1
|
Runtian Z, Wenqiang H, Zimeng S, Tianyu W, Jingquan Z. AEBP1 or ACLP, which is the key factor in inflammation and fibrosis? Int J Biol Macromol 2025; 310:143554. [PMID: 40294683 DOI: 10.1016/j.ijbiomac.2025.143554] [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: 02/01/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Adipocyte enhancer-binding protein 1 (AEBP1) and Aortic carboxypeptidase-like protein (ACLP) are two protein isoforms produced by the AEBP1 gene. AEBP1, originally discovered in preadipocytes, functions as a transcriptional repressor and is involved in promoting inflammation, proliferation, and migration through various signaling pathways. ACLP is an extracellular matrix protein linked to Ehlers-Danlos syndrome, a genetic disorder characterized by defective connective tissue development. Structurally, AEBP1 and ACLP share many similarities, and both participate in critical physiological or pathological processes, such as cancer and fibrosis, by influencing pathways like NK-κB, WNT, and TGF-β. In recent years, research on AEBP1 and ACLP has expanded to include major organs such as the brain, kidneys, and lungs, with a particular focus on the cardiovascular system, where they show potential as novel drug targets. However, most studies do not clearly distinguish between AEBP1 and ACLP. For instance, AEBP1 is implicated in myocardial fibrosis in hypertrophic cardiomyopathy models, whereas ACLP is associated with fibrosis in other organs. Additionally, literature on the relationship between AEBP1 and fibrosis is often contradictory. Clarifying the distinct roles of AEBP1 and ACLP and their different functions in various cell types would greatly benefit further research. Current research suggests that the AEBP1 gene encodes two proteins, AEBP1 and ACLP, which have been reported to exhibit distinct functions in different studies. However, many studies do not differentiate between these two proteins, potentially leading to misconceptions. Therefore, we have conducted a comprehensive review of the existing literature to elucidate the functions of the AEBP1 gene and its encoded proteins in detail.
Collapse
Affiliation(s)
- Zhang Runtian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Han Wenqiang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shen Zimeng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wang Tianyu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhong Jingquan
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China.
| |
Collapse
|
2
|
Majidiani H, Musavi M, Momtazi-Borojeni AA. New Roles of Artemisinins in Atherosclerosis Progression. Phytother Res 2025; 39:1847-1857. [PMID: 40200587 DOI: 10.1002/ptr.8483] [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: 04/25/2024] [Revised: 10/27/2024] [Accepted: 02/11/2025] [Indexed: 04/10/2025]
Abstract
Artemisinin is a natural compound derived from the Chinese plant Artemisia annua , which was officially approved by the FDA for its antimalarial effects. In recent years, a growing body of studies has shown the novel function of artemisinin in atherosclerosis therapy. In vivo studies have shown that artemisinin can inhibit the progression of atherosclerosis plaque. In the present review, the evidence showing the inhibitory effects of artemisinin on the progression of atherosclerosis plaque and its underlying mechanisms is discussed. Mechanistically, artemisinin and its derivatives act by modulating various atherosclerosis-mediating risk factors, including hyperlipidemia, inflammation, oxidative stress, and malfunctioning vascular smooth muscle cells (VSMCs). Notably, artesunate, but not artemisinin, can attenuate the plasma levels of TG, TC, VLDL-C, and LDL-c, along with a substantial decline in arterial lipid deposition through enhancing the LDPL activity via inducing the KFL2/NRF2/TCF7L2 axis. Artemisinin was found to ameliorate the atherosclerosis plaque inflammation by reducing monocyte adhesion and subsequent transmigration to the intima, via inhibiting the expression of ICAM-1 and VCAM-1, diminishing NLRP3 inflammasome activation, and reducing the expression of inflammatory factors such as IL-1β, IL-18, TNF-α, MCP-1, and TGF-β1 mechanistically and mainly via suppressing the by NF-κB activity. Artemisinin could exert antioxidant effects through activating the PI3K/Akt/eNOS signaling pathway and suppressing the ROS-mediated NF-κB signal pathway. Artemisinin could also improve the VSMC function in the atherosclerosis plaque. These findings can suggest artemisinin as a new therapeutic agent for treating atherosclerosis; however, future clinical trials are warranted to validate its therapeutic efficiency in patients with atherosclerosis.
Collapse
Affiliation(s)
- Hamidreza Majidiani
- Department of Basic Medical Sciences, Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Maryam Musavi
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Faculty of Medicine,Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Faculty of Medicine,Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
3
|
He W, Zhao H, Xue W, Luo Y, Yan M, Li J, Qing L, Wu W, Jin Z. Qingre Huoxue Decoction Alleviates Atherosclerosis by Regulating Macrophage Polarization Through Exosomal miR-26a-5p. Drug Des Devel Ther 2024; 18:6389-6411. [PMID: 39749190 PMCID: PMC11693966 DOI: 10.2147/dddt.s487476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Background Qingre Huoxue Decoction (QRHX) is a classical Chinese herbal prescription widely used in clinical practice for the treatment of atherosclerosis (AS). Our previous study demonstrated its efficacy in stabilizing plaque and improving prognosis, as well as its ability to regulate macrophage polarization. This study aimed to further investigate the effects of QRHX on AS and explore the underlying mechanisms. Methods ApoE-/- mice were fed a high-fat diet (HFD) for 8 weeks in order to establish an AS model. Oil Red O, H&E, Masson, and IHC staining were employed to assess lipid accumulation, plaque development, collagen loss and target of the aortas tissue. ELISA was employed to measure the levels of TNF-α and IL-10 in serum. Dual luciferase reporter assay was conducted to ascertain the connection between miR-26a-5p and PTGS2 in vitro. Western blot and RT-qPCR assay were conducted to assess the NF-κB signaling pathway and macrophage polarization. The effects of miR-26a-5p were tested after transfecting miR-26a-5p over-expressive lentivirus. Results QRHX attenuated HFD-induced plaque progression and inflammation of AS model mice. BMDM-derived exosomes (BMDM-exo) increased miR-26a-5p and decreased PTGS2 expressions, inhibited the NF-κB signaling pathway and regulated macrophage polarization in vivo. These effects of BMDM-exo were further enhanced after QRHX intervention. Dual luciferase reporter assay results showed that miR-26a-5p directly binds to the 3'-UTR of PTGS2 mRNA and regulates the expression of PTGS2. The miR-26a-5p of BMDM-exo played a key role in macrophage polarization. After overexpression of miR-26a-5p, the NF-κB signaling pathway was inhibited and macrophages were converted from M1 to M2 in vitro. Conclusion QRHX can exert anti-inflammatory and plaque-stabilizing effects through exosomal miR-26a-5p via inhibiting the PTGS2/NF-κB signaling pathway and regulating macrophage phenotype from M1 to M2 polarization in AS.
Collapse
Affiliation(s)
- Weifeng He
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Weiqi Xue
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Yuan Luo
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Mengyuan Yan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Junlong Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Lijin Qing
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| | - Zheng Jin
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, People’s Republic of China
| |
Collapse
|
4
|
Sabrie Z, Temiz-Resitoglu M, Kalkan T, Kilic B, Tunctan B, Malik KU, Sahan-Firat S. Protection by selective mTORC2 inhibition of Zymosan-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB activation. Prostaglandins Other Lipid Mediat 2024; 175:106918. [PMID: 39461547 DOI: 10.1016/j.prostaglandins.2024.106918] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Non-septic shock is a serious condition leading to multiple organ dysfunction. Although targeting the mammalian target of the rapamycin complex 1 (mTORC1) signaling pathway exerts potent anti-inflammatory activity, little is known about mTORC2's contribution to non-septic shock. Thus, our research aims to investigate mTORC2's contribution and associated changes of IκB kinase (IKKα)/inhibitor κB (IκB-α)/nuclear factor-ĸB (NF-κB) pathway on Zymosan (ZYM)-induced non-septic rat model using the novel mTORC2 selective inhibitor JR-AB2-011. Rats were given saline (4 ml/kg), dimethylsulfoxide (DMSO) (4 ml/kg), ZYM (500 mg/kg), and (or) JR-AB2-011 (1 mg/kg). Mean arterial pressure (MAP) and heart rate (HR) of rats were recorded. JR-AB2-011 reversed both ZYM-induced reduction in MAP and increase in HR. Protein expression and/or phosphorylation of rictor, protein kinase B (Akt), IκB-α, IKKα, NF-κB p65, inducible nitric oxide synthase (iNOS), nitrotyrosine, cyclooxygenase 2 (COX-2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, besides prostaglandin (PG) E2 levels were measured. The enhanced expression of the proteins mentioned above has been inhibited by JR-AB2-011. These data suggest mTORC2's promising role in ZYM-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB pathway.
Collapse
Affiliation(s)
- Zainab Sabrie
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Taskin Kalkan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Banu Kilic
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN, USA
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| |
Collapse
|
5
|
Su X, Zhang M, Yang G, Cui X, Yuan X, Du L, Pei Y. Bioinformatics and machine learning approaches reveal key genes and underlying molecular mechanisms of atherosclerosis: A review. Medicine (Baltimore) 2024; 103:e38744. [PMID: 39093811 PMCID: PMC11296484 DOI: 10.1097/md.0000000000038744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/07/2024] [Indexed: 08/04/2024] Open
Abstract
Atherosclerosis (AS) causes thickening and hardening of the arterial wall due to accumulation of extracellular matrix, cholesterol, and cells. In this study, we used comprehensive bioinformatics tools and machine learning approaches to explore key genes and molecular network mechanisms underlying AS in multiple data sets. Next, we analyzed the correlation between AS and immune fine cell infiltration, and finally performed drug prediction for the disease. We downloaded GSE20129 and GSE90074 datasets from the Gene expression Omnibus database, then employed the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts algorithm to analyze 22 immune cells. To enrich for functional characteristics, the black module correlated most strongly with T cells was screened with weighted gene co-expression networks analysis. Functional enrichment analysis revealed that the genes were mainly enriched in cell adhesion and T-cell-related pathways, as well as NF-κ B signaling. We employed the Lasso regression and random forest algorithms to screen out 5 intersection genes (CCDC106, RASL11A, RIC3, SPON1, and TMEM144). Pathway analysis in gene set variation analysis and gene set enrichment analysis revealed that the key genes were mainly enriched in inflammation, and immunity, among others. The selected key genes were analyzed by single-cell RNA sequencing technology. We also analyzed differential expression between these 5 key genes and those involved in iron death. We found that ferroptosis genes ACSL4, CBS, FTH1 and TFRC were differentially expressed between AS and the control groups, RIC3 and FTH1 were significantly negatively correlated, whereas SPON1 and VDAC3 were significantly positively correlated. Finally, we used the Connectivity Map database for drug prediction. These results provide new insights into AS genetic regulation.
Collapse
Affiliation(s)
- Xiaoxue Su
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Meng Zhang
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Guinan Yang
- Department of Urology, People’s Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Xuebin Cui
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | | | | | - Yuanmin Pei
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| |
Collapse
|
6
|
Wang Y, Wang B, Ling H, Li Y, Fu S, Xu M, Li B, Liu X, Wang Q, Li A, Zhang X, Liu M. Navigating the Landscape of Coronary Microvascular Research: Trends, Triumphs, and Challenges Ahead. Rev Cardiovasc Med 2024; 25:288. [PMID: 39228508 PMCID: PMC11366996 DOI: 10.31083/j.rcm2508288] [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: 12/25/2023] [Revised: 02/11/2024] [Accepted: 03/01/2024] [Indexed: 09/05/2024] Open
Abstract
Coronary microvascular dysfunction (CMD) refers to structural and functional abnormalities of the microcirculation that impair myocardial perfusion. CMD plays a pivotal role in numerous cardiovascular diseases, including myocardial ischemia with non-obstructive coronary arteries, heart failure, and acute coronary syndromes. This review summarizes recent advances in CMD pathophysiology, assessment, and treatment strategies, as well as ongoing challenges and future research directions. Signaling pathways implicated in CMD pathogenesis include adenosine monophosphate-activated protein kinase/Krüppel-like factor 2/endothelial nitric oxide synthase (AMPK/KLF2/eNOS), nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf2/ARE), Angiotensin II (Ang II), endothelin-1 (ET-1), RhoA/Rho kinase, and insulin signaling. Dysregulation of these pathways leads to endothelial dysfunction, the hallmark of CMD. Treatment strategies aim to reduce myocardial oxygen demand, improve microcirculatory function, and restore endothelial homeostasis through mechanisms including vasodilation, anti-inflammation, and antioxidant effects. Traditional Chinese medicine (TCM) compounds exhibit therapeutic potential through multi-targeted actions. Small molecules and regenerative approaches offer precision therapies. However, challenges remain in translating findings to clinical practice and developing effective pharmacotherapies. Integration of engineering with medicine through microfabrication, tissue engineering and AI presents opportunities to advance the diagnosis, prediction, and treatment of CMD.
Collapse
Affiliation(s)
- Yingyu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital,
Hengyang Medical School, University of South China, 410000 Changsha, Hunan, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Sunjing Fu
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Mengting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Bingwei Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Xueting Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Qin Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Ailing Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center,
Peking University First Hospital, 100005 Beijing, China
| | - Mingming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences &
Peking Union Medical College, 100005 Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, 100005 Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Science, 100005
Beijing, China
| |
Collapse
|
7
|
Zhou Y, Huang J, Mai W, Kuang W, Li X, Shi D, Yang Y, Wu J, Wu Z, Liao Y, Zhou Z, Qiu Z. The novel vaccines targeting interleukin-1 receptor type I. Int Immunopharmacol 2024; 132:111941. [PMID: 38554439 DOI: 10.1016/j.intimp.2024.111941] [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: 02/07/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/01/2024]
Abstract
OBJECTIVE There is mounting evidence indicating that atherosclerosis represents a persistent inflammatory process, characterized by the presence of inflammation at various stages of the disease. Interleukin-1 (IL-1) precisely triggers inflammatory signaling pathways by binding to interleukin-1 receptor type I (IL-1R1). Inhibition of this signaling pathway contributes to the prevention of atherosclerosis and myocardial infarction. The objective of this research is to develop therapeutic vaccines targeting IL-1R1 as a preventive measure against atherosclerosis and myocardial infarction. METHODS ILRQβ-007 and ILRQβ-008 vaccines were screened, prepared and then used to immunize high-fat-diet fed ApoE-/- mice and C57BL/6J mice following myocardial infarction. Progression of atherosclerosis in ApoE-/- mice was assessed primarily by oil-red staining of the entire aorta and aortic root, as well as by detecting the extent of macrophage infiltration. The post-infarction cardiac function in C57BL/6J mice were evaluated using cardiac ultrasound and histological staining. RESULTS ILRQβ-007 and ILRQβ-008 vaccines stimulated animals to produce high titers of antibodies that effectively inhibited the binding of interleukin-1β and interleukin-1α to IL-1R1. Both vaccines effectively reduced atherosclerotic plaque area, promoted plaque stabilization, decreased macrophage infiltration in plaques and influenced macrophage polarization, as well as decreasing levels of inflammatory factors in the aorta, serum, and ependymal fat in ApoE-/- mice. Furthermore, these vaccines dramatically improved cardiac function and macrophage infiltration in C57BL/6J mice following myocardial infarction. Notably, no significant immune-mediated damage was observed in immunized animals. CONCLUSION The vaccines targeting the IL-1R1 would be a novel and promising treatment for the atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Yanzhao Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jianwu Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wuqian Mai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenlong Kuang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dingyang Shi
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulu Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiacheng Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhijie Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Zhihua Qiu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
8
|
Snijckers RPM, Foks AC. Adaptive immunity and atherosclerosis: aging at its crossroads. Front Immunol 2024; 15:1350471. [PMID: 38686373 PMCID: PMC11056569 DOI: 10.3389/fimmu.2024.1350471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Adaptive immunity plays a profound role in atherosclerosis pathogenesis by regulating antigen-specific responses, inflammatory signaling and antibody production. However, as we age, our immune system undergoes a gradual functional decline, a phenomenon termed "immunosenescence". This decline is characterized by a reduction in proliferative naïve B- and T cells, decreased B- and T cell receptor repertoire and a pro-inflammatory senescence associated secretory profile. Furthermore, aging affects germinal center responses and deteriorates secondary lymphoid organ function and structure, leading to impaired T-B cell dynamics and increased autoantibody production. In this review, we will dissect the impact of aging on adaptive immunity and the role played by age-associated B- and T cells in atherosclerosis pathogenesis, emphasizing the need for interventions that target age-related immune dysfunction to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
9
|
Nascimento Júnior JXD, Sola-Penna M, Zancan P. Clotrimazole reverses macrophage M2 polarization by disrupting the PI3K/AKT/mTOR pathway. Biochem Biophys Res Commun 2024; 696:149455. [PMID: 38176247 DOI: 10.1016/j.bbrc.2023.149455] [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: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Macrophages switch among different activation phenotypes according to distinct environmental stimuli, varying from pro-inflammatory (M1) to alternative (also named resolutive; M2) activation forms. M1-and M2-activated macrophages represent the two extremes of the activation spectrum involving multiple species, which vary in terms of function and the cytokines secreted. The consensus is that molecular characterization of the distinct macrophage population and the signals driving their activation will help in explaining disease etiology and formulating therapies. For instance, myeloid cells residing in the tumor microenvironment are key players in tumor progression and usually display an M2-like phenotype, which help tumor cells to evade local inflammatory processes. Therefore, these specific cells have been proposed as targets for tumor therapies by changing their activation profile. Furthermore, M2 polarized macrophages are phagocytic cells promoting tissue repair and wound healing and are therefore potential targets to treat different diseases. We have already shown that clotrimazole (CTZ) decreases tumor cell viability and thus tumor growth. The mechanism by which CTZ exerts its effects remains to be determined, but this drug is an inhibitor of the PI3K/AKT/mTOR pathway. In this study, we show that CTZ downregulated M2-activation markers in macrophages polarized to the M2 profile. This effect occurred without interfering with the expression of M1-polarized markers or pro-inflammatory cytokines and signaling. Moreover, CTZ suppressed NFkB pathway intermediates and disrupted PI3K/AKT/mTOR signaling. We concluded that CTZ reverses macrophage M2 polarization by disrupting the PI3K/AKT/mTOR pathway, which results in the suppression of NFkB induction of M2 polarization. In addition, we find that CTZ represents a promising therapeutic tool as an antitumor agent.
Collapse
Affiliation(s)
- José Xavier do Nascimento Júnior
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Mauro Sola-Penna
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Patricia Zancan
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
10
|
Xing Z, Zhen Y, Chen J, Du M, Li D, Liu R, Zheng J. KPNA2 Silencing, Regulated by E3 Ubiquitin Ligase FBXW7, Alleviates Endothelial Dysfunction and Inflammation Through Inhibiting the Nuclear Translocation of p65 and IRF3: A Possible Therapeutic Approach for Atherosclerosis. Inflammation 2023; 46:2071-2088. [PMID: 37432596 DOI: 10.1007/s10753-023-01863-w] [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: 03/17/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023]
Abstract
Atherosclerosis (AS), characterized by a maladaptive inflammatory response, is one of the most common causes of death among the elderly. Karyopherin subunit alpha 2 (KPNA2), a member of the nuclear transport protein family, has been reported to play a pro-inflammatory role in various pathological processes by regulating the nuclear translocation of pro-inflammatory transcription factors. However, the function of KPNA2 in AS remains unknown. ApoE-/- mice were fed high-fat diets for 12 weeks to establish an AS mice model. Human umbilical vein endothelial cells (HUVECs) were treated with lipopolysaccharide (LPS) to establish an AS cell model. We found that KPNA2 was upregulated in the aortic roots of atherosclerotic mice and LPS-stimulated cells. KPNA2 knockdown inhibited LPS-induced secretion of pro-inflammatory factors and monocyte-endothelial adhesion in HUVECs, whereas KPNA2 overexpression exerted the opposite effects. p65 and interferon regulatory factor 3 (IRF3), the transcription factors known to regulate the transcription of pro-inflammatory genes, interacted with KPNA2, and their nuclear translocations were blocked following KPNA2 silencing. Furthermore, we found that KPNA2 protein level was decreased by E3 ubiquitin ligase F-box and WD repeat domain containing 7 (FBXW7), which was downregulated in the atherosclerotic mice. FBXW7 overexpression induced ubiquitination with subsequent proteasomal degradation of KPNA2. Meanwhile, the effects of KPNA2 deficiency on atherosclerotic lesions were further confirmed by in vivo experiments. Taken together, our study indicates that KPNA2 downregulation, regulated by FBXW7, may alleviate endothelial dysfunction and related inflammation in the progression of AS by suppressing the nuclear translocation of p65 and IRF3.
Collapse
Affiliation(s)
- Zeyu Xing
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Jie Chen
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Mingyang Du
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Dongdong Li
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Ruyin Liu
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China
| | - Jiahe Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, 110022, Liaoning, People's Republic of China.
| |
Collapse
|
11
|
Lin H, Ao H, Guo G, Liu M. The Role and Mechanism of Metformin in Inflammatory Diseases. J Inflamm Res 2023; 16:5545-5564. [PMID: 38026260 PMCID: PMC10680465 DOI: 10.2147/jir.s436147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Metformin is a classical drug used to treat type 2 diabetes. With the development of research on metformin, it has been found that metformin also has several advantages aside from its hypoglycemic effect, such as anti-inflammatory, anti-aging, anti-cancer, improving intestinal flora, and other effects. The prevention of inflammation is critical because chronic inflammation is associated with numerous diseases of considerable public health. Therefore, there has been growing interest in the role of metformin in treating various inflammatory conditions. However, the precise anti-inflammatory mechanisms of metformin were inconsistent in the reported studies. Thus, this review aims to summarize various currently known possible mechanisms of metformin involved in inflammatory diseases and provide references for the clinical application of metformin.
Collapse
Affiliation(s)
- Huan Lin
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Haiyong Ao
- Jiangxi Key Laboratory of Nanobiomaterials & School of Materials Science and Engineering, East China Jiaotong University, Nanchang, Jiangxi, People’s Republic of China
| | - Guanghua Guo
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Mingzhuo Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
12
|
Fonseca CG, Silvério V, Barata D, Giese W, Gerhardt H, Cardoso S, Franco CA. A 96-wells fluidic system for high-throughput screenings under laminar high wall shear stress conditions. MICROSYSTEMS & NANOENGINEERING 2023; 9:114. [PMID: 37719414 PMCID: PMC10504069 DOI: 10.1038/s41378-023-00589-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/14/2023] [Accepted: 07/26/2023] [Indexed: 09/19/2023]
Abstract
The ability of endothelial cells to respond to blood flow is fundamental for the correct formation and maintenance of a functional and hierarchically organized vascular network. Defective flow responses, in particular related to high flow conditions, have been associated with atherosclerosis, stroke, arteriovenous malformations, and neurodegenerative diseases. Yet, the molecular mechanisms involved in high flow response are still poorly understood. Here, we described the development and validation of a 96-wells fluidic system, with interchangeable cell culture and fluidics, to perform high-throughput screenings under laminar high-flow conditions. We demonstrated that endothelial cells in our newly developed 96-wells fluidic system respond to fluid flow-induced shear stress by aligning along the flow direction and increasing the levels of KLF2 and KLF4. We further demonstrate that our 96-wells fluidic system allows for efficient gene knock-down compatible with automated liquid handling for high-throughput screening platforms. Overall, we propose that this modular 96-well fluidic system is an excellent platform to perform genome-wide and/or drug screenings to identify the molecular mechanisms involved in the responses of endothelial cells to high wall shear stress.
Collapse
Affiliation(s)
- Catarina Gonçalves Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Vânia Silvério
- INESC Microsistemas and Nanotecnologias, INESC-MN, Lisboa, Portugal
- Department of Physics, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - David Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Wolfgang Giese
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Susana Cardoso
- INESC Microsistemas and Nanotecnologias, INESC-MN, Lisboa, Portugal
- Department of Physics, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Claudio Areias Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research Centre, Lisbon, Portugal
| |
Collapse
|
13
|
Matsumori A. Nuclear Factor-κB is a Prime Candidate for the Diagnosis and Control of Inflammatory Cardiovascular Disease. Eur Cardiol 2023; 18:e40. [PMID: 37456770 PMCID: PMC10345985 DOI: 10.15420/ecr.2023.10] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/17/2023] [Indexed: 07/18/2023] Open
Abstract
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is responsible for the regulation of genes involved in inflammation and immune responses. NF-κB may play an important role in cardiovascular diseases (CVDs), atherosclerosis and diabetes. Several therapeutic agents used for the treatment of CVDs and diabetes, such as pimobendan and sodium-glucose cotransporter 2 inhibitors, exert anti-inflammatory effects by inhibiting NF-κB activation; anti-inflammatory therapy may have beneficial effects in CVDs and diabetes. Several pharmacological agents and natural compounds may inhibit NF-κB, and these agents alone or in combination may be used to treat various inflammatory diseases. Immunoglobulin-free light chains could be surrogate biomarkers of NF-κB activation and may be useful for evaluating the efficacy of these agents. This review discusses recent advances in our understanding of how the NF-κB signalling pathway controls inflammation, metabolism and immunity, and how improved knowledge of these pathways may lead to better diagnostics and therapeutics for various human diseases.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization, Kyoto Medical Center Kyoto, Japan
| |
Collapse
|
14
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [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: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Pan Y, Feng X, Song W, Zhou X, Zhou Z, Chen G, Shen T, Zhang X. Effects and Potential Mechanism of Zhuyu Pill Against Atherosclerosis: Network Pharmacology and Experimental Validation. Drug Des Devel Ther 2023; 17:597-612. [PMID: 36866196 PMCID: PMC9970883 DOI: 10.2147/dddt.s398808] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is an immunoinflammatory disease associated with dyslipidemia. Zhuyu Pill (ZYP) is a classic Chinese herbal compound that has been shown to exhibit anti-inflammatory and lipid-lowering effects on AS in our previous studies. However, the underlying mechanisms by which ZYP ameliorates atherosclerosis have not yet been fully investigated. In this study, network pharmacology and in vivo experiments were conducted to explore the underlying pharmacological mechanisms of ZYP on ameliorating AS. METHODS The active ingredients of ZYP were acquired from our previous study. The putative targets of ZYP relevant to AS were obtained from TCMSP, SwissTargetPrediction, STITCH, DisGeNET, and GeneCards databases. Protein-protein interactions (PPI) network, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted using the Cytoscape software. Furthermore, in vivo experiments were carried out for target validation in apolipoprotein E (ApoE) -/- mice. RESULTS Animal experiments revealed that ZYP ameliorated AS mainly through lowering blood lipids, alleviating vascular inflammation, and decreasing the levels of vascular cell adhesion molecule-1 (VCAM1), intercellular adhesion molecule-1 (ICAM1), monocyte chemotactic protein-1 (MCP-1), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α). Additionally, the results of Real-Time quantitative PCR revealed that ZYP inhibited the gene expressions of mitogen-activated protein kinase (MAPK) p38, extracellular regulated protein kinases (ERK), c-Jun N-terminal kinase (JNK), and nuclear factor kappa-B (NF-κB) p65. The Immunohistochemistry and Western blot assays showed the inhibitory effect of ZYP on the proteins level of p38, p-p38, p65, and p-p65. CONCLUSION This study has provided valuable evidence on the pharmacological mechanisms of action of ZYP in ameliorating AS that will be useful for forming the rationale of future research studying the cardio-protection and anti-inflammation effects of ZYP.
Collapse
Affiliation(s)
- Yingying Pan
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xianrong Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wei Song
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xin Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Zhen Zhou
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Gaoyang Chen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaobo Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
16
|
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: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
17
|
Wang D, Tan Z, Yang J, Li L, Li H, Zhang H, Liu H, Liu Y, Wang L, Li Q, Guo H. Perfluorooctane sulfonate promotes atherosclerosis by modulating M1 polarization of macrophages through the NF-κB pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114384. [PMID: 36512850 DOI: 10.1016/j.ecoenv.2022.114384] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely used and distributed perfluorinated compounds and is reported to be harmful to cardiovascular health; however, the direct association between PFOS exposure and atherosclerosis and the underlying mechanisms remain unknown. Therefore, this study aimed to investigate the effects of PFOS exposure on the atherosclerosis progression and the underlying mechanisms. PFOS was administered through oral gavage to apolipoprotein E-deficient (ApoE-/-) mice for 12 weeks. PFOS exposure significantly increased pulse wave velocity (PWV) and intima-media thickness (IMT), increased aortic plaque burden and vulnerability, and elevated serum lipid and inflammatory cytokine levels. PFOS promoted aortic and RAW264.7 M1 macrophage polarization, which increased the secretion of nitric oxide synthase (iNOS) and pro-inflammatory factors (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]), and suppressed M2 macrophage polarization, which decreased the expression of CD206, arginine I (Arg-1), and interleukin-10 (IL-10). Moreover, PFOS activated nuclear factor-kappa B (NF-κB) in the aorta and macrophages. BAY11-7082 was used to inhibit NF-κB-alleviated M1 macrophage polarization and the inflammatory response induced by PFOS in RAW264.7 macrophages. Our results are the first to reveal the acceleratory effect of PFOS on the atherosclerosis progression in ApoE-/- mice, which is associated with the NF-κB activation of macrophages to M1 polarization to induce inflammation.
Collapse
Affiliation(s)
- Dan Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Zhenzhen Tan
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Longfei Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Haoran Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, China
| | - Heqiong Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang, China
| | - Qian Li
- Department of Physiology, Hebei Medical University, Shijiazhuang, China.
| | - Huicai Guo
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China.
| |
Collapse
|
18
|
Yuan J, Kong Y. MiR-7-5p attenuates vascular smooth muscle cell migration and intimal hyperplasia after vascular injury by NF-kB signaling. Biochem Biophys Rep 2022; 33:101394. [PMID: 36601516 PMCID: PMC9806680 DOI: 10.1016/j.bbrep.2022.101394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 01/01/2023] Open
Abstract
Background Atherosclerosis (AS) is the primary cause of coronary artery disease, which is featured by aberrant proliferation, differentiation, and migration of vascular smooth muscle cells (VSMCs). MicroRNAs play crucial roles in AS, but the function of miR-7-5p in AS remains unclear. Here, we aimed to explore the effect of miR-7-5p on AS and VSMCs in vitro and in vivo. Methods The in vivo rat AS model and apoE-/- mouse model were established. The carotid artery injury was checked by immunohistochemistry staining. The RNA levels of miR-7-5p and p65 were measured by qPCR assay. Protein levels were checked by western blotting. Cell apoptosis was evaluated by flow cytometry. Cell migration was checked by Transwell assay and wound healing assay. The potential interaction between miR-7-5p with p65 was checked by luciferase reporter gene assay. Results MiR-7-5p was downregulated and NF-κB p65 was upregulated in injured carotid arteries in rat model. The carotid artery injury in the AS rats and the treatment of miR-7-5p attenuated the phenotype in the model. Immunohistochemistry staining and Western blot analysis revealed that PCNA levels were increased in injured carotid arteries of the model rats and miR-7-5p could reverse the levels. The cell viability of VSMCs was induced by PDGF-BB but miR-7-5p blocked the phenotype. PDGF-BB decreased apoptosis of VSMCs, while miR-7-5p was able to restore the cell apoptosis in the model. PDGF-BB-induced migration of VSMCs was attenuated by miR-7-5p. miR-7-5p mimic remarkably repressed the luciferase activity of p65 in VSMCs. The levels of p65 were inhibited by miR-7-5p in the cells. The PDGF-BB-promoted cell viability and migration of VSMCs was repressed by miR-7-5p and p65 overexpression reversed the phenotype. Conclusion We concluded that miR-7-5p attenuates vascular smooth muscle cell migration and intimal hyperplasia after vascular injury by NF-kB signaling.
Collapse
Affiliation(s)
- Jixiang Yuan
- The First Affiliated Hospital of Northwest Minzu University, Yinchuan, 750002, Ningxia Hui Autonomous Region, China,Corresponding author.
| | - Yun Kong
- Beijing Bioscience Biomedical Technology Co., LTD, Beijing, 100010, China
| |
Collapse
|
19
|
Farina FM, Serio S, Hall IF, Zani S, Cassanmagnago GA, Climent M, Civilini E, Condorelli G, Quintavalle M, Elia L. The epigenetic enzyme DOT1L orchestrates vascular smooth muscle cell-monocyte crosstalk and protects against atherosclerosis via the NF-κB pathway. Eur Heart J 2022; 43:4562-4576. [PMID: 35292818 DOI: 10.1093/eurheartj/ehac097] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/21/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
AIMS Histone H3 dimethylation at lysine 79 is a key epigenetic mark uniquely induced by methyltransferase disruptor of telomeric silencing 1-like (DOT1L). We aimed to determine whether DOT1L modulates vascular smooth muscle cell (VSMC) phenotype and how it might affect atherosclerosis in vitro and in vivo, unravelling the related mechanism. METHODS AND RESULTS Gene expression screening of VSMCs stimulated with the BB isoform of platelet-derived growth factor led us to identify Dot1l as an early up-regulated epigenetic factor. Mouse and human atherosclerotic lesions were assessed for Dot1l expression, which resulted specifically localized in the VSMC compartment. The relevance of Dot1l to atherosclerosis pathogenesis was assessed through deletion of its gene in the VSMCs via an inducible, tissue-specific knock-out mouse model crossed with the ApoE-/- high-fat diet model of atherosclerosis. We found that the inactivation of Dot1l significantly reduced the progression of the disease. By combining RNA- and H3K79me2-chromatin immunoprecipitation-sequencing, we found that DOT1L and its induced H3K79me2 mark directly regulate the transcription of Nf-κB-1 and -2, master modulators of inflammation, which in turn induce the expression of CCL5 and CXCL10, cytokines fundamentally involved in atherosclerosis development. Finally, a correlation between coronary artery disease and genetic variations in the DOT1L gene was found because specific polymorphisms are associated with increased mRNA expression. CONCLUSION DOT1L plays a key role in the epigenetic control of VSMC gene expression, leading to atherosclerosis development. Results identify DOT1L as a potential therapeutic target for vascular diseases.
Collapse
Affiliation(s)
- Floriana Maria Farina
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU) München, D-80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, D-80336 Munich, Germany
| | - Simone Serio
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Humanitas University, Pieve Emanuele (MI), Italy
| | | | - Stefania Zani
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Humanitas University, Pieve Emanuele (MI), Italy
| | - Giada Andrea Cassanmagnago
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Humanitas University, Pieve Emanuele (MI), Italy
| | - Montserrat Climent
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy
| | - Efrem Civilini
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Humanitas University, Pieve Emanuele (MI), Italy
| | - Gianluigi Condorelli
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Humanitas University, Pieve Emanuele (MI), Italy
| | - Manuela Quintavalle
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Astrazeneca, V.le Decumano, 39, 20157 Milano (MI), Italy
| | - Leonardo Elia
- IRCCS Humanitas Research Hospital, Via Manzoni 113, 20089 Rozzano (MI), Italy.,Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
20
|
Zhang H, Huang Y, Li X, Chen W, Lun Y, Zhang J. Identifying Hub Genes and Immune Cell Infiltration for the Progression of Carotid Atherosclerotic Plaques in the Context of Predictive and Preventive Using Integrative Bioinformatics Approaches and Machine-Learning Strategies. J Immunol Res 2022; 2022:7657379. [PMID: 36304068 PMCID: PMC9596267 DOI: 10.1155/2022/7657379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/03/2022] Open
Abstract
Emerging evidence shows that carotid atherosclerosis is related to the activation of immune-related pathways and inflammatory cell infiltration. However, the immune-linked pathways that helped in the advancement of the carotid atherosclerotic plaque and the association of such plaques with the infiltration status of the body's immune cells still unclear. Here, the expression profiles of the genes expressed during the progression of the carotid atherosclerotic plaques were retrieved from the Gene Expression Omnibus database and 178 differentially expressed genes were examined. The Weighted Gene Coexpression Network Analysis technique identified one of the brown modules showed the greatest correlation with carotid atherosclerotic plaques. In total, 66 intersecting genes could be detected after combining the DEGs. LASSO regression analysis was subsequently performed to obtain five hub genes as potential biomarkers for carotid atherosclerotic plaques. The functional analysis emphasized the vital roles played by the inflammation- and immune system-related pathways in this disease. The immune cell infiltration results highlighted the significant correlation among the CD4+ T cells, B cells, macrophages, and CD8+ T cells. Thereafter, the gene expression levels and the diagnostic values related to every hub gene were further validated. The above results indicated that macrophages, B cells, CD4+ T cells, and CD8 + T cells were closely related to the formation of the advanced-stage carotid atherosclerotic plaques. Based on the results, it could be hypothesized that the expression of hub genes (C3AR1, SLAMF8, TMEM176A, FERMT3, and GIMAP4) assisted in the advancement of the early-stage to advanced-stage carotid atherosclerotic plaque through immune-related signaling pathways. This may help to provide novel strategies for the treatment of carotid plaque in the context of predictive, preventive, and personalized medicine.
Collapse
Affiliation(s)
- Han Zhang
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yinde Huang
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xin Li
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wenbin Chen
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
21
|
Qiu M, Chen J, Li X, Zhuang J. Intersection of the Ubiquitin–Proteasome System with Oxidative Stress in Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms232012197. [PMID: 36293053 PMCID: PMC9603077 DOI: 10.3390/ijms232012197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) present a major social problem worldwide due to their high incidence and mortality rate. Many pathophysiological mechanisms are involved in CVDs, and oxidative stress plays a vital mediating role in most of these mechanisms. The ubiquitin–proteasome system (UPS) is the main machinery responsible for degrading cytosolic proteins in the repair system, which interacts with the mechanisms regulating endoplasmic reticulum homeostasis. Recent evidence also points to the role of UPS dysfunction in the development of CVDs. The UPS has been associated with oxidative stress and regulates reduction–oxidation homeostasis. However, the mechanisms underlying UPS-mediated oxidative stress’s contribution to CVDs are unclear, especially the role of these interactions at different disease stages. This review highlights the recent research progress on the roles of the UPS and oxidative stress, individually and in combination, in CVDs, focusing on the pathophysiology of key CVDs, including atherosclerosis, ischemia–reperfusion injury, cardiomyopathy, and heart failure. This synthesis provides new insight for continued research on the UPS–oxidative stress interaction, in turn suggesting novel targets for the treatment and prevention of CVDs.
Collapse
Affiliation(s)
- Min Qiu
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jimei Chen
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaohong Li
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jian Zhuang
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Correspondence: ; Tel.: +86-020-83827812 (ext. 51050)
| |
Collapse
|
22
|
Zhang Q, Liu L, Hu Y, Shen L, Li L, Wang Y. Kv1.3 Channel Is Involved In Ox-LDL-induced Macrophage Inflammation Via ERK/NF-κB signaling pathway. Arch Biochem Biophys 2022; 730:109394. [PMID: 36100082 DOI: 10.1016/j.abb.2022.109394] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Macrophage inflammatory response is crucial for the initiation and progression of atherosclerosis. The voltage-gated potassium channel Kv1.3 plays an important role in the modulation of macrophage function. The aim of this study was to investigate the effect and possible mechanism of Kv1.3 on inflammation in oxidized low-density lipoprotein (ox-LDL)-induced RAW264.7 macrophages. Treatment with Kv1.3-siRNA attenuated the expression of IL-6 and TNF-α and reduced the phosphorylation of ERK1/2 and NF-κB in ox-LDL-induced macrophages. In contrast, overexpression of Kv1.3 with Lv-Kv1.3 promoted the expression of IL-6 and TNF-α, and increased ERK1/2 and NF-κB phosphorylation in macrophages. PD-98059, a specific inhibitor of ERK, reversed the expression of IL-6 and TNF-α in ox-LDL-treated macrophages. Kv1.3-siRNA did not inhibit inflammation any further when cells were treated with PD-98059. This suggests that ERK acts as a downstream regulator of the Kv1.3 channel. In conclusion, Kv1.3 may be an indispensable membrane protein in ox-LDL-induced RAW264.7 macrophage inflammation in atherosclerosis through the ERK/NF-κB pathway.
Collapse
Affiliation(s)
- Qiujie Zhang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Lin Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Yanyan Hu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Lin Shen
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Li Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Qilu Hospital of Shandong University, China
| | - Yuanyuan Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China.
| |
Collapse
|
23
|
Xu X, Wang Y, Li Y, Zhang B, Song Q. The future Landscape of macrophage research in cardiovascular disease: a bibliometric analysis. Curr Probl Cardiol 2022; 47:101311. [PMID: 35810847 DOI: 10.1016/j.cpcardiol.2022.101311] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/03/2022]
Abstract
Cardiovascular disease (CVD) refers to a group of diseases involving the heart or blood vessels and is currently the leading cause of morbidity and mortality in many countries around the world and poses a serious economic burden. Macrophages are key effectors of inflammatory and innate immune responses, and their aberrant expression contributes to the development of various types of CVD.This study retrieved articles published from 1990-2022 on macrophages in CVD from the Web of Science core collection, based on CiteSpace and VOSviewer on these literature The annual output, countries and regions, institutions, authors, core journals, keywords and co-cited literature were analyzed. A total of 7,197 articles and reviews were retrieved, with a general upward trend despite slight fluctuations in annual publications. Europe, the United States and Asia are the main countries and regions publishing articles, especially the United States, with the highest number of articles (2,581), citations (173,692) and H-index (197), which also has the world's largest number of elite institutions, professional The country also has the world's largest number of elite institutions, professional researchers and high-impact journals, and is the leading country in this field of research. Keywords "inflammation", "immunology", "autophagy", "lipid-peroxidation" are the main pathogenesis of CVD caused by macrophages. "NLRP3", "nf kappa b" and "TNF-α" are the most frequently studied signalling pathways. Atherosclerosis, myocarditis and myocardial injury are the most studied disease types in this field. In addition, the study of macrophage-related CVD induced by COVID-19 seems to be a recent hot topic, and the mechanisms involved are mainly macrophage polarization, inflammatory factor storm, ACE2 and so on. The present study reveals hot spots and new trends in research on macrophages in CVD, which can provide scholars with key information in this field of research and help further explore new research directions.
Collapse
Affiliation(s)
- Xia Xu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajiao Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
24
|
Cheng W, Cui C, Liu G, Ye C, Shao F, Bagchi AK, Mehta JL, Wang X. NF-κB, A Potential Therapeutic Target in Cardiovascular Diseases. Cardiovasc Drugs Ther 2022; 37:571-584. [PMID: 35796905 DOI: 10.1007/s10557-022-07362-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Atherosclerosis is the basis of major CVDs - myocardial ischemia, heart failure, and stroke. Among numerous functional molecules, the transcription factor nuclear factor κB (NF-κB) has been linked to downstream target genes involved in atherosclerosis. The activation of the NF-κB family and its downstream target genes in response to environmental and cellular stress, hypoxia, and ischemia initiate different pathological events such as innate and adaptive immunity, and cell survival, differentiation, and proliferation. Thus, NF-κB is a potential therapeutic target in the treatment of atherosclerosis and related CVDs. Several biologics and small molecules as well as peptide/proteins have been shown to regulate NF-κB dependent signaling pathways. In this review, we will focus on the function of NF-κB in CVDs and the role of NF-κB inhibitors in the treatment of CVDs.
Collapse
Affiliation(s)
- Weijia Cheng
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Can Cui
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fang Shao
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450046, China
| | - Ashim K Bagchi
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA.
| | - Xianwei Wang
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China. .,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
25
|
Ding S, Liu J, Han X, Ding W, Liu Z, Zhu Y, Zhan W, Wan Y, Gai S, Hou J, Wang X, Wu Y, Wu A, Li CY, Zheng Z, Tian XL, Cao H. ICAM-1-related noncoding RNA accelerates atherosclerosis by amplifying NF-κB signaling. J Mol Cell Cardiol 2022; 170:75-86. [PMID: 35714558 DOI: 10.1016/j.yjmcc.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/20/2022] [Accepted: 06/04/2022] [Indexed: 11/28/2022]
Abstract
Long noncoding RNAs (lncRNAs) are critical regulators of inflammation with great potential as new therapeutic targets. However, the role of lncRNAs in early atherosclerosis remains poorly characterized. This study aimed to identify the key lncRNA players in activated endothelial cells (ECs). The lncRNAs in response to pro-inflammatory factors in ECs were screened through RNA sequencing. ICAM-1-related non-coding RNA (ICR) was identified as the most potential candidate for early atherosclerosis. ICR is essential for intercellular adhesion molecule-1 (ICAM1) expression, EC adhesion and migration. In a high fat diet-induced atherosclerosis model in mice, ICR is upregulated in the development of atherosclerosis. After intravenous injection of adenovirus carrying shRNA for mouse ICR, the atherosclerotic plaque area was markedly reduced with the declined expression of ICR and ICAM1. Mechanistically, ICR stabilized the mRNA of ICAM1 in quiescent ECs; while under inflammatory stress, ICR upregulated ICAM1 in a nuclear factor kappa B (NF-κB) dependent manner. RNA-seq analysis showed pro-inflammatory targets of NF-κB were regulated by ICR. Furthermore, the chromatin immunoprecipitation assays showed that p65 binds to ICR promoter and facilitates its transcription. Interestingly, ICR, in turn, promotes p65 accumulation and activity, forming a positive feedback loop to amplify NF-κB signaling. Preventing the degradation of p65 using proteasome inhibitors rescued the expression of NF-κB targets suppressed by ICR. Taken together, ICR acts as an accelerator to amplify NF-κB signaling in activated ECs and suppressing ICR is a promising early intervention for atherosclerosis through ICR/p65 loop blockade.
Collapse
Affiliation(s)
- Shuangjin Ding
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jiankun Liu
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - XiaoRui Han
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Wanqiu Ding
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zhirui Liu
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - Ying Zhu
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - Wenxing Zhan
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - Yiqi Wan
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - Shujie Gai
- National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junjie Hou
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xiaoxia Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yixia Wu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Andong Wu
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China
| | - Chuan-Yun Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zhe Zheng
- National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Jiangxi, China.
| | - Huiqing Cao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
| |
Collapse
|
26
|
Soliman GF, Abdel-Maksoud OM, Khalifa MM, Rashed LA, Ibrahim W, Morsi H, Abdallah H, Bastawy N. Effect of nebivolol on altered skeletal and cardiac muscles induced by dyslipidemia in rats: impact on oxidative and inflammatory machineries. Arch Physiol Biochem 2022; 128:463-473. [PMID: 31876193 DOI: 10.1080/13813455.2019.1693599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AIM High cholesterol diet is greatly linked to deleterious health consequences. In this work we tried to explore direct effects of high cholesterol diet on striated (skeletal and cardiac) muscle tissues and the mechanisms by which nebivolol could improve such harmful effects. METHODS The study included 24 healthy adult male albino rats weighing 200-220 grams that were assigned into four groups: control group, control drug group, high cholesterol diet fed groups; one untreated the other was treated with nebivolol. RESULTS In the cholesterol fed group, we found decreased blood HDL and NO with elevated total cholesterol, triglycerides, myoglobin, CK, LDH, ALP, in addition to elevated muscle tissue levels of HIF-1, NF-kB, MDA, and decreased expression of both eNOS, reduced GSH. Wire hanging test time was shorter in the high cholesterol group than control group rats, which was confirmed histologically by increased striated muscle fibre thickness and cytochrome area %. Nebivolol treatment ameliorated the effects of high cholesterol diet. CONCLUSION High cholesterol diet caused myopathic changes in rat striated muscle tissues mostly due to oxidative stress associated with enhanced NF-kB and HIF-1 expression. Nebivolol appears beneficial in the management of hypercholesterolaemia-induced striated muscle injury.
Collapse
Affiliation(s)
| | | | | | | | - Walaa Ibrahim
- Department of Medical Biochemistry, Cairo University, Cairo, Egypt
| | - Heba Morsi
- Department of Medical Biochemistry, Cairo University, Cairo, Egypt
| | - Hanan Abdallah
- Department of Medical Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen Bastawy
- Department of Medical Physiology, Cairo University, Cairo, Egypt
| |
Collapse
|
27
|
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
|
28
|
Tomatoes: An Extensive Review of the Associated Health Impacts of Tomatoes and Factors That Can Affect Their Cultivation. BIOLOGY 2022; 11:biology11020239. [PMID: 35205105 PMCID: PMC8869745 DOI: 10.3390/biology11020239] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023]
Abstract
Simple Summary The research outlined in this review paper discusses potential health benefits associated with a diet enriched with tomatoes and tomato products. This includes details of previous studies investigating the anticancer properties of tomatoes, protection against cardiovascular and neurodegenerative diseases and diabetes, maintenance of a healthy gut microbiome, and improved skin health, fertility, immune response, and exercise recovery. The specific parts of a tomato fruit that contribute these health benefits are also outlined. The potential disadvantages to a tomato-rich diet are detailed, especially the consumption of supplements that contain compounds found in tomatoes, such as lycopene. This review also discusses how the cultivation of tomato plants can affect the nutritional value of the fruit harvested. Different environmental growing conditions such as light intensity, growing media, and temperature are explained in terms of the impact they have on the quality of fruit, its nutrient content, and hence the potential health benefits acquired from eating the fruit. Abstract This review outlines the health benefits associated with the regular consumption of tomatoes and tomato products. The first section provides a detailed account of the horticultural techniques that can impact the quality of the fruit and its nutritional properties, including water availability, light intensity, temperature, and growing media. The next section provides information on the components of tomato that are likely to contribute to its health effects. The review then details some of the health benefits associated with tomato consumption, including anticancer properties, cardiovascular and neurodegenerative diseases and skin health. This review also discusses the impact tomatoes can have on the gut microbiome and associated health benefits, including reducing the risk of inflammatory bowel diseases. Other health benefits of eating tomatoes are also discussed in relation to effects on diabetes, the immune response, exercise recovery, and fertility. Finally, this review also addresses the negative effects that can occur as a result of overconsumption of tomato products and lycopene supplements.
Collapse
|
29
|
Saiki P, Yoshihara M, Kawano Y, Miyazaki H, Miyazaki K. Anti-Inflammatory Effects of Heliangin from Jerusalem Artichoke (Helianthus tuberosus) Leaves Might Prevent Atherosclerosis. Biomolecules 2022; 12:biom12010091. [PMID: 35053238 PMCID: PMC8774036 DOI: 10.3390/biom12010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is considered the major cause of cardiovascular and cerebrovascular diseases, which are the leading causes of death worldwide. Excessive nitric oxide production and inflammation result in dysfunctional vascular endothelial cells, which are critically involved in the initiation and progression of atherosclerosis. The present study aimed to identify a bioactive compound from Jerusalem artichoke leaves with anti-inflammatory activity that might prevent atherosclerosis. We isolated bioactive heliangin that inhibited NO production in LPS-induced macrophage-like RAW 264.7 cells. Heliangin suppressed ICAM-1, VCAM-1, E-selectin, and MCP-1 expression, as well as NF-κB and IκBα phosphorylation, in vascular endothelial cells stimulated with TNF-α. These results suggested that heliangin suppresses inflammation by inhibiting excessive NO production in macrophages and the expression of the factors leading to the development of atherosclerosis via the NF-κB signaling pathway in vascular endothelial cells. Therefore, heliangin in Jerusalem artichoke leaves could function in the prevention of atherosclerosis that is associated with heart attacks and strokes.
Collapse
Affiliation(s)
- Papawee Saiki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
- Correspondence: ; Tel.: +81-29-861-4304
| | - Mizuki Yoshihara
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Yasuhiro Kawano
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| | - Hitoshi Miyazaki
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Koyomi Miyazaki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| |
Collapse
|
30
|
Razeghian-Jahromi I, Karimi Akhormeh A, Razmkhah M, Zibaeenezhad MJ. Immune system and atherosclerosis: Hostile or friendly relationship. Int J Immunopathol Pharmacol 2022; 36:3946320221092188. [PMID: 35410514 PMCID: PMC9009140 DOI: 10.1177/03946320221092188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
Coronary artery disease has remained a major health challenge despite enormous progress in prevention, diagnosis, and treatment strategies. Formation of atherosclerotic plaque is a chronic process that is developmentally influenced by intrinsic and extrinsic determinants. Inflammation triggers atherosclerosis, and the fundamental element of inflammation is the immune system. The immune system involves in the atherosclerosis process by a variety of immune cells and a cocktail of mediators. It is believed that almost all main components of this system possess a profound contribution to the atherosclerosis. However, they play contradictory roles, either protective or progressive, in different stages of atherosclerosis progression. It is evident that monocytes are the first immune cells appeared in the atherosclerotic lesion. With the plaque growth, other types of the immune cells such as mast cells, and T lymphocytes are gradually involved. Each cell releases several cytokines which cause the recruitment of other immune cells to the lesion site. This is followed by affecting the expression of other cytokines as well as altering certain signaling pathways. All in all, a mix of intertwined interactions determine the final outcome in terms of mild or severe manifestations, either clinical or subclinical. Therefore, it is of utmost importance to precisely understand the kind and degree of contribution which is made by each immune component in order to stop the growing burden of cardiovascular morbidity and mortality. In this review, we present a comprehensive appraisal on the role of immune cells in the atherosclerosis initiation and development.
Collapse
Affiliation(s)
| | - Ali Karimi Akhormeh
- Cardiovascular Research Center, Shiraz University of Medical
Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer
Research, Shiraz University of Medical
Sciences, Shiraz, Iran
| | | |
Collapse
|
31
|
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: 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: 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
|
32
|
Natural Compound Resveratrol Attenuates TNF-Alpha-Induced Vascular Dysfunction in Mice and Human Endothelial Cells: The Involvement of the NF-κB Signaling Pathway. Int J Mol Sci 2021; 22:ijms222212486. [PMID: 34830366 PMCID: PMC8620472 DOI: 10.3390/ijms222212486] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/07/2021] [Indexed: 01/04/2023] Open
Abstract
Resveratrol, a natural compound in grapes and red wine, has drawn attention due to potential cardiovascular-related health benefits. However, its effect on vascular inflammation at physiologically achievable concentrations is largely unknown. In this study, resveratrol in concentrations as low as 1 μm suppressed TNF-α-induced monocyte adhesion to human EA.hy926 endothelial cells (ECs), a key event in the initiation and development of atherosclerosis. Low concentrations of resveratrol (0.25–2 μm) also significantly attenuated TNF-α-stimulated mRNA expressions of MCP-1/CCL2 and ICAM-1, which are vital mediators of EC-monocyte adhesion molecules and cytokines for cardiovascular plaque formation. Additionally, resveratrol diminished TNF-α-induced IκB-α degradation and subsequent nuclear translocation of NF-κB p65 in ECs. In the animal study, resveratrol supplementation in diet significantly diminished TNF-α-induced increases in circulating levels of adhesion molecules and cytokines, monocyte adhesion to mouse aortic ECs, F4/80-positive macrophages and VCAM-1 expression in mice aortas and restored the disruption in aortic elastin fiber caused by TNF-α treatment. The animal study also confirmed that resveratrol blocks the activation of NF-κB In Vivo. In conclusion, resveratrol at physiologically achievable concentrations displayed protective effects against TNF-α-induced vascular endothelial inflammation in vitro and In Vivo. The ability of resveratrol in reducing inflammation may be associated with its role as a down-regulator of the NF-κB pathway.
Collapse
|
33
|
De Negri Atanasio G, Ferrari PF, Campardelli R, Perego P, Palombo D. Innovative nanotools for vascular drug delivery: the atherosclerosis case study. J Mater Chem B 2021; 9:8558-8568. [PMID: 34609399 DOI: 10.1039/d1tb01071a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are the leading cause of mortality in the Western world. Among them, atherosclerosis represents one of the most common diseases in the modern society due to a common sedentary lifestyle, high-fat diet, and smoking. In the near future, a new approach could potentially improve the therapy of vascular pathologies, where to date the non-specific treatments present several limitations, such as poor biodistribution, quick elimination from the body, and undesired side-effects. In this field, nanotechnology has a great potential for the therapy and diagnosis of atherosclerosis with more and more recent and innovative publications. This review is a critical analysis of the results reported in the literature regarding the different and possible new approaches for the therapy and diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Giulia De Negri Atanasio
- Department of Experimental Medicine, University of Genoa, via Leon Battista Alberti, 2, 16132 Genoa, Italy
| | - Pier Francesco Ferrari
- Department of Surgical and Integrated Diagnostic Sciences, University of Genoa, viale Benedetto XV, 6, 16132 Genoa, Italy
| | - Roberta Campardelli
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, via Opera Pia, 15, 16145 Genoa, Italy.
| | - Patrizia Perego
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, via Opera Pia, 15, 16145 Genoa, Italy. .,Research Center for Biologically Inspired Engineering in Vascular Medicine and Longevity, University of Genoa, via Montallegro, 1, 16145 Genoa, Italy
| | - Domenico Palombo
- Department of Surgical and Integrated Diagnostic Sciences, University of Genoa, viale Benedetto XV, 6, 16132 Genoa, Italy.,Research Center for Biologically Inspired Engineering in Vascular Medicine and Longevity, University of Genoa, via Montallegro, 1, 16145 Genoa, Italy.,Vascular and Endovascular Surgery Unit, IRCCS Ospedale Policlinico San Martino, largo Rosanna Benzi, 10, 16132 Genoa, Italy
| |
Collapse
|
34
|
Mishchenko EL, Mishchenko AM, Ivanisenko VA. Mechanosensitive molecular interactions in atherogenic regions of the arteries: development of atherosclerosis. Vavilovskii Zhurnal Genet Selektsii 2021; 25:552-561. [PMID: 34595377 PMCID: PMC8453358 DOI: 10.18699/vj21.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022] Open
Abstract
A terrible disease of the cardiovascular system, atherosclerosis, develops in the areas of bends and
branches of arteries, where the direction and modulus of the blood flow velocity vector change, and consequently
so does the mechanical effect on endothelial cells in contact with the blood flow. The review focuses on topical
research studies on the development of atherosclerosis – mechanobiochemical events that transform the proatherogenic
mechanical stimulus of blood flow – low and low/oscillatory arterial wall shear stress in the chains of biochemical
reactions in endothelial cells, leading to the expression of specific proteins that cause the progression
of the pathological process. The stages of atherogenesis, systemic risk factors for atherogenesis and its important
hemodynamic factor, low and low/oscillatory wall shear stress exerted by blood flow on the endothelial cells lining
the arterial walls, have been described. The interactions of cell adhesion molecules responsible for the development
of atherosclerosis under low and low/oscillating shear stress conditions have been demonstrated. The activation
of the regulator of the expression of cell adhesion molecules, the transcription factor NF-κB, and the factors
regulating its activation under these conditions have been described. Mechanosensitive signaling pathways leading
to the expression of NF-κB in endothelial cells have been described. Studies of the mechanobiochemical signaling
pathways and interactions involved in the progression of atherosclerosis provide valuable information for the
development of approaches that delay or block the development of this disease.
Key words: atherogenesis; shear stress; transcription factor NF-κB; RelA expression; mechanosensitive receptors;
cell adhesion molecules; signaling pathways; mechanotransduction.
Collapse
Affiliation(s)
- E L Mishchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - V A Ivanisenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
35
|
Kaltschmidt C, Greiner JFW, Kaltschmidt B. The Transcription Factor NF-κB in Stem Cells and Development. Cells 2021; 10:2042. [PMID: 34440811 PMCID: PMC8391683 DOI: 10.3390/cells10082042] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/26/2022] Open
Abstract
NF-κB (nuclear factor kappa B) belongs to a family of transcription factors known to regulate a broad range of processes such as immune cell function, proliferation and cancer, neuroprotection, and long-term memory. Upcoming fields of NF-κB research include its role in stem cells and developmental processes. In the present review, we discuss one role of NF-κB in development in Drosophila, Xenopus, mice, and humans in accordance with the concept of evo-devo (evolutionary developmental biology). REL domain-containing proteins of the NF-κB family are evolutionarily conserved among these species. In addition, we summarize cellular phenotypes such as defective B- and T-cell compartments related to genetic NF-κB defects detected among different species. While NF-κB proteins are present in nearly all differentiated cell types, mouse and human embryonic stem cells do not contain NF-κB proteins, potentially due to miRNA-dependent inhibition. However, the mesodermal and neuroectodermal differentiation of mouse and human embryonic stem cells is hampered upon the repression of NF-κB. We further discuss NF-κB as a crucial regulator of differentiation in adult stem cells such as neural crest-derived and mesenchymal stem cells. In particular, c-REL seems to be important for neuronal differentiation and the neuroprotection of human adult stem cells, while RELA plays a crucial role in osteogenic and mesodermal differentiation.
Collapse
Affiliation(s)
- Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
36
|
Corilagin ameliorates atherosclerosis by regulating MMP-1, -2, and -9 expression in vitro and in vivo. Eur J Pharmacol 2021; 906:174200. [PMID: 34062170 DOI: 10.1016/j.ejphar.2021.174200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022]
Abstract
Corilagin is a polyphenol has been identified anti-inflammatory properties. However, the anti-atherosclerotic effects of corilagin are not well understood. Here, we evaluated the anti-atherosclerotic effects and the underlying mechanisms of corilagin. We also verified whether corilagin can reverse atherosclerosis by regulating matrix metalloproteinase (MMP)-1, -2, and -9 in vitro and in vivo. An atherosclerosis model was established by feeding minipigs a high-fat diet combined with balloon injury, and the effects of different concentrations of corilagin on common carotid artery atherosclerosis in minipigs were monitored. Murine RAW264.7 macrophages were cultured and induced with oxidized low-density lipoprotein; fluorescence microscopy revealed the nuclear translocation of NF-κB. Furthermore, MMP-1, -2, and -9 expression in common carotid artery plaques and cellular models was detected by immunohistochemistry, western blotting, and RT-PCR. The pathological results suggested that the vascular intima of the model control group was significantly thickened, a large amount of collagen fibers was deposited, endothelial cells were damaged and detached, and plaque and foam cell formation occurred to varying degrees on the arterial wall, with lipid deposition. Corilagin treatment significantly reduced the degree of injury in the common carotid artery and decreased the number of lipid plaques and foam cells. Additionally, corilagin downregulated MMP-1, -2, and -9 expression in the common carotid artery plaques and cellular model. Moreover, corilagin significantly inhibited NF-κB nuclear translocation in vitro. Overall, corilagin exerted substantial therapeutic effects on experimental atherosclerotic minipigs via the downregulation of MMP-1, -2, and -9 expression.
Collapse
|
37
|
Erythorbyl laurate suppresses TNF-α-induced adhesion of monocytes to the vascular endothelium. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
38
|
Koushki K, Shahbaz SK, Mashayekhi K, Sadeghi M, Zayeri ZD, Taba MY, Banach M, Al-Rasadi K, Johnston TP, Sahebkar A. Anti-inflammatory Action of Statins in Cardiovascular Disease: the Role of Inflammasome and Toll-Like Receptor Pathways. Clin Rev Allergy Immunol 2021; 60:175-199. [PMID: 32378144 PMCID: PMC7985098 DOI: 10.1007/s12016-020-08791-9] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is one type of cardiovascular disease (CVD) in which activation of the NLRP3 inflammasome and toll-like receptor (TLR) pathways is implicated. One of the most effective treatments for atherosclerosis is the use of statin medications. Recent studies have indicated that statins, in addition to their lipid-lowering effects, exert inhibitory and/or stimulatory effects on the NLRP3 inflammasome and TLRs. Some of the statins lead to activation of the inflammasome and subsequently cause secretion of IL-1β and IL-18. Thus, these actions may further aggravate the disease. On the other hand, some statins cause inhibition of the inflammasome or TLRs and along with lipid-lowering, help to improve the disease by reducing inflammation. In this article, we discuss these contradictory studies and the mechanisms of action of statins on the NLRP3 inflammasome and TLR pathways. The dose-dependent effects of statins on the NLRP3 complex are related to their chemistry, pharmacokinetic properties, and danger signals. Lipophilic statins have more pleiotropic effects on the NLRP3 complex in comparison to hydrophilic statins. Statins can suppress TLR4/MyD88/NF-ĸB signaling and cause an immune response shift to an anti-inflammatory response. Furthermore, statins inhibit the NF-ĸB pathway by decreasing the expression of TLRs 2 and 4. Statins are cost-effective drugs, which should have a continued future in the treatment of atherosclerosis due to both their immune-modulating and lipid-lowering effects.
Collapse
Affiliation(s)
- Khadijeh Koushki
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sanaz Keshavarz Shahbaz
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Deris Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat, Oman
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
39
|
Screening of Hub Genes Associated with Pulmonary Arterial Hypertension by Integrated Bioinformatic Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6626094. [PMID: 33816621 PMCID: PMC8010527 DOI: 10.1155/2021/6626094] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022]
Abstract
Background Pulmonary arterial hypertension (PAH) is a disease or pathophysiological syndrome which has a low survival rate with abnormally elevated pulmonary artery pressure caused by known or unknown reasons. In addition, the pathogenesis of PAH is not fully understood. Therefore, it has become an urgent matter to search for clinical molecular markers of PAH, study the pathogenesis of PAH, and contribute to the development of new science-based PAH diagnosis and targeted treatment methods. Methods In this study, the Gene Expression Omnibus (GEO) database was used to downloaded a microarray dataset about PAH, and the differentially expressed genes (DEGs) between PAH and normal control were screened out. Moreover, we performed the functional enrichment analyses and protein-protein interaction (PPI) network analyses of the DEGs. In addition, the prediction of miRNA and transcriptional factor (TF) of hub genes and construction miRNA-TF-hub gene network were performed. Besides, the ROC curve was used to evaluate the diagnostic value of hub genes. Finally, the potential drug targets for the 5 identified hub genes were screened out. Results 69 DEGs were identified between PAH samples and normal samples. GO and KEGG pathway analyses revealed that these DEGs were mostly enriched in the inflammatory response and cytokine-cytokine receptor interaction, respectively. The miRNA-hub genes network was conducted subsequently with 131 miRNAs, 7 TFs, and 5 hub genes (CCL5, CXCL12, VCAM1, CXCR1, and SPP1) which screened out via constructing the PPI network. 17 drugs interacted with 5 hub genes were identified. Conclusions Through bioinformatic analysis of microarray data sets, 5 hub genes (CCL5, CXCL12, VCAM1, CXCR1, and SPP1) were identified from DEGs between control samples and PAH samples. Studies showed that the five hub genes might play an important role in the development of PAH. These 5 hub genes might be potential biomarkers for diagnosis or targets for the treatment of PAH. In addition, our work also indicated that paying more attention on studies based on these 5 hub genes might help to understand the molecular mechanism of the development of PAH.
Collapse
|
40
|
Akhigbe R, Ajayi A. The impact of reactive oxygen species in the development of cardiometabolic disorders: a review. Lipids Health Dis 2021; 20:23. [PMID: 33639960 PMCID: PMC7916299 DOI: 10.1186/s12944-021-01435-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress, an alteration in the balance between reactive oxygen species (ROS) generation and antioxidant buffering capacity, has been implicated in the pathogenesis of cardiometabolic disorders (CMD). At physiological levels, ROS functions as signalling mediators, regulates various physiological functions such as the growth, proliferation, and migration endothelial cells (EC) and smooth muscle cells (SMC); formation and development of new blood vessels; EC and SMC regulated death; vascular tone; host defence; and genomic stability. However, at excessive levels, it causes a deviation in the redox state, mediates the development of CMD. Multiple mechanisms account for the rise in the production of free radicals in the heart. These include mitochondrial dysfunction and uncoupling, increased fatty acid oxidation, exaggerated activity of nicotinamide adenine dinucleotide phosphate oxidase (NOX), reduced antioxidant capacity, and cardiac metabolic memory. The purpose of this study is to discuss the link between oxidative stress and the aetiopathogenesis of CMD and highlight associated mechanisms. Oxidative stress plays a vital role in the development of obesity and dyslipidaemia, insulin resistance and diabetes, hypertension via various mechanisms associated with ROS-led inflammatory response and endothelial dysfunction.
Collapse
Affiliation(s)
- Roland Akhigbe
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo State Nigeria
- Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun State Nigeria
- Department of Chemical Sciences, Kings University, Odeomu, Osun Nigeria
| | - Ayodeji Ajayi
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo State Nigeria
| |
Collapse
|
41
|
Kizilay Mancini O, Huynh DN, Menard L, Shum-Tim D, Ong H, Marleau S, Colmegna I, Servant MJ. Ex vivo Ikkβ ablation rescues the immunopotency of mesenchymal stromal cells from diabetics with advanced atherosclerosis. Cardiovasc Res 2021; 117:756-766. [PMID: 32339220 PMCID: PMC7898947 DOI: 10.1093/cvr/cvaa118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Diabetes is a conventional risk factor for atherosclerotic cardiovascular disease and myocardial infarction (MI) is the most common cause of death among these patients. Mesenchymal stromal cells (MSCs) in patients with type 2 diabetes mellitus (T2DM) and atherosclerosis have impaired ability to suppress activated T-cells (i.e. reduced immunopotency). This is mediated by an inflammatory shift in MSC-secreted soluble factors (i.e. pro-inflammatory secretome) and can contribute to the reduced therapeutic effects of autologous T2DM and atherosclerosis-MSC post-MI. The signalling pathways driving the altered secretome of atherosclerosis- and T2DM-MSC are unknown. Specifically, the effect of IκB kinase β (IKKβ) modulation, a key regulator of inflammatory responses, on the immunopotency of MSCs from T2DM patients with advanced atherosclerosis has not been studied. METHODS AND RESULTS MSCs were isolated from adipose tissue obtained from patients with (i) atherosclerosis and T2DM (atherosclerosis+T2DM MSCs, n = 17) and (ii) atherosclerosis without T2DM (atherosclerosis MSCs, n = 17). MSCs from atherosclerosis+T2DM individuals displayed an inflammatory senescent phenotype and constitutively expressed active forms of effectors of the canonical IKKβ nuclear factor-κB transcription factors inflammatory pathway. Importantly, this constitutive pro-inflammatory IKKβ signature resulted in an altered secretome and impaired in vitro immunopotency and in vivo healing capacity in an acute MI model. Notably, treatment with a selective IKKβ inhibitor or IKKβ knockdown (KD) (clustered regularly interspaced short palindromic repeats/Cas9-mediated IKKβ KD) in atherosclerosis+T2DM MSCs reduced the production of pro-inflammatory secretome, increased survival, and rescued their immunopotency both in vitro and in vivo. CONCLUSIONS Constitutively active IKKβ reduces the immunopotency of atherosclerosis+T2DM MSC by changing their secretome composition. Modulation of IKKβ in atherosclerosis+T2DM MSCs enhances their myocardial repair ability.
Collapse
Affiliation(s)
- Ozge Kizilay Mancini
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - David N Huynh
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Liliane Menard
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Dominique Shum-Tim
- Division of Cardiac Surgery Department of Surgery, McGill University, Montreal, QC H4A 3J1, Canada
- Division of Surgical Research, Department of Surgery, McGill University, Montreal, QC H4A 3J1, Canada
| | - Huy Ong
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Ines Colmegna
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Marc J Servant
- Faculty of Pharmacy, University of Montreal, C.P. 6128, Succursale Centre-Ville, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
42
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
43
|
Anti-inflammatory Effects of Statins in Lung Vascular Pathology: From Basic Science to Clinical Trials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:33-56. [PMID: 33788186 DOI: 10.1007/978-3-030-63046-1_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
HMG-CoA reductase inhibitors (or statins) are cholesterol-lowering drugs and are among the most widely prescribed medications in the United States. Statins exhibit pleiotropic effects that extend beyond cholesterol reduction including anti-atherosclerotic, antiproliferative, anti-inflammatory, and antithrombotic effects. Over the last 20 years, statins have been studied and examined in pulmonary vascular disorders, including both chronic pulmonary vascular disease such as pulmonary hypertension, and acute pulmonary vascular endothelial injury such as acute lung injury. In both research and clinical settings, statins have demonstrated promising vascular protection through modulation of the endothelium, attenuation of vascular leak, and promotion of endothelial repair following lung inflammation. This chapter provides a summary of the rapidly changing literature, summarizes the anti-inflammatory mechanism of statins on pulmonary vascular disorders, and explores clinical evidence for statins as a potential therapeutic approach to modulation of the endothelium as well as a means to broaden our understanding of pulmonary vasculopathy pathophysiology.
Collapse
|
44
|
Kamperidis N, Kamperidis V, Zegkos T, Kostourou I, Nikolaidou O, Arebi N, Karvounis H. Atherosclerosis and Inflammatory Bowel Disease-Shared Pathogenesis and Implications for Treatment. Angiology 2020; 72:303-314. [PMID: 33601945 DOI: 10.1177/0003319720974552] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Atherosclerosis and inflammatory bowel disease (IBD) are often regarded as 2 distinct entities. The commonest manifestation of atherosclerosis is ischemic heart disease (IHD), and an association between IHD and IBD has been reported. Atherosclerosis and IBD share common pathophysiological mechanisms in terms of their genetics, immunology, and contributing environmental factors. Factors associated with atherosclerosis are implicated in the development of IBD and vice versa. Therefore, treatments targeting the common pathophysiology pathways may be effective in both conditions. The current review considers the pathophysiological pathways that are shared between the 2 conditions and discusses the implications for treatment and research.
Collapse
Affiliation(s)
- Nikolaos Kamperidis
- 3749St Mark's Hospital, Harrow, London, United Kingdom.,* Nikolaos Kamperidis and Vasileios Kamperidis are sharing first authorship
| | - Vasileios Kamperidis
- 1st Cardiology Department, 37788AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.,* Nikolaos Kamperidis and Vasileios Kamperidis are sharing first authorship
| | - Thomas Zegkos
- 1st Cardiology Department, 37788AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Olga Nikolaidou
- Radiology Department, Pananikolaou General Hospital, Thessaloniki, Greece
| | - Naila Arebi
- 3749St Mark's Hospital, Harrow, London, United Kingdom
| | - Haralambos Karvounis
- 1st Cardiology Department, 37788AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
45
|
Karunakaran D, Nguyen MA, Geoffrion M, Vreeken D, Lister Z, Cheng HS, Otte N, Essebier P, Wyatt H, Kandiah JW, Jung R, Alenghat FJ, Mompeon A, Lee R, Pan C, Gordon E, Rasheed A, Lusis AJ, Liu P, Matic LP, Hedin U, Fish JE, Guo L, Kolodgie F, Virmani R, van Gils JM, Rayner KJ. RIPK1 Expression Associates With Inflammation in Early Atherosclerosis in Humans and Can Be Therapeutically Silenced to Reduce NF-κB Activation and Atherogenesis in Mice. Circulation 2020; 143:163-177. [PMID: 33222501 DOI: 10.1161/circulationaha.118.038379] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic activation of the innate immune system drives inflammation and contributes directly to atherosclerosis. We previously showed that macrophages in the atherogenic plaque undergo RIPK3 (receptor-interacting serine/threonine-protein kinase 3)-MLKL (mixed lineage kinase domain-like protein)-dependent programmed necroptosis in response to sterile ligands such as oxidized low-density lipoprotein and damage-associated molecular patterns and that necroptosis is active in advanced atherosclerotic plaques. Upstream of the RIPK3-MLKL necroptotic machinery lies RIPK1 (receptor-interacting serine/threonine-protein kinase 1), which acts as a master switch that controls whether the cell undergoes NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells)-dependent inflammation, caspase-dependent apoptosis, or necroptosis in response to extracellular stimuli. We therefore set out to investigate the role of RIPK1 in the development of atherosclerosis, which is driven largely by NF-κB-dependent inflammation at early stages. We hypothesize that, unlike RIPK3 and MLKL, RIPK1 primarily drives NF-κB-dependent inflammation in early atherogenic lesions, and knocking down RIPK1 will reduce inflammatory cell activation and protect against the progression of atherosclerosis. METHODS We examined expression of RIPK1 protein and mRNA in both human and mouse atherosclerotic lesions, and used loss-of-function approaches in vitro in macrophages and endothelial cells to measure inflammatory responses. We administered weekly injections of RIPK1 antisense oligonucleotides to Apoe-/- mice fed a cholesterol-rich (Western) diet for 8 weeks. RESULTS We find that RIPK1 expression is abundant in early-stage atherosclerotic lesions in both humans and mice. Treatment with RIPK1 antisense oligonucleotides led to a reduction in aortic sinus and en face lesion areas (47.2% or 58.8% decrease relative to control, P<0.01) and plasma inflammatory cytokines (IL-1α [interleukin 1α], IL-17A [interleukin 17A], P<0.05) in comparison with controls. RIPK1 knockdown in macrophages decreased inflammatory genes (NF-κB, TNFα [tumor necrosis factor α], IL-1α) and in vivo lipopolysaccharide- and atherogenic diet-induced NF-κB activation. In endothelial cells, knockdown of RIPK1 prevented NF-κB translocation to the nucleus in response to TNFα, where accordingly there was a reduction in gene expression of IL1B, E-selectin, and monocyte attachment. CONCLUSIONS We identify RIPK1 as a central driver of inflammation in atherosclerosis by its ability to activate the NF-κB pathway and promote inflammatory cytokine release. Given the high levels of RIPK1 expression in human atherosclerotic lesions, our study suggests RIPK1 as a future therapeutic target to reduce residual inflammation in patients at high risk of coronary artery disease.
Collapse
Affiliation(s)
- Denuja Karunakaran
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Dianne Vreeken
- Leiden University Medical Center, The Netherlands (D.V., J.M.v.G.)
| | - Zachary Lister
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Henry S Cheng
- Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C.)
| | - Nicola Otte
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Patricia Essebier
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Joshua W Kandiah
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Jung
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Francis J Alenghat
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Ana Mompeon
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, CA (R.L.)
| | - Calvin Pan
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Emma Gordon
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Aldons J Lusis
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Peter Liu
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Ljubica Perisic Matic
- Vascular Surgery Division, Department of Molecular Medicine and Surgery, Karolinska Institute, Sweden (L.P.M.)
| | | | - Jason E Fish
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Liang Guo
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Frank Kolodgie
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Renu Virmani
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | | | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| |
Collapse
|
46
|
Wang J, Hu X, Hu X, Gao F, Li M, Cui Y, Wei X, Qin Y, Zhang C, Zhao Y, Gao Y. MicroRNA-520c-3p targeting of RelA/p65 suppresses atherosclerotic plaque formation. Int J Biochem Cell Biol 2020; 131:105873. [PMID: 33166679 DOI: 10.1016/j.biocel.2020.105873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/19/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease, and it's the leading cause of death worldwide. Dysregulation of microRNAs (miRNAs) has been found to be associated with atherosclerosis. miR-520c-3p has been implicated in several types of cancer. However, little is known about the role of miR-520c-3p in atherosclerosis. In this study, we found that miR-520c-3p agomir decreased atherosclerotic plaque size, collagen content, the quantity of PCNA-positive cell and RelA/p65 expression of vascular smooth muscle cells (VSMCs) in the aortic valve of apoE-/- mice in vivo. The possible mechanisms of the protective effects of miR-520c-3p on atherosclerotic mice were then investigated in VSMCs. in vitro experiments showed that miR-520c-3p expressions were significantly reduced in human aortic vascular smooth muscle cell (HASMCs) treated with platelet-derived growth factor (PDGF-BB). miR-520c-3p mimics repress PDGF-BB-mediated the proliferation, migration and decrease in the percentage of cells in G2/M phase, which was associated with downregulation of RelA/p65. Mechanistically, miRNA pull-down, luciferase reporter and mRNA stability assays confirmed miR-520c-3p mimics was able to directly target 3'-UTR of RelA/p65 mRNA and decreased half-life of RelA/p65 mRNA in HASMCs. Overexpression of RelA/p65 reversed the inhibition of cell proliferation induced by miR-520c-3p mimics in HASMCs. In conclusion, our findings suggest that miR-520c-3p inhibits PDGF-BB-mediated the proliferation and migration of HASMCs by targeting RelA/p65, which may provide potential therapeutic strategies in atherosclerosis treatment.
Collapse
MESH Headings
- Animals
- Aortic Valve/metabolism
- Aortic Valve/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/therapy
- Becaplermin/pharmacology
- Cell Line
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Disease Models, Animal
- Gene Expression Regulation
- Genes, Reporter
- Humans
- Luciferases/genetics
- Luciferases/metabolism
- Mice
- Mice, Knockout, ApoE
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/therapy
- Primary Cell Culture
- Signal Transduction
- Transcription Factor RelA/genetics
- Transcription Factor RelA/metabolism
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
| | - Yuanhua Qin
- Department of Parasite, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chenghong Zhang
- Morphological Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China.
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China; Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China.
| |
Collapse
|
47
|
Che Man R, Sulaiman N, Ishak MF, Bt Hj Idrus R, Abdul Rahman MR, Yazid MD. The Effects of Pro-Inflammatory and Anti-Inflammatory Agents for the Suppression of Intimal Hyperplasia: An Evidence-Based Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217825. [PMID: 33114632 PMCID: PMC7672569 DOI: 10.3390/ijerph17217825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Anti-atherogenic therapy is crucial in halting the progression of inflammation-induced intimal hyperplasia. The aim of this concise review was to methodically assess the recent findings of the different approaches, mainly on the recruitment of chemokines and/or cytokine and its effects in combating the intimal hyperplasia caused by various risk factors. Pubmed and Scopus databases were searched, followed by article selection based on pre-set inclusion and exclusion criteria. The combination of keywords used were monocyte chemoattractant protein-1 OR MCP-1 OR TNF-alpha OR TNF-α AND hyperplasia OR intimal hyperplasia OR neointimal hyperplasia AND in vitro. These keywords combination was incorporated in the study and had successfully identified 77 articles, with 22 articles were acquired from Pubmed, whereas 55 articles were obtained from Scopus. However, after title screening, only twelve articles meet the requirements of defined inclusion criteria. We classified the data into 4 different approaches, i.e., utilisation of natural product, genetic manipulation and protein inhibition, targeted drugs in clinical setting, and chemokine and cytokines induction. Most of the articles are working on genetic manipulation targeted on specific pathway to inhibit the pro-inflammatory factors expression. We also found that the utilisation of chemokine- and cytokine-related treatments are emerging throughout the years. However, there is no study utilising the combination of approaches that might give a better outcome in combating intimal hyperplasia. Hopefully, this concise review will provide an insight regarding the usage of different novel approaches in halting the progression of intimal hyperplasia, which serves as a key factor for the development of atherosclerosis in cardiovascular disease.
Collapse
Affiliation(s)
- Rohaina Che Man
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-6995
| |
Collapse
|
48
|
Shi C, Deng J, Chiu M, Chen YX, O'Brien ER. Heat shock protein 27 immune complex altered signaling and transport (ICAST): Novel mechanisms of attenuating inflammation. FASEB J 2020; 34:14287-14301. [PMID: 32888229 DOI: 10.1096/fj.202001389rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 01/04/2023]
Abstract
Blood levels of heat shock protein (HSP27) and natural IgG auto-antibodies to HSP27 (AAbs) are higher in healthy controls compared to cardiovascular disease patients. Vaccination of mice with recombinant HSP25 (rHSP25, murine ortholog of human rHSP27) increased AAb levels, attenuated atherogenesis and reduced plaque inflammation and cholesterol content. We sought to determine if the HSP27 immune complex (IC) altered MΦ inflammation signaling (Toll Like Receptor 4; TLR4), and scavenger receptors involved in cholesterol uptake (SR-AI, CD-36). Combining a validated polyclonal IgG anti-HSP27 antibody (PAb) with rHSP27 enhanced binding to THP-1 MΦ cell membranes and activation of NF-κB signaling via TLR4, competing away LPS and effecting an anti-inflammatory cytokine profile. Similarly, adding the PAb with rHSP27 enhanced binding to SR-AI and CD-36, as well as lowered oxLDL binding in HEK293 cells separately transfected with SR-AI and CD-36, or THP-1 MΦ. Finally, the PAb enhanced the uptake and internalization of rHSP27 in THP-1 MΦ. Thus, the HSP27 IC potentiates HSP27 cell membrane signaling with receptors involved in modulating inflammation and cholesterol uptake, as well as HSP27 internalization. Going forward, we are focusing on the development of HSP27 Immune Complex Altered Signaling and Transport (ICAST) as a means of modulating inflammation.
Collapse
Affiliation(s)
- Chunhua Shi
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Jingti Deng
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Michael Chiu
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Yong-Xiang Chen
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Edward R O'Brien
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| |
Collapse
|
49
|
Li T, Safitri M, Zhang K, Wang Y, Huang L, Zhu Y, Daniel R, Wu LJ, Qiu J, Wang G. Downregulation of G3BP2 reduces atherosclerotic lesions in ApoE -/- mice. Atherosclerosis 2020; 310:64-74. [PMID: 32919187 DOI: 10.1016/j.atherosclerosis.2020.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/28/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is mainly caused by stress in arterial microenvironments, which results in the formation of stress granules as a consequence of the stress response. As the core protein of stress granules, GTPase-activating protein (SH3 domain)-binding protein 2 (G3BP2) is known to play pivotal roles in tumour initiation, viral infection and Alzheimer's disease, but the role of G3BP2 in atherosclerosis development is poorly understood. Previous studies have shown that vaccination with epitopes from self-antigens could reduce atherosclerotic lesions. Here, we investigated the effect of immunizing ApoE-/- mice with G3BP2 peptides, and whether this immunization exerted an anti-atherogenic effect. METHODS AND RESULTS In our study, ApoE-/- mice were fed a high-fat diet for 12 weeks from 8 to 20 weeks of age. Then, using a repetitive multiple site strategy, the mice were immunized with a Keyhole limpet haemocyanin (KLH) conjugated G3BP2 peptide for 2 weeks from weeks 16 to 18. High levels of G3BP2 antibodies were detectable before sacrifice. Histological analyses showed that the number of atherosclerotic lesions in ApoE-/- mice was significantly reduced following G3BP2 immunotherapy. The levels of pro-inflammatory cytokines and macrophages were also greatly decreased, while the collagen content of the plaques showed significant increase. Furthermore, knocking down G3BP2 in ApoE-/- mice reduced the number of lesions compared to ApoE-/- mice fed a high-fat diet for eight weeks. In vitro studies demonstrated that G3BP2 regulated ox-LDL-induced inflammation in HUVECs via controlling the localization of IκBα. CONCLUSIONS Immunization with the G3BP2 peptide antigen or knocking down of G3BP2 significantly decreased early atherosclerotic plaques in the ApoE-/- mouse model of atherosclerosis. G3BP2 is a promising potential target for atherosclerosis therapy.
Collapse
Affiliation(s)
- Tianhan Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Maharani Safitri
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Kang Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Lu Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Yuan Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Richard Daniel
- Biosciences Institute, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK.
| | - Ling Juan Wu
- Biosciences Institute, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK.
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
50
|
Reddy AT, Lakshmi SP, Maruthi Prasad E, Varadacharyulu NC, Kodidhela LD. Epigallocatechin gallate suppresses inflammation in human coronary artery endothelial cells by inhibiting NF-κB. Life Sci 2020; 258:118136. [PMID: 32726662 DOI: 10.1016/j.lfs.2020.118136] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022]
Abstract
The endothelium is a critical regulator of vascular homeostasis, controlling vascular tone and permeability as well as interactions of leukocytes and platelets with blood vessel walls. Consequently, endothelial dysfunction featuring inflammation and reduced vasodilation are considered central to cardiovascular disease (CVD) pathogenesis and have become a therapeutic area of focus. Type II endothelial cell (EC) activation by stress-related stimuli such as tumor necrosis factor-α (TNF-α) initiates the nuclear factor-κB (NF-κB) signaling pathway, a master regulator of inflammatory responses. Because dysregulated NF-κB signaling has been tightly linked to several CVDs, EC-specific inhibition of NF-κB represents an attractive pharmacological strategy. As accumulating evidence highlights the clinical benefits of tea catechin for multiple diseases including CVDs, we sought to determine whether the tea catechin epigallocatechin gallate (EGCG) that displays antioxidative, anti-inflammatory, hypolipidemic, anti-thrombogenic, and anti-hypertensive properties offers protection against CVDs by suppressing the canonical NF-κB pathway. Our findings indicate that EGCG downregulates multiple components of the TNF-α-induced NF-κB signaling pathway and thereby reduces the consequent increase in inflammatory gene transcription and protein expression. Furthermore, EGCG blocked type II EC activation, evidenced by diminished EC leakage and monocyte adhesion in EGCG-treated cells. In summary, our study advances knowledge of EGCG's anti-inflammatory effects on the NF-κB pathway and hence its benefits on endothelial health, supporting its therapeutic potential for CVDs.
Collapse
Affiliation(s)
- Aravind T Reddy
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India.
| | - Sowmya P Lakshmi
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India
| | - E Maruthi Prasad
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - N Ch Varadacharyulu
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India
| | - Lakshmi Devi Kodidhela
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India
| |
Collapse
|