1
|
Sun XF, Luo WC, Huang SQ, Zheng YJ, Xiao L, Zhang ZW, Liu RH, Zhong ZW, Song JQ, Nan K, Qiu ZX, Zhong J, Miao CH. Immune-cell signatures of persistent inflammation, immunosuppression, and catabolism syndrome after sepsis. MED 2025; 6:100569. [PMID: 39824181 DOI: 10.1016/j.medj.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/13/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Management of persistent inflammation, immunosuppression, and catabolism syndrome (PICS) after sepsis remains challenging for patients in the intensive care unit, experiencing poor quality of life and death. However, immune-cell signatures in patients with PICS after sepsis remain unclear. METHODS We determined immune-cell signatures of PICS after sepsis at single-cell resolution. Murine cecal ligation and puncture models of PICS were applied for validation. FINDINGS Immune functions of two enriched monocyte subpopulations, Mono1 and Mono4, were suppressed substantially in patients with sepsis and were partially restored in patients with PICS after sepsis and exhibited immunosuppressive and pro-apoptotic effects on B and CD8T cells. Patients with PICS and sepsis had reduced naive and memory B cells and proliferated plasma cells. Besides, naive and memory B cells in patients with PICS showed an active antigen processing and presentation gene signature compared to those with sepsis. PICS patients with better prognoses exhibited more active memory B cells and IGHA1-plasma cells. CD8TEMRA displayed signs of proliferation and immune dysfunction in the PICS-death group in contrast with the PICS-alive group. Megakaryocytes proliferation was more pronounced in patients with PICS and sepsis than in healthy controls, with notable changes in the anti-inflammatory and immunomodulatory effects observed in patients with PICS and verified in mice models. CONCLUSIONS Our study evaluated PICS after sepsis at the single-cell level, identifying the heterogeneity present within immune-cell subsets, facilitating the prediction of disease progression and the development of effective intervention. FUNDING This work was supported by the National Natural Science Foundation of China, Shanghai Municipal Health Commission "Yiyuan New Star" Youth Medical Talent Cultivating Program, and Shanghai Clinical Research Center for Anesthesiology.
Collapse
Affiliation(s)
- Xing-Feng Sun
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200438, China
| | - Wen-Chen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200438, China
| | - Yi-Jun Zheng
- Department of Critical Care and Pain Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lei Xiao
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhong-Wei Zhang
- Department of Critical Care and Pain Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Rong-Hua Liu
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zi-Wen Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jie-Qiong Song
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Zhi-Xin Qiu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China.
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China.
| | - Chang-Hong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Laboratory of Perioperative Stress and Protection, Shanghai 200032, China.
| |
Collapse
|
2
|
Lau C, Primus CP, Shabbir A, Chhetri I, Ono M, Masucci M, Bin Noorany Aubdool MA, Amarin J, Hamers AJ, Khan Z, Kumar NA, Montalvo Moreira SA, Nuredini G, Osman M, Whitear C, Godec T, Kapil V, Massimo G, Khambata RS, Rathod KS, Ahluwalia A. Accelerating inflammatory resolution in humans to improve endothelial function and vascular health: Targeting the non-canonical pathway for NO. Redox Biol 2025; 82:103592. [PMID: 40209616 PMCID: PMC12005330 DOI: 10.1016/j.redox.2025.103592] [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: 01/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chronic cardiovascular diseases (CVD) are characterised by low-grade systemic inflammation in part due to reduced nitric oxide (NO) bioavailability associated with endothelial dysfunction. Bioavailability of NO can be enhanced by activation of the non-canonical pathway, through increased dietary inorganic nitrate consumption with the potential to attenuate inflammation. METHODS We sought to determine whether dietary inorganic nitrate influences the inflammatory response in models of localised (cantharidin-induced blisters) and systemic inflammation (typhoid vaccine), in healthy male volunteers and conducted two clinical trials; Blister-NITRATE and Typhoid-NITRATE respectively. RESULTS We show that dietary nitrate attenuates endothelial dysfunction following typhoid vaccine administration and accelerates resolution of cantharidin-induced blisters. Both phenomena were associated with an increased level of pro-resolving mediators consequent to a reduction in the expression and activity of pro-inflammatory monocytes. Moreover, we show that leukocytes of the monocyte lineage express the nitrite reductase XOR, that may drive localised nitrite reduction to elevate NO (and cGMP) to drive the protective phenotype. CONCLUSIONS Inorganic nitrate improves endothelial function in the setting of systemic inflammation. Whilst the immediate inflammatory response appeared unaffected by inorganic nitrate treatment, during the resolution phase of the acute inflammatory response lower levels of pro-inflammatory classical inflammatory and intermediate monocytes and attenuated levels of inflammatory cytokines and chemokines were evident. We propose that this reflects a pro-resolution phenotype that may be of potential therapeutic benefit in patients with established CVD. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; unique identifiers NCT02715635, NCT03183830.
Collapse
Affiliation(s)
- Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christopher P Primus
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Masucci
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad Aadil Bin Noorany Aubdool
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Amarin
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jp Hamers
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zara Khan
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nitin Ajit Kumar
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Gani Nuredini
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Miski Osman
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Charlotte Whitear
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tom Godec
- Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vikas Kapil
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Cardiology, Barts Heart Centre, 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
3
|
Park HM, Kim CL, Kong D, Heo SH, Park HJ. Innovations in Vascular Repair from Mechanical Intervention to Regenerative Therapies. Tissue Eng Regen Med 2025:10.1007/s13770-024-00700-x. [PMID: 39921820 DOI: 10.1007/s13770-024-00700-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Vascular diseases, including atherosclerosis and thrombosis, are leading causes of morbidity and mortality worldwide, often resulting in vessel stenosis that impairs blood flow and leads to severe clinical outcomes. Traditional mechanical interventions, such as balloon angioplasty and bare-metal stents, provided initial solutions but were limited by restenosis and thrombosis. The advent of drug-eluting stents improved short-term outcomes by inhibiting vascular smooth muscle cell proliferation, however, they faced challenges including delayed reendothelialization and late-stage thrombosis. METHODS This review highlights the progression from mechanical to biological interventions in treating vascular stenosis and underscores the need for integrated approaches that combine mechanical precision with regenerative therapies. RESULTS To address long-term complications, bioresorbable stents were developed to provide temporary scaffolding that gradually dissolves, yet they still encounter challenges with mechanical integrity and optimal degradation rates. Consequently, emerging therapies now focus on biological approaches, such as gene therapy, extracellular vesicle treatments, and cell therapies, that aim to promote vascular repair at the cellular level. These strategies offer the potential for true vascular regeneration by enhancing endothelialization, modulating immune responses, and stimulating angiogenesis. CONCLUSION Integrating mechanical precision with regenerative biological therapies offers a promising future for treating vascular stenosis. A comprehensive approach combining these modalities could achieve sustainable vascular health.
Collapse
Affiliation(s)
- Hye-Min Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Chae-Lin Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Dasom Kong
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Seon-Hee Heo
- Department of Surgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Hyun-Ji Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
- Advanced College of Bio-Convergence Engineering, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
4
|
Zhang X, Liu S, Kong F, Shu L, Li Y, Wang D, Li L. Acidic polysaccharide from Ganoderma tsugae: Structural characterization and antiatherosclerotic related to macrophage polarization. Food Res Int 2025; 203:115913. [PMID: 40022418 DOI: 10.1016/j.foodres.2025.115913] [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: 10/24/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
Herein, a water-soluble Ganoderma tsugae acidic polysaccharide (GTP-2) was isolated and purified from the fruiting bodies of G. tsugae. GTP-2 has a molecular weight of 13.059 kDa, composed of →3)-β-d-Glcp-(1 → 3)-β-d-Glcp-(1 → 4)-GlcpUA-(1 → 4)-β-d-Glcp-1→ glucan backbone and branches ending with β-d-Glcp-(1 → 6)-β-d-Glcp-(1→, which is attached at C6 of →3,6)-β-d-Glcp-(1→. Subsequently, the antiatherosclerotic activity of GTP-2 was examined in apolipoprotein E deficient (ApoE-/-) mice fed with high-fat diet, and its potential mechanism of action was investigated. GTP-2 ameliorated blood lipid levels (total cholesterol, triglycerides, and low-density lipoprotein), while improving the serum levels of high-density lipoprotein. Furthermore, GTP-2 alleviated the atherosclerotic lesions and reduced levels of inflammatory cytokines. Analysis of the gut microbiota revealed that GTP-2 enhanced the abundance of beneficial bacteria (Lactobacillu and Akkermansia). The serum metabolite composition was further altered, with a significant reduction in octadecanoic acid level. GTP-2 regulated the nuclear factor kappa-B signaling pathway by inhibiting macrophage polarization to M1 phenotype. Collectively, these findings support the potential use of GTP-2 as an antiatherosclerotic therapy.
Collapse
Affiliation(s)
- Xin Zhang
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China.
| | - Shuai Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Lili Shu
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China.
| | - Yu Li
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866 China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118 China; College of Plant Protection, Jilin Agricultural University, Changchun 130118 China.
| |
Collapse
|
5
|
Khan AW, Jandeleit-Dahm KAM. Atherosclerosis in diabetes mellitus: novel mechanisms and mechanism-based therapeutic approaches. Nat Rev Cardiol 2025:10.1038/s41569-024-01115-w. [PMID: 39805949 DOI: 10.1038/s41569-024-01115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Atherosclerosis is a disease of large and medium arteries that can lead to life-threatening cardiovascular and cerebrovascular consequences, such as myocardial infarction and stroke. Moreover, atherosclerosis is a major contributor to cardiovascular-related mortality in individuals with diabetes mellitus. Diabetes aggravates the pathobiological mechanisms that underlie the development of atherosclerosis. Currently available anti-atherosclerotic drugs or strategies solely focus on optimal control of systemic risk factors, including hyperglycaemia and dyslipidaemia, but do not adequately target the diabetes-exacerbated mechanisms of atherosclerotic cardiovascular disease, highlighting the need for targeted, mechanism-based therapies. This Review focuses on emerging pathological mechanisms and related novel therapeutic targets in atherosclerotic cardiovascular disease in patients with diabetes.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Karin A M Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| |
Collapse
|
6
|
Gao Y, Ye F, Dong Y, Wang T, Xiong L, Chen T, Wang Y, Liu X, Zhang Y, Qiu Z, Jiang J, Liu X, Hu Q, Zhang C. Salvianic acid A ameliorates atherosclerosis through metabolic-dependent anti-EndMT pathway and repression of TGF-β/ALK5 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156307. [PMID: 39740380 DOI: 10.1016/j.phymed.2024.156307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/25/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) has been identified as a key factor to the initiation and progression of the pathogenesis of atherosclerosis (AS). Salvianic acid A (SAAS) is the primary water-soluble bioactive ingredient found in Salvia miltiorrhiza, is renowned for its therapeutic effects on cardiovascular diseases. However, the efficacy and mechanisms of SAAS in treating EndMT-induced AS remain underexplored. PURPOSE This study aimed to investigate the role SAAS in reversing EndMT process to impede AS development. METHODS We used a murine model of cholesterol-rich and high-fat diet-induced AS in ApoE-/- mice to evaluate the effect of SAAS on EndMT during AS progression in vivo. The biological effects of SAAS on EndMT-induced HUVEC cells were also detected by transcriptome sequencing (RNA-seq). Mechanistic exploration was carried out using omics data mining and screening, gene knockout experiments, gene expression, protein expression, and localization of key gene expression in animal lesion areas. RESULTS We found that SAAS treatment significantly alleviated EndMT injury in the AS mice model and also improved aortic root lesions and dyslipidemia. Furthermore, pre-treatment with SAAS effectively inhibited the EndMT in HUVEC cells, as evidenced by maintained endothelial cell morphology and reduced cell migration ability, as well as elevated CD31 and decreased α-SMA. RNA sequencing data indicated that key differentially expressed genes were mainly enriched in metabolism-related and TGF-β receptor signaling pathways. The metabolic regulator PDK4 and profibrotic TGF-β receptor ALK5 were identified specifically. Subsequently, RT-qPCR and western blot results demonstrated that SAAS notably increased metabolic regulator PDK4 and decreased profibrotic TGF-β receptor ALK5 in EndMT-induced HUVEC cells. Moreover, siRNA-directed PDK4 inhibition resulted in EndMT induction and SAAS mediated the suppression of EndMT in a PDK4-dependent manner. Additionally, SAAS partially reduced the TGF-β receptor ALK5 expression. Furthermore, ApoE-/- AS mice with SAAS treatment displayed downregulation of ALK5 and upregulation of PDK4 with reduced EndMT during AS. CONCLUSION This investigation demonstrated that SAAS improved AS through metabolic-dependent anti-EndMT pathway and repression of profibrotic TGF-β receptor signaling, thereby providing SAAS as a promising therapeutic candidate for managing AS and EndMT-related disorders.
Collapse
Affiliation(s)
- Yijun Gao
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Fei Ye
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Yafen Dong
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201200, PR China
| | - Tingfang Wang
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Liyan Xiong
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Ting Chen
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Yun Wang
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Xiaoyan Liu
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Yunan Zhang
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Zheng Qiu
- Shenzhen Medicines and Health Products IMP. & EXP. Co., Ltd, Shenzhen, PR China
| | - Jianfang Jiang
- Department of Infection Control, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, West Lake Rd 38, Hangzhou, 310009, PR China.
| | - Xijun Liu
- Shanghai 411 Hospital, Shanghai, 200081, PR China.
| | - Qingxun Hu
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China; Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, PR China.
| | - Chuan Zhang
- Shanghai 411 Hospital, School of Medicine, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
7
|
Annink ME, Kraaijenhof JM, Stroes ESG, Kroon J. Moving from lipids to leukocytes: inflammation and immune cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1446758. [PMID: 39161593 PMCID: PMC11330886 DOI: 10.3389/fcell.2024.1446758] [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: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most important cause of morbidity and mortality worldwide. While it is traditionally attributed to lipid accumulation in the vascular endothelium, recent research has shown that plaque inflammation is an important additional driver of atherogenesis. Though clinical outcome trials utilizing anti-inflammatory agents have proven promising in terms of reducing ASCVD risk, it is imperative to identify novel actionable targets that are more specific to atherosclerosis to mitigate adverse effects associated with systemic immune suppression. To that end, this review explores the contributions of various immune cells from the innate and adaptive immune system in promoting and mitigating atherosclerosis by integrating findings from experimental studies, high-throughput multi-omics technologies, and epidemiological research.
Collapse
Affiliation(s)
- Maxim E. Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
| |
Collapse
|
8
|
Ceccherini E, Persiani E, Cabiati M, Guiducci L, Del Ry S, Gisone I, Falleni A, Cecchettini A, Vozzi F. A Dynamic Cellular Model as an Emerging Platform to Reproduce the Complexity of Human Vascular Calcification In Vitro. Int J Mol Sci 2024; 25:7427. [PMID: 39000533 PMCID: PMC11242604 DOI: 10.3390/ijms25137427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Vascular calcification (VC) is a cardiovascular disease characterized by calcium salt deposition in vascular smooth muscle cells (VSMCs). Standard in vitro models used in VC investigations are based on VSMC monocultures under static conditions. Although these platforms are easy to use, the absence of interactions between different cell types and dynamic conditions makes these models insufficient to study key aspects of vascular pathophysiology. The present study aimed to develop a dynamic endothelial cell-VSMC co-culture that better mimics the in vivo vascular microenvironment. A double-flow bioreactor supported cellular interactions and reproduced the blood flow dynamic. VSMC calcification was stimulated with a DMEM high glucose calcification medium supplemented with 1.9 mM NaH2PO4/Na2HPO4 (1:1) for 7 days. Calcification, cell viability, inflammatory mediators, and molecular markers (SIRT-1, TGFβ1) related to VSMC differentiation were evaluated. Our dynamic model was able to reproduce VSMC calcification and inflammation and evidenced differences in the modulation of effectors involved in the VSMC calcified phenotype compared with standard monocultures, highlighting the importance of the microenvironment in controlling cell behavior. Hence, our platform represents an advanced system to investigate the pathophysiologic mechanisms underlying VC, providing information not available with the standard cell monoculture.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Elisa Persiani
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Manuela Cabiati
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Letizia Guiducci
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Silvia Del Ry
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Ilaria Gisone
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| | - Alessandra Falleni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Federico Vozzi
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.C.); (E.P.); (M.C.); (L.G.); (S.D.R.); (I.G.); (A.C.)
| |
Collapse
|
9
|
Wang D, Sun Z, Yin Y, Xiang J, Wei Y, Ma Y, Wang L, Liu G. Vitamin D and Atherosclerosis: Unraveling the Impact on Macrophage Function. Mol Nutr Food Res 2024; 68:e2300867. [PMID: 38864846 DOI: 10.1002/mnfr.202300867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/02/2024] [Indexed: 06/13/2024]
Abstract
Vitamin D plays a crucial role in preventing atherosclerosis and in the regulation of macrophage function. This review aims to provide a comprehensive summary of the clinical evidence regarding the impact of vitamin D on atherosclerotic cardiovascular disease, atherosclerotic cerebrovascular disease, peripheral arterial disease, and associated risk factors. Additionally, it explores the mechanistic studies investigating the influence of vitamin D on macrophage function in atherosclerosis. Numerous findings indicate that vitamin D inhibits monocyte or macrophage recruitment, macrophage cholesterol uptake, and esterification. Moreover, it induces autophagy of lipid droplets in macrophages, promotes cholesterol efflux from macrophages, and regulates macrophage polarization. This review particularly focuses on analyzing the molecular mechanisms and signaling pathways through which vitamin D modulates macrophage function in atherosclerosis. It claims that vitamin D has a direct inhibitory effect on the formation, adhesion, and migration of lipid-loaded monocytes, thus exerting anti-atherosclerotic effects. Therefore, this review emphasizes the crucial role of vitamin D in regulating macrophage function and preventing the development of atherosclerosis.
Collapse
Affiliation(s)
- Dongxia Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhen Sun
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yajuan Yin
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jingyi Xiang
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yuzhe Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Le Wang
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Gang Liu
- Department of Cardiology, Hebei International Joint Research Center for Structural Heart Disease, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| |
Collapse
|
10
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
11
|
Petrovič D, Letonja J, Petrovič D. SMAD3 rs17228212 Polymorphism Is Associated with Advanced Carotid Atherosclerosis in a Slovenian Population. Biomedicines 2024; 12:1103. [PMID: 38791063 PMCID: PMC11117620 DOI: 10.3390/biomedicines12051103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Smad proteins influence the TGFβ signaling pathway, which plays an important role in the progression of atherosclerosis. The aim of our study was to investigate the association between the rs17228212 polymorphism of the SMAD3 gene and advanced carotid atherosclerosis in Slovenian subjects and to investigate the effect of the rs17228212 SMAD3 polymorphism on the expression of SMAD3 in endarterectomy sequesters. In this cross-sectional case-control study, 881 unrelated Caucasians were divided into two groups. The first group included 308 patients with advanced carotid atherosclerosis of the common or internal carotid artery with stenosis greater than 75% that underwent a revascularization procedure (cases). The control group consisted of 573 subjects without hemodynamically significant carotid atherosclerosis. We analyzed the rs17228212 polymorphism of the SMAD3 gene using the StepOne real-time polymerase chain reaction system and TaqMan SNP genotyping assay. The results in the two genetic models showed a statistically significant association, codominant (OR 4.05; CI 1.10-17.75; p = 0.037) and dominant (OR 3.60; CI 1.15-15.45; p = 0.045). An immunohistochemical analysis of SMAD3 expression was conducted for 26 endarterectomy specimens. The T allele of the rs17228212 SMAD3 gene was shown to be associated with an increased numerical area density of SMAD3-positive cells in carotid plaques.
Collapse
Affiliation(s)
- David Petrovič
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
| | - Jernej Letonja
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
12
|
Redgrave RE, Singh E, Tual-Chalot S, Park C, Hall D, Bennaceur K, Smyth DJ, Maizels RM, Spyridopoulos I, Arthur HM. Exogenous Transforming Growth Factor-β1 and Its Helminth-Derived Mimic Attenuate the Heart's Inflammatory Response to Ischemic Injury and Reduce Mature Scar Size. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:562-573. [PMID: 37832870 DOI: 10.1016/j.ajpath.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/29/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023]
Abstract
Coronary reperfusion after acute ST-elevation myocardial infarction (STEMI) is standard therapy to salvage ischemic heart muscle. However, subsequent inflammatory responses within the infarct lead to further loss of viable myocardium. Transforming growth factor (TGF)-β1 is a potent anti-inflammatory cytokine released in response to tissue injury. The aim of this study was to investigate the protective effects of TGF-β1 after MI. In patients with STEMI, there was a significant correlation (P = 0.003) between higher circulating TGF-β1 levels at 24 hours after MI and a reduction in infarct size after 3 months, suggesting a protective role of early increase in circulating TGF-β1. A mouse model of cardiac ischemia reperfusion was used to demonstrate multiple benefits of exogenous TGF-β1 delivered in the acute phase. It led to a significantly smaller infarct size (30% reduction, P = 0.025), reduced inflammatory infiltrate (28% reduction, P = 0.015), lower intracardiac expression of inflammatory cytokines IL-1β and chemokine (C-C motif) ligand 2 (>50% reduction, P = 0.038 and 0.0004, respectively) at 24 hours, and reduced scar size at 4 weeks (21% reduction, P = 0.015) after reperfusion. Furthermore, a low-fibrogenic mimic of TGF-β1, secreted by the helminth parasite Heligmosomoides polygyrus, had an almost identical protective effect on injured mouse hearts. Finally, genetic studies indicated that this benefit was mediated by TGF-β signaling in the vascular endothelium.
Collapse
Affiliation(s)
- Rachael E Redgrave
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Esha Singh
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Simon Tual-Chalot
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Catherine Park
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Darroch Hall
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Karim Bennaceur
- Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Ioakim Spyridopoulos
- Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Helen M Arthur
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom.
| |
Collapse
|
13
|
Chen J, Wang L, Wang Y, Jin A, Wang S, Li X, Jiang YH. Effects of Banxia Baizhu Tianma Decoction in alleviating atherosclerosis based on the regulation of perivascular adipose. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117575. [PMID: 38103846 DOI: 10.1016/j.jep.2023.117575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The occurrence and development of atherosclerosis, a common chronic inflammatory vascular disease, are closely related to cardiovascular and cerebrovascular diseases. Banxia Baizhu Tianma Decoction (BBTD) is a representative traditional Chinese medicine formula that resolves phlegm, disperses wind, invigorates the spleen and eliminates dampness and is also a commonly used clinical medication for treating vascular diseases. AIM OF THE STUDY To explore the pharmacological mechanisms of BBTD in alleviating atherosclerosis, the present study was carried out by conducting an integrative analysis of aortic and perivascular adipose tissue (PVAT) proteomics and metabolomics. MATERIALS AND METHODS Eight-week-old ApoE-/- mice were randomly divided into the BBTD group and the model group, and nine age-matched C57BL/6J (C57) mice were used as the control group (n = 9). The C57 mice were fed a standard diet, while the ApoE-/- mice were fed a high-fat, high-cholesterol diet for 12 weeks. Mice in the BBTD group were transgastrically administered BBTD at a dose of 17.8 g/kg/day for 8 weeks, while the model group and control group mice received an equivalent volume of saline by gavage. Histomorphology of the aortas and PVAT was assessed by HE staining, oil red O staining, Masson staining, and α-SMA and CD68 immunohistochemical methods. An integrative analysis of aortic proteomics, PVAT proteomics and PVAT metabolomics was conducted to study the pharmacological mechanisms of BBTD. RESULTS Compared to the model group, mice treated with BBTD had thicker fibrous caps, increased collagen content, less erosion of smooth muscle cells and infiltration of macrophages, as well as a relatively low inflammatory response level, suggesting that BBTD treatment reduced plaque vulnerability. Omics analysis suggested that BBTD treatment demonstrated anti-atherosclerotic effects and increased plaque stability in the aorta by activating the TGF-beta pathway. Simultaneously, BBTD inhibited PVAT inflammation levels (decreased the levels of MCP and IL-6). Proteomics and metabolomics of PVAT suggested that the targets of BBTD included upregulation of the α-linolenic acid metabolic pathway and downregulation of multiple inflammatory pathways, such as the NF-kappa B signalling pathway, primary immunodeficiency and Th17 cell differentiation in PVAT. CONCLUSIONS BBTD reduces the vulnerability of atherosclerotic plaques and inhibits the inflammatory phenotype of perivascular adipose tissue.
Collapse
Affiliation(s)
- Jianqiao Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Lin Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yongcheng Wang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Ao Jin
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Sutong Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Xiao Li
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Yue-Hua Jiang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
14
|
Saita E, Kishimoto Y, Aoyama M, Ohmori R, Kondo K, Momiyama Y. Low Plasma Levels of Soluble Endoglin and Cardiovascular Events in Patients Undergoing Coronary Angiography. Biomedicines 2023; 11:2975. [PMID: 38001975 PMCID: PMC10669441 DOI: 10.3390/biomedicines11112975] [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: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
TGF-β is recognized as playing a protective role against atherosclerosis. Endoglin is a receptor for TGF-β, and its expression is upregulated in atherosclerotic plaques. Endoglin is secreted from the cell membrane into the circulation as a soluble form (sEng). We previously reported that plasma sEng levels were low in patients with coronary artery disease (CAD). However, the prognostic value of sEng levels has not been clarified. We investigated the association between plasma sEng levels and cardiovascular events in 403 patients who had an elective coronary angiography and were then followed up. Cardiovascular events were defined as cardiovascular death, myocardial infarction, unstable angina, heart failure, stroke, or coronary revascularization. Of the 403 patients, 209 (52%) had CAD. Plasma sEng levels were lower in patients with CAD than in those without CAD (median 4.26 vs. 4.41 ng/mL, p < 0.025). During a mean follow-up period of 7.5 ± 4.5 years, cardiovascular events occurred in 79 patients. Compared with 324 patients without events, 79 with events had lower sEng levels (3.95 vs. 4.39 ng/mL) and more often had an sEng level < 3.9 ng/mL (47% vs. 28%) (p < 0.02). A Kaplan-Meier analysis showed lower event-free survival in patients with sEng < 3.9 ng/mL than in those with ≥3.9 ng/mL (p < 0.02). In a multivariate Cox proportional hazards analysis, the sEng level (<3.9 ng/mL) was an independent predictor of cardiovascular events (hazard ratio: 1.59; 95%CI: 1.01-2.49). Furthermore, only among the 209 patients with CAD, the sEng level was also a predictor of further cardiovascular events (hazard ratio: 2.07; 95%CI: 1.24-3.45). Thus, low plasma sEng levels were found to be associated with an increased risk of cardiovascular events in patients with CAD and patients undergoing coronary angiography.
Collapse
Affiliation(s)
- Emi Saita
- Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Setsunan University, 45-1 Na-gaotouge-cho, Hirakata 573-0101, Japan
| | - Masayuki Aoyama
- Department of Cardiovascular Medicine, Toho University Graduate School of Medicine, 5-21-16 Omorinishi, Ota-ku, Tokyo 143-8540, Japan
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University, 350 Minecho, Tochigi 321-8505, Japan
| | - Kazuo Kondo
- Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan
| |
Collapse
|
15
|
Waldron C, Zafar MA, Ziganshin BA, Weininger G, Grewal N, Elefteriades JA. Evidence Accumulates: Patients with Ascending Aneurysms Are Strongly Protected from Atherosclerotic Disease. Int J Mol Sci 2023; 24:15640. [PMID: 37958625 PMCID: PMC10650782 DOI: 10.3390/ijms242115640] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Ascending thoracic aortic aneurysms may be fatal upon rupture or dissection and remain a leading cause of death in the developed world. Understanding the pathophysiology of the development of ascending thoracic aortic aneurysms may help reduce the morbidity and mortality of this disease. In this review, we will discuss our current understanding of the protective relationship between ascending thoracic aortic aneurysms and the development of atherosclerosis, including decreased carotid intima-media thickness, low-density lipoprotein levels, coronary and aortic calcification, and incidence of myocardial infarction. We also propose several possible mechanisms driving this relationship, including matrix metalloproteinase proteins and transforming growth factor-β.
Collapse
Affiliation(s)
- Christina Waldron
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT 06519, USA; (C.W.); (M.A.Z.); (B.A.Z.)
| | - Mohammad A. Zafar
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT 06519, USA; (C.W.); (M.A.Z.); (B.A.Z.)
| | - Bulat A. Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT 06519, USA; (C.W.); (M.A.Z.); (B.A.Z.)
- Department of Cardiovascular and Endovascular Surgery, Kazan State Medical University, 420012 Kazan, Russia
| | - Gabe Weininger
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT 06519, USA; (C.W.); (M.A.Z.); (B.A.Z.)
| | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands;
| | - John A. Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT 06519, USA; (C.W.); (M.A.Z.); (B.A.Z.)
| |
Collapse
|
16
|
Edsfeldt A, Singh P, Matthes F, Tengryd C, Cavalera M, Bengtsson E, Dunér P, Volkov P, Karadimou G, Gisterå A, Orho-Melander M, Nilsson J, Sun J, Gonçalves I. Transforming growth factor-β2 is associated with atherosclerotic plaque stability and lower risk for cardiovascular events. Cardiovasc Res 2023; 119:2061-2073. [PMID: 37200403 PMCID: PMC10478752 DOI: 10.1093/cvr/cvad079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/27/2023] [Accepted: 02/21/2023] [Indexed: 05/20/2023] Open
Abstract
AIMS Transforming growth factor-beta (TGF-β) exists in three isoforms TGF-β1, -β2, and -β3. TGF-β1 has been suggested to be important for maintaining plaque stability, yet the role of TGF-β2 and -β3 in atherosclerosis remains to be investigated.This study explores the association of the three isoforms of TGF-β with plaque stability in the human atherosclerotic disease. METHODS AND RESULTS TGF-β1, -β2, and -β3 proteins were quantified in 223 human carotid plaques by immunoassays. Indications for the endarterectomy were: symptomatic carotid plaque with stenosis >70% or without symptoms and >80% stenosis. Plaque mRNA levels were assessed by RNA sequencing. Plaque components and extracellular matrix were measured histologically and biochemically. Matrix metalloproteinases and monocyte chemoattractant protein-1 (MCP-1) was measured with immunoassays. The effect of TGF-β2 on inflammation and protease activity was investigated in vitro using THP-1 and RAW264.7 macrophages. Patients were followed longitudinally for cardiovascular (CV) events.TGF-β2 was the most abundant isoform and was increased at both protein and mRNA levels in asymptomatic plaques. TGF-β2 was the main determinant separating asymptomatic plaques in an Orthogonal Projections to Latent Structures Discriminant Analysis. TGF-β2 correlated positively to features of plaque stability and inversely to markers of plaque vulnerability. TGF-β2 was the only isoform inversely correlated to the matrix-degrading matrix metalloproteinase-9 and inflammation in the plaque tissue. In vitro, TGF-β2 pre-treatment reduced MCP-1 gene and protein levels as well as matrix metalloproteinase-9 gene levels and activity. Patients with plaques with high TGF-β2 levels had a lower risk to suffer from future CV events. CONCLUSIONS TGF-β2 is the most abundant TGF-β isoform in human plaques and may maintain plaque stability by decreasing inflammation and matrix degradation.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Pratibha Singh
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Frank Matthes
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | | | - Michele Cavalera
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms—Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Petr Volkov
- Department of Clinical Sciences, LUDC Bioinformatics Unit, Malmö, Lund University, Lund, Sweden
- Data Science and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Glykeria Karadimou
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine, Center for Molecular Medicine, Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Nilsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Jiangming Sun
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
17
|
Bazioti V, Halmos B, Westerterp M. T-cell Cholesterol Accumulation, Aging, and Atherosclerosis. Curr Atheroscler Rep 2023; 25:527-534. [PMID: 37395922 PMCID: PMC10471657 DOI: 10.1007/s11883-023-01125-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW The majority of leukocytes in advanced human atherosclerotic plaques are T-cells. T-cell subsets exert pro- or anti-atherogenic effects largely via the cytokines they secrete. Tregulatory cells (Tregs) are anti-inflammatory, but may lose these properties during atherosclerosis, proposed to be downstream of cholesterol accumulation. Aged T-cells also accumulate cholesterol. The effects of T-cell cholesterol accumulation on T-cell fate and atherosclerosis are not uniform. RECENT FINDINGS T-cell cholesterol accumulation enhances differentiation into pro-atherogenic cytotoxic T-cells and boosts their killing capacity, depending on the localization and extent of cholesterol accumulation. Excessive cholesterol accumulation induces T-cell exhaustion or T-cell apoptosis, the latter decreasing atherosclerosis but impairing T-cell functionality in terms of killing capacity and proliferation. This may explain the compromised T-cell functionality in aged T-cells and T-cells from CVD patients. The extent of T-cell cholesterol accumulation and its cellular localization determine T-cell fate and downstream effects on atherosclerosis and T-cell functionality.
Collapse
Affiliation(s)
- Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336, Munich, Germany
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands.
| |
Collapse
|
18
|
Tian Z, Yang S. Integrating the characteristic genes of macrophage pseudotime analysis in single-cell RNA-seq to construct a prediction model of atherosclerosis. Aging (Albany NY) 2023; 15:6361-6379. [PMID: 37421595 PMCID: PMC10373969 DOI: 10.18632/aging.204856] [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: 04/12/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Macrophages play an important role in the occurrence and development of atherosclerosis. However, few existing studies have deliberately analyzed the changes in characteristic genes in the process of macrophage phenotype transformation. METHOD Carotid atherosclerotic plaque single-cell RNA (scRNA) sequencing data were analyzed to define the cells involved and determine their transcriptomic characteristics. KEGG enrichment analysis, CIBERSORT, ESTIMATE, support vector machine (SVM), random forest (RF), and weighted correlation network analysis (WGCNA) were applied to bulk sequencing data. All data were downloaded from Gene Expression Omnibus (GEO). RESULT Nine cell clusters were identified. M1 macrophages, M2 macrophages, and M2/M1 macrophages were identified as three clusters within the macrophages. According to pseudotime analysis, M2/M1 macrophages and M2 macrophages can be transformed into M1 macrophages. The ROC curve values of the six genes in the test group were statistically significant (AUC (IL1RN): 0.899, 95% CI: 0.764-0.990; AUC (NRP1): 0.817, 95% CI: 0.620-0.971; AUC (TAGLN): 0.846, 95% CI: 0.678-0.971; AUC (SPARCL1): 0.825, 95% CI: 0.620-0.988; AUC (EMP2): 0.808, 95% CI: 0.630-0.947; AUC (ACTA2): 0.784, 95% CI: 0.591-0.938). The atherosclerosis prediction model showed significant statistical significance in both the train group (AUC: 0.909, 95% CI: 0.842-0.967) and the test group (AUC: 0.812, 95% CI: 0.630-0.966). CONCLUSIONS IL1RNHigh M1, NRP1High M2, ACTA2High M2/M1, EMP2High M1/M1, SPACL1High M2/M1 and TAGLNHigh M2/M1 macrophages play key roles in the occurrence and development of arterial atherosclerosis. These marker genes of macrophage phenotypic transformation can also be used to establish a model to predict the occurrence of atherosclerosis.
Collapse
Affiliation(s)
- Zemin Tian
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Shize Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| |
Collapse
|
19
|
Kumar V, Sethi B, Staller DW, Xin X, Ma J, Dong Y, Talmon GA, Mahato RI. Anti-miR-96 and Hh pathway inhibitor MDB5 synergistically ameliorate alcohol-associated liver injury in mice. Biomaterials 2023; 295:122049. [PMID: 36827892 PMCID: PMC9998370 DOI: 10.1016/j.biomaterials.2023.122049] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023]
Abstract
Alcohol-associated liver disease (ALD) and its complications are significant health problems worldwide. Several pathways in ALD are influenced by alcohol that drives inflammation, fatty acid metabolism, and fibrosis. Although miR-96 has become a key regulator in several liver diseases, its function in ALD remains unclear. In contrast, sonic hedgehog (SHH) signaling has a well-defined role in liver disease through influencing the activation of hepatic stellate cells (HSCs) and the inducement of liver fibrosis. In this study, we investigated the expression patterns of miR-96 and Hh molecules in mouse and human liver samples. We showed that miR-96 and Shh were upregulated in ethanol-fed mice. Furthermore, alcoholic hepatitis (AH) patient specimens also showed upregulated FOXO3a, TGF-β1, SHH, and GLI2 proteins. We then examined the effects of Hh inhibitor MDB5 and anti-miR-96 on inflammatory and extracellular matrix (ECM)-related genes. We identified FOXO3 and SMAD7 as direct target genes of miR-96. Inhibition of miR-96 decreased the expression of these genes in vitro in AML12 cells, HSC-T6 cells, and in vivo in ALD mice. Furthermore, MDB5 decreased HSCs activation and the expression of ECM-related genes, such as Gli1, Tgf-β1, and collagen. Lipid nanoparticles (LNPs) loaded with the combination of MDB5, and anti-miR-96 ameliorated ALD in mice. Our study demonstrated that this combination therapy could serve as a new therapeutic target for ALD.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bharti Sethi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dalton W Staller
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiaofei Xin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jingyi Ma
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey A Talmon
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
20
|
Lyu Q, Ley K. How Lymphatic Endothelial Cells Destabilize Regulatory T Cells. Arterioscler Thromb Vasc Biol 2023; 43:215-217. [PMID: 36579643 PMCID: PMC10108378 DOI: 10.1161/atvbaha.122.318849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Qingkang Lyu
- Immunology Center of Georgia (Q.L., K.L.), Augusta University
| | - Klaus Ley
- Immunology Center of Georgia (Q.L., K.L.), Augusta University.,Department of Physiology (K.L.), Augusta University
| |
Collapse
|
21
|
Liu H, Hallauer Hastings M, Kitchen R, Xiao C, Baldovino Guerra JR, Kuznetsov A, Rosenzweig A. Beneficial Effects of Moderate Hepatic Activin A Expression on Metabolic Pathways, Inflammation, and Atherosclerosis. Arterioscler Thromb Vasc Biol 2023; 43:330-349. [PMID: 36453275 DOI: 10.1161/atvbaha.122.318138] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFβ/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.
Collapse
Affiliation(s)
- Huan Liu
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Alexandra Kuznetsov
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| |
Collapse
|
22
|
Wang F, Stappenbeck F, Parhami F. Oxy210, a Semi-Synthetic Oxysterol, Inhibits Profibrotic Signaling in Cellular Models of Lung and Kidney Fibrosis. Pharmaceuticals (Basel) 2023; 16:114. [PMID: 36678611 PMCID: PMC9862207 DOI: 10.3390/ph16010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Oxy210, a semi-synthetic oxysterol derivative, displays cell-selective inhibition of Hedgehog (Hh) and transforming growth factor beta (TGF-β) signaling in epithelial cells, fibroblasts, and macrophages as well as antifibrotic and anti-inflammatory efficacy in models of liver fibrosis. In the present report, we examine the effects of Oxy210 in cellular models of lung and kidney fibrosis, such as human lung fibroblast cell lines IMR-90, derived from healthy lung tissue, and LL97A, derived from an idiopathic pulmonary fibrosis (IPF) patient. In addition, we examine the effects of Oxy210 in primary human renal fibroblasts, pericytes, mesangial cells, and renal tubular epithelial cells, known for their involvement in chronic kidney disease (CKD) and kidney fibrosis. We demonstrate in fibroblasts that the expression of several profibrotic TGF-β target genes, including fibronectin (FN), collagen 1A1 (COL1A1), and connective tissue growth factor (CTGF) are inhibited by Oxy210, both at the basal level and following TGF-β stimulation in a statistically significant manner. The inhibition of COL1A1 gene expression translated directly to significantly reduced COL1A1 protein expression. In human primary small airway epithelial cells (HSAECs) and renal tubular epithelial cells, Oxy210 significantly inhibited TGF-β target gene expression associated with epithelial-mesenchymal transition (EMT). Oxy210 also inhibited the proliferation of fibroblasts, pericytes, and mesangial cells in a dose-dependent and statistically significant manner.
Collapse
Affiliation(s)
| | | | - Farhad Parhami
- MAX BioPharma, Inc., 2870 Colorado Avenue, Santa Monica, CA 90404, USA
| |
Collapse
|
23
|
Alajbegovic A, Daoud F, Ali N, Kawka K, Holmberg J, Albinsson S. Transcription factor GATA6 promotes migration of human coronary artery smooth muscle cells in vitro. Front Physiol 2022; 13:1054819. [PMID: 36523548 PMCID: PMC9744938 DOI: 10.3389/fphys.2022.1054819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cell plasticity plays a pivotal role in the pathophysiology of vascular diseases. Despite compelling evidence demonstrating the importance of transcription factor GATA6 in vascular smooth muscle, the functional role of GATA6 remains poorly understood. The aim of this study was to elucidate the role of GATA6 on cell migration and to gain insight into GATA6-sensitive genes in smooth muscle. We found that overexpression of GATA6 promotes migration of human coronary artery smooth muscle cells in vitro, and that silencing of GATA6 in smooth muscle cells resulted in reduced cellular motility. Furthermore, a complete microarray screen of GATA6-sensitive gene transcription resulted in 739 upregulated and 248 downregulated genes. Pathways enrichment analysis showed involvement of transforming growth factor beta (TGF-β) signaling which was validated by measuring mRNA expression level of several members. Furthermore, master regulators prediction based on microarray data revealed several members of (mitogen activated protein kinase) MAPK pathway as a master regulators, reflecting involvement of MAPK pathway also. Our findings provide further insights into the functional role of GATA6 in vascular smooth muscle and suggest that targeting GATA6 may constitute as a new approach to inhibit vascular smooth muscle migration.
Collapse
Affiliation(s)
- Azra Alajbegovic
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fatima Daoud
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Neserin Ali
- Department of Clinical Sciences Lund, Orthopedics, Clinical Epidemiology Unit, Lund University, Lund, Sweden
| | - Katarzyna Kawka
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
24
|
Genome-Wide Transcriptional Profiling Reveals PHACTR1 as a Novel Molecular Target of Resveratrol in Endothelial Homeostasis. Nutrients 2022; 14:nu14214518. [DOI: 10.3390/nu14214518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease in which endothelial cells play an important role in maintaining vascular homeostasis. Endotheliitis caused by endothelial dysfunction (ED) is the key cause for the development of cardiovascular and cerebrovascular diseases as well as other vascular system diseases. Resveratrol (RES), a multi-functional polyphenol present in edible plants and fruits, prevents cardiovascular disease by regulating a variety of athero-relevant signaling pathways. By transcriptome profiling of RES-treated human umbilical vein endothelial cells (HUVECs) and in-depth bioinformatic analysis, we observed that differentially expressed genes (DEGs) were enriched in KEGG pathways of fluid shear stress and atherosclerosis, suggesting that the RES may serve as a good template for a shear stress mimetic drug that hold promise in combating atherosclerosis. A heat map and multiple datasets superimposed screening revealed that RES significantly down-regulated phosphatase and actin modulator 1 (PHACTR1), a pivotal coronary artery disease risk gene associated with endothelial inflammation and polyvascular diseases. We further demonstrate that RES down-regulated the gene and protein expression of PHACTR1 and inhibited TNF-α-induced adhesion of THP-1 monocytes to activated endothelial cells via suppressing the expression of PHACTR1. Taken together, our study reveals that PHACTR1 represents a new molecular target for RES to maintain endothelial cell homeostasis and prevent atherosclerotic cardiovascular disease.
Collapse
|
25
|
Jiang S, Ai Y, Ni L, Wu L, Huang X, Chen S. Platelet-derived TGF-β1 is related to portal vein thrombosis in cirrhosis by promoting hypercoagulability and endothelial dysfunction. Front Cardiovasc Med 2022; 9:938397. [PMID: 36225950 PMCID: PMC9548594 DOI: 10.3389/fcvm.2022.938397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundPortal vein thrombosis (PVT) is a serious complication of cirrhosis accompanied by unclear pathogenesis. Transforming growth factor-beta (TGF-β) has been implicated in atherosclerosis and venous thrombosis whereas study regarding its part in PVT is lacking. The aim of this study was to explore the role of cytokine TGF-β1 in PVT and the potential mechanism.Materials and methodsWe included patients with cirrhotic gastroesophageal varices and divided them into two groups according to the presence of PVT. Serum levels of TGF-β1 were detected using Cytometric Bead Array kit and compared between two groups. Coagulation status was assessed using thromboelastography (TEG). Primary liver sinusoidal endothelial cells were treated with TGF-β1 and evaluated for endothelial dysfunction by RT-PCR.ResultsOur results uncovered that TGF-β1 (6,866.55 vs. 3,840.60 pg/ml, P = 0.015) significantly increased in the PVT group. Splenectomy might promote PVT by increasing platelet-derived TGF-β1 levels. Other cytokines showed no difference between PVT and non-PVT groups. Besides, TGF-β1 was correlated with platelet, fibrinogen, TEG-CI, TEG-MA, and TEG-α (coef = 0.733, 0.494, 0.604, 0.608, and 0.511; P < 0.001, 0.027, 0.004, 0.004, and 0.021, respectively), which indicated a hypercoagulable state in PVT patients. RT-PCR of liver sinusoidal endothelial cells showed a markable increment of von Willebrand Factor (vWF), thrombomodulin(TM), intercellular adhesion moleclar-1(ICAM-1), and vascular endothelial growth factor(VEGF) after TGF-β1 treatment, suggesting the involvement of endothelial dysfunction.ConclusionElevated platelet-derived TGF-β1 exhibited association with hypercoagulability and promoting effect on endothelial dysfunction, closely related with PVT in cirrhotic patients.
Collapse
Affiliation(s)
- Siyu Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingjie Ai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liyuan Ni
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoquan Huang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Xiaoquan Huang,
| | - Shiyao Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
26
|
Wara AK, Rawal S, Yang X, Pérez-Cremades D, Sachan M, Chen J, Feinberg MW. KLF10 deficiency in CD4 + T cells promotes atherosclerosis progression by altering macrophage dynamics. Atherosclerosis 2022; 359:27-41. [PMID: 36174463 DOI: 10.1016/j.atherosclerosis.2022.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND AIMS Accumulating evidence supports a critical role for CD4+ T cells as drivers and modifiers of the chronic inflammatory response in atherosclerosis. Effector T cells have pro-atherogenic properties, whereas CD4+ regulatory T cells (Tregs) exert suppressive activity in atherosclerosis through increased secretion of inhibitory cytokines such as transforming growth factor-β or interleukin-10. In addition, Tregs have been shown to suppress inflammatory macrophages and promote the resolution of atherosclerosis plaques. Impaired Treg numbers and function have been associated with atherosclerosis plaque development. However, the underlying mechanisms remain unclear. METHODS AND RESULTS Here, we investigated a cell-autonomous role of a transcription factor, Krüppel-like factor 10 (KLF10), in CD4+ T cells in regulating atherosclerosis progression. Using CD4+ T-cell-specific KLF10 knockout (TKO) mice, we identified exaggerated plaque progression due to defects in immunosuppressive functions of Tregs on macrophages. TKO mice exhibited increased lesion size as well as higher CD4+ T cells and macrophage content compared to WT mice. TKO plaques also showed increased necrotic cores along with defective macrophage efferocytosis. In contrast, adoptive cellular therapy using WT Tregs abrogated the accelerated lesion progression and deleterious effects in TKO mice. Intriguingly, RNA-seq analyses of TKO lesions revealed increased chemotaxis and cell proliferation, and reduced phagocytosis compared to WT lesions. Mechanistically, TKO-Tregs impaired the efferocytosis capacity of macrophages in vitro and promoted a pro-inflammatory macrophage phenotype via increased IFN-γ and decreased TGF-β secretion. CONCLUSIONS Taken together, these findings establish a critical role for KLF10 in regulating CD4+ Treg-macrophage interactions and atherosclerosis.
Collapse
Affiliation(s)
- Akm Khyrul Wara
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Shruti Rawal
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xilan Yang
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of General Practice, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Daniel Pérez-Cremades
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Physiology, University of Valencia, and INCLIVA Biomedical Research Institute, Valencia, 46010, Spain
| | - Madhur Sachan
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jingshu Chen
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
27
|
Puppala S, Spradling-Reeves KD, Chan J, Birnbaum S, Newman DE, Comuzzie AG, Mahaney MC, VandeBerg JL, Olivier M, Cox LA. Hepatic transcript signatures predict atherosclerotic lesion burden prior to a 2-year high cholesterol, high fat diet challenge. PLoS One 2022; 17:e0271514. [PMID: 35925965 PMCID: PMC9352111 DOI: 10.1371/journal.pone.0271514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 07/04/2022] [Indexed: 11/19/2022] Open
Abstract
The purpose of this study was to identify molecular mechanisms by which the liver influences total lesion burden in a nonhuman primate model (NHP) of cardiovascular disease with acute and chronic feeding of a high cholesterol, high fat (HCHF) diet. Baboons (47 females, 64 males) were fed a HCHF diet for 2 years (y); liver biopsies were collected at baseline, 7 weeks (w) and 2y, and lesions were quantified in aortic arch, descending aorta, and common iliac at 2y. Unbiased weighted gene co-expression network analysis (WGCNA) revealed several modules of hepatic genes correlated with lesions at different time points of dietary challenge. Pathway and network analyses were performed to study the roles of hepatic module genes. More significant pathways were observed in males than females. In males, we found modules enriched for genes in oxidative phosphorylation at baseline, opioid signaling at 7w, and EIF2 signaling and HNF1A and HNF4A networks at baseline and 2y. One module enriched for fatty acid β oxidation pathway genes was found in males and females at 2y. To our knowledge, this is the first study of a large NHP cohort to identify hepatic genes that correlate with lesion burden. Correlations of baseline and 7w module genes with lesions at 2y were observed in males but not in females. Pathway analyses of baseline and 7w module genes indicate EIF2 signaling, oxidative phosphorylation, and μ-opioid signaling are possible mechanisms that predict lesion formation induced by HCHF diet consumption in males. Our findings of coordinated hepatic transcriptional response in male baboons but not female baboons indicate underlying molecular mechanisms differ between female and male primate atherosclerosis.
Collapse
Affiliation(s)
- Sobha Puppala
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Kimberly D. Spradling-Reeves
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jeannie Chan
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Shifra Birnbaum
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Deborah E. Newman
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | | | - Michael C. Mahaney
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, The University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, United States of America
| | - John L. VandeBerg
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, The University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, United States of America
| | - Michael Olivier
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Laura A. Cox
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
28
|
Kettunen S, Ruotsalainen AK, Ylä-Herttuala S. RNA interference-based therapies for the control of atherosclerosis risk factors. Curr Opin Cardiol 2022; 37:364-371. [PMID: 35731681 DOI: 10.1097/hco.0000000000000972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Atherosclerosis, characterized by lipid accumulation and chronic inflammation in the arterial wall, is the leading causes of death worldwide. The purpose of this article is to review the status of RNA interference (RNAi) based therapies in clinical trials for the treatment and prevention of atherosclerosis risk factors. RECENT FINDINGS There is a growing interest on using RNAi technology for the control of atherosclerosis risk factors. Current clinical trials utilizing RNAi for atherosclerosis are targeting lipid metabolism regulating genes including proprotein convertase subtilisin/kexin 9, apolipoprotein C-III, lipoprotein (a) and angiopoietin-like protein 3. Currently, three RNAi-based drugs have been approved by U.S. Food and Drug Administration, but there are several therapies in clinical trials at the moment, and potentially entering the market in near future. In addition, recent preclinical studies on regulating vascular inflammation have shown promising results. SUMMARY In recent years, RNAi based technologies and therapies have been intensively developed for the treatment of atherosclerosis risk factors, such as hyperlipidemia and vascular inflammation. Multiple potential therapeutic targets have emerged, and many of the reported clinical trials have already been successful in plasma lipid lowering. The scope of RNAi therapies is well recognized and recent approvals are encouraging for the treatment of cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
| | | | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
29
|
Bachmann JC, Baumgart SJ, Uryga AK, Bosteen MH, Borghetti G, Nyberg M, Herum KM. Fibrotic Signaling in Cardiac Fibroblasts and Vascular Smooth Muscle Cells: The Dual Roles of Fibrosis in HFpEF and CAD. Cells 2022; 11:1657. [PMID: 35626694 PMCID: PMC9139546 DOI: 10.3390/cells11101657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) and atherosclerosis-driven coronary artery disease (CAD) will have ongoing fibrotic remodeling both in the myocardium and in atherosclerotic plaques. However, the functional consequences of fibrosis differ for each location. Thus, cardiac fibrosis leads to myocardial stiffening, thereby compromising cardiac function, while fibrotic remodeling stabilizes the atherosclerotic plaque, thereby reducing the risk of plaque rupture. Although there are currently no drugs targeting cardiac fibrosis, it is a field under intense investigation, and future drugs must take these considerations into account. To explore similarities and differences of fibrotic remodeling at these two locations of the heart, we review the signaling pathways that are activated in the main extracellular matrix (ECM)-producing cells, namely human cardiac fibroblasts (CFs) and vascular smooth muscle cells (VSMCs). Although these signaling pathways are highly overlapping and context-dependent, effects on ECM remodeling mainly act through two core signaling cascades: TGF-β and Angiotensin II. We complete this by summarizing the knowledge gained from clinical trials targeting these two central fibrotic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kate M. Herum
- Research and Early Development, Novo Nordisk A/S, Novo Nordisk Park, 2760 Maaloev, Denmark; (J.C.B.); (S.J.B.); (A.K.U.); (M.H.B.); (G.B.); (M.N.)
| |
Collapse
|
30
|
Oxy210, a Semi-Synthetic Oxysterol, Exerts Anti-Inflammatory Effects in Macrophages via Inhibition of Toll-like Receptor (TLR) 4 and TLR2 Signaling and Modulation of Macrophage Polarization. Int J Mol Sci 2022; 23:ijms23105478. [PMID: 35628290 PMCID: PMC9141227 DOI: 10.3390/ijms23105478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammatory responses by the innate and adaptive immune systems protect against infections and are essential to health and survival. Many diseases including atherosclerosis, osteoarthritis, rheumatoid arthritis, psoriasis, and obesity involve persistent chronic inflammation. Currently available anti-inflammatory agents, including non-steroidal anti-inflammatory drugs, steroids, and biologics, are often unsafe for chronic use due to adverse effects. The development of effective non-toxic anti-inflammatory agents for chronic use remains an important research arena. We previously reported that oral administration of Oxy210, a semi-synthetic oxysterol, ameliorates non-alcoholic steatohepatitis (NASH) induced by a high-fat diet in APOE*3-Leiden.CETP humanized mouse model of NASH and inhibits expression of hepatic and circulating levels of inflammatory cytokines. Here, we show that Oxy210 also inhibits diet-induced white adipose tissue inflammation in APOE*3-Leiden.CETP mice, evidenced by the inhibition of adipose tissue expression of IL-6, MCP-1, and CD68 macrophage marker. Oxy210 and related analogs exhibit anti-inflammatory effects in macrophages treated with lipopolysaccharide in vitro, mediated through inhibition of toll-like receptor 4 (TLR4), TLR2, and AP-1 signaling, independent of cyclooxygenase enzymes or steroid receptors. The anti-inflammatory effects of Oxy210 are correlated with the inhibition of macrophage polarization. We propose that Oxy210 and its structural analogs may be attractive candidates for future therapeutic development for targeting inflammatory diseases.
Collapse
|
31
|
Moeinafshar A, Razi S, Rezaei N. Interleukin 17, the double-edged sword in atherosclerosis. Immunobiology 2022; 227:152220. [PMID: 35452921 DOI: 10.1016/j.imbio.2022.152220] [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: 06/04/2021] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022]
Abstract
Cardiovascular diseases, including atherosclerosis, are the number one cause of death worldwide. These diseases have taken the place of pneumonia and other infectious diseases in the epidemiological charts. Thus, their importance should not be underestimated. Atherosclerosis is an inflammatory disease. Therefore, immunological signaling molecules and immune cells carry out a central role in its etiology. One of these signaling molecules is interleukin (IL)-17. This relatively newly discovered signaling molecule might have a dual role as acting both pro-atherogenic and anti-atherogenic depending on the situation. The majority of articles have discussed IL-17 and its action in atherosclerosis, and it may be a new target for the treatment of patients with this disease. In this review, the immunological basis of atherosclerosis with an emphasis on the role of IL-17 and a brief explanation of the role of IL-17 on atherosclerogenic disorders will be discussed.
Collapse
Affiliation(s)
- Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Ampomah PB, Cai B, Sukka SR, Gerlach BD, Yurdagul A, Wang X, Kuriakose G, Darville LNF, Sun Y, Sidoli S, Koomen JM, Tall AR, Tabas I. Macrophages use apoptotic cell-derived methionine and DNMT3A during efferocytosis to promote tissue resolution. Nat Metab 2022; 4:444-457. [PMID: 35361955 PMCID: PMC9050866 DOI: 10.1038/s42255-022-00551-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/11/2022] [Indexed: 12/19/2022]
Abstract
Efferocytosis, the clearance of apoptotic cells (ACs) by macrophages, is critical for tissue resolution, with defects driving many diseases. Mechanisms of efferocytosis-mediated resolution are incompletely understood. Here, we show that AC-derived methionine regulates resolution through epigenetic repression of the extracellular signal-regulated kinase 1/2 (ERK1/2) phosphatase Dusp4. We focus on two key efferocytosis-induced pro-resolving mediators, prostaglandin E2 (PGE2) and transforming growth factor beta 1 (TGF-β1), and show that efferocytosis induces prostaglandin-endoperoxide synthase 2/cyclooxygenase 2 (Ptgs2/COX2), leading to PGE2 synthesis and PGE2-mediated induction of TGF-β1. ERK1/2 phosphorylation/activation by AC-activated CD36 is necessary for Ptgs2 induction, but this is insufficient owing to an ERK-DUSP4 negative feedback pathway that lowers phospho-ERK. However, subsequent AC engulfment and phagolysosomal degradation lead to Dusp4 repression, enabling enhanced p-ERK and induction of the Ptgs2-PGE2-TGF-β1 pathway. Mechanistically, AC-derived methionine is converted to S-adenosylmethionine, which is used by DNA methyltransferase-3A (DNMT3A) to methylate Dusp4. Bone-marrow DNMT3A deletion in mice blocks COX2/PGE2, TGF-β1, and resolution in sterile peritonitis, apoptosis-induced thymus injury and atherosclerosis. Knowledge of how macrophages use AC-cargo and epigenetics to induce resolution provides mechanistic insight and therapeutic options for diseases driven by impaired resolution.
Collapse
Affiliation(s)
- Patrick B Ampomah
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Shreveport, Shreveport, LA, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lancia N F Darville
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yan Sun
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alan R Tall
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
33
|
Ma J, Tan Q, Nie X, Zhou M, Wang B, Wang X, Cheng M, Ye Z, Xie Y, Wang D, Chen W. Longitudinal relationships between polycyclic aromatic hydrocarbons exposure and heart rate variability: Exploring the role of transforming growth factor-β in a general Chinese population. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:127770. [PMID: 34823955 DOI: 10.1016/j.jhazmat.2021.127770] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/16/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
We aim to investigate the long-term adverse effects of polycyclic aromatic hydrocarbons (PAHs) exposure on heart rate variability (HRV) reduction, and to assess the potential role of transforming growth factor-β1 (TGF-β1) in such relationship. We enrolled 2985 adult residents with 4100 observations who participated at baseline and 6-years follow-up from Wuhan-Zhuhai cohort. Ten detectable urinary PAHs metabolites and two HRV indices were repeatedly measured at baseline and follow-up; and plasma TGF-β1 levels were also determined for all subjects. We observed that both total urinary low molecular weight PAHs (ΣLWM OH-PAH) and total urinary high molecular weight PAHs (ΣHWM OH-PAH) were negatively associated with HRV reductions (P < 0.05). Subjects with persistent high levels of ΣHWM OH-PAH had a significant reduction in HRV over 6 years, and the incensement of TGF-β1 could aggravate above adverse effects in a dose-response manner. All kinds of PAHs were positively associated with plasma TGF-β1 elevation, which in turn, were negatively related to HRV indices. Increased TGF-β1 significant mediated 1.34-3.62% of PAHs-associated HRV reduction. Our findings demonstrated that long-term high levels of PAHs exposure could cause HRV reductions, and TGF-β1 may play an essential role in such association.
Collapse
Affiliation(s)
- Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qiyou Tan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiuquan Nie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xing Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Man Cheng
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zi Ye
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yujia Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dongming Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
34
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
35
|
Farahi L, Sinha SK, Lusis AJ. Roles of Macrophages in Atherogenesis. Front Pharmacol 2021; 12:785220. [PMID: 34899348 PMCID: PMC8660976 DOI: 10.3389/fphar.2021.785220] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that may ultimately lead to local proteolysis, plaque rupture, and thrombotic vascular disease, resulting in myocardial infarction, stroke, and sudden cardiac death. Circulating monocytes are recruited to the arterial wall in response to inflammatory insults and differentiate into macrophages which make a critical contribution to tissue damage, wound healing, and also regression of atherosclerotic lesions. Within plaques, macrophages take up aggregated lipoproteins which have entered the vessel wall to give rise to cholesterol-engorged foam cells. Also, the macrophage phenotype is influenced by various stimuli which affect their polarization, efferocytosis, proliferation, and apoptosis. The heterogeneity of macrophages in lesions has recently been addressed by single-cell sequencing techniques. This article reviews recent advances regarding the roles of macrophages in different stages of disease pathogenesis from initiation to advanced atherosclerosis. Macrophage-based therapies for atherosclerosis management are also described.
Collapse
Affiliation(s)
- Lia Farahi
- Monoclonal Antibody Research Center, Avicenna Research Institute, Tehran, Iran
| | - Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
36
|
Serum apolipoprotein A-I depletion is causative to silica nanoparticles-induced cardiovascular damage. Proc Natl Acad Sci U S A 2021; 118:2108131118. [PMID: 34716267 DOI: 10.1073/pnas.2108131118] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022] Open
Abstract
The rapid development of nanotechnology has greatly benefited modern science and engineering and also led to an increased environmental exposure to nanoparticles (NPs). While recent research has established a correlation between the exposure of NPs and cardiovascular diseases, the intrinsic mechanisms of such a connection remain unclear. Inhaled NPs can penetrate the air-blood barrier from the lung to systemic circulation, thereby intruding the cardiovascular system and generating cardiotoxic effects. In this study, on-site cardiovascular damage was observed in mice upon respiratory exposure of silica nanoparticles (SiNPs), and the corresponding mechanism was investigated by focusing on the interaction of SiNPs and their encountered biomacromolecules en route. SiNPs were found to collect a significant amount of apolipoprotein A-I (Apo A-I) from the blood, in particular when the SiNPs were preadsorbed with pulmonary surfactants. While the adsorbed Apo A-I ameliorated the cytotoxic and proinflammatory effects of SiNPs, the protein was eliminated from the blood upon clearance of the NPs. However, supplementation of Apo A-I mimic peptide mitigated the atherosclerotic lesion induced by SiNPs. In addition, we found a further declined plasma Apo A-I level in clinical silicosis patients than coronary heart disease patients, suggesting clearance of SiNPs sequestered Apo A-I to compromise the coronal protein's regular biological functions. Together, this study has provided evidence that the protein corona of SiNPs acquired in the blood depletes Apo A-I, a biomarker for prediction of cardiovascular diseases, which gives rise to unexpected toxic effects of the nanoparticles.
Collapse
|
37
|
Hui ST, Wang F, Stappenbeck F, French SW, Magyar CE, Parhami F, Lusis AJ. Oxy210, a novel inhibitor of hedgehog and TGF-β signalling, ameliorates hepatic fibrosis and hypercholesterolemia in mice. ENDOCRINOLOGY DIABETES & METABOLISM 2021; 4:e00296. [PMID: 34505423 PMCID: PMC8502222 DOI: 10.1002/edm2.296] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/19/2022]
Abstract
AIMS Non-alcoholic steatohepatitis (NASH) is associated with increased overall morbidity and mortality in non-alcoholic fatty liver disease (NAFLD) patients. Liver fibrosis is the strongest prognostic factor for clinical outcomes, liver-related mortality and liver transplantation. Currently, no single therapy or medication for NASH has been approved by the U.S. Food and Drug Administration (FDA). Oxy210, an oxysterol derivative, displays the unique property of antagonizing both Hedgehog (Hh) and transforming growth factor-beta (TGF-β) signalling in primary human hepatic stellate cells (HSC). We hypothesized that inhibition of both Hh and TGF-β signalling by Oxy210 could reduce hepatic fibrosis in NASH. In this study, we examined the therapeutic potential of Oxy210 on NASH in vivo. METHODS We examined the effect of Oxy210 treatment on Hh and TGF-β pathways in HSC. The efficacy of Oxy210 on liver fibrosis was tested in a 'humanized' hyperlipidemic mouse model of NASH that has high relevance to human pathology. APPROACH AND RESULTS We show that Oxy210 inhibits both Hh and TGF-β pathways in human HSC and attenuates baseline and TGF-β-induced expression of pro-fibrotic genes in vitro. Oral delivery of Oxy210 in food resulted in significant liver exposure and significantly reduced hepatic fibrosis in mice over the course of the 16-week study with no apparent safety issues. Additionally, we observed several benefits related to NASH phenotype: (a) reduced plasma pro-inflammatory cytokine and the corresponding hepatic gene expression; (b) reduced pro-fibrotic cytokine and inflammasome gene expression in the liver; (c) reduced apoptosis in the liver; (d) reduced hepatic unesterified cholesterol accumulation; and (e) reduced plasma total and unesterified cholesterol levels. CONCLUSIONS Oxy210 effectively ameliorated hepatic fibrosis and inflammation and improved hypercholesterolemia in mice. Our findings suggest that Oxy210 and related analogues are a new class of drug candidates that may serve as potential therapeutics candidates for NASH.
Collapse
Affiliation(s)
- Simon T Hui
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Feng Wang
- MAX BioPharma, Inc, Santa Monica, California, USA
| | | | - Samuel W French
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Clara E Magyar
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
38
|
Soliman AM, Das S, Mahakkanukrauh P. Inflammatory Molecular Mediators and Pathways Involved in Vascular Aging and Stroke: A Comprehensive Review. Curr Med Chem 2021; 29:5522-5542. [PMID: 34488579 DOI: 10.2174/0929867328666210901122359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/01/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
There is an increase in the incidence of cardiovascular diseases with aging and it is one of the leading causes of death worldwide. The main cardiovascular pathologies include atherosclerosis, stroke, myocardial infarction, hypertension and stroke. Chronic inflammation is one of the significant contributors to the age-related vascular diseases. Therefore, it is important to understand the molecular mechanisms of the persistent inflammatory conditions occurring in the blood vessels as well as the signaling pathways involved. Herein, we performed an extant search of literature involving PubMed, ISI, WoS and Scopus databases for retrieving all relevant articles with the most recent findings illustrating the potential role of various inflammatory mediators along with their proposed activated pathways in the pathogenesis and progression of vascular aging. We also highlight the major pathways contributing to age-related vascular disorders. The outlined molecular mechanisms, pathways and mediators of vascular aging represent potential drug targets that can be utilized to inhibit and/or slow the pathogenesis and progression of vascular aging.
Collapse
Affiliation(s)
- Amro M Soliman
- Department of Biological Sciences-Physiology, Cell and Developmental Biology, University of Alberta, Edmonton, AB T6G 2R3. Canada
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, P.C. 123, Al Khoud, Muscat. Oman
| | - Pasuk Mahakkanukrauh
- Department of Anatomy & Excellence center of Osteology Research and Training, Cadaveric Surgical and Training Center, Chiang Mai University, Chiang Mai 50200. Thailand
| |
Collapse
|
39
|
Tabares-Guevara JH, Villa-Pulgarin JA, Hernandez JC. Atherosclerosis: immunopathogenesis and strategies for immunotherapy. Immunotherapy 2021; 13:1231-1244. [PMID: 34382409 DOI: 10.2217/imt-2021-0009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory condition in which atheroma accumulates within the intima of the arterial wall, is a life-threatening manifestation of cardiovascular disease, due to atheroma rupture, chronic luminal narrowing and thrombosis. Current knowledge of the role of a protective immune response in atherosclerotic lesions has provided promising opportunities to develop new immunotherapeutic strategies. In particular, Tregs exert an atheroprotective role by releasing anti-inflammatory cytokines (IL-10/TGF-β) and suppressing autoreactive T lymphocytes. In vivo animal experiments have shown that this can be achieved by developing vaccines that stimulate immunological tolerance to atheroma antigens. Here, we present an overview of the current knowledge of the proatherogenic immune response, and we discuss the strategies currently used as immunoregulatory therapy.
Collapse
Affiliation(s)
| | - Janny A Villa-Pulgarin
- Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Juan C Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| |
Collapse
|
40
|
Muhammad K, Ayoub MA, Iratni R. Vascular Inflammation in Cardiovascular Disease: Is Immune System Protective or Bystander? Curr Pharm Des 2021; 27:2141-2150. [PMID: 33461451 DOI: 10.2174/1381612827666210118121952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/15/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Chronic atherosclerosis induced vascular inflammation and perturbation of lipid metabolism is believed to be a major cause of CVD. Interplay of innate and adaptive Immune system has been interwined with various risk factors associated with the initiation and progression of atherosclerosis in CVD. A large body of evidence indicates a correlation between immunity and atherosclerosis. Retention of plasma lipoproteins in arterial subendothelial wall triggers the T helper type 1 (Th1) cells and monocyte-derived macrophages to form atherosclerotic plaques. In the present review, we will discuss the pathogenesis of CVD in relation to atherosclerosis with a particular focus on pro-atherogenic role of immune cells. Recent findings have also suggested anti-atherogenic roles of different B cell subsets. Therapeutic approaches to target atherosclerosis risk factors have reduced the mortality, but a need exists for the novel therapies to treat arterial vascular inflammation. These insights into the immune pathogenesis of atherosclerosis can lead to new targeted therapeutics to abate cardiovascular mortality and morbidity.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed A Ayoub
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
41
|
Abstract
Atherosclerosis is the leading cause of acute cardiovascular events, and vascular calcification is an important pathological phenomenon in atherosclerosis. Recently, many studies have shown that immune cells are closely associated with the development of atherosclerosis and calcification, but there are many conflicting viewpoints because of immune system complications, such as the pro-atherosclerotic and atheroprotective effects of regulatory B cells (Bregs), T helper type 2 (Th2) cells and T helper type 17 (Th17) cells. In this review, we summarize the studies on the roles of immune cells, especially lymphocytes and macrophages, in atherosclerotic calcification. Furthermore, we prepared graphs showing the relationship between T cells, B cells and macrophages and atherosclerotic calcification. Finally, we highlight some potential issues that are closely associated with the function of immune cells in atherosclerotic calcification. Based on current research results, this review summarizes the relationship between immune cells and atherosclerotic calcification, and it will be beneficial to understand the relationship of immune cells and atherosclerotic calcification.
Collapse
Affiliation(s)
- Jingsong Cao
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jianghua Liu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.,Department of Metabolism and Endocrinology, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
42
|
Testai L, Brancaleone V, Flori L, Montanaro R, Calderone V. Modulation of EndMT by Hydrogen Sulfide in the Prevention of Cardiovascular Fibrosis. Antioxidants (Basel) 2021; 10:antiox10060910. [PMID: 34205197 PMCID: PMC8229400 DOI: 10.3390/antiox10060910] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelial mesenchymal transition (EndMT) has been described as a fundamental process during embryogenesis; however, it can occur also in adult age, underlying pathological events, including fibrosis. Indeed, during EndMT, the endothelial cells lose their specific markers, such as vascular endothelial cadherin (VE-cadherin), and acquire a mesenchymal phenotype, expressing specific products, such as α-smooth muscle actin (α-SMA) and type I collagen; moreover, the integrity of the endothelium is disrupted, and cells show a migratory, invasive and proliferative phenotype. Several stimuli can trigger this transition, but transforming growth factor (TGF-β1) is considered the most relevant. EndMT can proceed in a canonical smad-dependent or non-canonical smad-independent manner and ultimately regulate gene expression of pro-fibrotic machinery. These events lead to endothelial dysfunction and atherosclerosis at the vascular level as well as myocardial hypertrophy and fibrosis. Indeed, EndMT is the mechanism which promotes the progression of cardiovascular disorders following hypertension, diabetes, heart failure and also ageing. In this scenario, hydrogen sulfide (H2S) has been widely described for its preventive properties, but its role in EndMT is poorly investigated. This review is focused on the evaluation of the putative role of H2S in the EndMT process.
Collapse
Affiliation(s)
- Lara Testai
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
- Interdepartmental Center of Ageing, University of Pisa, 56126 Pisa, Italy
- Correspondence:
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (V.B.); (R.M.)
| | - Lorenzo Flori
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
| | - Rosangela Montanaro
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (V.B.); (R.M.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
- Interdepartmental Center of Ageing, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
43
|
Ye Y, Wang M, Huang H. Follicular regulatory T cell biology and its role in immune-mediated diseases. J Leukoc Biol 2021; 110:239-255. [PMID: 33938586 DOI: 10.1002/jlb.1mr0321-601rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Follicular regulatory T (Tfr) cells are recently found to be a special subgroup of regulatory T (Treg) cells. Tfr cells play an important role in regulating the germinal center (GC) response, especially modulating follicular helper T (Tfh) cells and GC-B cells, thereby affecting the production of antibodies. Tfr cells are involved in the generation and development of many immune-related and inflammatory diseases. This article summarizes the advances in several aspects of Tfr cell biology, with special focus on definition and phenotype, development and differentiation, regulatory factors, functions, and interactions with T/B cells and molecules involved in performance and regulation of Tfr function. Finally, we highlight the current understanding of Tfr cells involvement in autoimmunity and alloreactivity, and describe some drugs targeting Tfr cells. These latest studies have answered some basic questions in Tfr cell biology and explored the roles of Tfr cells in immune-mediated diseases.
Collapse
Affiliation(s)
- Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Mowang Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Levkovich TV, Pronko TP. ROLE OF THE TRANSFORMING GROWTH FACTOR β1 IN THE GENESIS OF ARTERIAL HYPERTENSION AND ITS COMPLICATIONS. JOURNAL OF THE GRODNO STATE MEDICAL UNIVERSITY 2021. [DOI: 10.25298/2221-8785-2021-19-1-16-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transforming growth factor beta 1 (TGFβ1) is an actively studied cytokine with rather contradictory effects. The article systematizes and summarizes the scientific data on TGFβ1 and its role in the development and progression of arterial hypertension, with an emphasis on arterial stiffness.
Collapse
|
45
|
Dhaouadi N, Nehme A, Faour WH, Feugier P, Cerutti C, Kacem K, Eid AH, Li JY, Zibara K. Transforming growth factor-β1 inhibits interleukin-1β-induced expression of inflammatory genes and Cathepsin S activity in human vascular smooth muscle cells. Fundam Clin Pharmacol 2021; 35:979-988. [PMID: 33683760 DOI: 10.1111/fcp.12666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE AND DESIGN This study investigated the opposite mechanisms by which IL-1β and TGF-β1 modulated the inflammatory and migratory phenotypes in cultured human intimal vascular smooth muscle cells vSMCs. MATERIALS AND TREATMENT Primary human vSMCs, obtained from twelve hypertensive patients who underwent carotid endarterectomy, were incubated for 24 hours with either 40 pM TGF-β1, or 1 nmol/L IL-1β, or their combination in presence or absence of anti-TGF-β neutralizing antibody. METHODS The expression levels of matrix metalloproteases and their inhibitors, and the elastolytic enzyme cathepsin S (CTSS) and its inhibitor cystatin C were evaluated with RT-PCR. CTSS activity was measured by fluorometry. RESULTS TGF-β1 reversed IL-1β-induced expression of iNOS, CXCL6, IL1R1, MMP12, and CTSS, while upregulated TIMP2 expression. Furthermore, anti-TGF-β neutralizing antibody abrogated TGF-β effects. Combination with IL-1β and TGF-β1 induced the expression of IL1α, IL1β, IL1R1, and CTSS, but suppressed CST3 expression. CTSS expression in the combination treatment was higher than that of cells treated with anti-TGF-β antibodies alone. Moreover, IL-1β-induced CTSS enzymatic activity was reduced when human vSMCs were co-treated with TGF-β, whereas this reduction was abrogated by anti-TGF-β neutralizing antibody. CONCLUSION TGF-β1 abrogated IL-1β-induced expression of inflammatory genes and elastolytic activity in cultured human vSMCs. Thus, TGF-β1 can play a crucial role in impairing IL-1β-induced vascular inflammation and damage involved in the etiology of cardiovascular diseases.
Collapse
Affiliation(s)
- Nedra Dhaouadi
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France.,Unité de Physiologie Intégrée, Laboratoire de Pathologies Vasculaires, Faculté des Sciences de Bizerte, Université de Carthage, Bizerte, Tunisia
| | - Ali Nehme
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France.,PRASE, Lebanese University, Beirut, Lebanon
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Patrick Feugier
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Catherine Cerutti
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Kamel Kacem
- Unité de Physiologie Intégrée, Laboratoire de Pathologies Vasculaires, Faculté des Sciences de Bizerte, Université de Carthage, Bizerte, Tunisia
| | - Ali H Eid
- Biomedical and Pharmaceutical Research Unit and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Jacques-Yuan Li
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon.,Department of Biology, Faculty of sciences - I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
46
|
Mohmmad‐Rezaei M, Arefnezhad R, Ahmadi R, Abdollahpour‐Alitappeh M, Mirzaei Y, Arjmand M, Ferns GA, Bashash D, Bagheri N. An overview of the innate and adaptive immune system in atherosclerosis. IUBMB Life 2021; 73:64-91. [DOI: 10.1002/iub.2425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
AbstractCardiovascular disease is the leading cause of death globally. Coronary artery disease (CAD) is a chronic inflammatory disease usually caused by atherosclerosis, in which the coronary arteries become narrowed by atheromatous plaque. Plaques in atherosclerosis are formed through the accumulation of lipids and various immune cells. Both adaptive and innate immune systems are involved in the pathogenesis of atherosclerosis and facilitate plaque formation and disease progression. Almost all immune system cells, including neutrophils, B cells, T cells monocytes, macrophages, foam cells, and dendritic cells (DCs), play a vital role in atherosclerotic plaque. Atherogenesis, the normal function of the endothelium, is initially disrupted and, then, cells of the immune system are recruited to the endothelium following increased expression of cell adhesion molecules. Accumulation of immune cells and lipids leads to the formation of a necrotic nucleus. As the disease progresses, smooth muscle cells form fibrous layers, whose rupture results in exposing the necrotic nucleus and thrombosis. Accordingly, the present review was conducted to determine the role of different cells in innate and adaptive immune systems in inhibition and progression of atherosclerosis.
Collapse
Affiliation(s)
- Mina Mohmmad‐Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | - Reza Arefnezhad
- Halal Research Center of IRI, FDA Tehran Iran
- Department of Anatomy, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | | | - Yousef Mirzaei
- Department of Biogeosciences, Scientific Research Center Soran University Soran Iraq
| | - Mohammad‐Hassan Arjmand
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
- Cancer Research Center Shahrekord University of Medical Sciences Shahrekord Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education Sussex United Kingdom
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| |
Collapse
|
47
|
Transforming Growth Factor-β Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts. Biomolecules 2020; 10:biom10121666. [PMID: 33322749 PMCID: PMC7763058 DOI: 10.3390/biom10121666] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is essential in embryo development and maintaining normal homeostasis. Extensive evidence shows that TGF-β activation acts on several cell types, including epithelial cells, fibroblasts, and immune cells, to form a pro-fibrotic environment, ultimately leading to fibrotic diseases. TGF-β is stored in the matrix in a latent form; once activated, it promotes a fibroblast to myofibroblast transition and regulates extracellular matrix (ECM) formation and remodeling in fibrosis. TGF-β signaling can also promote cancer progression through its effects on the tumor microenvironment. In cancer, TGF-β contributes to the generation of cancer-associated fibroblasts (CAFs) that have different molecular and cellular properties from activated or fibrotic fibroblasts. CAFs promote tumor progression and chronic tumor fibrosis via TGF-β signaling. Fibrosis and CAF-mediated cancer progression share several common traits and are closely related. In this review, we consider how TGF-β promotes fibrosis and CAF-mediated cancer progression. We also discuss recent evidence suggesting TGF-β inhibition as a defense against fibrotic disorders or CAF-mediated cancer progression to highlight the potential implications of TGF-β-targeted therapies for fibrosis and cancer.
Collapse
|
48
|
Li H, Zou J, Yu XH, Ou X, Tang CK. Zinc finger E-box binding homeobox 1 and atherosclerosis: New insights and therapeutic potential. J Cell Physiol 2020; 236:4216-4230. [PMID: 33275290 DOI: 10.1002/jcp.30177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/12/2020] [Indexed: 12/29/2022]
Abstract
Zinc finger E-box binding homeobox 1 (ZEB1), an important transcription factor belonging to the ZEB family, plays a crucial role in regulating gene expression required for both normal physiological and pathological processes. Accumulating evidence has shown that ZEB1 participates in the initiation and progression of atherosclerotic cardiovascular disease. Recent studies suggest that ZEB1 protects against atherosclerosis by regulation of endothelial cell angiogenesis, endothelial dysfunction, monocyte-endothelial cell interaction, macrophage lipid accumulation, macrophage polarization, monocyte-vascular smooth muscle cell (VSMC) interaction, VSMC proliferation and migration, and T cell proliferation. In this review, we summarize the recent progress of ZEB1 in the pathogenesis of atherosclerosis and provide insights into the prevention and treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China.,Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiang Ou
- Department of Endocrinology, The First Hospital of Changsha, Changsha, Hunan, China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
49
|
Lu X, Wang S, Feng S, Li H. CSE/H 2S system alleviates uremic accelerated atherosclerosis by regulating TGF-β/Smad3 pathway in 5/6 nephrectomy ApoE -/- mice. BMC Nephrol 2020; 21:527. [PMID: 33276745 PMCID: PMC7716493 DOI: 10.1186/s12882-020-02183-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/24/2020] [Indexed: 01/17/2023] Open
Abstract
Background Hydrogen sulfide (H2S) has been shown to inhibit the atherosclerosis development and progression. It is produced by cystathionine γ-lyase (CSE) in the cardiovascular system. In our previous study, it has been shown that CSE/H2S system plays a significant role in the changes of uremic accelerated atherosclerosis (UAAS), but the mechanism is not known clearly. Methods In this study, we explored the antagonism of CSE/H2S system in UAAS and identified its possible signaling molecules in ApoE−/− mice with 5/6 nephrectomy and fed with atherogenic diet. Mice were divided into sham operation group (sham group), UAAS group, sodium hydrosulfide group (UAAS+NaHS group) and propargylglycine group (UAAS+PPG group). Serum creatinine, urea nitrogen, lipid levels and lesion size of atherosclerotic plaque in the aortic roots were analyzed. Meanwhile, the expression of CSE, TGF-β and phosphorylation of Smad3 were detected. Results Compared with sham group, the aortic root of ApoE−/− mice in the UAAS group developed early atherosclerosis, the levels of total cholesterol, triglyceride, low-density lipoprotein-cholesterol, serum creatinine and urea nitrogen were also higher than that in the sham group. NaHS administration can inhibit the development of atherosclerosis, but PPG administration can accelerate the atherosclerosis development. Meanwhile, the protein expression levels of CSE and TGF-β and phosphorylation of Smad3 significantly decreased in the UAAS mice. Treatment of UAAS mice with NaHS inhibited TGF-β protein expression and Smad3 phosphorylation decrease, but PPG treatment had the opposite effect. Conclusions The CSE/H2S system is of great importance for treating atherosclerosis in patients with chronic kidney disease, and it may protect the vascular from atherosclerosis through the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Xiangxue Lu
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Shixiang Wang
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Sujuan Feng
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Han Li
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
50
|
Tengryd C, Nielsen SH, Cavalera M, Bengtsson E, Genovese F, Karsdal M, Dunér P, Orho-Melander M, Nilsson J, Edsfeldt A, Gonçalves I. The proteoglycan mimecan is associated with carotid plaque vulnerability and increased risk of future cardiovascular death. Atherosclerosis 2020; 313:88-95. [DOI: 10.1016/j.atherosclerosis.2020.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/31/2020] [Accepted: 09/10/2020] [Indexed: 01/28/2023]
|