1
|
Zhen Y, Li D, Meng Y, Xing Z, Zheng J. Unveiling the roles of HIPK2 in atherosclerosis: Insights into the β-catenin/STAT1 signaling cascade and the involvement of SENP1. Biochem Pharmacol 2025; 237:116911. [PMID: 40164339 DOI: 10.1016/j.bcp.2025.116911] [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: 11/29/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Atherosclerosis is a disorder of lipid metabolism, but its pathogenesis has not yet been fully elucidated. This study aimed to clarify the roles of homeodomain interacting protein kinase 2 (HIPK2) in atherosclerosis. Atherosclerotic model was constructed by feeding apolipoprotein E knockout (ApoE-/-) mice with a high-fat diet. Human THP-1 macrophages and mouse RAW 264.7 macrophages were stimulated with IFN-γ to establish an in vitro model. We showed an upregulation of HIPK2 in the aorta of atherosclerotic mice. HIPK2 knockdown reduced macrophage infiltration, M1 polarization, and attenuates atherosclerosis development. Downregulation of HIPK2 in macrophages led to a significant suppression in the expression of pro-inflammatory factors, which was accompanied by an enhancement in the phosphorylation and degradation of β-catenin, as well as the activation of the signal transducer and activator of transcription 1 (STAT1) signaling pathway. Silencing of HIPK2 alone in THP-1 macrophages resulted in anti-inflammatory effects and suppression of M1 macrophage polarization. However, simultaneous silencing of HIPK2 and β-catenin (CTNNB1) reversed these effects, counteracting the outcomes observed with HIPK2 silencing alone. We validated that small ubiquitin-like modifier (SUMO)-specific peptidase 1 (SENP1) regulated HIPK2 function by affecting the SUMOylation of HIPK2 at the K32 site. SENP1 knockdown promoted HIPK2 SUMOylation, impairing its protein stability. In the rescue experiments, IFN-γ-induced inflammation and M1 polarization were resumed upon restoration of HIPK2 expression in SENP1-silenced macrophages. Our work demonstrated that HIPK2 accelerated the progression of atherosclerosis by regulating β-catenin/STAT1 signaling cascade to promote macrophage infiltration and M1 polarization. HIPK2 was regulated by SENP1-mediated de-SUMOylation.
Collapse
Affiliation(s)
- Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Dongdong Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yulu Meng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zeyu Xing
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Jiahe Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| |
Collapse
|
2
|
Yang G, Tan X, Zhai Q, Wang Y, Zhang X, Zhao P, Liang F, Lu J, Bao L. Plasma Lipidomics, Gut Microbiota Profile, and Phenotype of Adipose Tissue in an ApoE-/- Mouse Model of Plaque Instability. FRONT BIOSCI-LANDMRK 2025; 30:27236. [PMID: 40152393 DOI: 10.31083/fbl27236] [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: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND An appropriate animal model that can simulate the pathological process of atherosclerosis is urgently needed to improve treatment strategies. This study aimed to develop a new atherosclerosis model using ApoE-/- mice and to characterize lipidomics, gut microbiota profiles, and phenotypic alterations in adipose tissue using this model. METHODS After a 14- or 18-week high-fat diet (HFD), male ApoE-/- mice were randomly divided into four groups and treated separately with or without short-term and strong co-stimulation, including ice water bath and intraperitoneal injection of lipopolysaccharide and phenylephrine. As a control group, C57BL/6 mice were fed with conventional chow. The serum lipid levels, aortic arch pathology, adipose tissue phenotypic changes, plasma lipidomics, and 16S rDNA gene sequencing of colon feces were investigated. RESULTS The serum lipid levels were significantly lowered following extended HFD feeding for four weeks. However, co-stimulation increased serum interleukin (IL)-1β levels but did not affect serum lipid profiles. Co-stimulation revealed typical vulnerable atherosclerotic plaque characteristics and defective adipose hypertrophy associated with peroxisome proliferator-activated receptor γ (PPARγ) regulation in adipose tissue and a reduction in mitochondrial uncoupling protein 1 (UCP1) within brown adipose tissue. Plasma lipidomic analysis showed that sphingomyelin (SM), ceramide (Cer), and monohexosylceramide (HexCer) levels in plasma were significantly elevated by HFD feeding, whereas co-stimulation further elevated HexCer levels. Additionally, glycerophosphocholines (16:0/16:0, 18:2/20:4, 18:1/18:1) and HexCer (C12:1, C16:0), Cer (d18:1/16:0), and SM (C16:0) were the most sensitive to co-stimulation. Combined co-stimulation and HFD-fed increased the abundance of Firmicutes, the abundance of f_Erysipelotrichaceae, and the Firmicutes/Bacteroidota ratio but decreased the abundance of microflora promoting bile acid metabolism and short-chain fatty acids (SCFAs) in mouse feces. The results were consistent with the findings of epidemiologic atherosclerotic cardiovascular disease studies. CONCLUSIONS This study established an ApoE-/- mouse atherosclerotic vulnerable plaque model using a multi-index evaluation method. Adipogenic disorders, dysregulation of lipid metabolism at the molecular level, and increasing harmful gut microbiota are significant risk factors for vulnerable plaques, with sphingolipid metabolism receiving the most attention.
Collapse
Affiliation(s)
- Guanlin Yang
- Laboratory of Pharmacology, Zaozhuang Thoracic Hospital, 277500 Zaozhuang, Shandong, China
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xin Tan
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Qiong Zhai
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xuan Zhang
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Pengwei Zhao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Fangyuan Liang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Jingkun Lu
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - LiLi Bao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Wang F, Liu J, Hernandez R, Park SH, Lai YJ, Wang S, Blumberg B, Zhou C. Adipocyte-Derived PXR Signaling Is Dispensable for Diet-Induced Obesity and Metabolic Disorders in Mice. Drug Metab Dispos 2023; 51:1207-1215. [PMID: 37230767 PMCID: PMC10449100 DOI: 10.1124/dmd.123.001311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/21/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Pregnane X receptor (PXR) is a xenobiotic receptor that can be activated by numerous chemicals including endogenous hormones, dietary steroids, pharmaceutical agents, and environmental chemicals. PXR has been established to function as a xenobiotic sensor to coordinately regulate xenobiotic metabolism by regulating the expression of many enzymes and transporters required for xenobiotic metabolism. Recent studies have implicated a potentially important role for PXR in obesity and metabolic disease beyond xenobiotic metabolism, but how PXR action in different tissues or cell types contributes to obesity and metabolic disorders remains elusive. To investigate the role of adipocyte PXR in obesity, we generated a novel adipocyte-specific PXR deficient mouse model (PXRΔAd). Notably, we found that loss of adipocyte PXR did not affect food intake, energy expenditure, and obesity in high-fat diet-fed male mice. PXRΔAd mice also had similar obesity-associated metabolic disorders including insulin resistance and hepatic steatosis as control littermates. PXR deficiency in adipocytes did not affect expression of key adipose genes in PXRΔAd mice. Our findings suggest that adipocyte PXR signaling may be dispensable in diet-induced obesity and metabolic disorders in mice. Further studies are needed to understand the role of PXR signaling in obesity and metabolic disorders in the future. SIGNIFICANCE STATEMENT: The authors demonstrate that deficiency of adipocyte pregnane X receptor (PXR) does not affect diet-induced obesity or metabolic disorders in mice and infers that adipocyte PXR signaling may not play a key role in diet-induced obesity. More studies are needed to understand the tissue-specific role of PXR in obesity.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Jingwei Liu
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Rebecca Hernandez
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Ying-Jing Lai
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Bruce Blumberg
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| |
Collapse
|
4
|
An HJ, Gwon MG, Gu H, Bae S, Leem J, Lee JB, Park KK. STAT3/NF‑κB decoy oligodeoxynucleotides inhibit atherosclerosis through regulation of the STAT/NF‑κB signaling pathway in a mouse model of atherosclerosis. Int J Mol Med 2023; 51:37. [PMID: 37026512 PMCID: PMC10094942 DOI: 10.3892/ijmm.2023.5240] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/11/2023] [Indexed: 04/03/2023] Open
Abstract
Atherosclerosis is a progressive chronic inflammatory condition that is the cause of most cardiovascular and cerebrovascular diseases. The transcription factor nuclear factor‑κB (NF‑κB) regulates a number of genes involved in the inflammatory responses of cells that are critical to atherogenesis, and signal transducer and activator of transcription (STAT)3 is a key transcription factor in immunity and inflammation. Decoy oligodeoxynucleotides (ODNs) bind to sequence‑specific transcription factors and limit gene expression by interfering with transcription in vitro and in vivo. The present study aimed to investigate the beneficial functions of STAT3/NF‑κB decoy ODNs in liposaccharide (LPS)‑induced atherosclerosis in mice. Atherosclerotic injuries of mice were induced via intraperitoneal injection of LPS and the mice were fed an atherogenic diet. Ring‑type STAT3/NF‑κB decoy ODNs were designed and administered via an injection into the tail vein of the mice. To investigate the effect of STAT3/NF‑κB decoy ODNs, electrophoretic mobility shift assay, western blot analysis, histological analysis with hematoxylin and eosin staining, Verhoeff‑Van Gieson and Masson's trichrome staining were performed. The results revealed that STAT3/NF‑κB decoy ODNs were able to suppress the development of atherosclerosis by attenuating morphological changes and inflammation in atherosclerotic mice aortae, and by reducing pro‑inflammatory cytokine secretion through inhibition of the STAT3/NF‑κB pathway. In conclusion, the present study provided novel insights into the antiatherogenic molecular mechanism of STAT3/NF‑κB decoy ODNs, which may serve as an additional therapeutic intervention to combat atherosclerosis.
Collapse
Affiliation(s)
- Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Immunology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Seongjae Bae
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jin Bae Lee
- Department of Cardiology, Daegu Catholic University Medical Center, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|
5
|
Hernandez R, Shi J, Liu J, Li X, Wu J, Zhao L, Zhou T, Chen Q, Zhou C. PANDORA-Seq unveils the hidden small noncoding RNA landscape in atherosclerosis of LDL receptor-deficient mice. J Lipid Res 2023; 64:100352. [PMID: 36871792 PMCID: PMC10119612 DOI: 10.1016/j.jlr.2023.100352] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/08/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Small noncoding RNAs (sncRNAs) play diverse roles in numerous biological processes. While the widely used RNA sequencing (RNA-Seq) method has advanced sncRNA discovery, RNA modifications can interfere with the complementary DNA library construction process, preventing the discovery of highly modified sncRNAs including transfer RNA-derived small RNAs (tsRNAs) and ribosomal RNA-derived small RNAs (rsRNAs) that may have important functions in disease development. To address this technical obstacle, we recently developed a novel PANDORA-Seq (Panoramic RNA Display by Overcoming RNA Modification Aborted Sequencing) method to overcome RNA modification-elicited sequence interferences. To identify novel sncRNAs associated with atherosclerosis development, LDL receptor-deficient (LDLR-/-) mice were fed a low-cholesterol diet or high-cholesterol diet (HCD) for 9 weeks. Total RNAs isolated from the intima were subjected to PANDORA-Seq and traditional RNA-Seq. By overcoming RNA modification-elicited limitations, PANDORA-Seq unveiled an rsRNA/tsRNA-enriched sncRNA landscape in the atherosclerotic intima of LDLR-/- mice, which was strikingly different from that detected by traditional RNA-Seq. While microRNAs were the dominant sncRNAs detected by traditional RNA-Seq, PANDORA-Seq substantially increased the reads of rsRNAs and tsRNAs. PANDORA-Seq also detected 1,383 differentially expressed sncRNAs induced by HCD feeding, including 1,160 rsRNAs and 195 tsRNAs. One of HCD-induced intimal tsRNAs, tsRNA-Arg-CCG, may contribute to atherosclerosis development by regulating the proatherogenic gene expression in endothelial cells. Overall, PANDORA-Seq revealed a hidden rsRNA and tsRNA population associated with atherosclerosis development. These understudied tsRNAs and rsRNAs, which are much more abundant than microRNAs in the atherosclerotic intima of LDLR-/- mice, warrant further investigations.
Collapse
Affiliation(s)
- Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Junchao Shi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Xiuchun Li
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jake Wu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Linlin Zhao
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Molecular Medicine Program, Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.
| |
Collapse
|
6
|
Yuan D, Chu J, Qian J, Lin H, Zhu G, Chen F, Liu X. New Concepts on the Pathophysiology of Acute Coronary Syndrome. Rev Cardiovasc Med 2023; 24:112. [PMID: 39076267 PMCID: PMC11273028 DOI: 10.31083/j.rcm2404112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 07/31/2024] Open
Abstract
Acute coronary syndrome (ACS) is the most severe form of ischemic heart disease. Although it is caused by atherosclerotic plaque thrombosis or nonatherosclerotic causes, its pathophysiological mechanism of ACS is not fully understood, and its concept is constantly updated and developed. At present, the main pathophysiological mechanisms include plaque rupture, plaque erosion, calcified nodules (CN) and non-atherosclerotic causes such as coronary vasospasm and myocardial bridging (MB). These mechanisms may overlap and coexist in some ACS patients. Therefore, the pathophysiological mechanism of ACS is complex, and is of great significance for the diagnosis and treatment of ACS. This review will discuss the pathophysiological mechanisms of ACS to provide new thoughts on the pathogenesis, diagnosis and treatment of ACS.
Collapse
Affiliation(s)
- Deqiang Yuan
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jiapeng Chu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jun Qian
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Hao Lin
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Guoqi Zhu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Fei Chen
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| |
Collapse
|
7
|
Gan J, Guo L, Zhang X, Yu Q, Yang Q, Zhang Y, Zeng W, Jiang X, Guo M. Anti-inflammatory therapy of atherosclerosis: focusing on IKKβ. J Inflamm (Lond) 2023; 20:8. [PMID: 36823573 PMCID: PMC9951513 DOI: 10.1186/s12950-023-00330-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Chronic low-grade inflammation has been identified as a major contributor in the development of atherosclerosis. Nuclear Factor-κappa B (NF-κB) is a critical transcription factors family of the inflammatory pathway. As a major catalytic subunit of the IKK complex, IκB kinase β (IKKβ) drives canonical activation of NF-κB and is implicated in the link between inflammation and atherosclerosis, making it a promising therapeutic target. Various natural product derivatives, extracts, and synthetic, show anti-atherogenic potential by inhibiting IKKβ-mediated inflammation. This review focuses on the latest knowledge and current research landscape surrounding anti-atherosclerotic drugs that inhibit IKKβ. There will be more opportunities to fully understand the complex functions of IKKβ in atherogenesis and develop new effective therapies in the future.
Collapse
Affiliation(s)
- Jiali Gan
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Guo
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaolu Zhang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qun Yu
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuyue Yang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yilin Zhang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- grid.459559.10000 0004 9344 2915Oncology department, Ganzhou People’s Hospital, Ganzhou, Jiangxi China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
8
|
Yu Q, Wei P, Xu L, Xia C, Li Y, Liu H, Song X, Tian K, Fu W, Wang R, Wang W, Bai L, Fan J, Liu E, Zhao S. Urotensin II Enhances Advanced Aortic Atherosclerosis Formation and Delays Plaque Regression in Hyperlipidemic Rabbits. Int J Mol Sci 2023; 24:ijms24043819. [PMID: 36835230 PMCID: PMC9963243 DOI: 10.3390/ijms24043819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Accumulated evidence shows that elevated urotensin II (UII) levels are associated with cardiovascular diseases. However, the role of UII in the initiation, progression, and regression of atherosclerosis remains to be verified. Different stages of atherosclerosis were induced in rabbits by a 0.3% high cholesterol diet (HCD) feeding, and either UII (5.4 μg/kg/h) or saline was chronically infused via osmotic mini-pumps. UII promoted atherosclerotic fatty streak formation in ovariectomized female rabbits (34% increase in gross lesion and 93% increase in microscopic lesion), and in male rabbits (39% increase in gross lesion). UII infusion significantly increased the plaque size of the carotid and subclavian arteries (69% increase over the control). In addition, UII infusion significantly enhanced the development of coronary lesions by increasing plaque size and lumen stenosis. Histopathological analysis revealed that aortic lesions in the UII group were characterized by increasing lesional macrophages, lipid deposition, and intra-plaque neovessel formation. UII infusion also significantly delayed the regression of atherosclerosis in rabbits by increasing the intra-plaque macrophage ratio. Furthermore, UII treatment led to a significant increase in NOX2 and HIF-1α/VEGF-A expression accompanied by increased reactive oxygen species levels in cultured macrophages. Tubule formation assays showed that UII exerted a pro-angiogenic effect in cultured endothelial cell lines and this effect was partly inhibited by urantide, a UII receptor antagonist. These findings suggest that UII can accelerate aortic and coronary plaque formation and enhance aortic plaque vulnerability, but delay the regression of atherosclerosis. The role of UII on angiogenesis in the lesion may be involved in complex plaque development.
Collapse
Affiliation(s)
- Qingqing Yu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Panpan Wei
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Liran Xu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Congcong Xia
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yafeng Li
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xiaojie Song
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Kangli Tian
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Weilai Fu
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Rong Wang
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Weirong Wang
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Liang Bai
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Tokyo 409-3898, Japan
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Correspondence: ; Tel.: +86-29-82655361
| |
Collapse
|
9
|
Liu J, Shi J, Hernandez R, Li X, Konchadi P, Miyake Y, Chen Q, Zhou T, Zhou C. Paternal phthalate exposure-elicited offspring metabolic disorders are associated with altered sperm small RNAs in mice. ENVIRONMENT INTERNATIONAL 2023; 172:107769. [PMID: 36709676 DOI: 10.1016/j.envint.2023.107769] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 05/10/2023]
Abstract
Exposure to ubiquitous plastic-associated endocrine disrupting chemicals (EDCs) is associated with the increased risk of many chronic diseases. For example, phthalate exposure is associated with cardiometabolic mortality in humans, with societal costs ∼ $39 billion/year or more. We recently demonstrated that several widely used plastic-associated EDCs increase cardiometabolic disease in appropriate mouse models. In addition to affecting adult health, parental exposure to EDCs has also been shown to cause metabolic disorders, including obesity and diabetes, in the offspring. While most studies have focused on the impact of maternal EDC exposure on the offspring's health, little is known about the effects of paternal EDC exposure. In the current study, we investigated the adverse impact of paternal exposure to a ubiquitous but understudied phthalate, dicyclohexyl phthalate (DCHP) on the metabolic health of F1 and F2 offspring in mice. Paternal DCHP exposure led to exacerbated insulin resistance and impaired insulin signaling in F1 offspring without affecting diet-induced obesity. We previously showed that sperm small non-coding RNAs including tRNA-derived small RNAs (tsRNAs) and rRNA-derived small RNAs (rsRNAs) contribute to the intergenerational transmission of paternally acquired metabolic disorders. Using a novel PANDORA-seq, we revealed that DCHP exposure can lead to sperm tsRNA/rsRNA landscape changes that were undetected by traditional RNA-seq, which may contribute to DCHP-elicited adverse effects. Lastly, we found that paternal DCHP can also cause sex-specific transgenerational adverse effects in F2 offspring and elicited glucose intolerance in female F2 descendants. Our results suggest that exposure to endocrine disrupting phthalates may have intergenerational and transgenerational adverse effects on the metabolic health of their offspring. These findings increase our understanding of the etiology of chronic human diseases originating from chemical-elicited intergenerational and transgenerational effects.
Collapse
Affiliation(s)
- Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Junchao Shi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Xiuchun Li
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Pranav Konchadi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Yuma Miyake
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, NV 89557, United States
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States.
| |
Collapse
|
10
|
Li S, Liu P, Feng X, Du M, Zhang Y, Wang Y, Wang J. Mechanism of Tao Hong Decoction in the treatment of atherosclerosis based on network pharmacology and experimental validation. Front Cardiovasc Med 2023; 10:1111475. [PMID: 36776258 PMCID: PMC9909180 DOI: 10.3389/fcvm.2023.1111475] [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: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Background Atherosclerosis (AS) has long been recognized as a cardiovascular disease and stroke risk factor. A well-known traditional Chinese medicine prescription, Tao Hong decoction (THD), has been proven effective in treating AS, but its mechanism of action is still unclear. Objective To assess the effects, explore THD's primary mechanism for treating AS, and provide a basis for rational interpretation of its prescription compatibility. Methods Based on network pharmacology, we evaluated the mechanism of THD on AS by data analysis, target prediction, the construction of PPI networks, and GO and KEGG analysis. AutoDockTools software to conduct Molecular docking. Then UPLC-Q-TOF-MS was used to identify significant constituents of THD. Furthermore, an AS mice model was constructed and intervened with THD. Immunofluorescence, RT-qPCR, and Western blot were used to verify the critical targets in animal experiments. Results The network pharmacology results indicate that eight core targets and seven core active ingredients play an essential role in this process. The GO and KEGG analysis results suggested that the mechanism is mainly involved in Fluid shear stress and atherosclerosis and Lipid and atherosclerosis. The molecular docking results indicate a generally strong affinity. The animal experiment showed that THD reduced plaque area, increased plaque stability, and decreased the levels of inflammatory cytokines (NF-κB, IL-1α, TNF-α, IL-6, IL-18, IL-1β) in high-fat diet -induced ApoE-/-mice. Decreased levels of PTGS2, HIF-1α, VEGFA, VEGFC, FLT-4, and the phosphorylation of PI3K, AKT, and p38 were detected in the THD-treated group. Conclusion THD plays a vital role in treating AS with multiple targets and pathways. Angiogenesis regulation, oxidative stress regulation, and immunity regulation consist of the crucial regulation cores in the mechanism. This study identified essential genes and pathways associated with the prognosis and pathogenesis of AS from new insights, demonstrating a feasible method for researching THD's chemical basis and pharmacology.
Collapse
|
11
|
Meng Z, Hernandez R, Liu J, Gwag T, Lu W, Hsiai TK, Kaul M, Zhou T, Zhou C. HIV Protein Tat Induces Macrophage Dysfunction and Atherosclerosis Development in Low-Density Lipoprotein Receptor-Deficient Mice. Cardiovasc Drugs Ther 2022; 36:201-215. [PMID: 33459922 PMCID: PMC8286272 DOI: 10.1007/s10557-021-07141-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE HIV infection is consistently associated with an increased risk of atherosclerotic cardiovascular disease, but the underlying mechanisms remain elusive. HIV protein Tat, a transcriptional activator of HIV, has been shown to activate NF-κB signaling and promote inflammation in vitro. However, the atherogenic effects of HIV Tat have not been investigated in vivo. Macrophages are one of the major cell types involved in the initiation and progression of atherosclerosis. We and others have previously revealed the important role of IκB kinase β (IKKβ), a central inflammatory coordinator through activating NF-κB, in the regulation of macrophage functions and atherogenesis. This study investigated the impact of HIV Tat exposure on macrophage functions and atherogenesis. METHODS To investigate the effects of Tat on macrophage IKKβ activation and atherosclerosis development in vivo, myeloid-specific IKKβ-deficient LDLR-deficient (IKKβΔMyeLDLR-/-) mice and their control littermates (IKKβF/FLDLR-/-) were exposed to recombinant HIV protein Tat. RESULTS Exposure to Tat significantly increased atherosclerotic lesion size and plaque vulnerability in IKKβF/FLDLR-/- but not IKKβΔMyeLDLR-/- mice. Deficiency of myeloid IKKβ attenuated Tat-elicited macrophage inflammatory responses and atherosclerotic lesional inflammation in IKKβΔMyeLDLR-/- mice. Further, RNAseq analysis demonstrated that HIV protein Tat affects the expression of many atherosclerosis-related genes in vitro in an IKKβ-dependent manner. CONCLUSIONS Our findings reveal atherogenic effects of HIV protein Tat in vivo and demonstrate a pivotal role of myeloid IKKβ in Tat-driven atherogenesis.
Collapse
Affiliation(s)
- Zhaojie Meng
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Weiwei Lu
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Tzung K Hsiai
- Departments of Medicine and Bioengineering, David Geffen School of Medicine, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Marcus Kaul
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.
| |
Collapse
|
12
|
Liu J, Hernandez R, Li X, Meng Z, Chen H, Zhou C. Pregnane X Receptor Mediates Atherosclerosis Induced by Dicyclohexyl Phthalate in LDL Receptor-Deficient Mice. Cells 2022; 11:1125. [PMID: 35406689 PMCID: PMC8997706 DOI: 10.3390/cells11071125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022] Open
Abstract
Plastic-associated endocrine disrupting chemicals (EDCs) have been implicated in the etiology of cardiovascular disease (CVD) in humans, but the underlying mechanisms remain elusive. Dicyclohexyl phthalate (DCHP) is a widely used phthalate plasticizer; whether and how exposure to DCHP elicits adverse effects in vivo is mostly unknown. We previously reported that DCHP is a potent ligand of the pregnane X receptor (PXR) which acts as a xenobiotic sensor to regulate xenobiotic metabolism. PXR also functions in macrophages to regulate atherosclerosis development in animal models. In the current study, LDL receptor-deficient mice with myeloid-specific PXR deficiency (PXRΔMyeLDLR-/-) and their control littermates (PXRF/FLDLR-/-) were used to determine the impact of DCHP exposure on macrophage function and atherosclerosis. Chronic exposure to DCHP significantly increased atherosclerotic lesion area in the aortic root and brachiocephalic artery of PXRF/FLDLR-/- mice by 65% and 77%, respectively. By contrast, DCHP did not affect atherosclerosis development in PXRΔMyeLDLR-/- mice. Exposure to DCHP led to elevated expression of the scavenger receptor CD36 in macrophages and increased macrophage form cell formation in PXRF/FLDLR-/- mice. Our findings provide potential mechanisms underlying phthalate-associated CVD risk and will ultimately stimulate further investigations and mitigation of the adverse effects of plastic-associated EDCs on CVD risk in humans.
Collapse
Affiliation(s)
- Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (J.L.); (R.H.); (X.L.); (Z.M.)
| | - Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (J.L.); (R.H.); (X.L.); (Z.M.)
| | - Xiuchun Li
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (J.L.); (R.H.); (X.L.); (Z.M.)
| | - Zhaojie Meng
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (J.L.); (R.H.); (X.L.); (Z.M.)
| | - Hong Chen
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA;
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (J.L.); (R.H.); (X.L.); (Z.M.)
| |
Collapse
|
13
|
Hernandez R, Zhou C. Recent Advances in Understanding the Role of IKKβ in Cardiometabolic Diseases. Front Cardiovasc Med 2021; 8:752337. [PMID: 34957242 PMCID: PMC8692734 DOI: 10.3389/fcvm.2021.752337] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiometabolic diseases, including cardiovascular disease, obesity, and diabetes, are the leading cause of mortality and morbidity worldwide. Cardiometabolic diseases are associated with many overlapping metabolic syndromes such as hypertension, hyperlipidemia, insulin resistance, and central adiposity. However, the underlying causes of cardiometabolic diseases and associated syndromes remain poorly understood. Within the past couple of decades, considerable progresses have been made to understand the role of inflammatory signaling in the pathogenesis of cardiometabolic diseases. The transcription factor, NF-κB, a master regulator of the innate and adaptive immune responses, is highly active in cardiometabolic diseases. IκB kinase β (IKKβ), the predominant catalytic subunit of the IKK complex, is required for canonical activation of NF-κB, and has been implicated as the critical molecular link between inflammation and cardiometabolic diseases. Recent studies have revealed that IKKβ has diverse and unexpected roles in mediating adiposity, insulin sensitivity, glucose homeostasis, vascular function, and atherogenesis through complex mechanisms. IKKβ has been demonstrated as a critical player in the development of cardiometabolic diseases and is implicated as a promising therapeutic target. This review summarizes current knowledge of the functions of IKKβ in mediating the development and progression of cardiometabolic diseases.
Collapse
Affiliation(s)
- Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | | |
Collapse
|
14
|
Lu W, Meng Z, Hernandez R, Zhou C. Fibroblast-specific IKKβ deficiency ameliorates angiotensin II-induced adverse cardiac remodeling in mice. JCI Insight 2021; 6:e150161. [PMID: 34324438 PMCID: PMC8492299 DOI: 10.1172/jci.insight.150161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/28/2021] [Indexed: 12/03/2022] Open
Abstract
Cardiac inflammation and fibrosis contribute significantly to hypertension-related adverse cardiac remodeling. IκB kinase β (IKK-β), a central coordinator of inflammation through activation of NF-κB, has been demonstrated as a key molecular link between inflammation and cardiovascular disease. However, the cell-specific contribution of IKK-β signaling toward adverse cardiac remodeling remains elusive. Cardiac fibroblasts are one of the most populous nonmyocyte cell types in the heart that play a key role in mediating cardiac fibrosis and remodeling. To investigate the function of fibroblast IKK-β, we generated inducible fibroblast-specific IKK-β–deficient mice. Here, we report an important role of IKK-β in the regulation of fibroblast functions and cardiac remodeling. Fibroblast-specific IKK-β–deficient male mice were protected from angiotensin II–induced cardiac hypertrophy, fibrosis, and macrophage infiltration. Ablation of fibroblast IKK-β inhibited angiotensin II–stimulated fibroblast proinflammatory and profibrogenic responses, leading to ameliorated cardiac remodeling and improved cardiac function in IKK-β–deficient mice. Findings from this study establish fibroblast IKK-β as a key factor regulating cardiac fibrosis and function in hypertension-related cardiac remodeling.
Collapse
Affiliation(s)
- Weiwei Lu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, United States of America
| | - Zhaojie Meng
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| | - Rebecca Hernandez
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| | - Changcheng Zhou
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| |
Collapse
|
15
|
Genkel VV, Kuznetsova AS, Lebedev EV, Shaposhnik II. Factors associated with atherosclerotic plaque echogenicity in patients aged 40-64 with carotid atherosclerosis. ACTA ACUST UNITED AC 2021; 61:35-40. [PMID: 34311686 DOI: 10.18087/cardio.2021.6.n1536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 11/18/2022]
Abstract
Aim To identify clinical and laboratory indexes related with the atherosclerotic plaque (ASP) echogenicity based on results of the analysis of grey-scale median (GSM) in patients aged 40-64 years.Material and methods The study included patients aged 40-64 years with carotid atherosclerosis. The carotid duplex scanning was performed for all patients. The GSM analysis of obtained images was performed with the Adobe Photoshop CS6 software.Results Atherosclerotic cardiovascular diseases were found in 31 (21.4 %) patients. Correlation analysis determined inverse interrelationships between GSM and the body weight index (BWI) (r=-0.359; p<0.0001), waist circumference (r=-0.357; p<0.0001), and levels of uric acid (r=-0.244; p=0.021) and glucose (r=-0.205; p=0.032). According to the regression, statistically significant correlations remained between GSM and BWI as well as the waist circumference after the adjustment for sex and age.Conclusion In patients with carotid atherosclerosis aged 40-64 years, the decrease in ASP GSM was associated with increases in BWI and waist circumference.
Collapse
Affiliation(s)
- V V Genkel
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - A S Kuznetsova
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - E V Lebedev
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - I I Shaposhnik
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| |
Collapse
|
16
|
Intermedin 1-53 attenuates atherosclerotic plaque vulnerability by inhibiting CHOP-mediated apoptosis and inflammasome in macrophages. Cell Death Dis 2021; 12:436. [PMID: 33934111 PMCID: PMC8088440 DOI: 10.1038/s41419-021-03712-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022]
Abstract
Atherosclerotic plaque vulnerability and rupture increase the risk of acute coronary syndromes. Advanced lesion macrophage apoptosis plays important role in the rupture of atherosclerotic plaque, and endoplasmic reticulum stress (ERS) has been proved to be a key mechanism of macrophage apoptosis. Intermedin (IMD) is a regulator of ERS. Here, we investigated whether IMD enhances atherosclerotic plaque stability by inhibiting ERS-CHOP-mediated apoptosis and subsequent inflammasome in macrophages. We studied the effects of IMD on features of plaque vulnerability in hyperlipemia apolipoprotein E-deficient (ApoE−/−) mice. Six-week IMD1-53 infusion significantly reduced atherosclerotic lesion size. Of note, IMD1-53 lowered lesion macrophage content and necrotic core size and increased fibrous cap thickness and vascular smooth muscle cells (VSMCs) content thus reducing overall plaque vulnerability. Immunohistochemical analysis indicated that IMD1-53 administration prevented ERS activation in aortic lesions of ApoE−/− mice, which was further confirmed in oxidized low-density lipoproteins (ox-LDL) induced macrophages. Similar to IMD, taurine (Tau), a non-selective ERS inhibitor significantly reduced atherosclerotic lesion size and plaque vulnerability. Moreover, C/EBP-homologous protein (CHOP), a pro-apoptosis transcription factor involved in ERS, was significantly increased in advanced lesion macrophages, and deficiency of CHOP stabilized atherosclerotic plaques in AopE−/− mice. IMD1-53 decreased CHOP level and apoptosis in vivo and in macrophages treated with ox-LDL. In addition, IMD1-53 infusion ameliorated NLRP3 inflammasome and subsequent proinflammatory cytokines in vivo and in vitro. IMD may attenuate the progression of atherosclerotic lesions and plaque vulnerability by inhibiting ERS-CHOP-mediated macrophage apoptosis, and subsequent NLRP3 triggered inflammation. The inhibitory effect of IMD on ERS-induced macrophages apoptosis was probably mediated by blocking CHOP activation.
Collapse
|
17
|
Sui Y, Meng Z, Park SH, Lu W, Livelo C, Chen Q, Zhou T, Zhou C. Myeloid-specific deficiency of pregnane X receptor decreases atherosclerosis in LDL receptor-deficient mice. J Lipid Res 2020; 61:696-706. [PMID: 32170024 DOI: 10.1194/jlr.ra119000122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
The pregnane X receptor (PXR) is a nuclear receptor that can be activated by numerous drugs and xenobiotic chemicals. PXR thereby functions as a xenobiotic sensor to coordinately regulate host responses to xenobiotics by transcriptionally regulating many genes involved in xenobiotic metabolism. We have previously reported that PXR has pro-atherogenic effects in animal models, but how PXR contributes to atherosclerosis development in different tissues or cell types remains elusive. In this study, we generated an LDL receptor-deficient mouse model with myeloid-specific PXR deficiency (PXRΔMyeLDLR-/-) to elucidate the role of macrophage PXR signaling in atherogenesis. The myeloid PXR deficiency did not affect metabolic phenotypes and plasma lipid profiles, but PXRΔMyeLDLR-/- mice had significantly decreased atherosclerosis at both aortic root and brachiocephalic arteries compared with control littermates. Interestingly, the PXR deletion did not affect macrophage adhesion and migration properties, but reduced lipid accumulation and foam cell formation in the macrophages. PXR deficiency also led to decreased expression of the scavenger receptor CD36 and impaired lipid uptake in macrophages of the PXRΔMyeLDLR-/- mice. Further, RNA-Seq analysis indicated that treatment with a prototypical PXR ligand affects the expression of many atherosclerosis-related genes in macrophages in vitro. These findings reveal a pivotal role of myeloid PXR signaling in atherosclerosis development and suggest that PXR may be a potential therapeutic target in atherosclerosis management.
Collapse
Affiliation(s)
- Yipeng Sui
- Department of Pharmacology and Nutritional Sciences,University of Kentucky, Lexington, KY 40536
| | - Zhaojie Meng
- Department of Pharmacology and Nutritional Sciences,University of Kentucky, Lexington, KY 40536; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences,University of Kentucky, Lexington, KY 40536
| | - Weiwei Lu
- Department of Pharmacology and Nutritional Sciences,University of Kentucky, Lexington, KY 40536
| | - Christopher Livelo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521
| | - Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences,University of Kentucky, Lexington, KY 40536; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521. mailto:
| |
Collapse
|
18
|
Lu W, Park SH, Meng Z, Wang F, Zhou C. Deficiency of Adipocyte IKKβ Affects Atherosclerotic Plaque Vulnerability in Obese LDLR Deficient Mice. J Am Heart Assoc 2019; 8:e012009. [PMID: 31203708 PMCID: PMC6645619 DOI: 10.1161/jaha.119.012009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Obesity‐associated chronic inflammation has been known to contribute to atherosclerosis development, but the underlying mechanisms remain elusive. Recent studies have revealed novel functions of IKKβ (inhibitor of NF‐κB [nuclear factor κB] kinase β), a key coordinator of inflammation through activation of NF‐κB, in atherosclerosis and adipose tissue development. However, it is not clear whether IKKβ signaling in adipocytes can also affect atherogenesis. This study aims to investigate the impact of adipocyte IKKβ expression on atherosclerosis development in lean and obese LDLR (low‐density lipoprotein receptor)–deficient (LDLR−/−) mice. Methods and Results To define the role of adipocyte IKKβ in atherogenesis, we generated adipocyte‐specific IKKβ‐deficient LDLR−/− (IKKβΔAdLDLR−/−) mice. Targeted deletion of IKKβ in adipocytes did not affect adiposity and atherosclerosis in lean LDLR−/− mice when fed a low‐fat diet. In response to high‐fat feeding, however, IKKβΔAdLDLR−/− mice had defective adipose remodeling and increased adipose tissue and systemic inflammation. Deficiency of adipocyte IKKβ did not affect atherosclerotic lesion sizes but resulted in enhanced lesional inflammation and increased plaque vulnerability in obese IKKβΔAdLDLR−/− mice. Conclusions These data demonstrate that adipocyte IKKβ signaling affects the evolution of atherosclerosis plaque vulnerability in obese LDLR−/− mice. This study suggests that the functions of IKKβ signaling in atherogenesis are complex, and IKKβ in different cell types or tissues may have different effects on atherosclerosis development.
Collapse
Affiliation(s)
- Weiwei Lu
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Se-Hyung Park
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Zhaojie Meng
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Fang Wang
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY
| | - Changcheng Zhou
- 1 Department of Pharmacology and Nutritional Sciences University of Kentucky Lexington KY.,2 Saha Cardiovascular Research Center University of Kentucky Lexington KY
| |
Collapse
|