51
|
Sallam M, Benotmane MA, Baatout S, Guns PJ, Aerts A. Radiation-induced cardiovascular disease: an overlooked role for DNA methylation? Epigenetics 2022; 17:59-80. [PMID: 33522387 PMCID: PMC8812767 DOI: 10.1080/15592294.2021.1873628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy in cancer treatment involves the use of ionizing radiation for cancer cell killing. Although radiotherapy has shown significant improvements on cancer recurrence and mortality, several radiation-induced adverse effects have been documented. Of these adverse effects, radiation-induced cardiovascular disease (CVD) is particularly prominent among patients receiving mediastinal radiotherapy, such as breast cancer and Hodgkin's lymphoma patients. A number of mechanisms of radiation-induced CVD pathogenesis have been proposed such as endothelial inflammatory activation, premature endothelial senescence, increased ROS and mitochondrial dysfunction. However, current research seems to point to a so-far unexamined and potentially novel involvement of epigenetics in radiation-induced CVD pathogenesis. Firstly, epigenetic mechanisms have been implicated in CVD pathophysiology. In addition, several studies have shown that ionizing radiation can cause epigenetic modifications, especially DNA methylation alterations. As a result, this review aims to provide a summary of the current literature linking DNA methylation to radiation-induced CVD and thereby explore DNA methylation as a possible contributor to radiation-induced CVD pathogenesis.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
52
|
Kang CM, Li WK, Yu KW, Li XH, Huang RY, Ke PF, Jin X, Cao SW, Yuan YS, Wang H, Yan J, Chen WY, Huang XZ, Zhao JJ. Long non‑coding RNA AL355711 promotes smooth muscle cell migration through the ABCG1/MMP3 pathway. Int J Mol Med 2021; 48:207. [PMID: 34608503 PMCID: PMC8510679 DOI: 10.3892/ijmm.2021.5040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis and related cardiovascular diseases pose severe threats to human health worldwide. There is evidence to suggest that at least 50% of foam cells in atheromas are derived from vascular smooth muscle cells (VSMCs); the first step in this process involves migration to human atherosclerotic lesions. Long non‑coding RNAs (lncRNAs) have been found to play significant roles in diverse biological processes. The present study aimed to investigate the role of lncRNAs in VSMCs. The expression of lncRNAs or mRNAs was detected using reverse transcription‑quantitative polymerase chain reaction. The Gene Expression Omnibus datasets in the NCBI portal were searched using the key words 'Atherosclerosis AND tissue AND Homo sapiens' and the GSE12288 dataset. Gene expression in circulating leukocytes was measured to identify patients with coronary artery disease (CAD) or controls, and used to analyze the correlation coefficient and expression profiles. The protein level of ATP‑binding cassette sub‑family G member 1 (ABCG1) and matrix metalloproteinase (MMP)3 was determined using immunohistochemistry and western blot analysis. The analysis of mouse aortic roots was performed using Masson's and Oil Red O staining. The expression of lncRNA AL355711, ABCG1 and MMP3 was found to be higher in human atherosclerotic plaques or in patients with atherosclerotic CAD. The correlation analysis revealed that ABCG1 may be involved in the regulation between lncRNA AL355711 and MMP3 in atherosclerotic CAD. The knockdown of lncRNA AL355711 inhibited ABCG1 transcription and smooth muscle cell migration. In addition, lncRNA AL355711 was found to regulate MMP3 expression through the ABCG1 pathway. The expression of ABCG1 and MMP3 was found to be high in an animal model of atherosclerosis. The results indicated that lncRNA AL355711 promoted VSMC migration and atherosclerosis partly via the ABCG1/MMP3 pathway. On the whole, the present study demonstrates that the inhibition of lncRNA AL355711 may serve as a novel therapeutic target for atherosclerosis. lncRNA AL355711 in circulating leukocytes may be a novel biomarker for atherosclerotic CAD.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cell Movement/genetics
- Cells, Cultured
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Male
- Matrix Metalloproteinase 3/genetics
- Matrix Metalloproteinase 3/metabolism
- Metabolic Networks and Pathways/genetics
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/physiology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- RNA, Long Noncoding/genetics
- Mice
Collapse
Affiliation(s)
- Chun-Min Kang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Wei-Kang Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Ke-Wei Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xue-Heng Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Rui-Ying Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Pei-Feng Ke
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xing Jin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Shun-Wang Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Ying-Shi Yuan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Heng Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Jun Yan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Wei-Ye Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xian-Zhang Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Jing-Jing Zhao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
53
|
Lin Y, Huang H, Yu Y, Zhu F, Xiao W, Yang Z, Shao L, Shen Z. Long non-coding RNA RP11-465L10.10 promotes vascular smooth muscle cells phenotype switching and MMP9 expression via the NF-κB pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1776. [PMID: 35071470 PMCID: PMC8756256 DOI: 10.21037/atm-21-6402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/17/2021] [Indexed: 01/02/2023]
Abstract
Background Thoracic aortic aneurysm/dissection (TAA/D) are complicated vascular disorders with rapid development and high mortality. Vascular smooth muscle cells (VSMCs) phenotype switching plays an important role in the pathological process of TAA/D. Previous studies have indicated a potential correlation between long non-coding RNA (lncRNA) RP11-465L10.10 and matrix metallopeptidase 9 (MMP9) involved in the development of TAA/D. This study aims to investigate the role of lncRNA RP11-465L10.10 in VSMCs phenotype switching and the molecular mechanism in regulating MMP9 expression. Methods The expression of RP11-465L10.10 in vascular tissues and in VMSCs was detected by RT-qPCR. To investigate the role of RP11-465L10.10 on VSMCs phenotype switching, an RP11-465L10.10-overexpressed lentiviral vector was constructed and transfected into VSMCs. Through EdU staining, migration assay, flow cytometry analysis, the roles of RP11-465L10.10 were estimated. Bioinformatics indicated that RP11-465L10.10 upregulating MMP9 expression via NF-κB signaling, and SN50 (a specific inhibitor of NF-κB pathway) was used to inhibit the NF-κB pathway activation, then the expression of MMP9 was detected in RP11-465L10.10 overexpressed VMSCs. Results In this study, we found RP11-465L10.10 and MMP9 were highly increased in TAD patient tissues, which was consistent in angiotensin II-induced VSMCs phenotype switching. RP11-465L10.10 overexpression facilitated VSMCs phenotype switching and MMP9 expression. Mechanismly, NF-κB signal pathway was involved in RP11-465L10.10 induced VSMCs phenotype switching and MMP9 expression by transcriptome data analysis and experimental confirm. Conclusion This study demonstrated that RP11-465L10.10 induces VSMCs phenotype switching and MMP9 expression via the NF-κB signal pathway, suggesting that RP11-465L10.10 might be a potential therapeutic target for TAA/D treatment.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - You Yu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Feng Zhu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weizhang Xiao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziying Yang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
54
|
Kremastiotis G, Handa I, Jackson C, George S, Johnson J. Disparate effects of MMP and TIMP modulation on coronary atherosclerosis and associated myocardial fibrosis. Sci Rep 2021; 11:23081. [PMID: 34848763 PMCID: PMC8632906 DOI: 10.1038/s41598-021-02508-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/12/2021] [Indexed: 11/12/2022] Open
Abstract
Matrix metalloproteinase (MMP) activity is tightly regulated by the endogenous tissue inhibitors (TIMPs), and dysregulated activity contributes to extracellular matrix remodelling. Accordingly, MMP/TIMP balance is associated with atherosclerotic plaque progression and instability, alongside adverse post-infarction cardiac fibrosis and subsequent heart failure. Here, we demonstrate that prolonged high-fat feeding of apolipoprotein (Apo)e-deficient mice triggered the development of unstable coronary artery atherosclerosis alongside evidence of myocardial infarction and progressive sudden death. Accordingly, the contribution of select MMPs and TIMPs to the progression of both interrelated pathologies was examined in Apoe-deficient mice with concomitant deletion of Mmp7, Mmp9, Mmp12, or Timp1 and relevant wild-type controls after 36-weeks high-fat feeding. Mmp7 deficiency increased incidence of sudden death, while Mmp12 deficiency promoted survival, whereas Mmp9 or Timp1 deficiency had no effect. While all mice harboured coronary disease, atherosclerotic burden was reduced in Mmp7-deficient and Mmp12-deficient mice and increased in Timp1-deficient animals, compared to relevant controls. Significant differences in cardiac fibrosis were only observed in Mmp-7-deficient mice and Timp1-deficient animals, which was associated with reduced capillary number. Adopting therapeutic strategies in Apoe-deficient mice, TIMP-2 adenoviral-overexpression or administration (delayed or throughout) of a non-selective MMP inhibitor (RS-130830) had no effect on coronary atherosclerotic burden or cardiac fibrosis. Taken together, our findings emphasise the divergent roles of MMPs on coronary plaque progression and associated post-MI cardiac fibrosis, highlighting the need for selective therapeutic approaches to target unstable atherosclerosis alongside adverse cardiac remodelling while negating detrimental adverse effects on either pathology, with targeting of MMP-12 seeming a suitable target.
Collapse
Affiliation(s)
- Georgios Kremastiotis
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Ishita Handa
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Christopher Jackson
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Sarah George
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Jason Johnson
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK.
| |
Collapse
|
55
|
Niu F, Liu Z, Liu P, Pan H, Bi J, Li P, Luo G, Chen Y, Zhang X, Dai X. Identification of novel genetic biomarkers and treatment targets for arteriosclerosis-related abdominal aortic aneurysm using bioinformatic tools. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:9761-9774. [PMID: 34814367 DOI: 10.3934/mbe.2021478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A large number of epidemiological studies have confirmed that arteriosclerosis (AS) is a risk factor for abdominal aortic aneurysm (AAA). However, the relationship between AS and AAA remains controversial. The objective of this work is to better understand the association between the two diseases by identifying the co-differentially expressed genes under both pathological conditions, so as to identify potential genetic biomarkers and treatment targets for atherosclerosis-related aneurysms. Differentially-expressed genes (DEGs) shared by both AS and AAA patients were identified by bioinformatics analyses of Gene Expression Omnibus (GEO) datasets GSE100927 and GSE7084. These DEGs were then subjected to bioinformatic analyses of protein-protein interaction (PPI), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Finally, the identified hub genes were further validated by qRT-PCR in AS (n = 4), AAA (n = 4), and healthy (n = 4) individuals. Differential expression analysis revealed a total of 169 and 37 genes that had increased and decreased expression levels, respectively, in both AS and AAA patients compared with healthy controls. The construction of a PPI network and key modules resulted in the identification of five hub genes (SPI1, TYROBP, TLR2, FCER1G, and MMP9) as candidate diagnostic biomarkers and treatment targets for patients with AS-related AAA. AS and AAA are indeed correlated; SPI1, TYROBP, TLR2, FCER1G and MMP9 genes are potential new genetic biomarkers for AS-related AAA.
Collapse
Affiliation(s)
- Fang Niu
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Zongwei Liu
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Peidong Liu
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongrui Pan
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Jiaxue Bi
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Peng Li
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Guangze Luo
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Yonghui Chen
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Xiaoxing Zhang
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Xiangchen Dai
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| |
Collapse
|
56
|
Okamori S, Ishii M, Asakura T, Suzuki S, Namkoong H, Kagawa S, Hegab AE, Yagi K, Kamata H, Kusumoto T, Ogawa T, Takahashi H, Yoda M, Horiuchi K, Hasegawa N, Fukunaga K. ADAM10 partially protects mice against influenza pneumonia by suppressing specific myeloid cell population. Am J Physiol Lung Cell Mol Physiol 2021; 321:L872-L884. [PMID: 34523355 DOI: 10.1152/ajplung.00619.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The influenza virus infection poses a serious health threat worldwide. Myeloid cells play pivotal roles in regulating innate and adaptive immune defense. A disintegrin and metalloproteinase (ADAM) family of proteins contributes to various immune responses; however, the role of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in influenza virus infection remains largely unknown. Herein, we investigated its role, focusing on myeloid cells, during influenza virus infection in mice. ADAM10 gene (Adam10)flox/flox/Lyz2-Cre (Adam10ΔLyz2) and control Adam10flox/flox mice were intranasally infected with 200 plaque-forming units of influenza virus A/H1N1/PR8/34. Adam10ΔLyz2 mice exhibited a significantly higher mortality rate, stronger lung inflammation, and a higher virus titer in the lungs than control mice. Macrophages and inflammatory cytokines, such as TNF-α, IL-1β, and CCL2, were increased in bronchoalveolar lavage fluid from Adam10ΔLyz2 mice following infection. CD11b+Ly6G-F4/80+ myeloid cells, which had an inflammatory monocyte/macrophage-like phenotype, were significantly increased in the lungs of Adam10ΔLyz2 mice. Adoptive transfer experiments suggested that these cells likely contributed to the poorer prognosis in Adam10ΔLyz2 mice. Seven days after infection, CD11b+Ly6G-F4/80+ lung cells exhibited significantly higher arginase-1 expression levels in Adam10ΔLyz2 mice than in control mice, whereas an arginase-1 inhibitor improved the prognosis of Adam10ΔLyz2 mice. Enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF)/GM-CSF receptor signaling likely contributed to this process. Collectively, these results indicate that myeloid ADAM10 protects against influenza virus pneumonia and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Satoshi Okamori
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Society of Promotion of Science, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shoji Suzuki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Medical Education Center, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Kazuma Yagi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Kusumoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takunori Ogawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Yoda
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Orthopedic Surgery, National Defence Medical College, Saitama, Japan
| | - Naoki Hasegawa
- Center for Infectious Diseases and Infection Control, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
57
|
Beck-Joseph J, Tabrizian M, Lehoux S. Molecular Interactions Between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front Cardiovasc Med 2021; 8:737934. [PMID: 34722670 PMCID: PMC8554018 DOI: 10.3389/fcvm.2021.737934] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is the largest contributor toward life-threatening cardiovascular events. Cellular activity and cholesterol accumulation lead to vascular remodeling and the formation of fatty plaques. Complications arise from blood clots, forming at sites of plaque development, which may detach and result in thrombotic occlusions. Vascular smooth muscle cells and macrophages play dominant roles in atherosclerosis. A firm understanding of how these cells influence and modulate each other is pivotal for a better understanding of the disease and the development of novel therapeutics. Recent studies have investigated molecular interactions between both cell types and their impact on disease progression. Here we aim to review the current knowledge. Intercellular communications through soluble factors, physical contact, and extracellular vesicles are discussed. We also present relevant background on scientific methods used to study the disease, the general pathophysiology and intracellular factors involved in phenotypic modulation of vascular smooth muscle cells. We conclude this review with a discussion of the current state, shortcomings and potential future directions of the field.
Collapse
Affiliation(s)
- Jahnic Beck-Joseph
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Stephanie Lehoux
- Department of Medicine, Lady Davis Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
58
|
Grootaert MOJ, Bennett MR. Vascular smooth muscle cells in atherosclerosis: time for a re-assessment. Cardiovasc Res 2021; 117:2326-2339. [PMID: 33576407 PMCID: PMC8479803 DOI: 10.1093/cvr/cvab046] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are key participants in both early and late-stage atherosclerosis. VSMCs invade the early atherosclerotic lesion from the media, expanding lesions, but also forming a protective fibrous cap rich in extracellular matrix to cover the 'necrotic' core. Hence, VSMCs have been viewed as plaque-stabilizing, and decreased VSMC plaque content-often measured by expression of contractile markers-associated with increased plaque vulnerability. However, the emergence of lineage-tracing and transcriptomic studies has demonstrated that VSMCs comprise a much larger proportion of atherosclerotic plaques than originally thought, demonstrate multiple different phenotypes in vivo, and have roles that might be detrimental. VSMCs down-regulate contractile markers during atherosclerosis whilst adopting alternative phenotypes, including macrophage-like, foam cell-like, osteochondrogenic-like, myofibroblast-like, and mesenchymal stem cell-like. VSMC phenotypic switching can be studied in tissue culture, but also now in the media, fibrous cap and deep-core region, and markedly affects plaque formation and markers of stability. In this review, we describe the different VSMC plaque phenotypes and their presumed cellular and paracrine functions, the regulatory mechanisms that control VSMC plasticity, and their impact on atherogenesis and plaque stability.
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrookes Hospital, CB2 0QQ Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrookes Hospital, CB2 0QQ Cambridge, UK
| |
Collapse
|
59
|
Nakagawa K, Tanaka M, Hahm TH, Nguyen HN, Matsui T, Chen YX, Nakashima Y. Accumulation of Plasma-Derived Lipids in the Lipid Core and Necrotic Core of Human Atheroma: Imaging Mass Spectrometry and Histopathological Analyses. Arterioscler Thromb Vasc Biol 2021; 41:e498-e511. [PMID: 34470476 DOI: 10.1161/atvbaha.121.316154] [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
[Figure: see text].
Collapse
Affiliation(s)
- Kazunori Nakagawa
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| | - Mitsuru Tanaka
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Tae-Hun Hahm
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Huu-Nghi Nguyen
- Department of Science and International Collaboration, Institute for Research and Development of Organic Products, Hanoi, Vietnam (H.-N.N.)
| | - Toshiro Matsui
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Yong-Xiang Chen
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan.,Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Alberta, Canada (Y.-X.C.)
| | - Yutaka Nakashima
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| |
Collapse
|
60
|
Gao H, Yu Z, Li Y, Wang X. miR-100-5p in human umbilical cord mesenchymal stem cell-derived exosomes mediates eosinophilic inflammation to alleviate atherosclerosis via the FZD5/Wnt/β-catenin pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1166-1176. [PMID: 34254638 DOI: 10.1093/abbs/gmab093] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Indexed: 01/05/2023] Open
Abstract
Exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSC-Ex) play important roles in immune and inflammation diseases. However, the role of hUCMSC-Ex in atherosclerosis has not been elucidated. In this study, the isolated exosomes were identified by transmission electron microscopy and nanoparticle tracking analysis. Exosome marker protein levels were increased in the hUCMSC-Ex compared with those in hUCMSC suspension, indicating that exosomes were successfully isolated from hUCMSCs. Furthermore, eosinophils were treated with oxidized low-density lipoprotein (ox-LDL) to construct inflammation model and then incubated with hUCMSC-Ex derived from hUCMSCs which were transfected with miR-100-5p mimic or miR-100-5p inhibitor. We found that hUCMSC-Ex increased miR-100-5p expression, inhibited cell migration, promoted cell apoptosis, and reduced inflammatory cytokine levels in ox-LDL-treated eosinophils, and miR-100-5p overexpression in hUCMSCs enhanced these effects, while miR-100-5p inhibition reversed these effects. Moreover, frizzled 5 (FZD5) was a target gene of miR-100-5p. FZD5 overexpression reversed the inhibitory effects of hUCMSC-Ex-miR-100-5p on cell progression and inflammation in eosinophils. Additionally, hUCMSC-Ex-miR-100-5p decreased the expression of cyclin D1 and β-catenin proteins. Wnt/β-catenin pathway activator BML-284 effectively reversed the effects of hUCMSC-Ex-miR-100-5p on cell progression and inflammation in eosinophils. ApoE-/- mice were fed with high-fat diet to construct an atherosclerosis mice model, and hUCMSC-Ex was injected into mice. hUCMSC-Ex reduced atherosclerotic plaque area and inflammation response in atherosclerosis mice. This study demonstrates that hUCMSC-Ex-miR-100-5p inhibits cell progression and inflammatory response in eosinophils via the FZD5/Wnt/β-catenin pathway, thereby alleviating atherosclerosis progression.
Collapse
Affiliation(s)
- Heng Gao
- Department of Emergency Internal Medicine, Shaanxi Provincial People’s Hospital, Xi’an 710068, China
| | - Zhanbiao Yu
- Department of Cardiovascular Medicine, Qingyang People’s Hospital, Qingyang 745000, China
| | - Yuanyuan Li
- Department of Emergency Surgery, Shaanxi Provincial People’s Hospital, Xi’an 710068, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
61
|
P 2Y 12 receptor blockers are anti-inflammatory drugs inhibiting both circulating monocytes and macrophages including THP-1 cells. Sci Rep 2021; 11:17459. [PMID: 34465804 PMCID: PMC8408182 DOI: 10.1038/s41598-021-95710-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/22/2021] [Indexed: 01/11/2023] Open
Abstract
P2Y12 blockade improves patient outcomes after myocardial infarction. As well as antithrombotic effects, anti-inflammatory effects may contribute to this beneficial clinical outcome. Here we aimed to identify potential anti-inflammatory effects of P2Y12 receptor blockers on monocytes and macrophages. Using flow cytometry, migration assays, flow chambers and RNA microarrays, we investigated the effects of adenosine diphosphate (ADP) and P2Y12 receptor blockers on blood monocytes, THP-1 monocytes and THP-1 monocytes after differentiation to macrophages. P2Y12 -expressing platelets can form aggregates with monocytes in circulating blood. Mediated by platelets, ADP results in activation of the integrin receptor Mac-1 on blood monocytes, as detected by the conformation-specific single-chain antibody MAN-1. Via the same association with platelets, THP-1 monocyte adhesion to the endothelial intercellular adhesion molecule 1 (ICAM-1) is induced by ADP. P2Y12 receptor blockers prevent these ADP effects on monocytes. Interestingly, in contrast to THP-1 monocytes, THP-1 monocytes, after differentiation to macrophages, directly expressed the P2Y12 receptor and consequently ADP was found to be a potent chemoattractant. Again, P2Y12 receptor blockers antagonised this effect. Accordingly, stimulation of THP-1 macrophages with ADP caused a substantial change in gene expression pattern and upregulation of several genes associated with inflammation and atherogenesis. These data establish novel anti-inflammatory effects of P2Y12 receptor blockers on monocytes and macrophages, which are expected to contribute to cardiovascular risk reduction.
Collapse
|
62
|
Zhang Y, Xu D, Huang P, Zhang Y, Li Q, Fan Z, Ren L. Essential role of protein kinase C βI in icariin-mediated protection against atherosclerosis. J Pharm Pharmacol 2021; 73:1169-1179. [PMID: 33822184 DOI: 10.1093/jpp/rgab040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study aimed to clarify the superior beneficial effects of icariin on atherosclerosis, as well as to explore the possible underlying mechanisms for its effect via the modulation of protein kinase C βI. METHODS Lipid profiles were determined while dissected aortas were prepared of ApoE-/- mice. The expression of protein kinase C βI and phosphorylation of protein kinase C βI were determined by immunohistochemistry analysis. Human vascular smooth muscle cells were subjected to ox-LDL stimulation. MTS assay was conducted to detect cell proliferation. A transwell migration assay was performed to evaluate migration capacity. Flow cytometric analysis was used to determine cell cycle progression. Quantitative real-time PCR and western blot were performed to assess gene expression. RESULTS Icariin significantly alleviated atherogenesis, as well as protein levels of protein kinase C βI and phosphorylated protein kinase C βI in the aorta. Icariin effectively suppressed cell proliferation and migration. protein kinase C βI, cyclin D1 and matrix metalloproteinase-9 were modulated in response to treatment with icariin. Protein kinase C activator reversed the protective effect of icariin on human vascular smooth muscle cells against ox- low-density lipoprotein, protein kinase C β inhibitor augmented the inhibitory effect of icariin. CONCLUSIONS Our findings highlight the probable application of icariin in atherosclerotic therapy and reveal that protein kinase C βI acts as a crucial regulator in the anti-atherosclerotic action of icariin.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Apolipoproteins E/metabolism
- Atherosclerosis/metabolism
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cyclin D1/metabolism
- Epimedium/chemistry
- Flavonoids/pharmacology
- Humans
- Lipoproteins, LDL/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Plant Extracts/pharmacology
- Protein Kinase C/metabolism
- Mice
Collapse
Affiliation(s)
- Yibing Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Dongsheng Xu
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Yang Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Qi Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, China
| |
Collapse
|
63
|
Krajnc MK, Hojs R, Holc I, Knez Ž, Pahor A. Accelerated atherosclerosis in premenopausal women with rheumatoid arthritis - 15-year follow-up. Bosn J Basic Med Sci 2021; 21:477-483. [PMID: 33259776 PMCID: PMC8292859 DOI: 10.17305/bjbms.2020.5176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with increased mortality and morbidity due to the higher cardiovascular risk in these patients. Traditional risk factors are not the only answer for the accelerated atherosclerosis. In a long-term prospective study, we investigated the relationship between asymptomatic atherosclerosis and traditional risk factors and inflammatory markers in patients with RA and matched healthy controls. We studied the laboratory test results, the concentrations of inflammatory mediators, matrix metalloproteases (MMP), and inflammation markers in a total of 70 (60 at follow-up) premenopausal healthy women with RA and 40 (34 at follow-up) matched controls. We used the B-mode ultrasound imaging of carotid arteries for the detection of asymptomatic atherosclerosis. Correlation with different factors was evaluated. Statistically significant higher values of inflammatory markers such as selective adhesion molecules ICAM and VCAM, interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-alpha), and MMP-3 in the patients group were found in the follow-up study. More plaques were found in the patients group (42.4% vs. 12.9%; p=0.005), as compared with the controls group. The patients had also higher values of cIMT (p=0.001). Using bivariate regression analysis only VCAM was found as a prognostic factor for plaque occurrence (r= 0. 341, p=0.016), but not for cIMT (r= -0.130, p=0.327) in premenopausal female patients with RA after the follow-up. Therefore, asymptomatic atherosclerosis is accelerated in premenopausal women with RA. The results of our follow-up study showed the association between inflammation and accelerated atherosclerosis. Furthermore, VCAM was found to have a statistically significant correlation with plaque occurrence in these patients.
Collapse
Affiliation(s)
- Metka Koren Krajnc
- Division of Internal Medicine, Department of Rheumatology, Maribor University Medical Centre, Ljubljana Slovenia
- Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Radovan Hojs
- Medical Faculty, University of Maribor, Maribor, Slovenia
- Division of Internal Medicine, Department of Nephrology, Maribor University Medical Centre, Maribor, Slovenia
| | - Iztok Holc
- Division of Internal Medicine, Department of Rheumatology, Maribor University Medical Centre, Ljubljana Slovenia
- Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Željko Knez
- Medical Faculty, University of Maribor, Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Artur Pahor
- Division of Internal Medicine, Department of Rheumatology, Maribor University Medical Centre, Ljubljana Slovenia
- Medical Faculty, University of Maribor, Maribor, Slovenia
| |
Collapse
|
64
|
Zhang F, Liu E, Radaic A, Yu X, Yang S, Yu C, Xiao S, Ye C. Diagnostic potential and future directions of matrix metalloproteinases as biomarkers in gingival crevicular fluid of oral and systemic diseases. Int J Biol Macromol 2021; 188:180-196. [PMID: 34339782 DOI: 10.1016/j.ijbiomac.2021.07.165] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023]
Abstract
Gingival crevicular fluid (GCF) is a physiological fluid and an inflammatory serum exudate derived from the gingival plexus of blood vessels and mixed with host tissues and subgingival plaque flows. In addition to proteins, GCF contains a diverse population of cells, including desquamated epithelial cells, cytokines, electrolytes, and bacteria from adjacent plaques. Recently, matrix metalloproteinases(MMPs), which are endopeptidases that are active against extracellular macromolecules, in GCF have been revealed as potential utility biomarkers for the diagnosis and follow-up of oral and systemic diseases, thereby facilitating the early evaluation of malignancy risk and the monitoring of disease progression and treatment response. Tissue inhibitors of metalloproteinases (TIMPs) are specific inhibitors of matrixins that participate in the regulation of local activities of MMPs in tissues. This review provides an overview of the latest findings on the diagnostic and prognostic values of MMPs and TIMPs in GCF of oral and systemic diseases, including periodontal disease, pulpitis, peri-implantitis and cardiovascular disease as well as the extraction, detection and analytical methods for GCF.
Collapse
Affiliation(s)
- Fan Zhang
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China; Physical Examination Center, West China Hospital, Sichuan University, Chengdu, China
| | - Enyan Liu
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Allan Radaic
- School of Dentistry, University of California San Francisco, San Francisco, CA, USA
| | - Xiaotong Yu
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuting Yang
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenhao Yu
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shimeng Xiao
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Changchang Ye
- State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China, Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
65
|
Buchler A, Munch M, Farber G, Zhao X, Al-Haddad R, Farber E, Rotstein BH. Selective Imaging of Matrix Metalloproteinase-13 to Detect Extracellular Matrix Remodeling in Atherosclerotic Lesions. Mol Imaging Biol 2021; 24:93-103. [PMID: 34231104 DOI: 10.1007/s11307-021-01626-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Overexpression and activation of matrix metalloproteinase-13 (MMP-13) within atheroma increases susceptibility to plaque rupture, a major cause of severe cardiovascular complications. In comparison to pan-MMP targeting [18F]BR-351, we evaluated the potential for [18F]FMBP, a selective PET radiotracer for MMP-13, to detect extracellular matrix (ECM) remodeling in vascular plaques possessing markers of inflammation. PROCEDURES [18F]FMBP and [18F]BR-351 were initially assessed in vitro by incubation with en face aortae from 8 month-old atherogenic ApoE-/- mice. Ex vivo biodistributions, plasma metabolite analyses, and ex vivo autoradiography were analogously performed 30 min after intravenous radiotracer administration in age-matched C57Bl/6 and ApoE-/- mice under baseline or homologous blocking conditions. En face aortae were subsequently stained with Oil Red O (ORO), sectioned, and subject to immunofluorescence staining for Mac-2 and MMP-13. RESULTS High-resolution autoradiographic image analysis demonstrated target specificity and regional concordance to lipid-rich lesions. Biodistribution studies revealed hepatobiliary excretion, low accumulation of radioactivity in non-excretory organs, and few differences between strains and conditions in non-target organs. Plasma metabolite analyses uncovered that [18F]FMBP exhibited excellent in vivo stability (≥74% intact) while [18F]BR-351 was extensively metabolized (≤37% intact). Ex vivo autoradiography and histology of en face aortae revealed that [18F]FMBP, relative to [18F]BR-351, exhibited 2.9-fold greater lesion uptake, substantial specific binding (68%), and improved sensitivity to atherosclerotic tissue (2.9-fold vs 2.1-fold). Immunofluorescent staining of aortic en face cross sections demonstrated elevated Mac-2 and MMP-13-positive areas within atherosclerotic lesions identified by [18F]FMBP ex vivo autoradiography. CONCLUSIONS While both radiotracers successfully identified atherosclerotic plaques, [18F]FMBP showed superior specificity and sensitivity for lesions possessing features of destructive plaque remodeling. The detection of ECM remodeling by selective targeting of MMP-13 may enable characterization of high-risk atherosclerosis featuring elevated collagenase activity.
Collapse
Affiliation(s)
- Ariel Buchler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Maxime Munch
- University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Gedaliah Farber
- University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Xiaoling Zhao
- University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Rami Al-Haddad
- University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Eadan Farber
- University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Benjamin H Rotstein
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada. .,University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada. .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.
| |
Collapse
|
66
|
Kirwin T, Gomes A, Amin R, Sufi A, Goswami S, Wang B. Mechanisms underlying the therapeutic potential of mesenchymal stem cells in atherosclerosis. Regen Med 2021; 16:669-682. [PMID: 34189963 DOI: 10.2217/rme-2021-0024] [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: 12/25/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory condition resulting in the formation of fibrofatty plaques within the intimal layer of arterial walls. The identification of resident stem cells in the vascular wall has led to significant investigation into their contributions to health and disease, as well as their therapeutic potential. Of these, mesenchymal stem cells (MSCs) are the most widely studied in human clinical trials, which have demonstrated a modulatory role in vascular physiology and disease. This review highlights the most recent knowledge surrounding the cell biology of MSCs, including their origin, identification markers and differentiation potential. The limitations concerning the implementation of MSC therapy are considered and novel solutions to overcome these are proposed.
Collapse
Affiliation(s)
- Thomas Kirwin
- Department of Medicine, Imperial College London, SW7 2BU, UK.,College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ana Gomes
- Department of Medicine, Imperial College London, SW7 2BU, UK
| | - Ravi Amin
- Department of Medicine, Imperial College London, SW7 2BU, UK
| | - Annam Sufi
- Department of Medicine, Imperial College London, SW7 2BU, UK.,GKT School of Medical Education, King's College London, London, SE1 1UL, UK
| | - Sahil Goswami
- Department of Medicine, Imperial College London, SW7 2BU, UK.,Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK
| | - Brian Wang
- Department of Medicine, Imperial College London, SW7 2BU, UK
| |
Collapse
|
67
|
Corilagin ameliorates atherosclerosis by regulating MMP-1, -2, and -9 expression in vitro and in vivo. Eur J Pharmacol 2021; 906:174200. [PMID: 34062170 DOI: 10.1016/j.ejphar.2021.174200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022]
Abstract
Corilagin is a polyphenol has been identified anti-inflammatory properties. However, the anti-atherosclerotic effects of corilagin are not well understood. Here, we evaluated the anti-atherosclerotic effects and the underlying mechanisms of corilagin. We also verified whether corilagin can reverse atherosclerosis by regulating matrix metalloproteinase (MMP)-1, -2, and -9 in vitro and in vivo. An atherosclerosis model was established by feeding minipigs a high-fat diet combined with balloon injury, and the effects of different concentrations of corilagin on common carotid artery atherosclerosis in minipigs were monitored. Murine RAW264.7 macrophages were cultured and induced with oxidized low-density lipoprotein; fluorescence microscopy revealed the nuclear translocation of NF-κB. Furthermore, MMP-1, -2, and -9 expression in common carotid artery plaques and cellular models was detected by immunohistochemistry, western blotting, and RT-PCR. The pathological results suggested that the vascular intima of the model control group was significantly thickened, a large amount of collagen fibers was deposited, endothelial cells were damaged and detached, and plaque and foam cell formation occurred to varying degrees on the arterial wall, with lipid deposition. Corilagin treatment significantly reduced the degree of injury in the common carotid artery and decreased the number of lipid plaques and foam cells. Additionally, corilagin downregulated MMP-1, -2, and -9 expression in the common carotid artery plaques and cellular model. Moreover, corilagin significantly inhibited NF-κB nuclear translocation in vitro. Overall, corilagin exerted substantial therapeutic effects on experimental atherosclerotic minipigs via the downregulation of MMP-1, -2, and -9 expression.
Collapse
|
68
|
Elevation of CD40/CD40L Inflammatory Pathway Molecules in Carotid Plaques from Moderate-and-Severe Obstructive Sleep Apnea Patients. Diagnostics (Basel) 2021; 11:diagnostics11060935. [PMID: 34067481 PMCID: PMC8224789 DOI: 10.3390/diagnostics11060935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 11/30/2022] Open
Abstract
A chronic inflammatory process characteristic of obstructive sleep apnea promotes vascular endothelial dysfunction and atherogenesis. This process can lead to destabilization and rupture of cardiovascular plaques, which clinically manifests as an acute coronary syndrome or stroke. The aim of this study was to investigate the inflammatory pathway leading to plaque destabilization in non-to-mild and moderate-to-severe groups of OSA patients. This prospective study involved enrollment of patients scheduled for endarterectomy. A sleep study was performed prior to surgery. Immunohistochemistry was performed on atherosclerotic plaques from carotid arteries obtained during standard open endarterectomy to determine levels of CD40, CD40L receptors, MCP-1, and MMP-9. The 46 patients included 14 controls, 13 with mild, 11 with moderate, and 8 with severe OSA. Increased expression of CD40, CD40L receptors, MCP-1, and MMP-9 were found to be proportionate with OSA severity. However, significant differences among groups were observed only for MCP-1 (p = 0.014). Increased expression of inflammatory markers (CD40, CD40L, MCP-1, MMP-9) is associated with increasing OSA severity. This suggests the CD40-CD4-L inflammatory pathway may contribute to plaque instability and rupture in OSA patients.
Collapse
|
69
|
Borén J, Chapman MJ, Krauss RM, Packard CJ, Bentzon JF, Binder CJ, Daemen MJ, Demer LL, Hegele RA, Nicholls SJ, Nordestgaard BG, Watts GF, Bruckert E, Fazio S, Ference BA, Graham I, Horton JD, Landmesser U, Laufs U, Masana L, Pasterkamp G, Raal FJ, Ray KK, Schunkert H, Taskinen MR, van de Sluis B, Wiklund O, Tokgozoglu L, Catapano AL, Ginsberg HN. Low-density lipoproteins cause atherosclerotic cardiovascular disease: pathophysiological, genetic, and therapeutic insights: a consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J 2021; 41:2313-2330. [PMID: 32052833 PMCID: PMC7308544 DOI: 10.1093/eurheartj/ehz962] [Citation(s) in RCA: 749] [Impact Index Per Article: 249.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/10/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M John Chapman
- Endocrinology-Metabolism Division, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France.,National Institute for Health and Medical Research (INSERM), Paris, France
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute and UCSF, Oakland, CA 94609, USA
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mat J Daemen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Linda L Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Denmark
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia.,Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Eric Bruckert
- INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hopital de la Pitie, Paris, France
| | - Sergio Fazio
- Departments of Medicine, Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK.,Institute for Advanced Studies, University of Bristol, Bristol, UK.,MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Luis Masana
- Research Unit of Lipids and Atherosclerosis, IISPV, CIBERDEM, University Rovira i Virgili, C. Sant Llorenç 21, Reus 43201, Spain
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Faculty of Medicine, Technische Universität München, Lazarettstr, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, and IRCCS MultiMedica, Milan, Italy
| | - Henry N Ginsberg
- Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| |
Collapse
|
70
|
Ten Cate V, Prochaska JH, Schulz A, Koeck T, Pallares Robles A, Lenz M, Eggebrecht L, Rapp S, Panova-Noeva M, Ghofrani HA, Meyer FJ, Espinola-Klein C, Lackner KJ, Michal M, Schuster AK, Strauch K, Zink AM, Laux V, Heitmeier S, Konstantinides SV, Münzel T, Andrade-Navarro MA, Leineweber K, Wild PS. Protein expression profiling suggests relevance of noncanonical pathways in isolated pulmonary embolism. Blood 2021; 137:2681-2693. [PMID: 33529319 PMCID: PMC9635523 DOI: 10.1182/blood.2019004571] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
Patients with isolated pulmonary embolism (PE) have a distinct clinical profile from those with deep vein thrombosis (DVT)-associated PE, with more pulmonary conditions and atherosclerosis. These findings suggest a distinct molecular pathophysiology and the potential involvement of alternative pathways in isolated PE. To test this hypothesis, data from 532 individuals from the Genotyping and Molecular Phenotyping of Venous ThromboEmbolism Project, a multicenter prospective cohort study with extensive biobanking, were analyzed. Targeted, high-throughput proteomics, machine learning, and bioinformatic methods were applied to contrast the acute-phase plasma proteomes of isolated PE patients (n = 96) against those of patients with DVT-associated PE (n = 276) or isolated DVT (n = 160). This resulted in the identification of shared molecular processes between PE phenotypes, as well as an isolated PE-specific protein signature. Shared processes included upregulation of inflammation, response to oxidative stress, and the loss of pulmonary surfactant. The isolated PE-specific signature consisted of 5 proteins: interferon-γ, glial cell line-derived neurotrophic growth factor, polypeptide N-acetylgalactosaminyltransferase 3, peptidyl arginine deiminase type-2, and interleukin-15 receptor subunit α. These proteins were orthogonally validated using cis protein quantitative trait loci. External replication in an independent population-based cohort (n = 5778) further validated the proteomic results and showed that they were prognostic for incident primary isolated PE in individuals without history of VTE (median time to event: 2.9 years; interquartile range: 1.6-4.2 years), supporting their possible involvement in the early pathogenesis. This study has identified molecular overlaps and differences between VTE phenotypes. In particular, the results implicate noncanonical pathways more commonly associated with respiratory and atherosclerotic disease in the acute pathophysiology of isolated PE.
Collapse
Affiliation(s)
- Vincent Ten Cate
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Center for Thrombosis and Hemostasis (CTH), and
| | - Jürgen H Prochaska
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Center for Thrombosis and Hemostasis (CTH), and
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
| | - Thomas Koeck
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
| | | | - Michael Lenz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lisa Eggebrecht
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Center for Thrombosis and Hemostasis (CTH), and
| | - Steffen Rapp
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
| | - Marina Panova-Noeva
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Center for Thrombosis and Hemostasis (CTH), and
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - H Ardeschir Ghofrani
- University Hospital Gießen and Marburg, Ambulance for Pulmonary Hypertension, Gießen, Germany
| | - F Joachim Meyer
- Lung Center Munich, Department of Pneumology and Pneumological Oncology, München Klinik Bogenhausen, München, Germany
| | | | | | | | | | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | - Stavros V Konstantinides
- Center for Thrombosis and Hemostasis (CTH), and
- Department of Cardiology, Democritus University of Thrace, University General Hospital, Greece; and
| | - Thomas Münzel
- Center for Thrombosis and Hemostasis (CTH), and
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Miguel A Andrade-Navarro
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology
- Center for Thrombosis and Hemostasis (CTH), and
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
71
|
Zhou S, Liu S, Liu X, Zhuang W. Bioinformatics Gene Analysis of Potential Biomarkers and Therapeutic Targets for Unstable Atherosclerotic Plaque-Related Stroke. J Mol Neurosci 2021; 71:1031-1045. [PMID: 33155176 DOI: 10.1007/s12031-020-01725-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/30/2020] [Indexed: 02/05/2023]
Abstract
Atherosclerotic plaque instability is a major cause of ischemic stroke. Researchers must develop novel strategies for the detection and treatment of unstable atherosclerotic plaque (UAP)-related stroke. We aimed to identify potential biomarkers and therapeutic targets of UAP-related stroke. Differentially expressed genes (DEGs) of UAP, ischemic stroke and smoking were identified by microarray analyses from the Gene Expression Omnibus. Gene Ontology (GO) and pathway functional enrichment analyses of DEGs were performed to analyze plaque destabilization and ischemic stroke physiopathology. An integrative analysis of UAP, ischemic stroke and smoking DEGs and functional annotations was performed to identify the underlying physiopathology and hub genes in UAP-related stroke and the relationship with smoking. Online search databases were applied to confirm hub gene biofunctions and their relationships with atherosclerosis and cerebrovascular diseases. Following integrative analysis, 18 co-DEGs of UAP and ischemic stroke, including 17 upregulated and one downregulated, were identified. Inflammation, immunity, extracellular matrix degradation, blood coagulation, apoptosis and nerve degeneration were the primary physiopathological processes in UAP-related stroke. Hub genes included MMP9, ITGAM, CCR1, NCF2 and CD163, among which MMP9 and ITGAM were top 10 genes for both UAP and stroke. Smoking may upregulate MMP9, NCF2, C5AR1 and ANPEP to accelerate plaque destabilization and UAP-related stroke. MMP9, ITGAM, CCR1, NCF2, CD163, hsa-miR-3123 and hsa-miR-144-3p are potential diagnostic and prognostic biomarkers of UAP-related stroke. MMP9 and ITGAM are potential therapeutic targets of UAP-related stroke, which will contribute to the development of novel management strategies.
Collapse
Affiliation(s)
- Shaojiong Zhou
- Shantou University Medical College, Shantou, Guangdong, China
- Neurology Department, First Affiliated Hospital of Shantou University Medical College, No.57, ChangPing Road, JinPing District, ShanTou City, 515041, Guangdong Province, China
| | - Shuo Liu
- Neurology Department, First Affiliated Hospital of Shantou University Medical College, No.57, ChangPing Road, JinPing District, ShanTou City, 515041, Guangdong Province, China
| | - Xiaoqiang Liu
- Neurology Department, First Affiliated Hospital of Shantou University Medical College, No.57, ChangPing Road, JinPing District, ShanTou City, 515041, Guangdong Province, China
| | - Weiduan Zhuang
- Neurology Department, First Affiliated Hospital of Shantou University Medical College, No.57, ChangPing Road, JinPing District, ShanTou City, 515041, Guangdong Province, China.
| |
Collapse
|
72
|
Ministrini S, Carbone F, Montecucco F. Updating concepts on atherosclerotic inflammation: From pathophysiology to treatment. Eur J Clin Invest 2021; 51:e13467. [PMID: 33259635 DOI: 10.1111/eci.13467] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Atherosclerosis is recognized as a systemic low-grade inflammatory disease. Furthermore, the dysregulation of the inflammatory response and its timely resolution is a pivotal process in determining the clinical manifestations of cardiac and cerebral acute ischaemia following atherothrombosis. METHODS This narrative review is based on the material searched on PubMed up to October 2020. The search terms we used were as follows: "atherosclerosis, inflammation, acute myocardial infarction and ischemic stroke" in combination with "biomarker, inflammatory cells and molecules, treatment." RESULTS The expected goal of addressing inflammation for the treatment of atherosclerosis and its acute ischaemic complications is reducing mortality and morbidity related to atherosclerotic cardiovascular disease, which are currently the first cause of death and disability worldwide. In this narrative review, we summarize the evidence about the main cellular and molecular mechanisms of inflammation in atherogenesis, atherothrombosis and acute ischaemic complications, with particular focus on the potential molecular targets for novel pharmacological treatments. CONCLUSION Although a large amount of evidence from animal models of atherothrombotic disease, and promising results of clinical trials, anti-inflammatory treatments against atherosclerosis are not yet recommended. A deepest understanding of pathophysiological mechanisms underlying the mechanisms driving resolution of the acute inflammation will probably allow to identify the optimal molecular target.
Collapse
Affiliation(s)
- Stefano Ministrini
- Department of Medicine, Internal Medicine, Università degli Studi di Perugia, Perugia, Italy.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
73
|
Abstract
Carbonic anhydrases (CAs) catalyze the synthesis of HCO3- from H2O and CO2. The dysfunction of CAs leads to aqueous humor secretion and high intraocular pressure to cause glaucoma pathogenesis. Methazolamide (MTZ), a CA inhibitor, can effectively treat glaucoma by reducing aqueous humor secretion. We previously reported that carbonic anhydrase I (CA1), a CA family member, was highly expressed in atherosclerotic tissues of the aorta and stimulated atherosclerosis (AS) by promoting calcification. MTZ showed therapeutic and preventive effects on AS in a mouse model. The above findings suggest a relationship between AS and glaucoma. This study explored the possible association between AS prevalence and glaucoma prevalence and the therapeutic effect of MTZ on AS by analyzing medical records. Among 10,751 patients with a primary diagnosis of glaucoma, 699 (6.5%) were also diagnosed with AS. However, the incidences of AS in patients with keratitis and scleritis, which are also ophthalmic diseases, were 2.5% (206/8383 patients) and 3.5% (46/1308 patients), respectively. In the population without ophthalmic records, the AS prevalence was only 1.9% (99,416/5,168,481 patients) (all p values between each group were below 0.001). Among 152,425 patients with a primary diagnosis of AS, 1245 (0.82%) were also diagnosed with glaucoma. Among 199,782 patients with a primary diagnosis of hypertension (excluding AS), 1149 (0.57%) were diagnosed with glaucoma, and among 5,313,433 patients without AS or hypertension, 9513 (0.18%) were diagnosed with glaucoma (all p values between each group were below 0.001). Additionally, among 14 patients who suffered from both AS and glaucoma and were treated with MTZ to cure their glaucoma, 9 of them showed reduced low-density lipoprotein (LDL) levels, the main index of AS, within 3 months after medication use (2.81 ± 0.61 mmol/L vs. 2.38 ± 0.58 mmol/L, p = 0.039). The above findings demonstrated a strong relation between AS and glaucoma and suggested that AS patients with glaucoma were more likely to suffer from angle-closure glaucoma.
Collapse
|
74
|
Park J, Choi H, Abekura F, Lim HS, Im JH, Yang WS, Hwang CW, Chang YC, Lee YC, Park NG, Kim CH. Avenanthramide C Suppresses Matrix Metalloproteinase-9 Expression and Migration Through the MAPK/NF- κB Signaling Pathway in TNF-α-Activated HASMC Cells. Front Pharmacol 2021; 12:621854. [PMID: 33841150 PMCID: PMC8027239 DOI: 10.3389/fphar.2021.621854] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/05/2021] [Indexed: 01/01/2023] Open
Abstract
In oat ingredients, flavonoids and phenolic acids are known to be the most important phenolic compounds. In phenolic compounds, wide-ranging biological responses, including antioxidative, anti-inflammatory, anti-allergic, and anti-cancer properties, were reported. Avenanthramide C (Avn C), a component of the phenolic compound of oats, has been reported to be highly antioxidant and anti-inflammatory, but its role in an anti-atherosclerosis response is unknown. The aim of this research was to assess the effect of Avn C on expression of MMP-9 on TNF-α-activated human arterial smooth-muscle cells (HASMC) and signaling involved in its anti-atherosclerosis activity. HASMC cells are known to produce inflammatory cytokines involving IL-6, IL-1β, and TNF-α during arteriosclerosis activity. Avn C specifically reduced IL-6 secretion in HASMC cells. Furthermore, we investigated whether Avn C could inhibit NF-κB nuclear protein translocation. Avn C suppressed nuclear protein translocation of NF-κB in TNF-α-stimulated HASMCs. The MMP-9 enzyme activity and expression are controlled through the MAPKs signaling path during the Avn C treatment. We confirmed that the levels of wound healing (p-value = 0.013, *p < 0.05) and migration (p-value = 0.007, **p < 0.01) are inhibited by 100 ng/ml TNF-α and 100 μM Avn C co-treated. Accordingly, Avn C inhibited the expression of MMP-9 and cell migration through the MAPK/NF-κB signaling pathway in TNF-α-activated HASMC. Therefore, Avn C can be identified and serve as disease prevention material and remedy for atherosclerosis.
Collapse
Affiliation(s)
- Junyoung Park
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Hyunju Choi
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Fukushi Abekura
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Hak-Seong Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Jong-Hwan Im
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, South Korea
| | | | - Cher-Won Hwang
- Department of AGEE, Handong Global University, Pohang, South Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, College of Medicine, Catholic University of Daegu, Daegu, South Korea
| | - Young-Choon Lee
- Faculty of Medicinal Biotechnology, Dong-A University, Busan, South Korea
| | - Nam Gyu Park
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, South Korea
| | - Cheorl-Ho Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
75
|
Henderson JM, Weber C, Santovito D. Beyond Self-Recycling: Cell-Specific Role of Autophagy in Atherosclerosis. Cells 2021; 10:cells10030625. [PMID: 33799835 PMCID: PMC7998923 DOI: 10.3390/cells10030625] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial vessel wall and underlies the development of cardiovascular diseases, such as myocardial infarction and ischemic stroke. As such, atherosclerosis stands as the leading cause of death and disability worldwide and intensive scientific efforts are made to investigate its complex pathophysiology, which involves the deregulation of crucial intracellular pathways and intricate interactions between diverse cell types. A growing body of evidence, including in vitro and in vivo studies involving cell-specific deletion of autophagy-related genes (ATGs), has unveiled the mechanistic relevance of cell-specific (endothelial, smooth-muscle, and myeloid cells) defective autophagy in the processes of atherogenesis. In this review, we underscore the recent insights on autophagy's cell-type-dependent role in atherosclerosis development and progression, featuring the relevance of canonical catabolic functions and emerging noncanonical mechanisms, and highlighting the potential therapeutic implications for prevention and treatment of atherosclerosis and its complications.
Collapse
Affiliation(s)
- James M. Henderson
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), D-80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, D-80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), D-80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, D-80336 Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), D-80336 Munich, Germany
- Correspondence: (C.W.); (D.S.)
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), D-80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, D-80336 Munich, Germany
- Institute for Genetic and Biomedical Research, UoS of Milan, National Research Council, I-09042 Milan, Italy
- Correspondence: (C.W.); (D.S.)
| |
Collapse
|
76
|
From Mitochondria to Atherosclerosis: The Inflammation Path. Biomedicines 2021; 9:biomedicines9030258. [PMID: 33807807 PMCID: PMC8000234 DOI: 10.3390/biomedicines9030258] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key process in metazoan organisms due to its relevance for innate defense against infections and tissue damage. However, inflammation is also implicated in pathological processes such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease of the arterial wall where unstable atherosclerotic plaque rupture causing platelet aggregation and thrombosis may compromise the arterial lumen, leading to acute or chronic ischemic syndromes. In this review, we will focus on the role of mitochondria in atherosclerosis while keeping inflammation as a link. Mitochondria are the main source of cellular energy. Under stress, mitochondria are also capable of controlling inflammation through the production of reactive oxygen species (ROS) and the release of mitochondrial components, such as mitochondrial DNA (mtDNA), into the cytoplasm or into the extracellular matrix, where they act as danger signals when recognized by innate immune receptors. Primary or secondary mitochondrial dysfunctions are associated with the initiation and progression of atherosclerosis by elevating the production of ROS, altering mitochondrial dynamics and energy supply, as well as promoting inflammation. Knowing and understanding the pathways behind mitochondrial-based inflammation in atheroma progression is essential to discovering alternative or complementary treatments.
Collapse
|
77
|
Yang L, Chen L, Fang Y, Ma S. Downregulation of GSK-3β Expression via Ultrasound-Targeted Microbubble Destruction Enhances Atherosclerotic Plaque Stability in New Zealand Rabbits. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:710-722. [PMID: 33261913 DOI: 10.1016/j.ultrasmedbio.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/14/2020] [Accepted: 11/01/2020] [Indexed: 06/12/2023]
Abstract
Accumulating evidence suggests that atherosclerosis (AS) is the underlying cause of vascular diseases, including heart disease and stroke. Ultrasound-targeted microbubble destruction (UTMD) technology provides a tolerable, efficient and effective system for drug delivery and gene transfection, which has broad application prospects in the treatment of AS. In addition, glycogen synthase kinase (GSK)-3β has been implicated as a potentially valuable therapeutic agent for AS treatment; however, the specific molecular mechanisms remain unknown. Therefore, this study was conducted to explore the effect of downregulation of GSK-3β expression via UTMD on atherosclerotic plaque stability. We established a THP-1 macrophage-derived foam cell model in vitro and an atherosclerotic plaque model in the right common carotid artery of New Zealand rabbits. We determined levels of the relevant vulnerable plaque stability elements. The results indicate that GSK-3β was upregulated in the foam cells and in atherosclerotic rabbits. Downregulation of GSK-3β expression by UTMD suppressed vulnerable plaque factors and inflammation in vitro and in vivo, changed the cytoskeleton of the foam cells in vitro, increased Young's modulus and decreased the peak intensity of atherosclerotic plaque in vivo. Moreover, GSK-3β inhibition by UTMD did not influence the viability of the foam cells. Collectively, our results indicate that GSK-3β could be a potential target for anti-atherogenic interventions and, in particular, can improve the stability of AS plaques in combination with UTMD.
Collapse
Affiliation(s)
- Lifei Yang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Lingzi Chen
- Ningbo University School of Medicine, Ningbo, China
| | - Ye Fang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Suya Ma
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China.
| |
Collapse
|
78
|
Song M, Meng L, Liu X, Yang Y. Feprazone Prevents Free Fatty Acid (FFA)-Induced Endothelial Inflammation by Mitigating the Activation of the TLR4/MyD88/NF-κB Pathway. ACS OMEGA 2021; 6:4850-4856. [PMID: 33644593 PMCID: PMC7905947 DOI: 10.1021/acsomega.0c05826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/18/2021] [Indexed: 06/12/2023]
Abstract
Increased levels of free fatty acid (FFA)-induced endothelial dysfunction play an important role in the initiation and development of atherosclerosis. Feprazone is a nonsteroidal anti-inflammatory compound. However, the beneficial effects of feprazone on FFA-induced endothelial dysfunction have not been reported before. In the current study, we found that treatment with feprazone ameliorated FFA-induced cell death of human aortic endothelial cells (HAECs) by restoring cell viability and reducing the release of lactate dehydrogenase (LDH). Importantly, we found that treatment with feprazone ameliorated FFA-induced oxidative stress by reducing the production of mitochondrial reactive oxygen species (ROS). In addition, feprazone prevented FFA-induced expression and secretion of proinflammatory cytokines and chemokines, such as chemokine ligand 5 (CCL5), interleukin-6 (IL-6), and interleukin-8 (IL-8). We also found that feprazone decreased the expression of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). Interestingly, we found that feprazone reduced the expression of cell adhesion molecules, such as vascular cell adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1). Our results also demonstrate that feprazone prevented FFA-induced activation of the toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor kappa-B (NF-κB) signaling pathway. These findings suggest that feprazone might serve as a potential agent for the treatment of atherosclerosis by improving the endothelial function.
Collapse
|
79
|
Hosseini V, Mallone A, Nasrollahi F, Ostrovidov S, Nasiri R, Mahmoodi M, Haghniaz R, Baidya A, Salek MM, Darabi MA, Orive G, Shamloo A, Dokmeci MR, Ahadian S, Khademhosseini A. Healthy and diseased in vitro models of vascular systems. LAB ON A CHIP 2021; 21:641-659. [PMID: 33507199 DOI: 10.1039/d0lc00464b] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Irregular hemodynamics affects the progression of various vascular diseases, such atherosclerosis or aneurysms. Despite the extensive hemodynamics studies on animal models, the inter-species differences between humans and animals hamper the translation of such findings. Recent advances in vascular tissue engineering and the suitability of in vitro models for interim analysis have increased the use of in vitro human vascular tissue models. Although the effect of flow on endothelial cell (EC) pathophysiology and EC-flow interactions have been vastly studied in two-dimensional systems, they cannot be used to understand the effect of other micro- and macro-environmental parameters associated with vessel wall diseases. To generate an ideal in vitro model of the vascular system, essential criteria should be included: 1) the presence of smooth muscle cells or perivascular cells underneath an EC monolayer, 2) an elastic mechanical response of tissue to pulsatile flow pressure, 3) flow conditions that accurately mimic the hemodynamics of diseases, and 4) geometrical features required for pathophysiological flow. In this paper, we review currently available in vitro models that include flow dynamics and discuss studies that have tried to address the criteria mentioned above. Finally, we critically review in vitro fluidic models of atherosclerosis, aneurysm, and thrombosis.
Collapse
Affiliation(s)
- Vahid Hosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Anna Mallone
- Institute of Regenerative Medicine, University of Zurich, Zurich CH-8952, Switzerland
| | - Fatemeh Nasrollahi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Serge Ostrovidov
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and Department of Radiological Sciences, University of California-Los Angeles, CA 90095, USA
| | - Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Department of Mechanical Engineering, Sharif University of Technology, Tehran 1136511155, Iran
| | - Mahboobeh Mahmoodi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd 8915813135, Iran
| | - Reihaneh Haghniaz
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Avijit Baidya
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA
| | - M Mehdi Salek
- School of Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain and Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01007, Spain
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 1136511155, Iran
| | - Mehmet R Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| |
Collapse
|
80
|
Liu MN, Luo G, Gao WJ, Yang SJ, Zhou H. miR-29 family: A potential therapeutic target for cardiovascular disease. Pharmacol Res 2021; 166:105510. [PMID: 33610720 DOI: 10.1016/j.phrs.2021.105510] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 01/11/2023]
Abstract
Cardiovascular disease (CVD), including heart failure, myocardial fibrosis and myocardial infarction, etc, remains one of the leading causes of mortality worldwide. Evidence shows that miRNA plays an important role in the pathogenesis of CVD. miR-29 family is one of miRNA, and over the past decades, many studies have demonstrated that miR-29 is involved in maintaining the integrity of arteries and in the regulation of atherosclerosis, especially in the process of myocardial fibrosis. Besides, heart failure, myocardial fibrosis and myocardial infarction are inseparable from the regulatory role of miR-29. Here, we comprehensively review recent studies regarding miR-29 and CVD, illustrate the possibility of miR-29 as a potential marker for prevention, treatment and prognostic observation.
Collapse
Affiliation(s)
- Meng-Nan Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Si-Jin Yang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China.
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China.
| |
Collapse
|
81
|
Shirakawa T, Fujisue K, Nakamura S, Yamamoto N, Oshima S, Matsumura T, Tsunoda R, Hirai N, Koide S, Tayama S, Kikuta K, Hirose T, Maruyama H, Fujimoto K, Kajiwara I, Sakamoto T, Nakao K, Sakaino N, Nagayoshi Y, Hokamaki J, Shimomura H, Sakamoto K, Yamamoto E, Izumiya Y, Kaikita K, Hokimoto S, Ogawa H, Tsujita K. Dose-Dependent Inhibitory Effect of Rosuvastatin in Japanese Patients with Acute Myocardial Infarction on Serum Concentration of Matrix Metalloproteinases-INVITATION Trial. J Atheroscler Thromb 2021; 29:229-241. [PMID: 33408315 PMCID: PMC8803556 DOI: 10.5551/jat.59477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Aim:
Matrix metalloproteinases (MMPs) play critical roles in acute myocardial infarction (AMI). This trial was conducted to determine the potential effects of higher-dose rosuvastatin on circulating MMP levels in patients with AMI.
Methods:
This was a multicenter, open-label, 1:1 randomized, parallel-group study. Patients with AMI were randomly assigned to the appropriate-dose group (10 mg rosuvastatin once daily) or the low-dose group (2.5 mg rosuvastatin once daily) within 24 hours after percutaneous coronary intervention. MMP-2 and MMP-9 levels were measured on day 1 and at week 4, 12, and 24 after enrollment. The primary endpoint was the change in MMP levels at 24 weeks after enrollment. The secondary endpoints were change in MMP levels at day 1 and weeks 4 and 12 after enrollment.
Results:
Between August 2017 and October 2018, 120 patients with AMI from 19 institutions were randomly assigned to either the appropriate-dose or the low-dose group. There were 109 patients who completed the 24-week follow-up. The primary endpoint for both MMP-2 and MMP-9 was not significantly different between the two groups. The change in the active/total ratio of MMP-9 at week 12 after baseline was significantly lower in the appropriate-dose group compared with the low-dose group (0.81 [−52.8–60.1]% vs. 70.1 [−14.5–214.2]%,
P
=0.004), while the changes in MMP-2 were not significantly different between the two groups during the study period.
Conclusions:
This study could not demonstrate the superiority of appropriate-dose of rosuvastatin in inhibiting serum MMPs levels in patients with AMI.
Collapse
Affiliation(s)
- Takuhiro Shirakawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University.,Department of Cardiovascular Medicine, Fukuoka Tokushukai Medical Center
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | - Shinichi Nakamura
- Division of Cardiology, Japan Community Health care Organization Hitoyoshi Medical Center
| | | | | | - Toshiyuki Matsumura
- Division of Cardiology, Japan Labor Health and Welfare Organization Kumamoto Rosai Hospital
| | - Ryusuke Tsunoda
- Division of Cardiology, Japanese Red Cross Kumamoto Hospital
| | - Nobutaka Hirai
- Division of Cardiology, Kumamoto Regional Medical Center
| | - Shunichi Koide
- Division of Cardiology, Health Insurance Kumamoto General Hospital
| | - Shinji Tayama
- Division of Cardiology, Health Insurance Kumamoto General Hospital
| | | | - Toyoki Hirose
- Division of Cardiology, Minamata City General Hospital & Medical Center
| | - Hideki Maruyama
- Division of Cardiology, Minamata City General Hospital & Medical Center
| | - Kazuteru Fujimoto
- Department of Cardiology, National Hospital Organization Kumamoto Medical Center
| | | | - Tomohiro Sakamoto
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center
| | - Koichi Nakao
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center
| | | | | | | | - Hideki Shimomura
- Department of Cardiovascular Medicine, Fukuoka Tokushukai Medical Center
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University.,Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | | | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University
| | | |
Collapse
|
82
|
Assallum H, Song TY, Aronow WS, Chandy D. Obstructive sleep apnoea and cardiovascular disease: a literature review. Arch Med Sci 2021; 17:1200-1212. [PMID: 34522249 PMCID: PMC8425247 DOI: 10.5114/aoms.2019.88558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022] Open
Abstract
As obesity becomes more common worldwide, the prevalence of obstructive sleep apnoea (OSA) continues to rise. Obstructive sleep apnoea is a well-known disorder that causes chronic intermittent hypoxia (CIH), which is considered a risk factor for atherosclerosis directly and indirectly. Ischaemic heart disease remains the leading cause of death. Most risk factors for atherosclerosis are well understood. However, other factors such as CIH are less well understood. Several studies have investigated the pathophysiology of CIH, attempting to uncover its link to atherosclerosis and to determine whether OSA treatment can be a therapeutic modality to modify the risk for atherosclerosis. In this article, we will review the pathophysiology of OSA as an independent risk factor for cardiovascular disease and discuss the most common markers that have been studied. We will also examine the potential impact of OSA management as a risk factor modifier on the reversibility of atherosclerosis.
Collapse
Affiliation(s)
- Hussein Assallum
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York Medical College, Valhalla, NY, USA
| | - Tian Yue Song
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York Medical College, Valhalla, NY, USA
| | | | - Dipak Chandy
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
83
|
Ben Braiek A, Chahed H, Dumont F, Abdelhak F, Hichem D, Gamra H, Baudin B. Identification of biomarker panels as predictors of severity in coronary artery disease. J Cell Mol Med 2020; 25:1518-1530. [PMID: 33381894 PMCID: PMC7875935 DOI: 10.1111/jcmm.16244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in atherosclerotic plaque rupture and recondition. Specific tissue inhibitors (TIMPs) control MMP functions. Both MMPs and TIMPs are potential biomarkers of plaque instability. Elevated Apo‐CII and CIII and Apo‐E levels are recognized as cardiovascular disease risk factors. We aimed to establish the best blood biomarker panel to evaluate the coronary artery disease (CAD) severity. Plasma levels of MMP‐3 and MMP‐9, TIMP‐1 and TIMP‐2, Apo‐CII, Apo‐CIII and Apo‐E were measured in 472 patients with CAD evaluated by coronary angiography and electrocardiography, and in 285 healthy controls. MMP‐3 and MMP‐9 plasma levels in CAD patients were significantly increased (P < 0.001) compared to controls (3.54‐ and 3.81‐fold, respectively). Furthermore, these increments are modulated by CAD severity as well as for Apo‐CII and Apo‐CIII levels (P < 0.001). TIMPs levels were decreased in CAD versus controls (P < 0.001) and in inverse correlation to MMPs. Standard ROC curve approach showed the importance of panels of biomarkers, including MMP‐3, MMP‐9, TIMP‐1, TIMP‐2, Apo‐CII and Apo‐CIII, for disease aggravation diagnosis. A high area under curve (AUC) value (0.995) was reached for the association of MMP‐9, TIMP‐2 and Apo‐CIII. The unbalance between MMPs and TIMPs in vascular wall and dyslipidaemia creates favourable conditions for plaque disruption. Our study suggests that the combination of MMP‐9, TIMP‐2 and Apo‐CIII values (‘CAD aggravation panel’) characterizes the severity of CAD, that is electrophysiological state, number of involved vessels, stent disposal and type of stent.
Collapse
Affiliation(s)
- Assia Ben Braiek
- Service de Biochimie, DMU BioGem, Hôpital Saint-Antoine, Paris, Sorbonne Université, Paris, France.,Molecular Biology Departments, Faculty of Pharmacy, Monastir University, Monastir, Tunisia.,UMS IPSIT - UFR Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Hinda Chahed
- Molecular Biology Departments, Faculty of Pharmacy, Monastir University, Monastir, Tunisia
| | - Florent Dumont
- UMS IPSIT - UFR Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Fodha Abdelhak
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia.,Department of Cardiology, Farhat Hached Hospital, Sousse, Tunisia
| | - Denguir Hichem
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia.,The Regional Hospital Mohamed Ben Sassi, Gabes, Tunisia
| | - Habib Gamra
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia
| | - Bruno Baudin
- Service de Biochimie, DMU BioGem, Hôpital Saint-Antoine, Paris, Sorbonne Université, Paris, France.,INSERM UMR 1193 - UFR Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
84
|
Mazuchová J, Halašová E, Mazuch J, Šarlinová M, Valentová V, Franeková M, Zelník Š, Krkošková K, Javorka K, Péč M, Grendár M. Investigation of association between genetic polymorphisms of MMP2, MMP8, MMP9 and TIMP2 and development of varicose veins in the Slovak Population - pilot study. Physiol Res 2020; 69:S443-S454. [PMID: 33471544 DOI: 10.33549/physiolres.934597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent metalloendopeptidases that degrades extracellular matrix (ECM) components. MMPs are associated with venous wall remodelling, proliferation, migration, phenotypic and functional transformation of vascular smooth muscle cells and ECM organization under the physiological and pathophysiological conditions. We investigated possible association of genetic promoter polymorphisms of MMP2 (rs243866), MMP8 (rs11225395), MMP9 (rs3918242) and TIMP2 (rs8179090) to varicose veins development in the Slovak population. Genomic DNA from 276 Slovak individuals (138 cases, 138 controls) was genotyped for selected SNPs (rs243866, rs11225395, rs3918242 and rs8179090) using the PCR-RFLP analysis. The data were analysed by chi-squared (chi2) test, logistic regression, and Mann-Whitney test. The risk of varicose veins development was evaluated in dominant, codominant and recessive genetic models. The statistical evaluation of selected polymorphisms in patients in all three genetic models has not shown a significant risk of varicose veins development. Our study has not shown the association between selected polymorphisms and increased risk of varicose veins development in Slovak population. More evidence with broaden sample size is needed.
Collapse
Affiliation(s)
- J Mazuchová
- Department of Medical Biology, Jessenius Faculty of Medicine, Martin, Slovakia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Lin Y, Liu M, Chen E, Jiang W, Shi W, Wang Z. Bone marrow-derived mesenchymal stem cells microvesicles stabilize atherosclerotic plaques by inhibiting NLRP3-mediated macrophage pyroptosis. Cell Biol Int 2020; 45:820-830. [PMID: 33325118 DOI: 10.1002/cbin.11526] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
Rupture of atherosclerotic plaques constitutes the major cause of thrombosis and acute ischemic coronary syndrome. Bone marrow-derived mesenchymal stem cells microvesicles (BMSCs-MVs) are reported to promote angiogenesis. This study investigated the role of BMSCs-MVs in stabilizing atherosclerotic plaques. BMSCs-MVs in mice were isolated and identified. The mouse model of atherosclerosis was established, and mice were injected with BMSCs-MVs via the tail vein. The macrophage model with high glucose and oxidative damage was established and then incubated with BMSCs-MVs. Nod-like receptor protein 3 (NLRP3) expression, pyroptosis-related proteins, and inflammatory factors were detected. Actinomycin D was used to inhibit the secretion of BMSCs-MVs to verify the source of microRNA-223 (miR-223). The binding relationship between miR-223 and NLRP3 was predicted and verified. BMSCs-MVs with knockdown of miR-223 were cocultured with bone marrow-derived macrophages with knockdown of NLRP3, and then levels of miR-223, NLRP3, pyroptosis-related proteins, and inflammatory factors were detected. BMSCs-MVs could reduce the vulnerability index of atherosclerotic plaques and intima-media thickness in mice, and inhibit pyroptosis and inflammation. BMSCs-MVs inhibited pyroptosis and inflammatory factors in macrophages. BMSCs-MVs carried miR-223 to inhibit NLRP3 expression and reduce macrophage pyroptosis, thereby stabilizing the atherosclerotic plaques.
Collapse
Affiliation(s)
- Yu Lin
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Meihan Liu
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Enqi Chen
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Jiang
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Weidong Shi
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiyuan Wang
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
86
|
Feng X, Yu F, Zhou X, Liu Z, Liao D, Huang Q, Li X, Jin X, Xia J. MMP9 rs17576 Is Simultaneously Correlated with Symptomatic Intracranial Atherosclerotic Stenosis and White Matter Hyperintensities in Chinese Population. Cerebrovasc Dis 2020; 50:4-11. [PMID: 33296906 DOI: 10.1159/000511582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/12/2020] [Indexed: 11/19/2022] Open
Abstract
PURPOSE The aim of this study was screening for single nucleotide polymorphisms (SNPs) associated with white matter hyperintensities (WMHs) in symptomatic intracranial atherosclerotic stenosis (sICAS) patients and exploring a possible connection in the genetic background between macrovascular disease and small vessel disease. METHODS There were 400 sICAS patients enrolled in the study. Fazekas scores were applied to WMH classification. Healthy controls were referred to 1,000 Genome Project and GeneSky company who provided 1,007 Chinese healthy controls. Fast target sequencing technology was used to select the SNPs of 102 genes related to the pathogenesis of sICAS in the sICAS patients. RESULTS The allele frequencies of 88 SNPs were significantly different between the sICAS group and the healthy controls (p < 0.05). The allele frequencies of 53 SNPs were significantly different between the sICAS patients with and without WMHs (p < 0.05). Further analysis found that matrix metalloproteinase 9 (MMP9) rs17576 was simultaneously related to sICAS and WMHs. The frequency of the rs17576 A allele was significantly lower in sICAS patients when compared to the normal controls (p = 0.03, OR [95% CI] = 0.75 [0.625-0.91]). Also, the frequency of the rs17576 genotypes was significantly different under codominant (p = 0.009), dominant (p = 0.014), and recessive (p= 0.023) models. The frequency of the rs17576 A allele was significantly higher in sICAS with WMH patients, compared to those without WMHs (p = 0.022, OR [95% CI] = 1.54 [1.06-2.22]); the frequency of the rs17576 genotypes was significantly different under codominant (p = 0.019) and recessive (p = 0.032) models. Logistic regression analysis showed that age, hypertension, and MMP9 rs17576 AA genotype were independent risk factors for sICAS with WMHs. CONCLUSION MMP9 rs17576 may be simultaneously associated with the risk of sICAS and WMHs.
Collapse
Affiliation(s)
- Xianjing Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Yu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoqing Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Di Liao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Jin
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China,
| |
Collapse
|
87
|
Yang K, Zeng L, Ge A, Pan X, Bao T, Long Z, Tong Q, Yuan M, Zhu X, Ge J, Huang Z. Integrating systematic biological and proteomics strategies to explore the pharmacological mechanism of danshen yin modified on atherosclerosis. J Cell Mol Med 2020; 24:13876-13898. [PMID: 33140562 PMCID: PMC7753997 DOI: 10.1111/jcmm.15979] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/14/2020] [Accepted: 09/24/2020] [Indexed: 02/05/2023] Open
Abstract
This research utilized the systematic biological and proteomics strategies to explore the regulatory mechanism of Danshen Yin Modified (DSYM) on atherosclerosis (AS) biological network. The traditional Chinese medicine database and HPLC was used to find the active compounds of DSYM, Pharmmapper database was used to predict potential targets, and OMIM database and GeneCards database were used to collect AS targets. String database was utilized to obtain the other protein of proteomics proteins and the protein-protein interaction (PPI) data of DSYM targets, AS genes, proteomics proteins and other proteins. The Cytoscape 3.7.1 software was utilized to construct and analyse the network. The DAVID database is used to discover the biological processes and signalling pathways that these proteins aggregate. Finally, animal experiments and proteomics analysis were used to further verify the prediction results. The results showed that 140 active compounds, 405 DSYM targets and 590 AS genes were obtained, and 51 differentially expressed proteins were identified in the DSYM-treated ApoE-/- mouse AS model. A total of 4 major networks and a number of their derivative networks were constructed and analysed. The prediction results showed that DSYM can regulate AS-related biological processes and signalling pathways. Animal experiments have also shown that DSYM has a therapeutic effect on ApoE-/-mouse AS model (P < .05). Therefore, this study proposed a new method based on systems biology, proteomics, and experimental pharmacology, and analysed the pharmacological mechanism of DSYM. DSYM may achieve therapeutic effects by regulating AS-related signalling pathways and biological processes found in this research.
Collapse
Affiliation(s)
- Kailin Yang
- The First Affiliated Hospital of Hunan University of Chinese MedicineChangshaChina
- Hunan University of Chinese MedicineChangshaChina
- Capital Medical UniversityBeijingChina
| | - Liuting Zeng
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Anqi Ge
- The First Affiliated Hospital of Hunan University of Chinese MedicineChangshaChina
- Hunan University of Chinese MedicineChangshaChina
| | - Xiaoping Pan
- Hunan University of Chinese MedicineChangshaChina
| | - Tingting Bao
- Guang'anmen Hospital, China Academy of Chinese Medical SciencesBeijingChina
- Beijing University of Chinese MedicineBeijingChina
| | | | | | | | - Xiaofei Zhu
- Xiangya School of MedicineCentral South UniversityChangsha CityChina
| | - Jinwen Ge
- Hunan University of Chinese MedicineChangshaChina
| | | |
Collapse
|
88
|
Basak S, Khare HA, Roursgaard M, Kempen PJ, Lee JH, Bazban-Shotorbani S, Kræmer M, Chernyy S, Andresen TL, Almdal K, Kamaly N. Simultaneous Cross-Linking and Cross-Polymerization of Enzyme Responsive Polyethylene Glycol Nanogels in Confined Aqueous Droplets for Reduction of Low-Density Lipoprotein Oxidation. Biomacromolecules 2020; 22:386-398. [PMID: 33125232 DOI: 10.1021/acs.biomac.0c01238] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A key initiating step in atherosclerosis is the accumulation and retention of apolipoprotein B complexing lipoproteins within the artery walls. In this work, we address this exact initiating mechanism of atherosclerosis, which results from the oxidation of low-density lipoproteins (oxLDL) using therapeutic nanogels. We present the development of biocompatible polyethylene glycol (PEG) cross-linked nanogels formed from a single simultaneous cross-linking and co-polymerization step in water without the requirement for an organic solvent, high temperature, or shear stress. The nanogel synthesis also incorporates in situ noncovalent electrostatically driven template polymerization around an innate anti-inflammatory and anti-oxidizing paraoxonase-1 (PON-1) enzyme payload-the release of which is triggered because of matrix metalloproteinase responsive elements instilled in the PEG cross-linker monomer. The results obtained demonstrate the potential of triggered release of the PON-1 enzyme and its efficacy against the production of ox-LDL, and therefore a reduction in macrophage foam cell and reactive oxygen species formation.
Collapse
Affiliation(s)
- Suman Basak
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Harshvardhan Ajay Khare
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, 2200 Copenhagen, Denmark
| | - Martin Roursgaard
- Department of Public Health, Section of Environmental Health, University of Copenhagen, 1014 Copenhagen K, Denmark
| | - Paul J Kempen
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jong Hyun Lee
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Salime Bazban-Shotorbani
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Martin Kræmer
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Sergey Chernyy
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kristoffer Almdal
- Department of Chemistry, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Nazila Kamaly
- Department of Health Technology, DTU Health Tech, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.,Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ London, U.K
| |
Collapse
|
89
|
The Role of Matrix Metalloproteinase-9 in Atherosclerotic Plaque Instability. Mediators Inflamm 2020; 2020:3872367. [PMID: 33082709 PMCID: PMC7557896 DOI: 10.1155/2020/3872367] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/10/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) belongs to the MMP family and has been widely investigated. Excessive MMP-9 expression can enhance extracellular matrix degradation and promote plaque instability. Studies have demonstrated that MMP-9 levels are higher in vulnerable plaques than in stable plaques. Additionally, several human studies have demonstrated that MMP-9 may be a predictor of atherosclerotic plaque instability and a risk factor for future adverse cardiovascular and cerebrovascular events. MMP-9 deficiency or blocking MMP-9 expression can inhibit plaque inflammation and prevent atherosclerotic plaque instability. All of these results suggest that MMP-9 may be a useful predictive biomarker for vulnerable atherosclerotic plaques, as well as a therapeutic target for preventing atherosclerotic plaque instability. In this review, we describe the structure, function, and regulation of MMP-9. We also discuss the role of MMP-9 in predicting and preventing atherosclerotic plaque instability.
Collapse
|
90
|
Hu A, Shuai Z, Liu J, Huang B, Luo Y, Deng J, Liu J, Yu L, Li L, Xu S. Ginsenoside Rg1 prevents vascular intimal hyperplasia involved by SDF-1α/CXCR4, SCF/c-kit and FKN/CX3CR1 axes in a rat balloon injury. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:113046. [PMID: 32504784 DOI: 10.1016/j.jep.2020.113046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng C. A. Mey. is a traditional tonic that has been used for thousands of years, and has positive effects on vascular diseases. Ginsenoside Rg1 (GS-Rg1) is one of the active ingredients of Panax ginseng C. A. Mey. and has been shown to have beneficial effects against ischemia/reperfusion injury. Our previously study has found that GS-Rg1 can mobilize bone marrow stem cells and inhibit vascular smooth muscle proliferation and phenotype transformation. However, pharmacological effects and mechanism of GS-Rg1 in inhibiting intimal hyperplasia is still unknown. AIM OF THE STUDY This study was aimed to investigate whether GS-Rg1 prevented vascular intimal hyperplasia, and the involvement of stromal cell-derived factor-1α (SDF-1α)/CXCR4, stem cell factor (SCF)/c-kit and fractalkine (FKN)/CX3CR1 axes. MATERIALS AND METHODS Rats were operated with carotid artery balloon injury. The treatment groups were injected with 4, 8 and 16 mg/kg of GS-Rg1 for 14 days. The degree of intimal hyperplasia was evaluated by histopathological examination. The expression of α-SMA (α-smooth muscle actin) and CD133 were detected by double-label immunofluorescence. Serum levels of SDF-1α, SCF and soluble FKN (sFKN) were detected by enzyme linked immunosorbent assay (ELISA). The protein expressions of SCF, SDF-1α and FKN, as well as the receptors c-kit, CXC chemokine receptor type 4 (CXCR4) and CX3C chemokine receptor type 1 (CX3CR1) were detected by immunochemistry. RESULTS GS-Rg1 reduced intimal hyperplasia by evidence of the values of NIA, the ratio of NIA/MA, and the ratio of NIA/IELA and the ratio of NIA/LA, especially in 16 mg/kg group. Furthermore, GS-Rg1 8 mg/kg group and 16 mg/kg group decreased the protein expressions of the SDF-1α/CXCR4, SCF/c-kit and FKN/CX3CR1 axes in neointima, meanwhile GS-Rg1 8 mg/kg group and 16 mg/kg group also attenuated the expressions of SDF-1α, SCF and sFKN in serum. In addition, the expression of α-SMA and CD133 marked smooth muscle progenitor cells (SMPCs) was decreased after GS-Rg1 treatment. CONCLUSIONS GS-Rg1 has a positive effect on inhibiting vascular intimal hyperplasia, and the underlying mechanism is related to inhibitory expression of SDF-1α/CXCR4, SCF/c-kit and FKN/CX3CR1 axes.
Collapse
MESH Headings
- Angioplasty, Balloon
- Animals
- CX3C Chemokine Receptor 1/metabolism
- Carotid Artery Injuries/etiology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/prevention & control
- Carotid Artery, Common/drug effects
- Carotid Artery, Common/metabolism
- Carotid Artery, Common/pathology
- Chemokine CX3CL1/metabolism
- Chemokine CXCL12/metabolism
- Disease Models, Animal
- Ginsenosides/pharmacology
- Hyperplasia
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Neointima
- Proto-Oncogene Proteins c-kit/metabolism
- Rats, Sprague-Dawley
- Receptors, CXCR4/metabolism
- Signal Transduction
- Stem Cell Factor/metabolism
Collapse
Affiliation(s)
- Anling Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China; State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, 550025, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, Guizhou, 550014, China.
| | - Zhiqin Shuai
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Jiajia Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Bo Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Yunmei Luo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Jiang Deng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Limei Yu
- State Key Laboratory of Cell Engineering of Guizhou Province, The Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, 563003, China.
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, China; State Key Laboratory of Cell Engineering of Guizhou Province, The Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, 563003, China.
| |
Collapse
|
91
|
Rashdan NA, Shrestha B, Pattillo CB. S-glutathionylation, friend or foe in cardiovascular health and disease. Redox Biol 2020; 37:101693. [PMID: 32912836 PMCID: PMC7767732 DOI: 10.1016/j.redox.2020.101693] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 12/27/2022] Open
Abstract
Glutathione is a low molecular weight thiol that is present at high levels in the cell. The high levels of glutathione in the cell make it one of the most abundant antioxidants contributing to cellular redox homeostasis. As a general rule, throughout cardiovascular disease and progression there is an imbalance in redox homeostasis characterized by reactive oxygen species overproduction and glutathione underproduction. As research into these imbalances continues, glutathione concentrations are increasingly being observed to drive various physiological and pathological signaling responses. Interestingly in addition to acting directly as an antioxidant, glutathione is capable of post translational modifications (S-glutathionylation) of proteins through both chemical interactions and enzyme mediated events. This review will discuss both the chemical and enzyme-based S-glutathionylation of proteins involved in cardiovascular pathologies and angiogenesis.
Collapse
Affiliation(s)
- N A Rashdan
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA
| | - B Shrestha
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA
| | - C B Pattillo
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
92
|
Zhou K, Guo T, Xu Y, Guo R. Correlation Between Plasma Matrix Metalloproteinase-28 Levels and Severity of Calcific Aortic Valve Stenosis. Med Sci Monit 2020; 26:e925260. [PMID: 32950995 PMCID: PMC7526340 DOI: 10.12659/msm.925260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease is a common cardiovascular disorder worldwide. This study aimed to investigate the correlation between plasma matrix metalloproteinase-28 (MMP-28) levels and the severity of calcific aortic valve stenosis. MATERIAL AND METHODS Calcific aortic valve stenosis patients who were admitted to the heart center of our hospital between January 2016 and January 2019 to undergo surgery were successively enrolled in this study (55 males and 24 females with an average age of 58.5±9.6). Information on echocardiography, plasma MMP-28 levels, and other clinical data of the patients was retrospectively collected. RESULTS The average plasma MMP-28 level was 2.43±2.22 ng/mL (range, 0.22-8.27 ng/mL). Plasma MMP-28 levels in patients with mild (n=24), moderate (n=31), or severe (n=24) aortic valve stenosis were 0.74 (0.25-2.23), 1.46 (0.50-3.22), and 4.13 (1.54-6.18) ng/mL, respectively, indicating that the patients with severe aortic valve stenosis had significantly higher MMP-28 levels than the patients with moderate or mild aortic valve stenosis (both P<0.01). Regression analysis using the general linear model further revealed that plasma MMP-28 level was correlated with the peak blood flow velocity and mean pressure gradient of the transaortic valve, and the correlations were statistically significant (both P<0.01). CONCLUSIONS MMP-28 level is significantly elevated in severe cases of calcific aortic valve stenosis. Moreover, plasma MMP-28 levels are positively correlated with the mean pressure gradients and peak blood flow velocity of the transaortic valve.
Collapse
Affiliation(s)
- Ke Zhou
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Ting Guo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China (mainland)
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Rong Guo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
93
|
Chen Z, Yan Y, Wu J, Qi C, Liu J, Wang J. Expression level and diagnostic value of exosomal NEAT1/miR-204/MMP-9 in acute ST-segment elevation myocardial infarction. IUBMB Life 2020; 72:2499-2507. [PMID: 32916037 DOI: 10.1002/iub.2376] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/21/2022]
Abstract
Acute myocardium infarction (AMI) is one of the main causes of cardiovascular death, and timely intervention and diagnosis are essential. Owing to the irreversible apoptosis and death of myocardial cells, which ultimately causes heart failure, the problem of myocardial repair after myocardial infarction needs to be urgently addressed. Exosomes can act as messengers between cells, delivering large amounts of proteins, RNA, and lipids to receptor cells, and regulating target cell functions. Studies have shown that exosomes can repair infarcted myocardium. We aimed to investigate the relationship between long non-coding RNA NEAT1 in serum exosomes of patients and AMI and its underlying mechanism. Subjects were divided into control, UA, and STEMI groups. RNA was extracted from the serum exosomes, and the expressions of lncRNA NEAT1 and miR-204 were detected by qRT-PCR. MMP-9 was detected by western blot, Spearman test was used to analyze the correlation among the three. Logistic regression and Receiver-operating characteristic curve (ROC) were used to evaluate the prediction of acute myocardial infarction. The expressions of NEAT1 and MMP-9 in serum exosomes of patients with acute ST-segment elevation myocardial infarction were up-regulated and positively correlated, miR-204 expression was down-regulated, there were no correlations between miR-204 with NEAT1, or MMP-9. Exosomal NEAT1, miR-204, and MMP-9 displayed potent biomarkers for diagnosis of acute ST-segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chao Qi
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
94
|
Nagata N, Ohta H, Yamada A, Teoh YB, Ichii O, Morishita K, Sasaki N, Takiguchi M. Activities of matrix metalloproteinase-2, matrix metalloproteinase-9, and serine proteases in samples of the colorectal mucosa of Miniature Dachshunds with inflammatory colorectal polyps. Am J Vet Res 2020; 81:572-580. [PMID: 32584177 DOI: 10.2460/ajvr.81.7.572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the activities of gelatinases (matrix metalloproteinase [MMP]-2 and MMP-9) and serine proteases in the colorectal mucosa of Miniature Dachshunds (MDs) with inflammatory colorectal polyps (ICRPs). ANIMALS 15 MDs with ICRPs and 5 dogs with non-ICRP-related large bowel diarrhea (controls). PROCEDURES Zymographic methods were used to evaluate the activities of MMP-2, MMP-9, latent forms of MMP-2 and MMP-9 (pro-MMP-2 and pro-MMP-9), and serine proteases in inflamed and noninflamed tissue samples from MDs with ICRPs and in noninflamed tissue samples from control dogs. The associations of serine protease activities with MMP-2 or MMP-9 activity were also analyzed. RESULTS Activities of pro-MMP-2 and pro-MMP-9 were detected in most tissue samples, regardless of the tissue type, whereas activities of MMP-2 and MMP-9 were not detected in control tissue samples. In the inflamed tissue samples from MDs with ICRPs, the activities of MMP-2, pro-MMP-9, and MMP-9 were significantly higher than those in the noninflamed tissue samples from those dogs. Serine protease activities were significantly higher in the inflamed and noninflamed tissue samples from MDs with ICRP, compared with findings for control tissue samples. A weak correlation was detected between serine protease activities and MMP-9 activity. CONCLUSIONS AND CLINICAL RELEVANCE Study results suggested that gelatinase and serine protease activities are upregulated in the colorectal mucosa of MDs with ICRPs, possibly contributing to the pathogenesis of this disease through the functions of these enzymes in degradation of extracellular matrix and promotion of inflammatory cell migration and inflammatory responses.
Collapse
|
95
|
Zipfel P, Rochais C, Baranger K, Rivera S, Dallemagne P. Matrix Metalloproteinases as New Targets in Alzheimer's Disease: Opportunities and Challenges. J Med Chem 2020; 63:10705-10725. [PMID: 32459966 DOI: 10.1021/acs.jmedchem.0c00352] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although matrix metalloproteinases (MMPs) are implicated in the regulation of numerous physiological processes, evidence of their pathological roles have also been obtained in the last decades, making MMPs attractive therapeutic targets for several diseases. Recent discoveries of their involvement in central nervous system (CNS) disorders, and in particular in Alzheimer's disease (AD), have paved the way to consider MMP modulators as promising therapeutic strategies. Over the past few decades, diverse approaches have been undertaken in the design of therapeutic agents targeting MMPs for various purposes, leading, more recently, to encouraging developments. In this article, we will present recent examples of inhibitors ranging from small molecules and peptidomimetics to biologics. We will also discuss the scientific knowledge that has led to the development of emerging tools and techniques to overcome the challenges of selective MMP inhibition.
Collapse
Affiliation(s)
- Pauline Zipfel
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Christophe Rochais
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Kévin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Patrick Dallemagne
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| |
Collapse
|
96
|
Wang JC, Chien WC, Chung CH, Lin CY, Hsu CW, Lin CS, Tsai SH. Association between surgical repair of aortic aneurysms and the diagnosis of subsequent cardiovascular diseases. J Cardiol 2020; 75:621-627. [DOI: 10.1016/j.jjcc.2019.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/25/2019] [Accepted: 12/08/2019] [Indexed: 11/27/2022]
|
97
|
Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int J Mol Sci 2020; 21:ijms21113946. [PMID: 32486345 PMCID: PMC7313469 DOI: 10.3390/ijms21113946] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases responsible for tissue remodeling and degradation of extracellular matrix (ECM) proteins. MMPs may modulate various cellular and signaling pathways in atherosclerosis responsible for progression and rupture of atherosclerotic plaques. The effect of MMPs polymorphisms and the expression of MMPs in both the atherosclerotic plaque and plasma was shown. They are independent predictors of atherosclerotic plaque instability in stable coronary heart disease (CHD) patients. Increased levels of MMPs in patients with advanced cardiovascular disease (CAD) and acute coronary syndrome (ACS) was associated with future risk of cardiovascular events. These data confirm that MMPs may be biomarkers in plaque instability as they target in potential drug therapies for atherosclerosis. They provide important prognostic information, independent of traditional risk factors, and may turn out to be useful in improving risk stratification.
Collapse
|
98
|
Circulating Matrix Metalloproteinase-28 Levels Are Related to GRACE Scores and Short-Term Outcomes in Patients with Acute Myocardial Infarction. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9206703. [PMID: 32596395 PMCID: PMC7273487 DOI: 10.1155/2020/9206703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 01/14/2023]
Abstract
Objective To investigate the relationship between the level of matrix metalloproteinase-28 (MMP-28) in patients with acute myocardial infarction (AMI) and the global registry of acute coronary events (GRACE) scores as well as their short-term prognosis. Methods Two hundred eleven patients with AMI were enrolled, and their basic clinical characteristics were collected for determining the GRACE score. We measured the plasma levels of MMP-28 and other biomarkers in the study population. The association of MMP-28 levels with cardiac events and cardiac deaths occurring within 30 days of discharge was evaluated with multivariable Cox proportional hazard models. Results The MMP-28 levels were significantly higher in patients with acute ST-elevation myocardial infarction (STEMI) than in patients with non-ST-elevation myocardial infarction (NSTEMI) (P < 0.01). Correlation analysis showed that the level of MMP-28 was positively correlated with the GRACE score in patients with AMI (R2 = 0.366, P < 0.05). Cox multivariate regression results showed that MMP-28 was associated with cardiovascular events during the hospitalization and 30 days after discharge (P < 0.01). In addition, Kaplan–Meier analysis showed that cardiac events and deaths were significantly higher in patients with MMP-28 ≥ 1.21 ng/mL (all P < 0.01). Conclusion There is a correlation between the plasma MMP-28 level and GRACE score in patients with AMI. MMP-28 is also associated with cardiovascular events and cardiovascular deaths during the hospitalization of patients and within 30 days of discharge.
Collapse
|
99
|
Effect of 5-year continuous positive airway pressure treatment on MMPs and TIMPs: implications for OSA comorbidities. Sci Rep 2020; 10:8609. [PMID: 32451401 PMCID: PMC7248085 DOI: 10.1038/s41598-020-65029-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/27/2020] [Indexed: 01/30/2023] Open
Abstract
Continuous positive airway pressure (CPAP) treatment results in nearly complete remission of symptoms of obstructive sleep apnoea (OSA); however, its effect on OSA comorbidities including cardiovascular diseases remains contradictory. Here we investigated the short- and long-term effect of CPAP treatment on matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in patients with severe OSA. Serum levels of 7 MMPs and 3 TIMPs were followed in OSA patients (n = 28) with an apnoea-hypopnoea index of ≥30 events/h at the time of diagnosis and at control visits (2 months, 6 months and 5 years) after initiation of fixed-pressure CPAP treatment. The first few months of CPAP therapy resulted in significant decrease of MMP-8 and MMP-9 levels (MMP-8: 146 (79-237) vs. 287 (170-560) pg/mL; MMP-9: 10.1 (7.1-14.1) vs. 12.7 (10.4-15.6) ng/mL, p < 0.05 for each at 2 months), while the rest of the panel remained unchanged as compared to baseline values. In contrast, at 5 years, despite of uninterrupted CPAP treatment and excellent adherence the levels of MMP-8, MMP-9 and TIMPs significantly increased (p < 0.05). Our data suggest that initiation of CPAP therapy leads to a decrease in the level of key MMPs in the short-term; however, this effect is not sustained over the long-term.
Collapse
|
100
|
do Prado AF, Bannwart CM, Shinkai VMT, de Souza Lima IM, Meschiari CA. Phyto-derived Products as Matrix Metalloproteinases Inhibitors in Cardiovascular Diseases. Curr Hypertens Rev 2020; 17:47-58. [PMID: 32386496 DOI: 10.2174/1573402116666200510011356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/22/2020] [Accepted: 03/09/2020] [Indexed: 11/22/2022]
Abstract
Matrix metalloproteinases (MMPs) are enzymes that present a metallic element in their structure. These enzymes are ubiquitously distributed and function as extracellular matrix (ECM) remodelers. MMPs play a broad role in cardiovascular biology regulating processes such as cell adhesion and function, cellular communication and differentiation, integration of mechanical force and force transmission, tissue remodeling, modulation of damaged-tissue structural integrity, cellular survival or apoptosis and regulation of inflammation-related cytokines and growth factors. MMPs inhibition and downregulation are correlated with minimization of cardiac damage, i.e., Chinese herbal medicine has shown to stabilize abdominal aorta aneurysm due to its antiinflammatory, antioxidant and MMP-2 and 9 inhibitory properties. Thus phyto-derived products rise as promising sources for novel therapies focusing on MMPs inhibition and downregulation to treat or prevent cardiovascular disorders.
Collapse
Affiliation(s)
- Alejandro F do Prado
- Structural Biology Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Cahy M Bannwart
- Structural Biology Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Victoria M T Shinkai
- Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | | | - César A Meschiari
- Health and Sports Science Center, Federal University of Acre, Rio Branco, AC, Brazil
| |
Collapse
|