel L, Hovingh GK, Conde-Knape K, Hedin U, Matic L, Nyberg M. A neutralizing antibody against DKK1 does not reduce plaque formation in classical murine models of atherosclerosis: Is the therapeutic potential lost in translation? Atherosclerosis 2020; 314:1-9. [PMID: 33129080 DOI: 10.1016/j.atherosclerosis.2020.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/10/2020] [Accepted: 10/02/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS Clinical interventions targeting nonlipid risk factors are needed given the high residual risk of atherothrombotic events despite effective control of dyslipidemia. Dickkopf-1 (DKK1) plays a lipid-independent role in vascular pathophysiology but its involvement in atherosclerosis development and its therapeutic attractiveness remain to be established. METHODS Patient data, in vitro studies and pharmacological intervention in murine models of atherosclerosis were utilized. RESULTS In patients' material (n = 127 late stage plaque specimens and n = 10 control vessels), DKK1 mRNA was found to be higher in atherosclerotic plaques versus control arteries. DKK1 protein was detected in the luminal intimal area and in the necrotic core of plaques. DKK1 was released from isolated primary human platelets (~12 - 21-fold) and endothelial cells (~1.4-2.5-fold) upon stimulation with different pathophysiological stimuli. In ApoE-/- and Ldlr-/- mice, plasma DKK1 concentrations were similar to those observed in humans, whereas DKK1 expression in different atheroprone arterial segments was very low/absent. Chronic treatment with a neutralizing DKK1 antibody effectively reduced plasma concentrations, however, plaque lesion area was not reduced in ApoE-/- and Ldlr-/- mice fed a western diet for 14 and 16 weeks. Anti-DKK1 treatment increased bone volume and bone mineral content. CONCLUSIONS Functional inhibition of DKK1 with an antibody does not alter atherosclerosis progression in classical murine models. This may reflect the absence of DKK1 expression in plaques and more advanced animal disease models could be needed to evaluate the role and therapeutic attractiveness of DKK1 in late stage complications such as plaque destabilization, calcification, rupture and thrombosis.
Collapse
Affiliation(s)
| | - Bidda Rolin
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | | | | | | | | | | | - Maj Petersen
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | | | - Karin Hultman
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Valentina Manfe
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Niels Blume
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Stefan Zahn
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden; Technical University of Munich, Klinikum Rechts der Isar, Department for Vascular and Endovascular Surgery, Munich, Germany
| | - G Kees Hovingh
- Chief Medical Office, Novo Nordisk A/S, Soeborg, Denmark; Department of Vascular Medicine, Academisch Medisch Centrum, Amsterdam, Netherlands
| | | | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Michael Nyberg
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark.
| |
Collapse
|
47
|
Ma R, Qi Y, Zhao X, Li X, Sun X, Niu P, Li Y, Guo C, Chen R, Sun Z. Amorphous silica nanoparticles accelerated atherosclerotic lesion progression in ApoE -/- mice through endoplasmic reticulum stress-mediated CD36 up-regulation in macrophage. Part Fibre Toxicol 2020; 17:50. [PMID: 33008402 PMCID: PMC7531166 DOI: 10.1186/s12989-020-00380-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Background The biosafety concern of silica nanoparticles (SiNPs) is rapidly expanding alongside with its mass production and extensive applications. The cardiovascular effects of SiNPs exposure have been gradually confirmed, however, the interaction between SiNPs exposure and atherosclerosis, and the underlying mechanisms still remain unknown. Thereby, this study aimed to explore the effects of SiNPs on the progression of atherosclerosis, and to investigate related mechanisms. Results We firstly investigated the in vivo effects of SiNPs exposure on atherosclerosis via intratracheal instillation of ApoE−/− mice fed a Western diet. Ultrasound microscopy showed a significant increase of pulse wave velocity (PWV) compared to the control group, and the histopathological investigation reflected a greater plaque burden in the aortic root of SiNPs-exposed ApoE−/− mice. Compared to the control group, the serum levels of total triglycerides (TG) and low-density lipoprotein cholesterol (LDL-C) were elevated after SiNPs exposure. Moreover, intensified macrophage infiltration and endoplasmic reticulum (ER) stress was occurred in plaques after SiNPs exposure, as evidenced by the upregulated CD68 and CHOP expressions. Further in vitro, SiNPs was confirmed to activate ER stress and induce lipid accumulation in mouse macrophage, RAW264.7. Mechanistic analyses showed that 4-PBA (a classic ER stress inhibitor) pretreatment greatly alleviated SiNPs-induced macrophage lipid accumulation, and reversed the elevated CD36 expression induced by SiNPs. Conclusions Our results firstly revealed the acceleratory effect of SiNPs on the progression of atherosclerosis in ApoE−/− mice, which was related to lipid accumulation caused by ER stress-mediated upregulation of CD36 expression in macrophage. Graphical abstract ![]()
Collapse
Affiliation(s)
- Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yi Qi
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xinying Zhao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Xueyan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xuejing Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China. .,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Rui Chen
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
48
|
Vedder VL, Aherrahrou Z, Erdmann J. Dare to Compare. Development of Atherosclerotic Lesions in Human, Mouse, and Zebrafish. Front Cardiovasc Med 2020; 7:109. [PMID: 32714944 PMCID: PMC7344238 DOI: 10.3389/fcvm.2020.00109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases, such as atherosclerosis, are the leading cause of death worldwide. Although mice are currently the most commonly used model for atherosclerosis, zebrafish are emerging as an alternative, especially for inflammatory and lipid metabolism studies. Here, we review the history of in vivo atherosclerosis models and highlight the potential for future studies on inflammatory responses in lipid deposits in zebrafish, based on known immune reactions in humans and mice, in anticipation of new zebrafish models with more advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Viviana L Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
49
|
Zhao Y, Qu H, Wang Y, Xiao W, Zhang Y, Shi D. Small rodent models of atherosclerosis. Biomed Pharmacother 2020; 129:110426. [PMID: 32574973 DOI: 10.1016/j.biopha.2020.110426] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/08/2020] [Accepted: 06/13/2020] [Indexed: 12/30/2022] Open
Abstract
The ease of breeding, low cost of maintenance, and relatively short period for developing atherosclerosis make rodents ideal for atherosclerosis research. However, none of the current models accurately model human lipoprotein profile or atherosclerosis progression since each has its advantages and disadvantages. The advent of transgenic technologies much supports animal models' establishment. Notably, two classic transgenic mouse models, apoE-/- and Ldlr-/-, constitute the primary platforms for studying underlying mechanisms and development of pharmaceutical approaches. However, there exist crucial differences between mice and humans, such as the unhumanized lipoprotein profile, and the different plaque progression and characteristics. Among rodents, hamsters and guinea pigs might be the more realistic models in atherosclerosis research based on the similarities in lipoprotein metabolism to humans. Studies involving rat models, a rodent with natural resistance to atherosclerosis, have revealed evidence of atherosclerotic plaques under dietary induction and genetic manipulation by novel technologies, notably CRISPR-Cas9. Ldlr-/- hamster models were established in recent years with severe hyperlipidemia and atherosclerotic lesion formation, which could offer an alternative to classic transgenic mouse models. In this review, we provide an overview of classic and innovative small rodent models in atherosclerosis researches, including mice, rats, hamsters, and guinea pigs, focusing on their lipoprotein metabolism and histopathological changes.
Collapse
Affiliation(s)
- Yihan Zhao
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Hua Qu
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, Health Science Center, Peking University, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Wenli Xiao
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Dazhuo Shi
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
50
|
Di Gregoli K, Somerville M, Bianco R, Thomas AC, Frankow A, Newby AC, George SJ, Jackson CL, Johnson JL. Galectin-3 Identifies a Subset of Macrophages With a Potential Beneficial Role in Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:1491-1509. [PMID: 32295421 PMCID: PMC7253188 DOI: 10.1161/atvbaha.120.314252] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Galectin-3 (formerly known as Mac-2), encoded by the LGALS3 gene, is proposed to regulate macrophage adhesion, chemotaxis, and apoptosis. We investigated the role of galectin-3 in determining the inflammatory profile of macrophages and composition of atherosclerotic plaques. Approach and Results: We observed increased accumulation of galectin-3-negative macrophages within advanced human, rabbit, and mouse plaques compared with early lesions. Interestingly, statin treatment reduced galectin-3-negative macrophage accrual in advanced plaques within hypercholesterolemic (apolipoprotein E deficient) Apoe-/- mice. Accordingly, compared with Lgals3+/+:Apoe-/- mice, Lgals3-/-:Apoe-/- mice displayed altered plaque composition through increased macrophage:smooth muscle cell ratio, reduced collagen content, and increased necrotic core area, characteristics of advanced plaques in humans. Additionally, macrophages from Lgals3-/- mice exhibited increased invasive capacity in vitro and in vivo. Furthermore, loss of galectin-3 in vitro and in vivo was associated with increased expression of proinflammatory genes including MMP (matrix metalloproteinase)-12, CCL2 (chemokine [C-C motif] ligand 2), PTGS2 (prostaglandin-endoperoxide synthase 2), and IL (interleukin)-6, alongside reduced TGF (transforming growth factor)-β1 expression and consequent SMAD signaling. Moreover, we found that MMP12 cleaves macrophage cell-surface galectin-3 resulting in the appearance of a 22-kDa fragment, whereas plasma levels of galectin-3 were reduced in Mmp12-/-:Apoe-/- mice, highlighting a novel mechanism where MMP12-dependent cleavage of galectin-3 promotes proinflammatory macrophage polarization. Moreover, galectin-3-positive macrophages were more abundant within plaques of Mmp12-/-:Apoe-/- mice compared with Mmp12+/+:Apoe-/- animals. CONCLUSIONS This study reveals a prominent protective role for galectin-3 in regulating macrophage polarization and invasive capacity and, therefore, delaying plaque progression.
Collapse
Affiliation(s)
- Karina Di Gregoli
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Michelle Somerville
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Rosaria Bianco
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Anita C. Thomas
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Aleksandra Frankow
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Andrew C. Newby
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Sarah J. George
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Christopher L. Jackson
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Jason L. Johnson
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| |
Collapse
|