1
|
Wang Y, Zhang S, Ge Y, Miao C, Liu B, Yang T, Qiu X, Ou W. Serum untargeted metabolomics analysis of mice after myocardial infarction affected by qiliqiangxin capsule. J Pharm Biomed Anal 2025; 252:116516. [PMID: 39405786 DOI: 10.1016/j.jpba.2024.116516] [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: 08/26/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 11/07/2024]
Abstract
Qiliqiangxin (QLQX) capsule consists of 11 herbs, namely Huang qi (astragalus membranaceus), Ren shen (ginseng), Fu zi (radix aconiti carmichaeli), Dan shen (salvia miltiorrhiza), Ting li zi (lepidium seed), Ze xie (rhizoma alismatis), Yu zhu (radix polygonati officinalis), Gui zhi (cassia twig), Hong hua (carthamus tinctorious), Xiang jia Pi (cortex periplocae), Chen Pi (pericarpium citri reticulatae), and it is a standardized commercial formula designed to address yang deficiency and to restore the balance of qi in the heart. QLQX is also known to invigorate the blood and promote the circulation of the blood and to promote the use of fluids to relieve water retention and edema, and can be used in cardiovascular diseases such as mild to moderate congestive heart failure resulting from coronary artery disease and hypertension. The further research on the effect of QLQX on cardiac function in mice after myocardial infarction was manipulated. QLQX was given to mice in myocardial infarction model by gavage with appropriate dosage and the samples were analyzed at the end of the animal experiments through the UHPLC-Q-Exactive LC-MS. The liquid mass spectrometry was used to collect and followed by further analysis of the corresponding metabolites and metabolic pathways using metabolomics analysis. As a result, 9 differential metabolites were identified, with 15 being up-regulated and 4 down-regulated following intervention with QLQX. Then the metabolic pathways by KEGG enrichment pathway bubble diagram was analyzed, and 4 metabolic pathways were obtained, and combined with the metabolites that had been screened and analyzed together, finally the two differential metabolites, 2,5-Dihydroxybenzenesulfonic Acid and o-Cresol sulfate were found. The Glycerophospholipid metabolism pathway was closely related to the remaining seven differential metabolites, and the pathway might be an important pathway related to the effects of QLQX on cardiac function in mice.
Collapse
Affiliation(s)
- Yingfei Wang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China
| | - Shijiao Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Yingying Ge
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China
| | - Chunxia Miao
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China
| | - Benrong Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China
| | - Tao Yang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China
| | - Xiangjun Qiu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, PR China.
| | - Wenchao Ou
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, PR China.
| |
Collapse
|
2
|
Yu H, Jin Y, Jeon H, Kim L, Heo KS. Protective effect of 6'-Sialyllactose on LPS-induced macrophage inflammation via regulating Nrf2-mediated oxidative stress and inflammatory signaling pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:503-513. [PMID: 39467714 PMCID: PMC11519721 DOI: 10.4196/kjpp.2024.28.6.503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 10/30/2024]
Abstract
Macrophages play a central role in cardiovascular diseases, like atherosclerosis, by accumulating in vessel walls and inducing sustained local inflammation marked by the release of chemokines, cytokines, and matrix-degrading enzymes. Recent studies indicate that 6'-sialyllactose (6'-SL) may mitigate inflammation by modulating the immune system. Here, we examined the impact of 6'-SL on lipopolysaccharide (LPS)-induced acute inflammation using RAW 264.7 cells and a mouse model. In vivo, ICR mice received pretreatment with 100 mg/kg 6'-SL for 2 h, followed by intraperitoneal LPS injection (10 mg/kg) for 6 h. In vitro, RAW 264.7 cells were preincubated with 6'-SL before LPS stimulation. Mechanistic insights were gained though Western blotting, qRT-PCR, and immunofluorescence analysis, while reactive oxygen species (ROS) production was assessed via DHE assay. 6'-SL effectively attenuated LPS-induced p38 MAPK and Akt phosphorylation, as well as p65 nuclear translocation. Additionally, 6'-SL inhibited LPS-induced expression of tissue damage marker MMP9, IL-1β, and MCP-1 by modulating NF-κB activation. It also reduced ROS levels, mediated by p38 MAPK and Akt pathways. Moreover, 6'-SL restored LPS-suppressed Nrf2 and HO-1 akin to specific inhibitors SB203580 and LY294002. Consistent with in vitro results, 6'-SL decreased oxidative stress, MMP9, and MCP-1 expression in mouse endothelium following LPS-induced macrophage activation. In summary, our findings suggest that 6'-SL holds promise in mitigating atherosclerosis by dampening LPS-induced acute macrophage inflammation.
Collapse
Affiliation(s)
- Hami Yu
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea
| | - Yujin Jin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea
| | - Hyesu Jeon
- Department of Cancer AI & Digital Health, National Cancer Center, Goyang 10408, Korea
| | - Lila Kim
- GeneChem Inc., Daejeon 34025, Korea
| | - Kyung-Sun Heo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
3
|
Luo Y, Hu S, Li Y, Ma L. Inflammation environment-adaptive matrix confinement for three-dimensional modulation of macrophages. Biomater Sci 2024; 12:5324-5336. [PMID: 39248106 DOI: 10.1039/d4bm00939h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
The balance of macrophages in immune reactions is crucial for tissue repair. Despite some studies on responsive surfaces for immunomodulation regulation of macrophage phenotypes via external stimuli, 2D and manual interventions are limited. Herein, to address these limitations, we developed an inflammation environment-responsive macrophage-laden hydrogel-filled scaffold for investigating the impact of matrix confinement on macrophage phenotypes adaptively. We fabricated gelatin scaffolds with a controllable pore size and found that macrophages within smaller pores tended to have an anti-inflammation phenotype. We prepared poly(vinyl alcohol) (PVA)-based hydrogels crosslinked with phenylboronic acid (PBA)-based linkers. The hydrogels possessed shear-thinning, cell-loading, and ROS-sensitive-degradation abilities. Subsequently, a macrophage-laden hydrogel-filled scaffold was fabricated by filling the hydrogels into the porous scaffold under vacuum. With the degradation of the hydrogels under the overexpression of ROS in an inflammation environment, the macrophages were transferred from a state with strong matrix confinement to that with a weaker one. Meanwhile, with the change in matrix confinement, the macrophages upregulated the expressions of Arg-1 and IL-10 and downregulated the expressions of IL-1β, TNF-α, and IL-6, indicating polarization toward the anti-inflammatory phenotype. The inflammation environment-adaptive modulation of macrophage phenotypes in 3D provides a smart and biomimetic strategy for immunomodulation and regenerative medicine.
Collapse
Affiliation(s)
- Yilun Luo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
- MOE Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Sentao Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Yan Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
4
|
Hage Z, Madeira MM, Koliatsis D, Tsirka SE. Convergence of endothelial dysfunction, inflammation and glucocorticoid resistance in depression-related cardiovascular diseases. BMC Immunol 2024; 25:61. [PMID: 39333855 PMCID: PMC11428380 DOI: 10.1186/s12865-024-00653-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Major Depressive Disorder, or depression, has been extensively linked to dysregulated HPA axis function, chronic inflammation and cardiovascular diseases. While the former two have been studied in depth, the mechanistic connection between depression and cardiovascular disease is unclear. As major mediators of vascular homeostasis, vascular pathology and immune activity, endothelial cells represent an important player connecting the diseases. Exaggerated inflammation and glucocorticoid function are important topics to explore in the endothelial response to MDD. Glucocorticoid resistance in several cell types strongly promotes inflammatory signaling and results in worsened severity in many diseases. However, endothelial health and inflammation in chronic stress and depression are rarely considered from the perspective of glucocorticoid signaling and resistance. In this review, we aim to discuss (1) endothelial dysfunction in depression, (2) inflammation in depression, (3) general glucocorticoid resistance in depression and (4) endothelial glucocorticoid resistance in depression co-morbid inflammatory diseases. We will first describe vascular pathology, inflammation and glucocorticoid resistance separately in depression and then describe their potential interactions with one another in depression-relevant diseases. Lastly, we will hypothesize potential mechanisms by which glucocorticoid resistance in endothelial cells may contribute to vascular disease states in depressed people. Overall, endothelial-glucocorticoid signaling may play an important role in connecting depression and vascular pathology and warrants further study.
Collapse
Affiliation(s)
- Zachary Hage
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Miguel M Madeira
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Dimitris Koliatsis
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
5
|
He Z, Chen W, Hu K, Luo Y, Zeng W, He X, Li T, Ouyang J, Li Y, Xie L, Zhang Y, Xu Q, Yang S, Guo M, Zou W, Li Y, Huang L, Chen L, Zhang X, Saiding Q, Wang R, Zhang MR, Kong N, Xie T, Song X, Tao W. Resolvin D1 delivery to lesional macrophages using antioxidative black phosphorus nanosheets for atherosclerosis treatment. NATURE NANOTECHNOLOGY 2024; 19:1386-1398. [PMID: 38898135 DOI: 10.1038/s41565-024-01687-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The buildup of plaques in atherosclerosis leads to cardiovascular events, with chronic unresolved inflammation and overproduction of reactive oxygen species (ROS) being major drivers of plaque progression. Nanotherapeutics that can resolve inflammation and scavenge ROS have the potential to treat atherosclerosis. Here we demonstrate the potential of black phosphorus nanosheets (BPNSs) as a therapeutic agent for the treatment of atherosclerosis. BPNSs can effectively scavenge a broad spectrum of ROS and suppress atherosclerosis-associated pro-inflammatory cytokine production in lesional macrophages. We also demonstrate ROS-responsive, targeted-peptide-modified BPNS-based carriers for the delivery of resolvin D1 (an inflammation-resolving lipid mediator) to lesional macrophages, which further boosts the anti-atherosclerotic efficacy. The targeted nanotherapeutics not only reduce plaque areas but also substantially improve plaque stability in high-fat-diet-fed apolipoprotein E-deficient mice. This study presents a therapeutic strategy against atherosclerosis, and highlights the potential of BPNS-based therapeutics to treat other inflammatory diseases.
Collapse
Affiliation(s)
- Zhongshan He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kuan Hu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaoyao Luo
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wanqin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xi He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Qin Xu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuping Yang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mengran Guo
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zou
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanfei Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lingjing Huang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tian Xie
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
7
|
Allaeys I, Lemaire G, Leclercq M, Lacasse E, Fleury M, Dubuc I, Gudimard L, Puhm F, Tilburg J, Stone A, Machlus KR, Droit A, Flamand L, Boilard E. SARS-CoV-2 infection modifies the transcriptome of the megakaryocytes in the bone marrow. Blood Adv 2024; 8:2777-2789. [PMID: 38522092 PMCID: PMC11176959 DOI: 10.1182/bloodadvances.2023012367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 03/26/2024] Open
Abstract
ABSTRACT Megakaryocytes (MKs), integral to platelet production, predominantly reside in the bone marrow (BM) and undergo regulated fragmentation within sinusoid vessels to release platelets into the bloodstream. Inflammatory states and infections influence MK transcription, potentially affecting platelet functionality. Notably, COVID-19 has been associated with altered platelet transcriptomes. In this study, we investigated the hypothesis that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection could affect the transcriptome of BM MKs. Using spatial transcriptomics to discriminate subpopulations of MKs based on proximity to BM sinusoids, we identified ∼19 000 genes in MKs. Machine learning techniques revealed that the transcriptome of healthy murine BM MKs exhibited minimal differences based on proximity to sinusoid vessels. Furthermore, at peak SARS-CoV-2 viremia, when the disease primarily affected the lungs, MKs were not significantly different from those from healthy mice. Conversely, a significant divergence in the MK transcriptome was observed during systemic inflammation, although SARS-CoV-2 RNA was never detected in the BM, and it was no longer detectable in the lungs. Under these conditions, the MK transcriptional landscape was enriched in pathways associated with histone modifications, MK differentiation, NETosis, and autoimmunity, which could not be explained by cell proximity to sinusoid vessels. Notably, the type I interferon signature and calprotectin (S100A8/A9) were not induced in MKs under any condition. However, inflammatory cytokines induced in the blood and lungs of COVID-19 mice were different from those found in the BM, suggesting a discriminating impact of inflammation on this specific subset of cells. Collectively, our data indicate that a new population of BM MKs may emerge through COVID-19-related pathogenesis.
Collapse
Affiliation(s)
- Isabelle Allaeys
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Guillaume Lemaire
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Mickaël Leclercq
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Emile Lacasse
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Maude Fleury
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Leslie Gudimard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Florian Puhm
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Julia Tilburg
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA
| | - Andrew Stone
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA
| | - Arnaud Droit
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| |
Collapse
|
8
|
Teuwen JTJ, van der Vorst EPC, Maas SL. Navigating the Maze of Kinases: CaMK-like Family Protein Kinases and Their Role in Atherosclerosis. Int J Mol Sci 2024; 25:6213. [PMID: 38892400 PMCID: PMC11172518 DOI: 10.3390/ijms25116213] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating low-density lipoprotein (LDL) levels are a major risk factor for cardiovascular diseases (CVD), and even though current treatment strategies focusing on lowering lipid levels are effective, CVD remains the primary cause of death worldwide. Atherosclerosis is the major cause of CVD and is a chronic inflammatory condition in which various cell types and protein kinases play a crucial role. However, the underlying mechanisms of atherosclerosis are not entirely understood yet. Notably, protein kinases are highly druggable targets and represent, therefore, a novel way to target atherosclerosis. In this review, the potential role of the calcium/calmodulin-dependent protein kinase-like (CaMKL) family and its role in atherosclerosis will be discussed. This family consists of 12 subfamilies, among which are the well-described and conserved liver kinase B1 (LKB1) and 5' adenosine monophosphate-activated protein kinase (AMPK) subfamilies. Interestingly, LKB1 plays a key role and is considered a master kinase within the CaMKL family. It has been shown that LKB1 signaling leads to atheroprotective effects, while, for example, members of the microtubule affinity-regulating kinase (MARK) subfamily have been described to aggravate atherosclerosis development. These observations highlight the importance of studying kinases and their signaling pathways in atherosclerosis, bringing us a step closer to unraveling the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jules T. J. Teuwen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
9
|
Hachuła M, Kosowski M, Basiak M, Okopień B. Influence of Dulaglutide on Serum Biomarkers of Atherosclerotic Plaque Instability: An Interventional Analysis of Cytokine Profiles in Diabetic Subjects-A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:908. [PMID: 38929525 PMCID: PMC11205508 DOI: 10.3390/medicina60060908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: The rise in global diabetes cases, reaching a staggering 529 million in 2021 from 108 million in 1980, underscores the urgency of addressing its complications, notably macrovascular ones like coronary artery, cerebrovascular, and peripheral artery diseases, which contribute to over 50% of diabetes mortality. Atherosclerosis, linked to hyperglycemia-induced endothelial dysfunction, is pivotal in cardiovascular disease development. Cytokines, including pentraxin 3 (PTX3), copeptin, lipoprotein(a) [Lp(a)], and matrix metalloproteinase-9 (MMP-9), influence atherosclerosis progression and plaque vulnerability. Inhibiting atherosclerosis progression is crucial, especially in diabetic individuals. Glucagon-like peptide 1 receptor agonists (GLP-1 RAs), increasingly used for type 2 diabetes, show promise in reducing the cardiovascular risk, sparking interest in their effects on atherogenesis. This study sought to examine the effects of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) on biomarkers that indicate the instability of atherosclerotic plaques. These biomarkers include pentraxin 3 (PTX3), copeptin (CPC), matrix metalloproteinase-9 (MMP-9), and lipoprotein(a) [Lp(a)]. Materials and Methods: A total of 34 participants, ranging in age from 41 to 81 years (with an average age of 61), who had been diagnosed with type 2 diabetes mellitus (with a median HbA1c level of 8.8%), dyslipidemia, and verified atherosclerosis using B-mode ultrasonography, were included in the study. All subjects were eligible to initiate treatment with a GLP-1 RA-dulaglutide. Results: Significant reductions in anthropometric parameters, blood pressure, fasting glucose levels, and HbA1c levels were observed posttreatment. Moreover, a notable decrease in biochemical markers associated with atherosclerotic plaque instability, particularly PTX3 and MMP-9 (p < 0.001), as well as Lp(a) (p < 0.05), was evident following the GLP-1 RA intervention. Conclusions: These findings underscore the potential of GLP-1 RAs in mitigating atherosclerosis progression and plaque vulnerability, thus enhancing cardiovascular outcomes in individuals with type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Marcin Basiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (M.H.); (M.K.); (B.O.)
| | | |
Collapse
|
10
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Ma Y, Wang Q, Du S, Luo J, Sun X, Jia B, Ge J, Dong J, Jiang S, Li Z. Multipathway Regulation for Targeted Atherosclerosis Therapy Using Anti-miR-33-Loaded DNA Origami. ACS NANO 2024. [PMID: 38321605 DOI: 10.1021/acsnano.3c10213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Given the multifactorial pathogenesis of atherosclerosis (AS), a chronic inflammatory disease, combination therapy arises as a compelling approach to effectively address the complex interplay of pathogenic mechanisms for a more desired treatment outcome. Here, we present cRGD/ASOtDON, a nanoformulation based on a self-assembled DNA origami nanostructure for the targeted combination therapy of AS. cRGD/ASOtDON targets αvβ3 integrin receptors overexpressed on pro-inflammatory macrophages and activated endothelial cells in atherosclerotic lesions, alleviates the oxidative stress induced by extracellular and endogenous reactive oxygen species, facilitates the polarization of pro-inflammatory macrophages toward the anti-inflammatory M2 phenotype, and inhibits foam cell formation by promoting cholesterol efflux from macrophages by downregulating miR-33. The antiatherosclerotic efficacy and safety profile of cRGD/ASOtDON, as well as its mechanism of action, were validated in an AS mouse model. cRGD/ASOtDON treatment reversed AS progression and restored normal morphology and tissue homeostasis of the diseased artery. Compared to probucol, a clinical antiatherosclerotic drug with a similar mechanism of action, cRGD/ASOtDON enabled the desired therapeutic outcome at a notably lower dosage. This study demonstrates the benefits of targeted combination therapy in AS management and the potential of self-assembled DNA nanoformulations in addressing multifactorial inflammatory conditions.
Collapse
Affiliation(s)
- Yuxuan Ma
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Qi Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Shiyu Du
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingwei Luo
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Xiaolei Sun
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Bin Jia
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingru Ge
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P. R. China
| | - Shuoxing Jiang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Zhe Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| |
Collapse
|
12
|
Ehteshami A, Shirban F, Bagherniya M, Sathyapalan T, Jamialahmadi T, Sahebkar A. The Association between High-density Lipoproteins and Periodontitis. Curr Med Chem 2024; 31:6407-6428. [PMID: 37493158 DOI: 10.2174/0929867331666230726140736] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/27/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023]
Abstract
Periodontitis is one of the most typical chronic dental diseases. This inflammatory disease can change various functions of immune cells and impair lipid metabolism through proinflammatory cytokines. High-Density Lipoprotein (HDL) is considered protective of the cardiovascular system. It has anti-thrombotic and anti-inflammatory effects. In this article, we have reviewed the association between periodontitis and HDL. Various studies have demonstrated a reverse relationship between inflammatory cytokines and HDL. HDL contains antioxidative enzymes and proteins, whereas periopathogens impair HDL's antioxidant function. The presence of periodontal bacteria is associated with a low HDL level in patients with periodontitis. Genetic variants in the interleukin- 6 (IL)-6 gene and cytochrome (CYP)1A1 rs1048943 gene polymorphism are associated with HDL levels and periodontal status. Studies showed that HDL levels improve after treatment for periodontitis. On the one hand, periodontal pathogenic bacteria and their metabolites and pro-inflammatory cytokines from periodontal infection can result in various disorders of lipid metabolism and lipid peroxidation. On the other hand, hyperlipidemia and lipid peroxidation stimulate proinflammatory cytokines, resulting in oxidative stress and delayed wound healing, making individuals susceptible to periodontitis.
Collapse
Affiliation(s)
- Ailin Ehteshami
- Department of Orthodontics, Dental Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farinaz Shirban
- Department of Orthodontics, Dental Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes Endocrinology and Metabolism, Hull York Medical School, University of Hull, UK of Great Britain and Northern Ireland, Hull, UK
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Shi X, Tao T, Wang Y, Han Y, Xu X, Yin Q, Wang F, Liu R, Liu X. Heavy macrophage infiltration identified by optical coherence tomography relates to plaque rupture. Ann Clin Transl Neurol 2023; 10:2334-2346. [PMID: 37822283 PMCID: PMC10723231 DOI: 10.1002/acn3.51923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/31/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVE Risk stratification plays a critical role in patients with asymptomatic carotid atherosclerotic stenosis. Heavy macrophage infiltration (HMC) is an important factor of plaque destabilization. However, in vivo imaging technologies and screening criteria for HMC remain limited. We aimed to (i) introduce algorithms for in vivo detection of macrophage infiltrations using optical coherence tomography (OCT) and (ii) to investigate the threshold of HMC and its association with plaque vulnerability. METHODS Ex vivo OCT images were co-registered with histopathology in 282 cross-sectional pairs from 19 carotid endarterectomy specimens. Of these, 197 randomly selected pairs were employed to define the parameters, and the remaining 85 pairs were used to evaluate the accuracy of the OCT-based algorithm in detecting macrophage infiltrations. Clinical analysis included 93 patients receiving carotid OCT evaluation. The prevalence and burden of macrophage infiltration were analyzed. Multivariable and subgroup analysis were performed to investigate the association between HMC and plaque rupture. RESULTS The sensitivity and specificity of algorithm for detecting macrophage infiltration were 88.0% and 74.9%, respectively. Of 93 clinical patients, ruptured plaques exhibited higher prevalence of macrophage infiltration than nonruptured plaques (83.7% [36/43] vs 32.0% [16/50], p < 0.001). HMC was identified when the macrophage index was greater than 60.2 (sensitivity = 74.4%, specificity = 84.0%). Multivariable analysis showed that HMC and multiple calcification were independent risk factors for non-lipid-rich plaque rupture. INTERPRETATION This study provides a novel approach and screening criteria for HMC, which might be valuable for atherosclerotic risk stratification.
Collapse
Affiliation(s)
- Xuan Shi
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yi Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yunfei Han
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Xiaohui Xu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Qin Yin
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Fang Wang
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Xinfeng Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Stroke Center and Department of Neurology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
14
|
Yu J, Xu W, Dong Q, Ji Q, Cheng M, Hu D, Cai Y, Zeng Q, Yu K. Latency-associated peptide (LAP) +CD4 + regulatory T cells prevent atherosclerosis by modulating macrophage polarization. Clin Immunol 2023; 255:109767. [PMID: 37689092 DOI: 10.1016/j.clim.2023.109767] [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: 06/14/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023]
Abstract
RATIONALE A persistent autoimmune and inflammatory response plays a critical role in the progression of atherosclerosis. The transcription factor forkhead box P3 (Foxp3)+CD4+ regulatory T cells (Foxp3+ Tregs) attenuate atherosclerosis. Latency-associated peptide (LAP)+CD4+ T cells are a new class of Tregs whose role in atherosclerosis is unknown. OBJECTIVE To investigate the function of CD4+LAP+ Tregs in inhibiting inflammation and preventing atherosclerosis. METHODS AND RESULTS Depletion of CD4+LAP+ Tregs results in aggravated inflammation and atherosclerotic lesions. Mechanistically, CD4+LAP+ Treg depletion was associated with decreased M2-like macrophages and increased Th1 and Th17 cells, characterized by increased unstable plaque promotion and decreased expression of inflammation-resolving factors in both arteries and immune organs. In contrast, adoptive transfer of CD4+LAP+ Tregs to ApoE-/- mice or CD4-/-ApoE-/- mice led to decreased atherosclerotic lesions. Compared with control animals, adoptive transfer of CD4+LAP+ Tregs induced M2-like macrophage differentiation within the atherosclerotic lesion and spleen, associated with increased collagen and α-SMA in plaques and decreased expression of MMP-2 and MMP-9. Mechanistic studies reveal that isolated CD4+LAP+ Tregs exhibit a tolerance phenotype, with increased expression of inhibitory cytokines and coinhibitory molecules. After coculture with CD4+LAP+ Tregs, monocytes/macrophages display typical features of M2 macrophages, including upregulated expression of CD206 and Arg-1 and decreased production of MCP-1, IL-6, IL-1β and TNF-α, which was almost abrogated by transwell and partially TGF-β1 neutralization. RNA-seq analysis showed different gene expression profiles between CD4+LAP+ Tregs and LAP-CD4+ T cells and between CD4+LAP+ Tregs of ApoE-/- mice and CD4+LAP+ Tregs of C57BL/6 mice, of which Fancd2 and IL4i1 may contribute to the powerful inhibitory properties of CD4+LAP+ Tregs. Furthermore, the number and the suppressive properties of CD4+LAP+ Tregs were impaired by oxLDL. CONCLUSIONS Our data indicate that the remaining CD4+LAP+ Tregs play a protective role in atherosclerosis by modulating monocyte/macrophage differentiation and regulatory factors, which may partly explain the protective effect of T cells tolerance in atherosclerosis. Moreover, adoptive transfer of CD4+LAP+ Tregs constitutes a novel approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Jian Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wenbin Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qian Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Cai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
15
|
Hammaréus F, Nilsson L, Ong KL, Kristenson M, Festin K, Lundberg AK, Chung RWS, Swahn E, Alfredsson J, Holm Nielsen S, Jonasson L. Plasma type I collagen α1 chain in relation to coronary artery disease: findings from a prospective population-based cohort and an acute myocardial infarction prospective cohort in Sweden. BMJ Open 2023; 13:e073561. [PMID: 37714678 PMCID: PMC10510861 DOI: 10.1136/bmjopen-2023-073561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 09/01/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVES To investigate the association between type I collagen α1 chain (COL1α1) levels and coronary artery disease (CAD) by using absolute quantification in plasma. Also, to investigate the correlates of COL1α1 to clinical characteristics and circulating markers of collagen metabolism. DESIGN Life conditions, Stress and Health (LSH) study: prospective cohort study, here with a nested case-control design.Assessing Platelet Activity in Coronary Heart Disease (APACHE) study: prospective cohort study. SETTING LSH: primary care setting, southeast Sweden.APACHE: cardiology department, university hospital, southeast Sweden. PARTICIPANTS LSH: 1007 randomly recruited individuals aged 45-69 (50% women). Exclusion criteria was serious disease. After 13 years of follow-up, 86 cases with primary endpoint were identified and sex-matched/age-matched to 184 controls. APACHE 125 patients with myocardial infarction (MI), 73 with ST-elevation MI and 52 with non-ST-elevation MI. EXCLUSION CRITERIA Intervention study participation, warfarin treatment and short life expectancy. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcome was the association between baseline COL1α1 and first-time major event of CAD, defined as fatal/non-fatal MI or coronary revascularisation after 13 years. Secondary outcomes were the association between the collagen biomarkers PRO-C1 (N-terminal pro-peptide of type I collagen)/C1M (matrix metalloproteinase-mediated degradation of type I collagen) and CAD; temporal change of COL1α1 after acute MI up to 6 months and lastly, correlates between COL1α1 and patient characteristics along with circulating markers of collagen metabolism. RESULTS COL1α1 levels were associated with CAD, both unadjusted (HR=0.69, 95% CI=0.56 to 0.87) and adjusted (HR=0.55, 95% CI=0.41 to 0.75). PRO-C1 was associated with CAD, unadjusted (HR=0.62, 95% CI=0.47 to 0.82) and adjusted (HR=0.61, 95% CI=0.43 to 0.86), while C1M was not. In patients with MI, COL1α1 remained unchanged up to 6 months. COL1α1 was correlated to PRO-C1, but not to C1M. CONCLUSIONS Plasma COL1α1 was independently and inversely associated with CAD. Furthermore, COL1α1 appeared to reflect collagen synthesis but not degradation. Future studies are needed to confirm whether COL1α1 is a clinically useful biomarker of CAD.
Collapse
Affiliation(s)
- Filip Hammaréus
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Lennart Nilsson
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Kwok-Leung Ong
- Faculty of Medicine and Health, NHMRC Clinical Trials Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Margareta Kristenson
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Karin Festin
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Anna K Lundberg
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Rosanna W S Chung
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Eva Swahn
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Joakim Alfredsson
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | - Signe Holm Nielsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
- Nordic Bioscience, Herlev, Denmark
| | - Lena Jonasson
- Department of Health Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| |
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
|
Alonso-Herranz L, Albarrán-Juárez J, Bentzon JF. Mechanisms of fibrous cap formation in atherosclerosis. Front Cardiovasc Med 2023; 10:1254114. [PMID: 37671141 PMCID: PMC10475556 DOI: 10.3389/fcvm.2023.1254114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
The fibrous cap is formed by smooth muscle cells that accumulate beneath the plaque endothelium. Cap rupture is the main cause of coronary thrombosis, leading to infarction and sudden cardiac death. Therefore, the qualities of the cap are primary determinants of the clinical outcome of coronary and carotid atherosclerosis. In this mini-review, we discuss current knowledge about the formation of the fibrous cap, including cell recruitment, clonal expansion, and central molecular signaling pathways. We also examine the differences between mouse and human fibrous caps and explore the impact of anti-atherosclerotic therapies on the state of the fibrous cap. We propose that the cap should be understood as a neo-media to substitute for the original media that becomes separated from the surface endothelium during atherogenesis and that embryonic pathways involved in the development of the arteria media contribute to cap formation.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julián Albarrán-Juárez
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus and Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Basiak M, Hachula M, Kosowski M, Machnik G, Maliglowka M, Dziubinska-Basiak M, Krysiak R, Okopien B. The Effect of PCSK9 Inhibition on the Stabilization of Atherosclerotic Plaque Determined by Biochemical and Diagnostic Imaging Methods. Molecules 2023; 28:5928. [PMID: 37570897 PMCID: PMC10421011 DOI: 10.3390/molecules28155928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Atherosclerosis is a multifactorial, progressive, chronic inflammatory disease. Ultrasound and magnetic resonance imaging are the most accurate predictors of atherosclerotic plaque instability (MRI). Cytokines such as osteopontin, osteoprotegerin, and metalloproteinase 9 could be used as the most recent markers to identify and track the efficacy of anti-atherosclerotic therapy. Patients with USG and MRI-verified unstable atherosclerotic plaque were included in the study. Biomarker concentrations were measured and compared before and after PCSK9 inhibitor therapy. Additionally, concentrations prior to treatment were correlated with MRI images of the carotid artery. After treatment with alirocumab, the concentrations of MMP-9 (p < 0.01) and OPN, OPG (p < 0.05) decreased significantly. Furthermore, the results of OPN, OPG, and MMP 9 varied significantly depending on the type of atherosclerotic plaque in the MRI assay. In stable atherosclerotic plaques, the concentrations of OPN and OPG were greater (p < 0.01), whereas the concentration of MMP9 correlated with the instability of the plaque (p < 0.05). We demonstrated, probably for the first time, that alirocumab therapy significantly decreased the serum concentration of atherosclerotic plaque markers. In addition, we demonstrated the relationship between the type of atherosclerotic plaque as determined by carotid MRI and the concentration of these markers.
Collapse
Affiliation(s)
- Marcin Basiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Marcin Hachula
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Michal Kosowski
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Mateusz Maliglowka
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | | | - Robert Krysiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Boguslaw Okopien
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| |
Collapse
|
19
|
Zhang X, Kang Z, Yin D, Gao J. Role of neutrophils in different stages of atherosclerosis. Innate Immun 2023; 29:97-109. [PMID: 37491844 PMCID: PMC10468622 DOI: 10.1177/17534259231189195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/20/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
Neutrophils constitute the first line of defense in human immunity and can be attracted to inflamed and infected sites by various chemokines. As essential players in immune processes, neutrophils theoretically play integral roles in the course of chronic inflammation-induced atherosclerosis. However, because neutrophils are rarely found in atherosclerotic lesions, their involvement in the pathophysiological progression of atherosclerosis has been largely underestimated or ignored. Recent research has revealed convincing evidence showing the presence of neutrophils in atherosclerotic lesions and has revealed neutrophil contributions to different atherosclerosis stages in mice and humans. This review describes the underlying mechanisms of neutrophils in different stages of atherosclerosis and highlights potential neutrophil-targeted therapeutic strategies relevant to atherosclerosis. An in-depth understanding of neutrophils' roles in atherosclerosis pathology will promote exploration of new methods for the prevention and treatment of atherogenesis and atherothrombosis.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Zhanfang Kang
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Dazhong Yin
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Jun Gao
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
20
|
Gao DL, Johal MS. LRP-1 Binds Fibrinogen in a Sialylation-Dependent Manner: A Quartz Crystal Microbalance Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:10375-10382. [PMID: 37459110 DOI: 10.1021/acs.langmuir.3c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in the United States. Atherosclerosis, the dominant condition leading to CVD, is characterized by fibrofatty plaque formation. Fibrinogen, an important clotting factor, has been known to promote atherogenesis as it retains the ability to trigger smooth muscle cell proliferation, localize in areas crucial to plaque progression, and bind both platelets and leukocytes. Yet, these consequences can be suppressed through anti-inflammatory receptors like LRP-1─an endocytic receptor part of the LDLR family responsible for the endocytosis of cell debris and protein degradation products. However, the continual progression of atherosclerosis in many patients indicates that such clearance mechanisms, deemed efferocytosis, are impaired during atherosclerosis. Using the quartz crystal microbalance with dissipation monitoring (QCM-D) as a platform to investigate receptor-ligand interactions, we identify fibrinogen to be a ligand of LRP-1 and characterize its binding with LRP-1. By examining a key player in atherosclerosis development─the effect of sialidase on receptor efficacy─we found that the desialylation of LRP-1 reduces its ability to bind fibrinogen. Protein docking simulations highlighted the N-terminus portion of fibrinogen's α domain as the LRP-1 docking site. The sialylated O-linked glycans at T894 and T935 have the potential to mediate direct binding of LRP-1 to fibrinogen and support the tertiary structure of LRP-1. These phenomena are important in showing a probable cause of defective efferocytosis that occurs readily during atherosclerosis.
Collapse
Affiliation(s)
- Daniel L Gao
- Department of Chemistry, Pomona College, 645 N College Avenue, Claremont, California 91711 United States
| | - Malkiat S Johal
- Department of Chemistry, Pomona College, 645 N College Avenue, Claremont, California 91711 United States
| |
Collapse
|
21
|
Galindo CL, Khan S, Zhang X, Yeh YS, Liu Z, Razani B. Lipid-laden foam cells in the pathology of atherosclerosis: shedding light on new therapeutic targets. Expert Opin Ther Targets 2023; 27:1231-1245. [PMID: 38009300 PMCID: PMC10843715 DOI: 10.1080/14728222.2023.2288272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
INTRODUCTION Lipid-laden foam cells within atherosclerotic plaques are key players in all phases of lesion development including its progression, necrotic core formation, fibrous cap thinning, and eventually plaque rupture. Manipulating foam cell biology is thus an attractive therapeutic strategy at early, middle, and even late stages of atherosclerosis. Traditional therapies have focused on prevention, especially lowering plasma lipid levels. Despite these interventions, atherosclerosis remains a major cause of cardiovascular disease, responsible for the largest numbers of death worldwide. AREAS COVERED Foam cells within atherosclerotic plaques are comprised of macrophages, vascular smooth muscle cells, and other cell types which are exposed to high concentrations of lipoproteins accumulating within the subendothelial intimal layer. Macrophage-derived foam cells are particularly well studied and have provided important insights into lipid metabolism and atherogenesis. The contributions of foam cell-based processes are discussed with an emphasis on areas of therapeutic potential and directions for drug development. EXERT OPINION As key players in atherosclerosis, foam cells are attractive targets for developing more specific, targeted therapies aimed at resolving atherosclerotic plaques. Recent advances in our understanding of lipid handling within these cells provide insights into how they might be manipulated and clinically translated to better treat atherosclerosis.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Saifur Khan
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Yu-Sheng Yeh
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Ziyang Liu
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
| |
Collapse
|
22
|
Chen C, Wang J, Liu C, Hu J. Cardiac resident macrophages: key regulatory mediators in the aftermath of myocardial infarction. Front Immunol 2023; 14:1207100. [PMID: 37457720 PMCID: PMC10348646 DOI: 10.3389/fimmu.2023.1207100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Acute myocardial infarction (MI) is a prevalent and highly fatal global disease. Despite significant reduction in mortality rates with standard treatment regimens, the risk of heart failure (HF) remains high, necessitating innovative approaches to protect cardiac function and prevent HF progression. Cardiac resident macrophages (cMacs) have emerged as key regulators of the pathophysiology following MI. cMacs are a heterogeneous population composed of subsets with different lineage origins and gene expression profiles. Several critical aspects of post-MI pathophysiology have been shown to be regulated by cMacs, including recruitment of peripheral immune cells, clearance and replacement of damaged myocardial cells. Furthermore, cMacs play a crucial role in regulating cardiac fibrosis, risk of arrhythmia, energy metabolism, as well as vascular and lymphatic remodeling. Given the multifaceted roles of cMacs in post-MI pathophysiology, targeting cMacs represents a promising therapeutic strategy. Finally, we discuss novel treatment strategies, including using nanocarriers to deliver drugs to cMacs or using cell therapies to introduce exogenous protective cMacs into the heart.
Collapse
|
23
|
Kim BY, Son Y, Kim BJ, Chung SW, Lee D, Eo SK, Kim K. Atheroma-Relevant 7-Oxysterols Differentially Upregulate Cd14 Expression. Int J Mol Sci 2023; 24:10542. [PMID: 37445719 DOI: 10.3390/ijms241310542] [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: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The expression of CD14 in monocytic cells is elevated in atherosclerotic lesions where 7-oxyterols are abundant. However, it remains unknown whether atheroma-relevant 7-oxysterols are involved in receptor expression. Therefore, we investigated the effects of 7α-hydroxycholesterol (7αOHChol), 7β-hydroxycholesterol (7βOHChol), and 7-ketocholesterol (7K) on CD14 levels in THP-1 cells. The three 7-oxysterols increased CD14 transcript levels at a distinct time point, elevated cellular CD14 protein levels, and promoted the release of soluble CD (sCD14) from THP-1 cells. Our data revealed that CD14 expression was most strongly induced after treatment with 7αOHChol. Moreover, 7αOHChol alone upregulated membrane-bound CD14 levels and enhanced responses to lipopolysaccharides, as determined by CCL2 production and monocytic cell migration. The 7-oxysterols also increased the gelatinolytic activity of MMP-9, and a cell-permeable, reversible MMP-9 inhibitor, MMP-9 inhibitor I, significantly impaired sCD14 release. These results indicate that 7-oxysterols differentially induce CD14 expression in vascular cells and contribute to the monocytic cell expression of CD14 via overlapping, but distinct, mechanisms.
Collapse
Affiliation(s)
- Bo-Young Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Joon Kim
- Kim Byoung Joon Ledas Varicose Vein Clinic, Busanjin-gu, Busan 47256, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Sung Woon Chung
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
24
|
Chen H, Zhang L, Mi S, Wang H, Wang C, Jia W, Gong L, Dong H, Xu B, Jing Y, Ge P, Pei Z, Zhong L, Yang J. FURIN suppresses the progression of atherosclerosis by promoting macrophage autophagy. FASEB J 2023; 37:e22933. [PMID: 37093709 DOI: 10.1096/fj.202201762rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 04/25/2023]
Abstract
FURIN, a member of the mammalian proprotein convertases (PCs) family, can promote the proteolytic maturation of proproteins. It has been shown that FURIN plays an important role in the progression of atherosclerosis (AS). Current evidence indicates that autophagy widely participates in atherogenesis. This study aimed to explore whether FURIN could affect atherogenesis via autophagy. The effect of FURIN on autophagy was studied using aortic tissues from aortic dissection patients who had BENTALL surgery, as well as macrophages and ApoE-/- mice. In atherosclerotic plaques of aortic tissues from patients, FURIN expression and autophagy were elevated. In macrophages, FURIN-shRNA and FURIN-overexpression lentivirus were used to intervene in FURIN expression. The results showed that FURIN overexpression accelerated LC3 formation in macrophages during the autophagosome formation phase. Furthermore, FURIN-induced autophagy resulted in lower lipid droplet concentrations in macrophages. The western blot revealed that FURIN regulated autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway. In vivo, FURIN overexpression resulted in increased macrophage LC3 formation in ApoE-/- mice atherosclerotic plaques, confirming that FURIN could inhibit the progression of AS by promoting macrophage autophagy. The present study demonstrated that FURIN suppressed the progression of AS by promoting macrophage autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway, which attenuated atherosclerotic lesion formation. Based on this data, current findings add to our understanding of the complexity of AS.
Collapse
Affiliation(s)
- Hongping Chen
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lihui Zhang
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Shaohua Mi
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Hua Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Chunxiao Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Wenjuan Jia
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lei Gong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Haibin Dong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Bowen Xu
- The 2nd Medical Colloge, Binzhou Medical University, Yantai, China
| | - Yanyan Jing
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Peipei Ge
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Zhigang Pei
- Department of Vascular Surgery, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, China
| | - Lin Zhong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Jun Yang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| |
Collapse
|
25
|
Yuan D, Chu J, Qian J, Lin H, Zhu G, Chen F, Liu X. New Concepts on the Pathophysiology of Acute Coronary Syndrome. Rev Cardiovasc Med 2023; 24:112. [PMID: 39076267 PMCID: PMC11273028 DOI: 10.31083/j.rcm2404112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 07/31/2024] Open
Abstract
Acute coronary syndrome (ACS) is the most severe form of ischemic heart disease. Although it is caused by atherosclerotic plaque thrombosis or nonatherosclerotic causes, its pathophysiological mechanism of ACS is not fully understood, and its concept is constantly updated and developed. At present, the main pathophysiological mechanisms include plaque rupture, plaque erosion, calcified nodules (CN) and non-atherosclerotic causes such as coronary vasospasm and myocardial bridging (MB). These mechanisms may overlap and coexist in some ACS patients. Therefore, the pathophysiological mechanism of ACS is complex, and is of great significance for the diagnosis and treatment of ACS. This review will discuss the pathophysiological mechanisms of ACS to provide new thoughts on the pathogenesis, diagnosis and treatment of ACS.
Collapse
Affiliation(s)
- Deqiang Yuan
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jiapeng Chu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jun Qian
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Hao Lin
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Guoqi Zhu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Fei Chen
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| |
Collapse
|
26
|
Hong SY, Jiang HC, Xu WC, Zeng HS, Wang SG, Qin BL. Bioinformatics analysis reveals the potential role of matrix metalloproteinases in immunity and urolithiasis. Front Immunol 2023; 14:1158379. [PMID: 37006258 PMCID: PMC10050583 DOI: 10.3389/fimmu.2023.1158379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundThe pathogenesis of urolithiasis remains unclear, making the development of medications for treatment and prevention stagnant. Randall’s plaques (RPs) begin as interstitial calcium phosphate crystal deposits, grow outward and breach the renal papillary surface, acting as attachment for CaOx stones. Since matrix metalloproteinases (MMPs) can degrade all components of extracellular matrix (ECM), they might participate in the breach of RPs. Besides, MMPs can modulate the immune response and inflammation, which were confirmed to be involved in urolithiasis. We aimed to investigate the role of MMPs in the development of RPs and stone formation.MethodsThe public dataset GSE73680 was mined to identify differentially expressed MMPs (DEMMPs) between normal tissues and RPs. WGCNA and three machine learning algorithms were performed to screen the hub DEMMPs. In vitro experiments were conducted for validation. Afterwards, RPs samples were classified into clusters based on the hub DEMMPs expression. Differentially expressed genes (DEGs) between clusters were identified and functional enrichment analysis and GSEA were applied to explore the biological role of DEGs. Moreover, the immune infiltration levels between clusters were evaluated by CIBERSORT and ssGSEA.ResultsFive DEMMPs, including MMP1, MMP3, MMP9, MMP10, and MMP12, were identified between normal tissues and RPs, and all of them were elevated in RPs. Based on WGCNA and three machine learning algorithms, all of five DEMMPs were regarded as hub DEMMPs. In vitro validation found the expression of hub DEMMPs also increased in renal tubular epithelial cells under lithogenic environment. RPs samples were divided into two clusters and cluster A exhibited higher expression of hub DEMMPs compared to cluster B. Functional enrichment analysis and GSEA found DEGs were enriched in immune-related functions and pathways. Moreover, increased infiltration of M1 macrophages and enhanced levels of inflammation were observed in cluster A by immune infiltration analysis.ConclusionWe assumed that MMPs might participate in RPs and stone formation through ECM degradation and macrophages-mediated immune response and inflammation. Our findings offer a novel perspective on the role of MMPs in immunity and urolithiasis for the first time, and provide potential biomarkers to develop targets for treatment and prevention.
Collapse
Affiliation(s)
- Sen-Yuan Hong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Cheng Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Chao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - He-Song Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Gang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shao-Gang Wang, ; Bao-Long Qin,
| | - Bao-Long Qin
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shao-Gang Wang, ; Bao-Long Qin,
| |
Collapse
|
27
|
Bonavita CM, White TM, Francis J, Farrell HE, Davis-Poynter NJ, Cardin RD. The Viral G-Protein-Coupled Receptor Homologs M33 and US28 Promote Cardiac Dysfunction during Murine Cytomegalovirus Infection. Viruses 2023; 15:711. [PMID: 36992420 PMCID: PMC10054303 DOI: 10.3390/v15030711] [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: 12/29/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen that infects the majority of the world population and causes lifelong latent infection. HCMV has been shown to exacerbate cardiovascular diseases, including myocarditis, vascular sclerosis, and transplant vasculopathy. Recently, we have shown that murine CMV (MCMV) recapitulates the cardiovascular dysfunction observed in patients with HCMV-induced myocarditis. To understand the viral mechanisms involved in CMV-induced heart dysfunction, we further characterized cardiac function in response to MCMV and examined virally encoded G-protein-coupled receptor homologs (vGPCRs) US28 and M33 as potential factors that promote infection in the heart. We hypothesized that the CMV-encoded vGPCRs could exacerbate cardiovascular damage and dysfunction. Three viruses were used to evaluate the role of vGPCRs in cardiac dysfunction: wild-type MCMV, a M33-deficient virus (∆M33), and a virus with the M33 open reading frame (ORF) replaced with US28, an HCMV vGPCR (i.e., US28+). Our in vivo studies revealed that M33 plays a role in promoting cardiac dysfunction by increasing viral load and heart rate during acute infection. During latency, ΔM33-infected mice demonstrated reduced calcification, altered cellular gene expression, and less cardiac hypertrophy compared with wild-type MCMV-infected mice. Ex vivo viral reactivation from hearts was less efficient in ΔM33-infected animals. HCMV protein US28 expression restored the ability of the M33-deficient virus to reactivate from the heart. US28+ MCMV infection caused damage to the heart comparable with wild-type MCMV infection, suggesting that the US28 protein is sufficient to complement the function of M33 in the heart. Altogether, these data suggest a role for vGPCRs in viral pathogenesis in the heart and thus suggest that vGPCRs promote long-term cardiac damage and dysfunction.
Collapse
Affiliation(s)
- Cassandra M. Bonavita
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Timothy M. White
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Joseph Francis
- Department of Comparative Biological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Helen E. Farrell
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | | | - Rhonda D. Cardin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
28
|
Arnhold J. Host-Derived Cytotoxic Agents in Chronic Inflammation and Disease Progression. Int J Mol Sci 2023; 24:ijms24033016. [PMID: 36769331 PMCID: PMC9918110 DOI: 10.3390/ijms24033016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
At inflammatory sites, cytotoxic agents are released and generated from invading immune cells and damaged tissue cells. The further fate of the inflammation highly depends on the presence of antagonizing principles that are able to inactivate these host-derived cytotoxic agents. As long as the affected tissues are well equipped with ready-to-use protective mechanisms, no damage by cytotoxic agents occurs and resolution of inflammation is initiated. However, long-lasting and severe immune responses can be associated with the decline, exhaustion, or inactivation of selected antagonizing principles. Hence, cytotoxic agents are only partially inactivated and contribute to damage of yet-unperturbed cells. Consequently, a chronic inflammatory process results. In this vicious circle of permanent cell destruction, not only novel cytotoxic elements but also novel alarmins and antigens are liberated from affected cells. In severe cases, very low protection leads to organ failure, sepsis, and septic shock. In this review, the major classes of host-derived cytotoxic agents (reactive species, oxidized heme proteins and free heme, transition metal ions, serine proteases, matrix metalloproteases, and pro-inflammatory peptides), their corresponding protective principles, and resulting implications on the pathogenesis of diseases are highlighted.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
29
|
Jin Z, Zhao H, Luo Y, Li X, Cui J, Yan J, Yang P. Identification of core genes associated with the anti-atherosclerotic effects of Salvianolic acid B and immune cell infiltration characteristics using bioinformatics analysis. BMC Complement Med Ther 2022; 22:190. [PMID: 35842645 PMCID: PMC9288713 DOI: 10.1186/s12906-022-03670-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/06/2022] [Indexed: 12/17/2022] Open
Abstract
Background Atherosclerosis (AS) is the greatest contributor to pathogenesis of atherosclerotic cardiovascular disease (ASCVD), which is associated with increased mortality and reduced quality of life. Early intervention to mitigate AS is key to prevention of ASCVD. Salvianolic acid B (Sal B) is mainly extracted from root and rhizome of Salvia Miltiorrhiza Bunge, and exerts anti-atherosclerotic effect. The purpose of this study was to screen for anti-AS targets of Sal B and to characterize immune cell infiltration in AS. Methods We identified targets of Sal B using SEA (http://sea.bkslab.org/) and SIB (https://www.sib.swiss/) databases. GSE28829 and GSE43292 datasets were obtained from Gene Expression Omnibus database. We identified differentially expressed genes (DEGs) and performed enrichment analysis. Weighted gene co-expression network analysis (WGCNA) was used to determine the most relevant module associated with atherosclerotic plaque stability. Intersecting candidate genes were evaluated by generating receiver operating characteristic (ROC) curves and molecular docking. Then, immune cell types were identified using CIBERSOFT and single-sample gene set enrichment analysis (ssGSEA), the relationship between candidate genes and immune cell infiltration was evaluated. Finally, a network-based approach to explore the candidate genes relationship with microRNAs (miRNAs) and Transcription factors (TFs). Results MMP9 and MMP12 were been selected as candidate genes from 64 Sal B-related genes, 81 DEGs and turquoise module with 220 genes. ROC curve results showed that MMP9 (AUC = 0.815, P<0.001) and MMP12 (AUC = 0.763, P<0.001) were positively associated with advanced atherosclerotic plaques. The results of immune infiltration showed that B cells naive, B cells memory, Plasma cells, T cells CD8, T cells CD4 memory resting, T cells CD4 memory activated, T cells regulatory (Tregs), T cells gamma delta, NK cells activated, Monocytes, and Macrophages M0 may be involved in development of AS, and the candidate genes MMP9 and MMP12 were associated with these immune cells to different degrees. What’ s more, miR-34a-5p and FOXC1, JUN maybe the most important miRNA and TFs. Conclusion The anti-AS effects of Sal B may be related to MMP9 and MMP12 and associated with immune cell infiltration, which is expected to be used in the early intervention of AS. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03670-6.
Collapse
|
30
|
Batty M, Bennett MR, Yu E. The Role of Oxidative Stress in Atherosclerosis. Cells 2022; 11:3843. [PMID: 36497101 PMCID: PMC9735601 DOI: 10.3390/cells11233843] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the vascular system and is the leading cause of cardiovascular diseases worldwide. Excessive generation of reactive oxygen species (ROS) leads to a state of oxidative stress which is a major risk factor for the development and progression of atherosclerosis. ROS are important for maintaining vascular health through their potent signalling properties. However, ROS also activate pro-atherogenic processes such as inflammation, endothelial dysfunction and altered lipid metabolism. As such, considerable efforts have been made to identify and characterise sources of oxidative stress in blood vessels. Major enzymatic sources of vascular ROS include NADPH oxidases, xanthine oxidase, nitric oxide synthases and mitochondrial electron transport chains. The production of ROS is balanced by ROS-scavenging antioxidant systems which may become dysfunctional in disease, contributing to oxidative stress. Changes in the expression and function of ROS sources and antioxidants have been observed in human atherosclerosis while in vitro and in vivo animal models have provided mechanistic insight into their functions. There is considerable interest in utilising antioxidant molecules to balance vascular oxidative stress, yet clinical trials are yet to demonstrate any atheroprotective effects of these molecules. Here we will review the contribution of ROS and oxidative stress to atherosclerosis and will discuss potential strategies to ameliorate these aspects of the disease.
Collapse
Affiliation(s)
| | | | - Emma Yu
- Section of Cardiorespiratory Medicine, University of Cambridge, Cambridge CB2 0BB, UK
| |
Collapse
|
31
|
Li W, Jin K, Luo J, Xu W, Wu Y, Zhou J, Wang Y, Xu R, Jiao L, Wang T, Yang G. NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front Cardiovasc Med 2022; 9:988266. [PMID: 36204587 PMCID: PMC9530249 DOI: 10.3389/fcvm.2022.988266] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a common cardiovascular disease with complex pathogenesis, in which multiple pathways and their interweaving regulatory mechanism remain unclear. The primary transcription factor NF-κB plays a critical role in AS via modulating the expression of a series of inflammatory mediators under various stimuli such as cytokines, microbial antigens, and intracellular stresses. Endoplasmic reticulum (ER) stress, caused by the disrupted synthesis and secretion of protein, links inflammation, metabolic signals, and other cellular processes via the unfolded protein response (UPR). Both NF-κB and ER stress share the intersection regarding their molecular regulation and function and are regarded as critical individual contributors to AS. In this review, we summarize the multiple interactions between NF-κB and ER stress activation, including the UPR, NLRP3 inflammasome, and reactive oxygen species (ROS) generation, which have been ignored in the pathogenesis of AS. Given the multiple links between NF-κB and ER stress, we speculate that the integrated network contributes to the understanding of molecular mechanisms of AS. This review aims to provide an insight into these interactions and their underlying roles in the progression of AS, highlighting potential pharmacological targets against the atherosclerotic inflammatory process.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liqun Jiao,
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Tao Wang,
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Tao Wang,
| |
Collapse
|
32
|
Keul P, Peters S, von Wnuck Lipinski K, Schröder NH, Nowak MK, Duse DA, Polzin A, Weske S, Gräler MH, Levkau B. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Aggravates Atherosclerosis and Induces Plaque Rupture in ApoE−/− Mice. Int J Mol Sci 2022; 23:ijms23179606. [PMID: 36077004 PMCID: PMC9455951 DOI: 10.3390/ijms23179606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Altered plasma sphingosine-1-phosphate (S1P) concentrations are associated with clinical manifestations of atherosclerosis. However, whether long-term elevation of endogenous S1P is pro- or anti-atherogenic remains unclear. Here, we addressed the impact of permanently high S1P levels on atherosclerosis in cholesterol-fed apolipoprotein E-deficient (ApoE−/−) mice over 12 weeks. This was achieved by pharmacological inhibition of the S1P-degrading enzyme S1P lyase with 4-deoxypyridoxine (DOP). DOP treatment dramatically accelerated atherosclerosis development, propagated predominantly unstable plaque phenotypes, and resulted in frequent plaque rupture with atherothrombosis. Macrophages from S1P lyase-inhibited or genetically deficient mice had a defect in cholesterol efflux to apolipoprotein A-I that was accompanied by profoundly downregulated cholesterol transporters ATP-binding cassette transporters ABCA1 and ABCG1. This was dependent on S1P signaling through S1PR3 and resulted in dramatically enhanced atherosclerosis in ApoE−/−/S1PR3−/− mice, where DOP treatment had no additional effect. Thus, high endogenous S1P levels promote atherosclerosis, compromise cholesterol efflux, and cause genuine plaque rupture.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Susann Peters
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H. Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K. Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Dragos A. Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Heinrich Heine University Medical Center Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care and Center for Molecular Biomedicine, University Hospital Jena, 07743 Jena, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-88-12611
| |
Collapse
|
33
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [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] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
34
|
Liu Q, Pan J, Bao L, Xu C, Qi Y, Jiang B, Wang D, Zhu X, Li X, Zhang H, Bai H, Yang Q, Ma J, Wiemer EAC, Ben J, Chen Q. Major Vault Protein Prevents Atherosclerotic Plaque Destabilization by Suppressing Macrophage ASK1-JNK Signaling. Arterioscler Thromb Vasc Biol 2022; 42:580-596. [PMID: 35387478 DOI: 10.1161/atvbaha.121.316662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Macrophages are implicated in atherosclerotic plaque instability by inflammation and degradation of extracellular matrix. However, the regulatory mechanisms driving these macrophage-associated processes are not well understood. Here, we aimed to identify the plaque destabilization-associated cytokines and signaling pathways in macrophages. METHODS The atherosclerotic models of myeloid-specific MVP (major vault protein) knockout mice and control mice were generated. Atherosclerotic instability, macrophage inflammatory signaling, and active cytokines released by macrophages were examined in vivo and in vitro by using cellular and molecular biological approaches. RESULTS MVP deficiency in myeloid cells exacerbated murine plaque instability by increasing production of both MMP (matrix metallopeptidase)-9 and proinflammatory cytokines in artery wall. Mechanistically, expression of MMP-9 was mediated via ASK1 (apoptosis signal-regulating kinase 1)-MKK-4 (mitogen-activated protein kinase kinase 4)-JNK (c-Jun N-terminal kinase) signaling in macrophages. MVP and its α-helical domain could bind with ASK1 and inhibit its dimerization and phosphorylation. A 62 amino acid peptide (MVP-[686-747]) in the α-helical domain of MVP showed a crucial role in preventing macrophage MMP-9 production and plaque instability. CONCLUSIONS MVP may act as an inhibitor for ASK1-JNK signaling-mediated MMP-9 production in macrophages and, thereby, attenuate unstable plaque formation. Our findings suggest that suppression of macrophage ASK1-JNK signaling may be a useful strategy antagonizing atherosclerotic diseases.
Collapse
Affiliation(s)
- Qingling Liu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junlu Pan
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Linrui Bao
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Chunxiang Xu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Yu Qi
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Dongdong Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junqing Ma
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands (E.A.C.W.)
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| |
Collapse
|
35
|
Park DY, Park JY, Lee D, Hwang I, Kim HS. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells 2022; 11:1079. [PMID: 35406643 PMCID: PMC8997512 DOI: 10.3390/cells11071079] [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: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
The oral cavity is the gateway for microorganisms into your body where they disseminate not only to the directly connected respiratory and digestive tracts but also to the many remote organs. Oral microbiota, travelling to the end of the intestine and circulating in our bodies through blood vessels, not only affect a gut microbiome profile but also lead to many systemic diseases. By gathering information accumulated from the era of focal infection theory to the age of revolution in microbiome research, we propose a pivotal role of "leaky gum", as an analogy of "leaky gut", to underscore the importance of the oral cavity in systemic health. The oral cavity has unique structures, the gingival sulcus (GS) and the junctional epithelium (JE) below the GS, which are rarely found anywhere else in our body. The JE is attached to the tooth enamel and cementum by hemidesmosome (HD), which is structurally weaker than desmosome and is, thus, vulnerable to microbial infiltration. In the GS, microbial biofilms can build up for life, unlike the biofilms on the skin and intestinal mucosa that fall off by the natural process. Thus, we emphasize that the GS and the JE are the weakest leaky point for microbes to invade the human body, making the leaky gum just as important as, or even more important than, the leaky gut.
Collapse
Affiliation(s)
- Do-Young Park
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Jin Young Park
- Department of Gastrointestinal Endoscopy, Apple Tree Healthcare Center, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| | - Inseong Hwang
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Hye-Sung Kim
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| |
Collapse
|
36
|
Gorisse L, Jaisson S, Piétrement C, Gillery P. Carbamylated Proteins in Renal Disease: Aggravating Factors or Just Biomarkers? Int J Mol Sci 2022; 23:574. [PMID: 35008998 PMCID: PMC8745352 DOI: 10.3390/ijms23010574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Carbamylation is a nonenzymatic post-translational modification resulting from the reaction between cyanate, a urea by-product, and proteins. In vivo and in vitro studies have demonstrated that carbamylation modifies protein structures and functions, triggering unfavourable molecular and cellular responses. An enhanced formation of carbamylation-derived products (CDPs) is observed in pathological contexts, especially during chronic kidney disease (CKD), because of increased blood urea. Significantly, studies have reported a positive correlation between serum CDPs and the evolutive state of renal failure. Further, serum concentrations of carbamylated proteins are characterized as strong predictors of mortality in end-stage renal disease patients. Over time, it is likely that these modified compounds become aggravating factors and promote long-term complications, including cardiovascular disorders and inflammation or immune system dysfunctions. These poor clinical outcomes have led researchers to consider strategies to prevent or slow down CDP formation. Even if growing evidence suggests the involvement of carbamylation in the pathophysiology of CKD, the real relevance of carbamylation is still unclear: is it a causal phenomenon, a metabolic consequence or just a biological feature? In this review, we discuss how carbamylation, a consequence of renal function decline, may become a causal phenomenon of kidney disease progression and how CDPs may be used as biomarkers.
Collapse
Affiliation(s)
- Laëtitia Gorisse
- MEDyC Unit CNRS UMR n° 7369, Faculty of Medicine, University of Reims Champagne-Ardenne, 51092 Reims, France; (L.G.); (S.J.); (C.P.)
| | - Stéphane Jaisson
- MEDyC Unit CNRS UMR n° 7369, Faculty of Medicine, University of Reims Champagne-Ardenne, 51092 Reims, France; (L.G.); (S.J.); (C.P.)
- Biochemistry Department, University Hospital of Reims, 51092 Reims, France
| | - Christine Piétrement
- MEDyC Unit CNRS UMR n° 7369, Faculty of Medicine, University of Reims Champagne-Ardenne, 51092 Reims, France; (L.G.); (S.J.); (C.P.)
- Pediatrics Department, University Hospital of Reims, 51092 Reims, France
| | - Philippe Gillery
- MEDyC Unit CNRS UMR n° 7369, Faculty of Medicine, University of Reims Champagne-Ardenne, 51092 Reims, France; (L.G.); (S.J.); (C.P.)
- Biochemistry Department, University Hospital of Reims, 51092 Reims, France
| |
Collapse
|
37
|
Huang S, Wu X, Zhang L, Wu J, He Y, Lai M, Xu J, Li Z. Assessment of Carotid Plaque Stability Using Contrast-Enhanced Ultrasound and Its Correlation With the Expression of CD147 and MMP-9 in the Plaque. Front Comput Neurosci 2021; 15:778946. [PMID: 34924986 PMCID: PMC8672308 DOI: 10.3389/fncom.2021.778946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
This study aims to investigate the correlation between the enhancement degree of contrast-enhanced ultrasound (CEUS) and the expression of CD147 and MMP-9 in carotid atherosclerotic plaques in patients with carotid endarterectomy and evaluate the diagnostic efficacy of CEUS using pathological results as the gold standard. Thirty-eight patients who underwent carotid endarterectomy (CEA) for carotid stenosis in the Department of Neurovascular Surgery of the Second People’s Hospital of Shenzhen from July 2019 to June 2020 were selected. Preoperatively, two-dimensional (2D) ultrasound scan was performed on all patients to assess the characteristics of the plaque and degree of stenosis, and CEUS was used to evaluate the surface morphology of the plaque and the distribution of neovascularization. Postoperatively, pathological sections and immunohistochemical analysis of CD147 and MMP-9 levels in the plaque were performed on the stripped plaque tissue, and the results were analyzed against the CEUS grading and pathological results. Among the 38 patients, pathological results showed that 10 and 28 were in the stable and vulnerable plaque groups, respectively. There were more smokers in the vulnerable plaque group than in the stable plaque group, with higher intraplaques CD147 and MMP-9. The difference in ultrasound plaque surface morphology grading and CEUS grading between the two groups was statistically significant. There was no significant difference in age, sex, incidence of complications such as hypertension, diabetes, and coronary heart disease between the two groups. CD147 was higher in the CEUS grade IV group than in the grades I (P = 0.040) and II (P = 0.010) groups. MMP-9 was higher in the CEUS grade IV group than in the grade II group (P = 0.017); MMP-9 was higher in the grade III group than in the grade II group (P = 0.015). Intraplaque contrast enhancement intensity was positively correlated with CD147 (r = 0.462, P = 0.003) and MMP-9 (r = 0.382, P = 0.018) levels. There was moderate consistency between the assessment of plaque vulnerability by 2D-ultrasound and by histopathological hematoxylin-eosin (HE) (kappa = 0.457, P > 0.05). 2D diagnosis of vulnerable plaque had a sensitivity of 85.7%, a specificity of 60.0%, a positive predictive value of 85.7%, a negative predictive value of 60.0%, and an accuracy of 78.0%. There was a strong consistency between the assessment of plaque vulnerability by CEUS and histopathological HE (kappa = 0.671, P < 0.01). CEUS had a sensitivity of 89.2%, a specificity of 80.0%, a positive predictive value of 92.6%, a negative predictive value of 72.7%, and an accuracy of 86.8% for the diagnosis of vulnerable plaques; CEUS is a reliable, non-invasive test that can show the distribution of neovascularization within vulnerable plaques, evaluate the vulnerability and risk of intraplaque hemorrhage, with a high consistency with pathological findings. The degree of intraplaque enhancement and the levels of CD147 and MMP-9 in the tissue were positively correlated.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinyin Wu
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Linlin Zhang
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianming Wu
- Department of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yi He
- Department of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Manlin Lai
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jiaqi Xu
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhenzhou Li
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
38
|
Abdelwahed YS, Nelles G, Frick C, Seppelt C, Meteva D, Stähli BE, Rai H, Riedel M, Skurk C, Rauch-Kröhnert U, Haghikia A, Sinning D, Dreger H, Knebel F, Trippel T, Krisper M, Klotsche J, Joner M, Landmesser U, Leistner DM. Coexistence of calcified- and lipid-containing plaque components and their association with incidental rupture points in acute coronary syndrome-causing culprit lesions: results from the prospective OPTICO-ACS study. Eur Heart J Cardiovasc Imaging 2021; 23:1598-1605. [PMID: 34904655 DOI: 10.1093/ehjci/jeab247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Rupture of the fibrous cap (RFC) represents the main pathophysiological mechanism causing acute coronary syndromes (ACS). Destabilization due to plaque biomechanics is considered to be importantly involved, exact mechanisms triggering plaque ruptures are, however, unknown. This study aims at characterizing the relation between plaque components and rupture points at ACS-causing culprit lesions in a large cohort of ACS-patients assessed by high-resolution intracoronary imaging. METHODS AND RESULTS Within the prospective, multicentric OPTICO-ACS study program, the ACS-causing culprit plaques of 282 consecutive patients were investigated following a standardized optical coherence tomography (OCT) imaging protocol. Each pullback was assessed on a frame-by-frame basis for the presence of lipid components (LC), calcium components (CC), and coexistence of both LC and CC (LCC) by two independent OCT-core labs. Of the 282 ACS-patients, 204 patients (72.3%) presented with ACS caused by culprit lesions with rupture of the fibrous cap (RFC-ACS) and 27.7% patients had ACS caused by culprit lesions with intact fibrous cap (IFC-ACS). When comparing RFC-ACS to IFC-ACS, a preferential occurrence of all three plaque components (LC, CC, and LCC) in RFC-ACS became apparent (P < 0.001). Within ruptured culprit lesions, the zone straight at the rupture point [extended rupture zone (RZ)] was characterized by similar (24.7% vs. 24.0%; P = ns) calcium content when compared with the proximal and distal border of the culprit lesion [border zone (BZ)]. The RZ displayed a significantly higher amount of both, LC (100% vs. 69.8%; P < 0.001) and LCC (22.7% vs. 6.8%; P < 0.001), when compared with the BZ. The relative component increase towards the RZ was particularly evident for LCC (+233.8%), while LC showed only a modest increase (+43.3%). CONCLUSIONS Calcified- and lipid-containing components characterize ruptured fibrous cap ACS-causing culprit lesions. Their coexistence is accelerated directly at the ruptured point, suggesting a pathophysiological contribution in the development of RFC-ACS.
Collapse
Affiliation(s)
- Youssef S Abdelwahed
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Gregor Nelles
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Clara Frick
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany
| | - Claudio Seppelt
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Denitsa Meteva
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Barbara E Stähli
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Himanshu Rai
- Department of Cardiology and ISAR Research Center, German Heart Center, 80636 Munich, Germany
| | - Matthias Riedel
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - David Sinning
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany
| | - Henryk Dreger
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Campus Mitte (CCM), 10117 Berlin, Germany
| | - Fabian Knebel
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Campus Mitte (CCM), 10117 Berlin, Germany
| | - Tobias Trippel
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Campus Virchow Clinic (CVK), 13353 Berlin, Germany
| | - Maximillian Krisper
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Campus Virchow Clinic (CVK), 13353 Berlin, Germany
| | - Jens Klotsche
- German Rheumatism Research Center Berlin, and Institute for Social Medicine, Epidemiology und Heath Economy, Charité University Medicine Berlin, Campus Charité Mitte, 10117 Berlin, Germany
| | - Michael Joner
- Department of Cardiology, Charité University Medicine Berlin, Campus Mitte (CCM), 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Munch, 80636, Munich, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2 D-10178 Berlin, Germany
| | - David M Leistner
- Department of Cardiology, Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203 Berlin, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2 D-10178 Berlin, Germany
| |
Collapse
|
39
|
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
|
40
|
Tubular lysosomes harbor active ion gradients and poise macrophages for phagocytosis. Proc Natl Acad Sci U S A 2021; 118:2113174118. [PMID: 34607961 PMCID: PMC8522270 DOI: 10.1073/pnas.2113174118] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Lysosomes are organelles that also act as cell-signaling hubs. They regulate functions ranging from antigen presentation to autophagy. Spherical lysosomes can spontaneously elongate into tubules in starving or inflamed immune cells. We describe a DNA-based reagent, denoted Tudor, that tubulates lysosomes in macrophages without triggering either an immune response or autophagy. Chemical imaging revealed that tubular lysosomes differ from vesicular ones in terms of their pH, calcium, and proteolytic activity. Tudor revealed a role for tubular lysosomes in that they enhance MMP9 secretion and phagocytosis in resting macrophages. The ability to tubulate lysosomes in resting immune cells without starving or inflaming them may help reveal new insights into how tubular lysosomes function. Lysosomes adopt dynamic, tubular states that regulate antigen presentation, phagosome resolution, and autophagy. Tubular lysosomes are studied either by inducing autophagy or by activating immune cells, both of which lead to cell states where lysosomal gene expression differs from the resting state. Therefore, it has been challenging to pinpoint the biochemical properties lysosomes acquire upon tubulation that could drive their functionality. Here we describe a DNA-based assembly that tubulates lysosomes in macrophages without activating them. Proteolytic activity maps at single-lysosome resolution revealed that tubular lysosomes were less degradative and showed proximal to distal luminal pH and Ca2+ gradients. Such gradients had been predicted but never previously observed. We identify a role for tubular lysosomes in promoting phagocytosis and activating MMP9. The ability to tubulate lysosomes without starving or activating immune cells may help reveal new roles for tubular lysosomes.
Collapse
|
41
|
Peng J, Le CY, Xia B, Wang JW, Liu JJ, Li Z, Zhang QJ, Zhang Q, Wang J, Wan CW. Research on the correlation between activating transcription factor 3 expression in the human coronary artery and atherosclerotic plaque stability. BMC Cardiovasc Disord 2021; 21:356. [PMID: 34320932 PMCID: PMC8317287 DOI: 10.1186/s12872-021-02161-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/06/2021] [Indexed: 12/26/2022] Open
Abstract
Background Activating transcription factor 3 (ATF3) is an early response gene that is activated in response to atherosclerotic stimulation and may be an important factor in inhibiting the progression of atherosclerosis. In this study, we directly measured the expression of ATF3 and inflammatory factors in human coronary atherosclerotic plaques to examine the relationship between ATF3 expression, inflammation and structural stability in human coronary atherosclerotic plaques. Methods A total of 68 coronary artery specimens were collected from the autopsy group, including 36 cases of sudden death from coronary heart disease (SCD group) and 32 cases of acute death caused by mechanical injury with coronary atherosclerosis (CHD group). Twenty-two patients who had no coronary heart disease were collected as the control group (Con group). The histological structure of the coronary artery was observed under a light microscope after routine HE staining, and the intimal and lesion thicknesses, thickness of the fibrous cap, thickness of necrosis core, degree of lumen stenosis were assessed by image analysis software. Western blotting and immunohistochemistry were used to measure the expression and distribution of ATF3, inflammatory factors (CD45, IL-1β, TNF-α) and matrix metalloproteinase-9 (MMP-9) and vascular cell adhesion molecule 1 (VCAM1) in the coronary artery. The Pearson correlation coefficient was used to analyse the correlation between ATF3 protein expression and inflammatory factors and between ATF3 protein expression and structure-related indexes in the lesion group. Results Compared with those in the control group, the intima and necrotic core in the coronary artery were thickened, the fibrous cap became thin and the degree of vascular stenosis was increased in the lesion group, while the intima and necrotic core became thicker and the fibrous cap became thinner in the SCD group than in the CHD group (P < 0.05). There was no or low expression of ATF3, inflammatory factors, VCAM1 and MMP-9 in the control group, and the expression of inflammatory factors, VCAM1 and MMP-9 in the SCD group was higher than that in CHD group, while the expression of ATF3 in the SCD group was significantly lower than that in CHD group (P < 0.05). In the lesion group, the expression of ATF3 was negatively correlated with intimal and necrotic focus thickness, positively correlated with fibrous cap thickness (P < 0.01), and negatively correlated with inflammatory factors, VCAM1 and MMP-9 (P < 0.01). Conclusions The expression of ATF3 may be related to the progression and stability of atherosclerotic plaques, and may affect the structural stability of atherosclerotic plaques by regulating the inflammatory response, thus participating in the regulation of atherosclerotic progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02161-9.
Collapse
Affiliation(s)
- J Peng
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - C Y Le
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - B Xia
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - J W Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - J J Liu
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Z Li
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Q J Zhang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Q Zhang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - J Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| | - C W Wan
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| |
Collapse
|
42
|
Tillie RJHA, Theelen TL, van Kuijk K, Temmerman L, de Bruijn J, Gijbels M, Betsholtz C, Biessen EAL, Sluimer JC. A Switch from Cell-Associated to Soluble PDGF-B Protects against Atherosclerosis, despite Driving Extramedullary Hematopoiesis. Cells 2021; 10:1746. [PMID: 34359916 PMCID: PMC8308020 DOI: 10.3390/cells10071746] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/30/2022] Open
Abstract
Platelet-derived growth factor B (PDGF-B) is a mitogenic, migratory and survival factor. Cell-associated PDGF-B recruits stabilizing pericytes towards blood vessels through retention in extracellular matrix. We hypothesized that the genetic ablation of cell-associated PDGF-B by retention motif deletion would reduce the local availability of PDGF-B, resulting in microvascular pericyte loss, microvascular permeability and exacerbated atherosclerosis. Therefore, Ldlr-/-Pdgfbret/ret mice were fed a high cholesterol diet. Although plaque size was increased in the aortic root of Pdgfbret/ret mice, microvessel density and intraplaque hemorrhage were unexpectedly unaffected. Plaque macrophage content was reduced, which is likely attributable to increased apoptosis, as judged by increased TUNEL+ cells in Pdgfbret/ret plaques (2.1-fold) and increased Pdgfbret/ret macrophage apoptosis upon 7-ketocholesterol or oxidized LDL incubation in vitro. Moreover, Pdgfbret/ret plaque collagen content increased independent of mesenchymal cell density. The decreased macrophage matrix metalloproteinase activity could partly explain Pdgfbret/ret collagen content. In addition to the beneficial vascular effects, we observed reduced body weight gain related to smaller fat deposition in Pdgfbret/ret liver and adipose tissue. While dampening plaque inflammation, Pdgfbret/ret paradoxically induced systemic leukocytosis. The increased incorporation of 5-ethynyl-2'-deoxyuridine indicated increased extramedullary hematopoiesis and the increased proliferation of circulating leukocytes. We concluded that Pdgfbret/ret confers vascular and metabolic effects, which appeared to be protective against diet-induced cardiovascular burden. These effects were unrelated to arterial mesenchymal cell content or adventitial microvessel density and leakage. In contrast, the deletion drives splenic hematopoiesis and subsequent leukocytosis in hypercholesterolemia.
Collapse
Affiliation(s)
- Renée J. H. A. Tillie
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
| | - Thomas L. Theelen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
| | - Kim van Kuijk
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
| | - Jenny de Bruijn
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
| | - Marion Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden;
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
| | - Judith C. Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (R.J.H.A.T.); (T.L.T.); (K.v.K.); (L.T.); (J.d.B.); (M.G.); (E.A.L.B.)
- BHF Centre for Cardiovascular Sciences (CVS), University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
43
|
Garoffolo G, Pesce M. Vascular dysfunction and pathology: focus on mechanical forces. VASCULAR BIOLOGY 2021; 3:R69-R75. [PMID: 34291191 PMCID: PMC8284946 DOI: 10.1530/vb-21-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 11/08/2022]
Abstract
The role of mechanical forces is emerging as a new player in the pathophysiologic programming of the cardiovascular system. The ability of the cells to 'sense' mechanical forces does not relate only to perception of movement or flow, as intended traditionally, but also to the biophysical properties of the extracellular matrix, the geometry of the tissues, and the force distribution inside them. This is also supported by the finding that cells can actively translate mechanical cues into discrete gene expression and epigenetic programming. In the present review, we will contextualize these new concepts in the vascular pathologic programming.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, Milan, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, Milan, Italy
| |
Collapse
|
44
|
Liao B, Geng L, Zhang F, Shu L, Wei L, Yeung PKK, Lam KSL, Chung SK, Chang J, Vanhoutte PM, Xu A, Wang K, Hoo RLC. Adipocyte fatty acid-binding protein exacerbates cerebral ischaemia injury by disrupting the blood-brain barrier. Eur Heart J 2021; 41:3169-3180. [PMID: 32350521 PMCID: PMC7556749 DOI: 10.1093/eurheartj/ehaa207] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/12/2019] [Accepted: 03/11/2020] [Indexed: 01/06/2023] Open
Abstract
Aims Adipocyte fatty acid-binding protein (A-FABP) is an adipokine implicating in various metabolic diseases. Elevated circulating levels of A-FABP correlate positively with poor prognosis in ischaemic stroke (IS) patients. No information is available concerning the role of A-FABP in the pathogenesis of IS. Experiments were designed to determine whether or not A-FABP mediates blood–brain barrier (BBB) disruption, and if so, to explore the molecular mechanisms underlying this deleterious effects. Methods and results Circulating A-FABP and its cerebral expression were increased in mice after middle cerebral artery occlusion. Genetic deletion and pharmacological inhibition of A-FABP alleviated cerebral ischaemia injury with reduced infarction volume, cerebral oedema, neurological deficits, and neuronal apoptosis; BBB disruption was attenuated and accompanied by reduced degradation of tight junction proteins and induction of matrix metalloproteinases-9 (MMP-9). In patients with acute IS, elevated circulating A-FABP levels positively correlated with those of MMP-9 and cerebral infarct volume. Mechanistically, ischaemia-induced elevation of A-FABP selectively in peripheral blood monocyte-derived macrophages and cerebral resident microglia promoted MMP-9 transactivation by potentiating JNK/c-Jun signalling, enhancing degradation of tight junction proteins and BBB leakage. The detrimental effects of A-FABP were prevented by pharmacological inhibition of MMP-9. Conclusion A-FABP is a key mediator of cerebral ischaemia injury promoting MMP-9-mediated BBB disruption. Inhibition of A-FABP is a potential strategy to improve IS outcome. ![]()
Collapse
Affiliation(s)
- Boya Liao
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Guangzhou 510120, China
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ling Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Anhui Province Key Laboratory on Cognition and Neuropsychiatric Disorders, No. 218, Jixi Road, Hefei, Anhui Province 230022, China
| | - Patrick K K Yeung
- Department of Anatomy, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sookja K Chung
- Department of Anatomy, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Macau University of Science and Technology, Avenida Wai Long, Taipa 999078, Macau, China
| | - Junlei Chang
- Centre for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Blvd, Xili Nanshan, Shenzhen 518055, China
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Anhui Province Key Laboratory on Cognition and Neuropsychiatric Disorders, No. 218, Jixi Road, Hefei, Anhui Province 230022, China
| | - Ruby L C Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
45
|
Lu L, Huang J, Xue X, Wang T, Huang Z, Li J. Berberine Regulated miR150-5p to Inhibit P2X7 Receptor, EMMPRIN and MMP-9 Expression in oxLDL Induced Macrophages. Front Pharmacol 2021; 12:639558. [PMID: 33959010 PMCID: PMC8093865 DOI: 10.3389/fphar.2021.639558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/25/2021] [Indexed: 02/05/2023] Open
Abstract
Elevated extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 (MMP-9) in oxidized low density lipoprotein (oxLDL)-induced macrophages leads to the progression of vulnerable plaques by degradation of the extracellular matrix. Our previous report showed that berberine regulates the expression of both EMMPRIN and MMP-9. In addition, P2X7 receptor (P2X7R) upregulation plays a crucial role in the development of atherosclerosis. However, it is unclear whether berberine regulated P2X7R level to inhibit both EMMPRIN and MMP-9 expession in macrophages. In the present study, we investigated the impact of berberine on P2X7R expression and the regulation of P2X7R in the expression of EMMPRIN and MMP-9 in oxLDL-induced macrophages. We found that P2X7R expression was increased, miR150-5p was reduced in oxLDL-induced macrophages, relatively. And A-438079 (a P2X7R inhibitor) or miR150-5p mimic treatment greatly reversed the upregulation of EMMPRIN and MMP-9 expression. Moreover, A-438079 significantly reduced oxLDL-induced AMP-activated protein kinase-α (AMPK-α) phosphorylation and reversed the activation of mitogen-activated protein kinase (MAPK), which in turn decreased the expression of EMMPRIN and MMP-9. These findings illustrate that P2X7R suppresses EMMPRIN and MMP-9 expression by inhibiting the AMPK-α/MAPK pathway in oxLDL-induced macrophages. Accordingly, exposure to berberine markedly upregulated miR150-5p, decreased P2X7R expression and downregulated MMP-9 and EMMPRIN levels in oxLDL-induced macrophages, resulting in AMPK-α/MAPK (JNK, p38, and ERK) inactivation. Overall, these results indicate that berberine increased miR150-5p level, subsequently inhibits P2X7R-mediated EMMPRIN and MMP-9 expression by suppressing AMPK-α and MAPK signaling in oxLDL-induced macrophages.
Collapse
Affiliation(s)
- Lin Lu
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, China
| | - Jianjian Huang
- Department of Anesthesiology, Wenzhou Medical University, Wenzhou, China
| | - Xia Xue
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, China
| | - Ting Wang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, China
| | - Zhouqing Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Fisetin Attenuates Lipopolysaccharide-Induced Inflammatory Responses in Macrophage. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5570885. [PMID: 33954178 PMCID: PMC8057890 DOI: 10.1155/2021/5570885] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 12/30/2022]
Abstract
Several studies have reported the efficacy and safety of polyphenols in human health; however, the verification of their efficacy remains insufficient. The aim of this study was to examine whether fisetin, one of flavonoids prevalently present in fruits and vegetables, could suppress lipopolysaccharide- (LPS-) induced inflammatory responses in macrophages. LPS increased proinflammatory mRNA abundance (MCP 1, IL-1β, and iNOS) but were suppressed by fisetin. The increment of nitric oxide by LPS, an oxidative stress factor, was attenuated by fisetin. In addition, LPS-enhanced phosphorylation of mitogen-activated protein kinase (ERK and JNK) was reduced. Finally, fisetin attenuated the expression or activity of uPA, uPAR, MMP-2, and MMP-9, which are known as associated factors of macrophage recruitment or infiltration. In conclusion, fisetin is a promising therapeutic agent for macrophage-related inflammation diseases, like sepsis and atherosclerosis.
Collapse
|
47
|
Sun R, Qiao Y, Yan G, Wang D, Zuo W, Ji Z, Zhang X, Yao Y, Ma G, Tang C. Association between serum adipsin and plaque vulnerability determined by optical coherence tomography in patients with coronary artery disease. J Thorac Dis 2021; 13:2414-2425. [PMID: 34012589 PMCID: PMC8107545 DOI: 10.21037/jtd-21-259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Early identification of vulnerable plaques is important for patients with coronary artery disease (CAD) to reduce acute coronary events and improve their prognosis. We sought to examine the relationship between adipsin, an adipokine secreted from adipocytes, and plaque vulnerability in CAD patients. Methods A total of 103 plaques from 99 consecutive patients who underwent coronary angiography were assessed by optical coherence tomography. The serum level of adipsin was measured using enzyme-linked immunosorbent assay (ELISA). The accuracy of adipsin for detecting thin-cap fibroatheroma (TCFA) was determined by the area under the receiver operating characteristic curve (AUC). Results Of the 99 patients, 49 were classified into the low adipsin group and 50 into the high adipsin group according to the median level of serum adipsin (2.43 µg/mL). The plaques from the high adipsin group exhibited a greater lipid index (2,700.0 vs. 1,975.9° × mm, P=0.015) and an increased proportion of TCFAs (41.2% vs. 21.2%, P=0.028) compared with the low adipsin group. Serum adipsin was found to be negatively correlated with fibrous cap thickness (ρ=−0.322, P=0.002), while it was positively correlated with average lipid arc (ρ=0.253, P=0.015), maximum lipid arc (ρ=0.211, P=0.044), lipid core length (ρ=0.241, P=0.021), lipid index (ρ=0.335, P=0.001), and vulnerability score (ρ=0.254, P=0.014). Furthermore, adipsin had a significant association with TCFAs (OR: 1.290, 95% CI: 1.048–1.589, P=0.016) in the multivariate analysis, while having a moderate diagnostic accuracy for TCFAs (AUC: 0.710, 95% CI: 0.602–0.817, P<0.001). Conclusions Our findings suggest that serum adipsin is significantly and positively correlated with the incidence of TCFAs. The application of adipsin as a biomarker may offer improvement in the diagnosis of vulnerable plaques and clinical benefits for CAD patients.
Collapse
Affiliation(s)
- Renhua Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Department of Cardiology, Yancheng No.1 People's Hospital, Yancheng, China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaoguo Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
48
|
Close Association of Matrix Metalloproteinase-9 Levels With the Presence of Thin-Cap Fibroatheroma in Acute Coronary Syndrome Patients: Assessment by Optical Coherence Tomography and Intravascular Ultrasonography. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 32:5-10. [PMID: 33485858 DOI: 10.1016/j.carrev.2020.12.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Thin-cap fibroatheroma (TCFA) has been suggested as a precursor lesion of coronary plaque rupture. As elevated plasma matrix metalloproteinase-9 (MMP-9) levels have been documented in patients with acute coronary syndrome (ACS), we sought to determine whether the presence of TCFA is linked to MMP-9 levels in these patients. METHODS We evaluated 51 ACS patients with de novo culprit lesions who were examined via optical coherence tomography and intravascular ultrasound. Blood samples were obtained from the peripheral vein (PV) and the ostium and culprit lesion of the infarct-related coronary artery (CA) in the acute phase of ACS and from the PV in the chronic phase (8 months after ACS). RESULTS The plasma MMP-9 level in the acute phase was significantly higher than that in the chronic phase. Plasma MMP-9 levels at the culprit lesion of the infarct-related CA were significantly higher than, but positively correlated with those in the PV (10.9 (5.9-16.1) ng/mL and 8.9 (5.6-13.0) ng/mL, p < 0.0001, respectively; Spearman ρ = 0.84, p < 0.0001). Significantly higher PV plasma MMP-9 levels were observed in patients with TCFA than in patients without TCFA (12.1 (7.0-13.5) and 5.7 (4.0-8.2) ng/ml, p<0.0001, respectively). Further, plasma MMP-9 levels in the PV were positively correlated with the remodeling index (Spearman ρ = 0.29, p = 0.039) and negatively correlated with fibrous cap thickness (Spearman ρ = -0.42, p = 0.0021). Receiver operating characteristic curve analysis showed that the plasma MMP-9 levels in the PV could predict the presence of TCFA at a cut-off value of 9.9 ng/mL. CONCLUSIONS Plasma MMP-9 levels were closely associated with MMP-9 levels in the CA and were further linked with TCFA in patients with ACS.
Collapse
|
49
|
Silva H, Francisco R, Saraiva A, Francisco S, Carrascosa C, Raposo A. The Cardiovascular Therapeutic Potential of Propolis-A Comprehensive Review. BIOLOGY 2021; 10:biology10010027. [PMID: 33406745 PMCID: PMC7823408 DOI: 10.3390/biology10010027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
Simple Summary Propolis, also described as bee glue, is a natural component made up of a resinous mixture of honeybee compounds from multiple botanical sources. The literature has demonstrated a variety of medicinal properties attributed to propolis due to its chemical complexity. However, the positive effects of propolis on cardiovascular health have gained little coverage. Therefore, we aimed to provide an accurate and up-to-date review of the main cardiovascular health benefits of propolis. In particular, we intend to establish the key varieties of propolis and pharmacological compounds with the therapeutic effects that are most encouraging, as well as the physiological processes by which those advantages are accomplished. The Brazilian green and red varieties reveal the greatest number of beneficial activities among the varieties of propolis studied. While much of the cardiovascular beneficial effects appear to derive from the cumulative actions of several compounds working via multiple signaling mechanisms, some individual compounds that may enhance the existing therapeutic arsenal have also shown significant results. It is also worth exploring the prospect of using propolis as food supplements. Abstract Owing to its chemical richness, propolis has a myriad of therapeutic properties. To the authors’ knowledge, this is the first comprehensive review paper on propolis to focus exclusively on its major effects for cardiovascular health. The propolis compound varieties with the most promising therapeutic benefits and their respective physiological mechanisms will be discussed. Propolis displays an anti-atherosclerotic activity, attained through modulation of the plasma lipid profile and through stabilization of the fatty plaque by inhibiting macrophage apoptosis, vascular smooth muscle proliferation and metalloproteinase activity. The antihypertensive effects of propolis probably arise through the combination of several mechanisms, including the suppression of catecholamine synthesis, stimulation of endothelium-dependent vasorelaxation and vascular anti-inflammatory activity. The anti-hemostatic activity of propolis is attributed to the inhibition of platelet plug formation and antifibrinolytic activity. By inhibiting the secretion of proangiogenic factors, propolis suppresses endothelial cell migration and tubulogenesis, exerting antiangiogenic activity. The antioxidant and anti-inflammatory activities are responsible for protection against vascular endothelial and cardiomyocyte dysfunction, mostly by the prevention of oxidative stress. Among the reviewed propolis varieties, the Brazilian green and red varieties show the largest number of beneficial activities. Further research, especially preclinical, should be conducted to assess the cardiovascular benefits of the given varieties with different compositions.
Collapse
Affiliation(s)
- Henrique Silva
- Informetrics Research Group, Ton Duc Thang University, Ho Chi Minh City 758307, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 758307, Vietnam
- Correspondence: (H.S.); (A.R.)
| | - Rafaela Francisco
- Pharmacological Sciences Department, Faculty of Pharmacy, Universidade de Lisboa, Av Prof Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Ariana Saraiva
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain; (A.S.); (C.C.)
| | - Simone Francisco
- Faculty of Medicine, Nutrition Lab—Universidade de Lisboa, 1649-028 Lisboa, Portugal;
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain; (A.S.); (C.C.)
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
- Correspondence: (H.S.); (A.R.)
| |
Collapse
|
50
|
Wang Z, Guo X, Zhang Q, Du G, Zeng Z, Zheng C, Wei Y. Elimination of Ox-LDL through the liver inhibits advanced atherosclerotic plaque progression. Int J Med Sci 2021; 18:3652-3664. [PMID: 34790037 PMCID: PMC8579296 DOI: 10.7150/ijms.63065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/31/2021] [Indexed: 01/02/2023] Open
Abstract
Aim: In the late stage of atherosclerosis, the endothelial barrier of plaque is destroyed. The rapid deposition of oxidized lipids in the circulation leads to migration of numerous smooth muscle cells and macrophages, as well as foaming necrosis. The plaque progresses rapidly, and vulnerable plaques can easily induce adverse cardiovascular events. Here, we take the principle of gene editing to transfer the liver to express the LOX-1 receptor which is more sensitive to Ox-LDL by using AAV8 containing a liver-specific promoter. In this way, we want to explore whether the progress of advanced atherosclerosis and the stability of advanced plaque can be improved when the liver continues to clear Ox-LDL from the circulation. Methods and Results: In order to explore the effect of the physiological and continuous elimination of Ox-LDL through the liver on advanced atherosclerosis, we chose ApoE-/- mice in high-fat diet for 20 weeks. After 16 weeks of high-fat diet, the baseline group was sacrificed and the specimens were collected. The virus group and the control group were injected with the same amount of virus dilution and normal saline through the tail vein, and continued to feed until 20 weeks of high-fat diet, and then sacrificed to collect specimens. The results showed that LOX-1 was ectopically and functionally expressed in the liver as an Ox-LDL receptor, reducing the content of it in circulation. Compared with the control group, the degree of plaque progression in the virus group was significantly reduced, similar to the baseline group, the plaque necrosis core decreased, and the collagen fiber content increased. In addition, there are more contractile smooth muscle cells in the plaques of the virus group instead of synthetic ones, and the content of macrophages was also reduced. These data suggested that the virus group mice have greatly increased advanced plaque stability compared with the control group mice. Conclusions: Due to the destruction of endothelial barrier in advanced plaques, rapid deposition of Ox-LDL can result in fast plaque progression, increased necrotic cores, and decreased stability. Our research shows that the use of AAV8 through gene editing allows the liver to express LOX-1 receptors that are more sensitive to Ox-LDL, so that it can continue to bind Ox-LDL in the circulation and exploit the liver's strong lipid metabolism ability to physiologically clear Ox-LDL, which can inhibit the rapid progress of advanced plaque and increase the stability of plaque.
Collapse
Affiliation(s)
- Zhiwen Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaohui Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuanglin Zeng
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|