1
|
Nakahara T, Fujimoto S, Jinzaki M. Molecular imaging of cardiovascular disease: Current status and future perspective. J Cardiol 2025:S0914-5087(25)00017-6. [PMID: 39922562 DOI: 10.1016/j.jjcc.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/15/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Advancements in knowledge of cardiovascular disease, pharmacology, and chemistry have led to the development of newer radiopharmaceuticals and targets for new and more suitable molecules. Molecular imaging encompasses multiple imaging techniques for identifying the characteristics of key components involved in disease. Despite its limitations in spatial resolution, the affinity for key molecules compensates for disadvantages in diagnosing diseases and elucidating their pathophysiology. This review introduce established molecular tracers involved in clinical practice and emerging tracers already applied in clinical studies, classifying the key component in A: artery, specifically those vulnerable plaque (A-I) inflammatory cells [18F-FDG]; A-II) lipid/fatty acid; A-III) hypoxia; A-IV) angiogenesis; A-V) protease [18F/68Ga-FAPI]; A-VI) thrombus/hemorrhage; A-VII) apoptosis and A-VIII) microcalcification [18F-NaF]) and B: myocardium, including myocardial ischemia, infarction and myocardiopathy (B-I) myocardial ischemia; B-II) myocardial infarction (myocardial damage and fibrosis); B-III) myocarditis and endocarditis; B-IV) sarcoidosis; B-V) amyloidosis; B-VI) metabolism; B-VII) innervation imaging). In addition to cardiovascular-specific tracers tested in animal models, many radiotracers may have been developed in other areas, such as oncology imaging or neuroimaging. While this review does not cover all available tracers, some of them hold potential for future use assessing cardiovascular disease. Advances in molecular biology, pharmaceuticals, and imaging sciences will facilitate the identification of precise disease mechanisms, enabling precise diagnoses, better assessment of disease status, and enhanced therapeutic evaluation in this multi-modality era.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - Shinichiro Fujimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Higashi T, Hashimoto K, Mai Y, Naganuma F, Yoshikawa T. Cystine transporter SLC7A11 regulates sensitivity to unsaturated carbonyl compounds in mouse macrophage cell lines. J Pharmacol Sci 2025; 157:96-103. [PMID: 39828398 DOI: 10.1016/j.jphs.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025] Open
Abstract
Cytotoxic effects of cigarette smoke are thought to be causes of cigarette smoking-related diseases such as respiratory infection, chronic obstructive pulmonary disease, and atherosclerosis. Unsaturated carbonyl compounds are major cytotoxic factors in the gas phase of cigarette smoke. Cell death induced by unsaturated carbonyl compounds in cigarette smoke is PKC-dependent ferroptosis. Although the cell sensitivity to unsaturated carbonyl compounds varies by cell types, the molecular mechanisms underlying this sensitivity remain unclear. In this study, we have examined the factors involved in determining sensitivity to unsaturated carbonyl compounds. We found that two mouse macrophage cell lines exhibited different sensitivities; J774.1 macrophages were sensitive to unsaturated carbonyl compounds, whereas RAW264.7 macrophages were resistant. Glutathione synthesis inhibitor increased the sensitivity of RAW264.7 macrophages to unsaturated carbonyl compounds. Quantitative RT-PCR revealed that the expression level of the cystine transporter SLC7A11 was higher in RAW264.7 macrophages than in J774.1 macrophages. Inhibition of SLC7A11 activity increased sensitivity to unsaturated carbonyl compounds, while overexpression of SLC7A11 enhances resistance to these compounds. The current results suggest that the SLC7A11 level is a key factor in determining the macrophage sensitivity to unsaturated carbonyl compounds.
Collapse
Affiliation(s)
- Tsunehito Higashi
- Department of Cellular Pharmacology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Konoka Hashimoto
- Department of Cellular Pharmacology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Yosuke Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Fumito Naganuma
- Department of Neuropharmacology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Takeo Yoshikawa
- Department of Cellular Pharmacology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan; Department of Neuropharmacology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Nishi 7, Kita 15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
3
|
Cheng WH, Wang Y. Inflammatory Pathways in Coronary Artery Disease: Which Ones to Target for Secondary Prevention? Cells 2025; 14:153. [PMID: 39936945 PMCID: PMC11817712 DOI: 10.3390/cells14030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Coronary artery disease (CAD), the build-up of atherosclerotic plaques on the wall of blood vessels, causes adverse cardiovascular events. Secondary prevention focuses on treating patients with existing plaques to prevent disease progression. Recent studies have shown that inflammation is an independent risk factor that drives disease progression, and targeting inflammation could be an effective therapeutic strategy for secondary prevention. In this review, we highlighted the roles of several inflammatory pathways in rupture and erosion, two major processes through which established plaques lead to adverse cardiovascular events. In the past 15 years, numerous clinical trials have tested the therapeutic potential of targeting these pathways, including neutralizing inflammatory cytokines and blocking signaling transduction of the inflammatory pathways. Only colchicine was approved for clinical use in patients with CAD. This is primarily due to the multifaceted roles of inflammatory pathways in disease progression. Commonly used pre-clinical models provided robust information for the onset of early disease but limited understanding of the inflammatory network in established plaques. This review will summarize lessons learned from successful and failed clinical trials to advocate for assessing inflammation in established plaques before designing therapeutics for secondary prevention.
Collapse
Affiliation(s)
- Wan-Hei Cheng
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
4
|
Pervaiz N, Mehmood R, Aithabathula RV, Kathuria I, Ahn W, Le BT, Kim KS, Singh UP, Csanyi G, Singla B. Smooth muscle cell-specific CD47 deletion suppresses atherosclerosis. Life Sci 2025; 361:123315. [PMID: 39675550 PMCID: PMC11740882 DOI: 10.1016/j.lfs.2024.123315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Recent smooth muscle cell (SMC)-lineage tracing and single-cell RNA sequencing (scRNA-seq) experiments revealed a significant role of SMC-derived cells in atherosclerosis development. Further, thrombospondin-1 (TSP1), a matricellular protein, and activation of its receptor cluster of differentiation (CD) 47 have been linked with atherosclerosis. However, the role of vascular SMC TSP1-CD47 signaling in regulating VSMC phenotype and atherogenesis remains unknown. METHODS We investigated the role of SMC CD47 activation by TSP1 in regulating VSMC phenotype and atherosclerosis development using various in vitro cell-based assays, molecular biological techniques, immunohistological approaches, reanalysis of publicly available scRNA-seq data, and cell-specific knockout mice. RESULTS We observed elevated TSP1 expression in human atherosclerotic vascular tissues and VSMCs. TSP1-treated VSMCs exhibited decreased expression of contractile SMC markers (ACTA2, CNN1, and TAGLN) and increased proliferation. Additional experiments and reanalysis of the scRNA-seq dataset showed CD47 as the major TSP1 receptor in VSMCs, with its expression increased in SMC-derived modulated cells of murine atherosclerotic arteries. Knockdown of CD47 gene in human VSMCs upregulated expression of contractile SMC markers and abrogated TSP1's effects on these genes. SMC-specific Cd47 deletion in mice suppressed atherosclerotic lesion formation, reduced macrophage accumulation, and decreased necrotic area. However, no significant differences were observed in weight gain, liver and adipose tissue mass, plasma total cholesterol, and fasting blood glucose between control and SMC-restricted Cd47-deficient mice. Further experiments demonstrated increased efferocytosis of apoptotic CD47-silenced VSMCs by macrophages. CONCLUSIONS These findings suggest that CD47 plays a crucial role in regulating VSMC phenotype, and SMC-specific-Cd47 deletion suppresses atherosclerosis. NEW AND NOTEWORTHY VSMC phenotypic switching contributes to atherosclerosis development. The present study reports the novel observations that Cd47 levels are upregulated in phenotypically modulated SMCs within atherosclerotic arteries and targeted deletion of Cd47 specifically in SMCs attenuates atherosclerosis. Mechanistic in vitro investigations further showed that TSP1-CD47 signaling regulates VSMC phenotype. Therefore, targeting SMC CD47 represents a promising therapeutic target to suppress atherogenesis.
Collapse
Affiliation(s)
- Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rashid Mehmood
- Department of Hematology, St. Jude Children's Hospital, Memphis, TN, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - WonMo Ahn
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Britney-Thuy Le
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ki-Suk Kim
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gabor Csanyi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
5
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it commonly results from atherosclerotic plaque progression. One of the increasingly recognized drivers of atherosclerosis is dysfunctional efferocytosis, a homeostatic mechanism responsible for the clearance of dead cells and the resolution of inflammation. In atherosclerosis, the capacity of phagocytes to participate in efferocytosis is hampered, leading to the accumulation of apoptotic and necrotic tissue within the plaque, which results in enlargement of the necrotic core, increased luminal stenosis and plaque inflammation, and predisposition to plaque rupture or erosion. In this Review, we describe the different forms of programmed cell death that can occur in the atherosclerotic plaque and highlight the efferocytic machinery that is normally implicated in cardiovascular physiology. We then discuss the mechanisms by which efferocytosis fails in atherosclerosis and other cardiovascular and cardiometabolic diseases, including myocardial infarction and diabetes mellitus, and discuss therapeutic approaches that might reverse this pathological process.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
| |
Collapse
|
6
|
Niida T, Usui E, Suzuki K, Kinoshita D, Yuki H, Fujimoto D, Covani M, Dey D, Lee H, McNulty I, Ferencik M, Yonetsu T, Kakuta T, Jang IK. Differences in total plaque burden between plaque rupture and plaque erosion: A combined computed tomography angiography and optical coherence tomography study. J Cardiovasc Comput Tomogr 2024; 18:568-574. [PMID: 39322513 DOI: 10.1016/j.jcct.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/06/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUNDS Coronary computed tomography angiography (CTA) allows for the assessment of atherosclerotic plaque burden across the entire coronary vasculature. No studies have examined the relationship between the underlying pathology of the culprit lesion and total plaque burden in patients with acute coronary syndromes. The aim of this study was to compare the total plaque burden between patients with plaque rupture versus plaque erosion. METHODS A total of 232 patients who presented with their first non-ST-segment elevation acute coronary syndrome and underwent both CTA and optical coherence tomography imaging before intervention were selected. Quantitative analysis was performed using semi-automated software (Autoplaque version 3.0, Cedars-Sinai Medical Center). An attenuation of <30 Hounsfield units defined low-density non-calcified plaque (LDNCP). All 3 vessels were assessed using the modified 17-segment American Heart Association model for coronary segment classification. RESULTS Among 232 patients, 125 (53.9%) had plaque rupture and 107 (46.1%) had plaque erosion. Total plaque burden (48.2 [39.8-54.9] % vs. 44.1 [38.6-50.0] %, P = 0.006), total non-calcified plaque (NCP) burden (46.6 [39.1-53.3] % vs. 43.0 [37.6-49.2] %, P = 0.013), total LDNCP burden (2.3 [1.4-3.0] % vs. 1.7 [1.2-2.6] %, P = 0.016), and total calcified plaque (CP) burden (0.8 [0.1-1.6] % vs. 0.4 [0.0-1.4] %, P = 0.047) were significantly greater in patients with culprit plaque rupture than in those with culprit plaque erosion. CONCLUSION Patients with plaque rupture, compared with those with plaque erosion, had a greater total plaque burden, NCP burden, LDNCP burden, and CP burden. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04523194.
Collapse
Affiliation(s)
- Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eisuke Usui
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daichi Fujimoto
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Covani
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Taishi Yonetsu
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Mori M, Sakamoto A, Kawakami R, Guo L, Slenders L, Mosquera JV, Ghosh SKB, Wesseling M, Shiraki T, Bellissard A, Shah P, Weinkauf CC, Konishi T, Sato Y, Cornelissen A, Kawai K, Jinnouchi H, Xu W, Vozenilek AE, Williams D, Tanaka T, Sekimoto T, Kelly MC, Fernandez R, Grogan A, Coslet AJ, Fedotova A, Kurse A, Mokry M, Romero ME, Kolodgie FD, Pasterkamp G, Miller CL, Virmani R, Finn AV. CD163 + Macrophages Induce Endothelial-to-Mesenchymal Transition in Atheroma. Circ Res 2024; 135:e4-e23. [PMID: 38860377 DOI: 10.1161/circresaha.123.324082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Cell phenotype switching is increasingly being recognized in atherosclerosis. However, our understanding of the exact stimuli for such cellular transformations and their significance for human atherosclerosis is still evolving. Intraplaque hemorrhage is thought to be a major contributor to plaque progression in part by stimulating the influx of CD163+ macrophages. Here, we explored the hypothesis that CD163+ macrophages cause plaque progression through the induction of proapoptotic endothelial-to-mesenchymal transition (EndMT) within the fibrous cap. METHODS Human coronary artery sections from CVPath's autopsy registry were selected for pathological analysis. Athero-prone ApoE-/- and ApoE-/-/CD163-/- mice were used for in vivo studies. Human peripheral blood mononuclear cell-induced macrophages and human aortic endothelial cells were used for in vitro experiments. RESULTS In 107 lesions with acute coronary plaque rupture, 55% had pathological evidence of intraplaque hemorrhage in nonculprit vessels/lesions. Thinner fibrous cap, greater CD163+ macrophage accumulation, and a larger number of CD31/FSP-1 (fibroblast specific protein-1) double-positive cells and TUNEL (terminal deoxynucleotidyl transferase-dUTP nick end labeling) positive cells in the fibrous cap were observed in nonculprit intraplaque hemorrhage lesions, as well as in culprit rupture sections versus nonculprit fibroatheroma sections. Human aortic endothelial cells cultured with supernatants from hemoglobin/haptoglobin-exposed macrophages showed that increased mesenchymal marker proteins (transgelin and FSP-1) while endothelial markers (VE-cadherin and CD31) were reduced, suggesting EndMT induction. Activation of NF-κB (nuclear factor kappa β) signaling by proinflammatory cytokines released from CD163+ macrophages directly regulated the expression of Snail, a critical transcription factor during EndMT induction. Western blot analysis for cleaved caspase-3 and microarray analysis of human aortic endothelial cells indicated that apoptosis was stimulated during CD163+ macrophage-induced EndMT. Additionally, CD163 deletion in athero-prone mice suggested that CD163 is required for EndMT and plaque progression. Using single-cell RNA sequencing from human carotid endarterectomy lesions, a population of EndMT was detected, which demonstrated significant upregulation of apoptosis-related genes. CONCLUSIONS CD163+ macrophages provoke EndMT, which may promote plaque progression through fibrous cap thinning.
Collapse
MESH Headings
- Humans
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Animals
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Macrophages/metabolism
- Macrophages/pathology
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Mice
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Male
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Apoptosis
- Female
- Epithelial-Mesenchymal Transition
- Coronary Vessels/pathology
- Coronary Vessels/metabolism
Collapse
Affiliation(s)
- Masayuki Mori
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Atsushi Sakamoto
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- Hamamatsu University School of Medicine, Shizuoka, Japan (A.S.)
| | - Rika Kawakami
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Liang Guo
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Lotte Slenders
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Jose Verdezoto Mosquera
- Department of Public Health Sciences, Department of Biochemistry and Molecular Genetics, Center for Public Health Genomics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Saikat Kumar B Ghosh
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Marian Wesseling
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Tatsuya Shiraki
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Arielle Bellissard
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Palak Shah
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | | | - Takao Konishi
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Yu Sato
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Anne Cornelissen
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Kenji Kawai
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Weili Xu
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Aimee E Vozenilek
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Desiree Williams
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Takamasa Tanaka
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Teruo Sekimoto
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Raquel Fernandez
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Alyssa Grogan
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - A J Coslet
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Alisa Fedotova
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Anjali Kurse
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Michal Mokry
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Maria E Romero
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Frank D Kolodgie
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Gerard Pasterkamp
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Clint L Miller
- Department of Public Health Sciences, Department of Biochemistry and Molecular Genetics, Center for Public Health Genomics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- University of Maryland School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
8
|
Zhou ZY, Wu L, Liu YF, Tang MY, Tang JY, Deng YQ, Liu L, Nie BB, Zou ZK, Huang L. IRE1α: from the function to the potential therapeutic target in atherosclerosis. Mol Cell Biochem 2024; 479:1079-1092. [PMID: 37310588 DOI: 10.1007/s11010-023-04780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023]
Abstract
Inositol requiring enzyme 1 (IRE1) is generally thought to control the most conserved pathway in the unfolded protein response (UPR). Two isoforms of IRE1, IRE1α and IRE1β, have been reported in mammals. IRE1α is a ubiquitously expressed protein whose knockout shows marked lethality. In contrast, the expression of IRE1β is exclusively restricted in the epithelial cells of the respiratory and gastrointestinal tracts, and IRE1β-knockout mice are phenotypically normal. As research continues to deepen, IRE1α was showed to be tightly linked to inflammation, lipid metabolism regulation, cell death and so on. Growing evidence also suggests an important role for IRE1α in promoting atherosclerosis (AS) progression and acute cardiovascular events through disrupting lipid metabolism balance, facilitating cells apoptosis, accelerating inflammatory responses and promoting foam cell formation. In addition, IRE1α was recognized as novel potential therapeutic target in AS prevention. This review provides some clues about the relationship between IRE1α and AS, hoping to contribute to further understanding roles of IRE1α in atherogenesis and to be helpful for the design of novel efficacious therapeutics agents targeting IRE1α-related pathways.
Collapse
Affiliation(s)
- Zheng-Yang Zhou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Li Wu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yi-Fan Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mu-Yao Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jing-Yi Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Anaesthesiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Ya-Qian Deng
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Lei Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bin-Bin Nie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zi-Kai Zou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Liang Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
9
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Jia Y, Cheng L, Yang J, Mao J, Xie Y, Yang X, Zhang X, Wang D, Zhao Z, Schober A, Wei Y. miR-223-3p Prevents Necroptotic Macrophage Death by Targeting Ripk3 in a Negative Feedback Loop and Consequently Ameliorates Advanced Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:218-237. [PMID: 37970714 DOI: 10.1161/atvbaha.123.319776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND The formation of large necrotic cores results in vulnerable atherosclerotic plaques, which can lead to severe cardiovascular diseases. However, the specific regulatory mechanisms underlying the development of necrotic cores remain unclear. METHODS To evaluate how the modes of lesional cell death are reprogrammed during the development of atherosclerosis, the expression levels of key proteins that are involved in the necroptotic, apoptotic, and pyroptotic pathways were compared between different stages of plaques in humans and mice. Luciferase assays and loss-of-function studies were performed to identify the microRNA-mediated regulatory mechanism that protects foamy macrophages from necroptotic cell death. The role of this mechanism in atherosclerosis was determined by using a knockout mouse model with perivascular drug administration and tail vein injection of microRNA inhibitors in Apoe-/- mice. RESULTS Here, we demonstrate that the necroptotic, rather than the apoptotic or pyroptotic, pathway is more activated in advanced unstable plaques compared with stable plaques in both humans and mice, which closely correlates with necrotic core formation. The upregulated expression of Ripk3 (receptor-interacting protein kinase 3) promotes the C/EBPβ (CCAAT/enhancer binding protein beta)-dependent transcription of the microRNA miR-223-3p, which conversely inhibits Ripk3 expression and forms a negative feedback loop to regulate the necroptosis of foamy macrophages. The knockout of the Mir223 gene in bone marrow cells accelerates atherosclerosis in Apoe-/- mice, but this effect can be rescued by Ripk3 deficiency or treatment with the necroptosis inhibitors necrostatin-1 and GSK-872. Like the Mir223 knockout, treating Apoe-/- mice with miR-223-3p inhibitors increases atherosclerosis. CONCLUSIONS Our study suggests that miR-223-3p expression in macrophages protects against atherosclerotic plaque rupture by limiting the formation of necrotic cores, thus providing a potential microRNA therapeutic candidate for atherosclerosis.
Collapse
Affiliation(s)
- Yunhui Jia
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Lianping Cheng
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Jiaxuan Yang
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Jiaqi Mao
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Yuhuai Xie
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Xian Yang
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Xin Zhang
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Dingxin Wang
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
| | - Zhen Zhao
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, China (Z.Z.)
- Vascular Center of Shanghai Jiaotong University, China (Z.Z.)
| | - Andreas Schober
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
- Experimental Vascular Medicine (EVM), Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Germany (A.S.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Germany (A.S.)
| | - Yuanyuan Wei
- Department of Immunology, School of Basic Medical Sciences, and Department of Rheumatology, Zhongshan Hospital (Y.J., L.C., J.Y., J.M., Y.X., X.Y., X.Z., D.W., Y.W.), Fudan University, China
- Shanghai Key Laboratory of Bioactive Small Molecules and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences (Y.W.), Fudan University, China
| |
Collapse
|
11
|
Hajibabaie F, Abedpoor N, Mohamadynejad P. Types of Cell Death from a Molecular Perspective. BIOLOGY 2023; 12:1426. [PMID: 37998025 PMCID: PMC10669395 DOI: 10.3390/biology12111426] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
The former conventional belief was that cell death resulted from either apoptosis or necrosis; however, in recent years, different pathways through which a cell can undergo cell death have been discovered. Various types of cell death are distinguished by specific morphological alterations in the cell's structure, coupled with numerous biological activation processes. Various diseases, such as cancers, can occur due to the accumulation of damaged cells in the body caused by the dysregulation and failure of cell death. Thus, comprehending these cell death pathways is crucial for formulating effective therapeutic strategies. We focused on providing a comprehensive overview of the existing literature pertaining to various forms of cell death, encompassing apoptosis, anoikis, pyroptosis, NETosis, ferroptosis, autophagy, entosis, methuosis, paraptosis, mitoptosis, parthanatos, necroptosis, and necrosis.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| | - Navid Abedpoor
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| |
Collapse
|
12
|
Sahoo G, Samal D, Khandayataray P, Murthy MK. A Review on Caspases: Key Regulators of Biological Activities and Apoptosis. Mol Neurobiol 2023; 60:5805-5837. [PMID: 37349620 DOI: 10.1007/s12035-023-03433-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Caspases are proteolytic enzymes that belong to the cysteine protease family and play a crucial role in homeostasis and programmed cell death. Caspases have been broadly classified by their known roles in apoptosis (caspase-3, caspase-6, caspase-7, caspase-8, and caspase-9 in mammals) and in inflammation (caspase-1, caspase-4, caspase-5, and caspase-12 in humans, and caspase-1, caspase-11, and caspase-12 in mice). Caspases involved in apoptosis have been subclassified by their mechanism of action as either initiator caspases (caspase-8 and caspase-9) or executioner caspases (caspase-3, caspase-6, and caspase-7). Caspases that participate in apoptosis are inhibited by proteins known as inhibitors of apoptosis (IAPs). In addition to apoptosis, caspases play a role in necroptosis, pyroptosis, and autophagy, which are non-apoptotic cell death processes. Dysregulation of caspases features prominently in many human diseases, including cancer, autoimmunity, and neurodegenerative disorders, and increasing evidence shows that altering caspase activity can confer therapeutic benefits. This review covers the different types of caspases, their functions, and their physiological and biological activities and roles in different organisms.
Collapse
Affiliation(s)
- Gayatri Sahoo
- Department of Zoology, PSSJ College, Banarpal, 759128, Odisha, India
| | - Dibyaranjan Samal
- Department of Biotechnology, Academy of Management and Information Technology (AMIT, affiliated to Utkal University), Khurda, 752057, Odisha, India
| | | | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
13
|
Yarovinsky TO, Su M, Chen C, Xiang Y, Tang WH, Hwa J. Pyroptosis in cardiovascular diseases: Pumping gasdermin on the fire. Semin Immunol 2023; 69:101809. [PMID: 37478801 PMCID: PMC10528349 DOI: 10.1016/j.smim.2023.101809] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/13/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023]
Abstract
Pyroptosis is a form of programmed cell death associated with activation of inflammasomes and inflammatory caspases, proteolytic cleavage of gasdermin proteins (forming pores in the plasma membrane), and selective release of proinflammatory mediators. Induction of pyroptosis results in amplification of inflammation, contributing to the pathogenesis of chronic cardiovascular diseases such as atherosclerosis and diabetic cardiomyopathy, and acute cardiovascular events, such as thrombosis and myocardial infarction. While engagement of pyroptosis during sepsis-induced cardiomyopathy and septic shock is expected and well documented, we are just beginning to understand pyroptosis involvement in the pathogenesis of cardiovascular diseases with less defined inflammatory components, such as atrial fibrillation. Due to the danger that pyroptosis represents to cells within the cardiovascular system and the whole organism, multiple levels of pyroptosis regulation have evolved. Those include regulation of inflammasome priming, post-translational modifications of gasdermins, and cellular mechanisms for pore removal. While pyroptosis in macrophages is well characterized as a dramatic pro-inflammatory process, pyroptosis in other cell types within the cardiovascular system displays variable pathways and consequences. Furthermore, different cells and organs engage in local and distant crosstalk and exchange of pyroptosis triggers (oxidized mitochondrial DNA), mediators (IL-1β, S100A8/A9) and antagonists (IL-9). Development of genetic tools, such as Gasdermin D knockout animals, and small molecule inhibitors of pyroptosis will not only help us fully understand the role of pyroptosis in cardiovascular diseases but may result in novel therapeutic approaches inhibiting inflammation and progression of chronic cardiovascular diseases to reduce morbidity and mortality from acute cardiovascular events.
Collapse
Affiliation(s)
- Timur O Yarovinsky
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Meiling Su
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Chaofei Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Yaozu Xiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Kowloon, the Hong Kong Special Administrative Region of China
| | - John Hwa
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Neels JG, Gollentz C, Chinetti G. Macrophage death in atherosclerosis: potential role in calcification. Front Immunol 2023; 14:1215612. [PMID: 37469518 PMCID: PMC10352763 DOI: 10.3389/fimmu.2023.1215612] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Cell death is an important aspect of atherosclerotic plaque development. Insufficient efferocytosis of death cells by phagocytic macrophages leads to the buildup of a necrotic core that impacts stability of the plaque. Furthermore, in the presence of calcium and phosphate, apoptotic bodies resulting from death cells can act as nucleation sites for the formation of calcium phosphate crystals, mostly in the form of hydroxyapatite, which leads to calcification of the atherosclerotic plaque, further impacting plaque stability. Excessive uptake of cholesterol-loaded oxidized LDL particles by macrophages present in atherosclerotic plaques leads to foam cell formation, which not only reduces their efferocytosis capacity, but also can induce apoptosis in these cells. The resulting apoptotic bodies can contribute to calcification of the atherosclerotic plaque. Moreover, other forms of macrophage cell death, such as pyroptosis, necroptosis, parthanatos, and ferroptosis can also contribute by similar mechanisms to plaque calcification. This review focuses on macrophage death in atherosclerosis, and its potential role in calcification. Reducing macrophage cell death and/or increasing their efferocytosis capacity could be a novel therapeutic strategy to reduce the formation of a necrotic core and calcification and thereby improving atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Jaap G. Neels
- Université Côte d’Azur, Institut national de la santé et de la recherche médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Claire Gollentz
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Giulia Chinetti
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| |
Collapse
|
15
|
Galindo CL, Khan S, Zhang X, Yeh YS, Liu Z, Razani B. Lipid-laden foam cells in the pathology of atherosclerosis: shedding light on new therapeutic targets. Expert Opin Ther Targets 2023; 27:1231-1245. [PMID: 38009300 PMCID: PMC10843715 DOI: 10.1080/14728222.2023.2288272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
INTRODUCTION Lipid-laden foam cells within atherosclerotic plaques are key players in all phases of lesion development including its progression, necrotic core formation, fibrous cap thinning, and eventually plaque rupture. Manipulating foam cell biology is thus an attractive therapeutic strategy at early, middle, and even late stages of atherosclerosis. Traditional therapies have focused on prevention, especially lowering plasma lipid levels. Despite these interventions, atherosclerosis remains a major cause of cardiovascular disease, responsible for the largest numbers of death worldwide. AREAS COVERED Foam cells within atherosclerotic plaques are comprised of macrophages, vascular smooth muscle cells, and other cell types which are exposed to high concentrations of lipoproteins accumulating within the subendothelial intimal layer. Macrophage-derived foam cells are particularly well studied and have provided important insights into lipid metabolism and atherogenesis. The contributions of foam cell-based processes are discussed with an emphasis on areas of therapeutic potential and directions for drug development. EXERT OPINION As key players in atherosclerosis, foam cells are attractive targets for developing more specific, targeted therapies aimed at resolving atherosclerotic plaques. Recent advances in our understanding of lipid handling within these cells provide insights into how they might be manipulated and clinically translated to better treat atherosclerosis.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Saifur Khan
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Yu-Sheng Yeh
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Ziyang Liu
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
| |
Collapse
|
16
|
Giannarelli C. Single-Point Vulnerabilities in Atherosclerotic Plaque. J Am Coll Cardiol 2023; 81:2228-2230. [PMID: 37286251 DOI: 10.1016/j.jacc.2023.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 06/09/2023]
Affiliation(s)
- Chiara Giannarelli
- Department of Medicine, Division of Cardiology, New York University (NYU) Cardiovascular Research Center, New York, New York, USA; Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA.
| |
Collapse
|
17
|
Son H, Choi HS, Baek SE, Kim YH, Hur J, Han JH, Moon JH, Lee GS, Park SG, Woo CH, Eo SK, Yoon S, Kim BS, Lee D, Kim K. Shear stress induces monocyte/macrophage-mediated inflammation by upregulating cell-surface expression of heat shock proteins. Biomed Pharmacother 2023; 161:114566. [PMID: 36963359 DOI: 10.1016/j.biopha.2023.114566] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
The loss of endothelial cells is associated with the accumulation of monocytes/macrophages underneath the surface of the arteries, where cells are prone to mechanical stimulation, such as shear stress. However, the impact of mechanical stimuli on monocytic cells remains unclear. To assess whether mechanical stress affects monocytic cell function, we examined the expression of inflammatory molecules and surface proteins, whose levels changed following shear stress in human THP-1 cells. Shear stress increased the inflammatory chemokine CCL2, which enhanced the migration of monocytic cells and tumor necrosis factor (TNF)-α and interleukin (IL)- 1β at transcriptional and protein levels. We identified that the surface levels of heat shock protein 70 (HSP70), HSP90, and HSP105 increased using mass spectrometry-based proteomics, which was confirmed by western blot analysis, flow cytometry, and immunofluorescence. Treatment with HSP70/HSP105 and HSP90 inhibitors suppressed the expression and secretion of CCL2 and monocytic cell migration, suggesting an association between HSPs and inflammatory responses. We also demonstrated the coexistence and colocalization of increased HSP90 immunoreactivity and CD68 positive cells in atherosclerotic plaques of ApoE deficient mice fed a high-fat diet and human femoral artery endarterectomy specimens. These results suggest that monocytes/macrophages affected by shear stress polarize to a pro-inflammatory phenotype and increase surface protein levels involved in inflammatory responses. The regulation of the abovementioned HSPs upregulated on the monocytes/macrophages surface may serve as a novel therapeutic target for inflammation due to shear stress.
Collapse
Affiliation(s)
- Hyojae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Seon Choi
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seung Eun Baek
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yun-Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Department of Bioinformatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jin Hur
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jung-Hwa Han
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.
| | - Chang-Hoon Woo
- Department of Pharmacology, Yeungnam University College of Medicine, Daegu 49415, Republic of Korea
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea.
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
18
|
Kluck GE, Qian AS, Sakarya EH, Quach H, Deng YD, Trigatti BL. Apolipoprotein A1 Protects Against Necrotic Core Development in Atherosclerotic Plaques: PDZK1-Dependent High-Density Lipoprotein Suppression of Necroptosis in Macrophages. Arterioscler Thromb Vasc Biol 2023; 43:45-63. [PMID: 36353992 PMCID: PMC9762725 DOI: 10.1161/atvbaha.122.318062] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic disease affecting artery wall and a major contributor to cardiovascular diseases. Large necrotic cores increase risk of plaque rupture leading to thrombus formation. Necrotic cores are rich in debris from dead macrophages. Programmed necrosis (necroptosis) contributes to necrotic core formation. HDL (high-density lipoprotein) exerts direct atheroprotective effects on different cells within atherosclerotic plaques. Some of these depend on the SR-B1 (scavenger receptor class B type I) and the adapter protein PDZK1 (postsynaptic density protein/Drosophila disc-large protein/Zonula occludens protein containing 1). However, a role for HDL in protecting against necroptosis and necrotic core formation in atherosclerosis is not completely understood. METHODS Low-density lipoprotein receptor-deficient mice engineered to express different amounts of ApoA1 (apolipoprotein A1), or to lack PDZK1 were fed a high fat diet for 10 weeks. Atherosclerotic plaque areas, necrotic cores, and key necroptosis mediators, RIPK3 (receptor interacting protein kinase 3), and MLKL (mixed lineage kinase domain-like protein) were characterized. Cultured macrophages were treated with HDL to determine its effects, as well as the roles of SR-B1, PDZK1, and the PI3K (phosphoinositide 3-kinase) signaling pathway on necroptotic cell death. RESULTS Genetic overexpression reduced, and ApoA1 knockout increased necrotic core formation and RIPK3 and MLKL within atherosclerotic plaques. Macrophages were protected against necroptosis by HDL and this protection required SR-B1, PDZK1, and PI3K/Akt pathway. PDZK1 knockout increased atherosclerosis in LDLRKO mice, increasing necrotic cores and phospho-MLKL; both of which were reversed by restoring PDZK1 in BM-derived cells. CONCLUSIONS Our findings demonstrate that HDL in vitro and ApoA1, in vivo, protect against necroptosis in macrophages and necrotic core formation in atherosclerosis, suggesting a pathway that could be a target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- George E.G. Kluck
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Alexander S. Qian
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Emmanuel H. Sakarya
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Henry Quach
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Yak D. Deng
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, and Hamilton Health Sciences, Ontario, Canada
| |
Collapse
|
19
|
Colombo G, Altomare A, Astori E, Landoni L, Garavaglia ML, Rossi R, Giustarini D, Lionetti MC, Gagliano N, Milzani A, Dalle-Donne I. Effects of Physiological and Pathological Urea Concentrations on Human Microvascular Endothelial Cells. Int J Mol Sci 2022; 24:ijms24010691. [PMID: 36614132 PMCID: PMC9821335 DOI: 10.3390/ijms24010691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 01/03/2023] Open
Abstract
Urea is the uremic toxin accumulating with the highest concentration in the plasma of chronic kidney disease (CKD) patients, not being completely cleared by dialysis. Urea accumulation is reported to exert direct and indirect side effects on the gastrointestinal tract, kidneys, adipocytes, and cardiovascular system (CVS), although its pathogenicity is still questioned since studies evaluating its side effects lack homogeneity. Here, we investigated the effects of physiological and pathological urea concentrations on a human endothelial cell line from the microcirculation (Human Microvascular Endothelial Cells-1, HMEC-1). Urea (5 g/L) caused a reduction in the proliferation rate after 72 h of exposure and appeared to be a potential endothelial-to-mesenchymal transition (EndMT) stimulus. Moreover, urea induced actin filament rearrangement, a significant increase in matrix metalloproteinases 2 (MMP-2) expression in the medium, and a significant up- or down-regulation of other EndMT biomarkers (keratin, fibrillin-2, and collagen IV), as highlighted by differential proteomic analysis. Among proteins whose expression was found to be significantly dysregulated following exposure of HMEC-1 to urea, dimethylarginine dimethylaminohydrolase (DDAH) and vasorin turned out to be down-regulated. Both proteins have been directly linked to cardiovascular diseases (CVD) by in vitro and in vivo studies. Future experiments will be needed to deepen their role and investigate the signaling pathways in which they are involved to clarify the possible link between CKD and CVD.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Emanuela Astori
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Lucia Landoni
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Maria Lisa Garavaglia
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018–2022), University of Siena, 53100 Siena, Italy
| | - Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018–2022), University of Siena, 53100 Siena, Italy
| | - Maria Chiara Lionetti
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Aldo Milzani
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
| | - Isabella Dalle-Donne
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, 20133 Milan, Italy
- Correspondence:
| |
Collapse
|
20
|
Xu XD, Chen JX, Zhu L, Xu ST, Jiang J, Ren K. The emerging role of pyroptosis-related inflammasome pathway in atherosclerosis. Mol Med 2022; 28:160. [PMID: 36544112 PMCID: PMC9773468 DOI: 10.1186/s10020-022-00594-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis (AS), a chronic sterile inflammatory disorder, is one of the leading causes of mortality worldwide. The dysfunction and unnatural death of plaque cells, including vascular endothelial cells (VEC), macrophages, and vascular smooth muscle cells (VSMC), are crucial factors in the progression of AS. Pyroptosis was described as a form of cell death at least two decades ago. It is featured by plasma membrane swelling and rupture, cell lysis, and consequent robust release of cytosolic contents and pro-inflammatory mediators, including interleukin-1β (IL-1β), IL-18, and high mobility group box 1 (HMGB1). Pyroptosis of plaque cells is commonly observed in the initiation and development of AS, and the levels of pyroptosis-related proteins are positively correlated with plaque instability, indicating the crucial contribution of pyroptosis to atherogenesis. Furthermore, studies have also identified some candidate anti-atherogenic agents targeting plaque cell pyroptosis. Herein, we summarize the research progress in understating (1) the discovery and definition of pyroptosis; (2) the characterization and molecular mechanisms of pyroptosis; (3) the regulatory mechanisms of pyroptosis in VEC, macrophage, and VSMC, as well as their potential role in AS progression, aimed at providing therapeutic targets for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Xiao-Dan Xu
- grid.412679.f0000 0004 1771 3402Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui People’s Republic of China
| | - Jia-Xian Chen
- grid.443397.e0000 0004 0368 7493Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| | - Lin Zhu
- grid.252251.30000 0004 1757 8247College of Nursing, Anhui University of Chinese Medicine, Hefei, 230012 Anhui People’s Republic of China
| | - Shu-Ting Xu
- grid.411971.b0000 0000 9558 1426Department of Nephrology, The Affiliated Hospital of Dalian Medical University, Dalian, 116044 Liaoning People’s Republic of China
| | - Jian Jiang
- grid.443397.e0000 0004 0368 7493Department of Organ Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| | - Kun Ren
- grid.252251.30000 0004 1757 8247College of Nursing, Anhui University of Chinese Medicine, Hefei, 230012 Anhui People’s Republic of China ,grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| |
Collapse
|
21
|
Zhang Y, Zhou XA, Liu C, Shen Q, Wu Y. Vitamin B6 Inhibits High Glucose-Induced Islet β Cell Apoptosis by Upregulating Autophagy. Metabolites 2022; 12:1048. [PMID: 36355132 PMCID: PMC9695582 DOI: 10.3390/metabo12111048] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 07/27/2023] Open
Abstract
Vitamin B6 may alleviate diabetes by regulating insulin secretion and increasing insulin sensitivity, but its mechanism remains to be explored. In this study, vitamin B6-mediated autophagy and high glucose-induced apoptosis were tested to investigate the mechanism by which vitamin B6 regulates insulin release. The results showed that 20 mM glucose increased the apoptosis rate from 10.39% to 22.44%. Vitamin B6 reduced the apoptosis rate of RIN-m5F cells from 22.44% to 11.31%. Our data also showed that the vitamin B6 content in processed eggs was decreased and that the hydrothermal process did not affect the bioactivity of vitamin B6. Vitamin B6 increased the number of autophagosomes and the ratio of autophagosome marker protein microtubule associated protein 1 light chain 3 beta to microtubule associated protein 1 light chain 3 alpha (LC3-II/LC3-I). It also decreased the amount of sequetosome 1 (SQSTM1/p62) and inhibited the phosphorylation of p70 ribosomal protein S6 kinase (p70S6K) under normal and high glucose stress. Another study showed that vitamin B6 inhibited the apoptosis rate, whereas the autophagy inhibitor 3-methyladenine (3-MA) blocked the protective effect of vitamin B6 against apoptosis induced by high glucose. The hydrothermal process decreased the vitamin B6 content in eggs but had no effect on the cytoprotective function of vitamin B6 in RIN-m5f cells. In conclusion, we demonstrated that vitamin B6-mediated autophagy protected RIN-m5f cells from high glucose-induced apoptosis might via the mTOR-dependent pathway. Our data also suggest that low temperatures and short-term hydrothermal processes are beneficial for dietary eggs.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Horticulture and Landscape College, Hunan Agricultural University, Changsha 410128, China
| | - Xi-an Zhou
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Horticulture and Landscape College, Hunan Agricultural University, Changsha 410128, China
| | - Chuxin Liu
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Horticulture and Landscape College, Hunan Agricultural University, Changsha 410128, China
| | - Qingwu Shen
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Horticulture and Landscape College, Hunan Agricultural University, Changsha 410128, China
| | - Yanyang Wu
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Horticulture and Landscape College, Hunan Agricultural University, Changsha 410128, China
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha 410128, China
| |
Collapse
|
22
|
Singh B, Li K, Cui K, Peng Q, Cowan DB, Wang DZ, Chen K, Chen H. Defective efferocytosis of vascular cells in heart disease. Front Cardiovasc Med 2022; 9:1031293. [PMID: 36247464 PMCID: PMC9561431 DOI: 10.3389/fcvm.2022.1031293] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
The efficient phagocytic clearance of dying cells and apoptotic cells is one of the processes that is essential for the maintenance of physiologic tissue function and homeostasis, which is termed "efferocytosis." Under normal conditions, "find me" and "eat me" signals are released by apoptotic cells to stimulate the engulfment and efferocytosis of apoptotic cells. In contrast, abnormal efferocytosis is related to chronic and non-resolving inflammatory diseases such as atherosclerosis. In the initial steps of atherosclerotic lesion development, monocyte-derived macrophages display efficient efferocytosis that restricts plaque progression; however, this capacity is reduced in more advanced lesions. Macrophage reprogramming as a result of the accumulation of apoptotic cells and augmented inflammation accounts for this diminishment of efferocytosis. Furthermore, defective efferocytosis plays an important role in necrotic core formation, which triggers plaque rupture and acute thrombotic cardiovascular events. Recent publications have focused on the essential role of macrophage efferocytosis in cardiac pathophysiology and have pointed toward new therapeutic strategies to modulate macrophage efferocytosis for cardiac tissue repair. In this review, we discuss the molecular and cellular mechanisms that regulate efferocytosis in vascular cells, including macrophages and other phagocytic cells and detail how efferocytosis-related molecules contribute to the maintenance of vascular hemostasis and how defective efferocytosis leads to the formation and progression of atherosclerotic plaques.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Kathryn Li
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Qianman Peng
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Douglas B. Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, United States
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
23
|
Petrossian G, Ozdemir D, Galougahi KK, Scheiner J, Thomas SV, Shlofmitz R, Shlofmitz E, Jeremias A, Ali ZA. Role of Intracoronary Imaging in Acute Coronary Syndromes. US CARDIOLOGY REVIEW 2022; 16:e15. [PMID: 39600836 PMCID: PMC11588184 DOI: 10.15420/usc.2022.03] [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: 01/26/2022] [Accepted: 04/12/2022] [Indexed: 11/04/2022] Open
Abstract
Intravascular imaging with optical coherence tomography (OCT) and intravascular ultrasound provides superior visualization of the culprit plaques for acute coronary syndromes (ACS) compared with coronary angiography. Combined with angiography, intravascular imaging can be used to instigate 'precision therapy' for ACS. Post-mortem histopathology identified atherothrombosis at the exposed surface of a ruptured fibrous cap as the main cause of ACS. Further histopathological studies identified intact fibrous caps and calcified nodules as other culprit lesions for ACS. These plaque types were subsequently also identified on intravascular imaging, particularly with the high-resolution OCT. The less-common non-atherothrombotic causes of ACS are coronary artery spasm, coronary artery dissection, and coronary embolism. In this review, the authors provide an overview of clinical studies using intravascular imaging with OCT in the diagnosis and management of ACS.
Collapse
Affiliation(s)
| | - Denizhan Ozdemir
- Division of Cardiology, Columbia University Irving Medical Center/NewYork-Presbyterian HospitalNew York, NY
| | - Keyvan Karimi Galougahi
- Department of Cardiology, Royal Prince Alfred HospitalSydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of SydneySydney, Australia
- Heart Research InstituteSydney, Australia
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
| | - Jonathan Scheiner
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
| | - Susan V Thomas
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
| | - Richard Shlofmitz
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
| | - Evan Shlofmitz
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
| | - Allen Jeremias
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
- Clinical Trials Center, Cardiovascular Research FoundationNew York, NY
| | - Ziad A Ali
- DeMatteis Cardiovascular Institute, St Francis Hospital – The Heart CenterRoslyn, NY
- Clinical Trials Center, Cardiovascular Research FoundationNew York, NY
| |
Collapse
|
24
|
Jarr KU, Kojima Y, Weissman IL, Leeper NJ. 2021 Jeffrey M. Hoeg Award Lecture: Defining the Role of Efferocytosis in Cardiovascular Disease: A Focus on the CD47 (Cluster of Differentiation 47) Axis. Arterioscler Thromb Vasc Biol 2022; 42:e145-e154. [PMID: 35387480 PMCID: PMC9183217 DOI: 10.1161/atvbaha.122.317049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023]
Abstract
A key feature of atherogenesis is the accumulation of diseased and dying cells within the lesional necrotic core. While the burden of intraplaque apoptotic cells may be driven in part by an increase in programmed cell death, mounting evidence suggests that their presence may primarily be dictated by a defect in programmed cell removal, or efferocytosis. In this brief review, we will summarize the evidence suggesting that inflammation-dependent changes within the plaque render target cells inedible and reduce the appetite of lesional phagocytes. We will present the genetic causation studies, which indicate these phenomena promote lesion expansion and plaque vulnerability, and the interventional data which suggest that these processes can be reversed. Particular emphasis is provided related to the antiphagocytic CD47 (cluster of differentiation 47) do not eat me axis, which has emerged as a novel antiatherosclerotic translational target that is predicted to provide benefit independent of traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
25
|
Puylaert P, Van Praet M, Vaes F, Neutel CHG, Roth L, Guns PJ, De Meyer GRY, Martinet W. Gasdermin D Deficiency Limits the Transition of Atherosclerotic Plaques to an Inflammatory Phenotype in ApoE Knock-Out Mice. Biomedicines 2022; 10:biomedicines10051171. [PMID: 35625908 PMCID: PMC9138554 DOI: 10.3390/biomedicines10051171] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Gasdermin D (GSDMD) is the key executor of pyroptotic cell death. Recent studies suggest that GSDMD-mediated pyroptosis is involved in atherosclerotic plaque destabilization. We report that cleaved GSDMD is expressed in macrophage- and smooth muscle cell-rich areas of human plaques. To determine the effects of GSDMD deficiency on atherogenesis, ApoE−/− Gsdmd−/− (n = 16) and ApoE−/−Gsdmd+/+ (n = 18) mice were fed a western-type diet for 16 weeks. Plaque initiation and formation of stable proximal aortic plaques were not altered. However, plaques in the brachiocephalic artery (representing more advanced lesions compared to aortic plaques) of ApoE−/− Gsdmd−/− mice were significantly smaller (115 ± 18 vs. 186 ± 16 × 103 µm2, p = 0.006) and showed features of increased stability, such as decreased necrotic core area (19 ± 4 vs. 37 ± 7 × 103 µm2, p = 0.03) and increased αSMA/MAC3 ratio (1.6 ± 0.3 vs. 0.7 ± 0.1, p = 0.01), which was also observed in proximal aortic plaques. Interestingly, a significant increase in TUNEL positive cells was observed in brachiocephalic artery plaques from ApoE−/− Gsdmd−/− mice (141 ± 25 vs. 62 ± 8 cells/mm2, p = 0.005), indicating a switch to apoptosis. This switch from pyroptosis to apoptosis was also observed in vitro in Gsdmd−/− macrophages. In conclusion, targeting GSDMD appears to be a promising approach for limiting the transition to an inflammatory, vulnerable plaque phenotype.
Collapse
|
26
|
Verma S, Kumar A, Narang R, Bisoi AK, Mitra DK. Signature transcriptome analysis of stage specific atherosclerotic plaques of patients. BMC Med Genomics 2022; 15:99. [PMID: 35488341 PMCID: PMC9055692 DOI: 10.1186/s12920-022-01250-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Inflammation plays an important role in all the stages of atherosclerotic plaque development. The current study aimed at assessing the altered expression of genes functioning in inflammation within the early stage (ES) and advanced stage (AS) atherosclerotic plaques obtained from patients undergoing coronary artery bypass grafting (CABG) surgery and identifying biomarker panel/s that may detect the status of plaque stages using peripheral blood samples. METHODS A section of ES and AS plaques and normal left internal mammary arteries (LIMA) were obtained from 8 patients undergoing the CABG surgery. Total RNA isolated was analyzed for mRNA and miRNA expression profile by Affymetrix arrays. A significant number of mRNAs was found to be differentially expressed in ES and AS plaque tissues relative to LIMA. The pathway analysis of differentially expressed mRNAs in the two plaque stages was also performed using DAVID Bioinformatics Database. RESULTS The mRNAs were found to be involved in critical inflammatory processes such as the toll-like receptor signaling pathway and cytokine-cytokine receptor interaction. Few miRNAs targeting these mRNAs were also altered in the two plaque conditions. QRT-PCR results showed a similar expression pattern of a few of the mRNAs and miRNAs in peripheral blood of the same patients relative to healthy controls. CONCLUSION Changes in mRNA and miRNA expression associated with various inflammatory processes occur in different atherosclerotic stage plaques as well as peripheral blood. Detection of such variations in patients' blood can be used as a possible prognostic tool to detect and/or predict the risk and stage of atherosclerosis.
Collapse
Affiliation(s)
- Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Abhay Kumar
- Department of Microbiology, Indira Gandhi Institute of Medical Sciences, Patna, India
| | - Rajiv Narang
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Akshya K Bisoi
- Department of Cardiothoracic and Vascular Surgery, Cardio, and Neurosciences Center, AIIMS, New Delhi, India
| | - Dipendra K Mitra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences (AIIMS), Room No-75, New Delhi, 110029, India.
| |
Collapse
|
27
|
Zhang M, Xie Z, Long H, Ren K, Hou L, Wang Y, Xu X, Lei W, Yang Z, Ahmed S, Zhang H, Zhao G. Current advances in the imaging of atherosclerotic vulnerable plaque using nanoparticles. Mater Today Bio 2022; 14:100236. [PMID: 35341094 PMCID: PMC8943324 DOI: 10.1016/j.mtbio.2022.100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 01/29/2023]
Abstract
Vulnerable atherosclerotic plaques of the artery wall that pose a significant risk of cardio-cerebral vascular accidents remain the global leading cause of morbidity and mortality. Thus, early delineation of vulnerable atherosclerotic plaques is of clinical importance for prevention and treatment. The currently available imaging technologies mainly focus on the structural assessment of the vascular wall. Unfortunately, several disadvantages in these strategies limit the improvement in imaging effect. Nanoparticle technology is a novel diagnostic strategy for targeting and imaging pathological biomarkers. New functionalized nanoparticles that detect hallmarks of vulnerable plaques are promising for advance further control of this critical illness. The review aims to address the current opportunities and challenges for the use of nanoparticle technology in imagining vulnerable plaques.
Collapse
|
28
|
Lin L, Zhang MX, Zhang L, Zhang D, Li C, Li YL. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front Cell Dev Biol 2022; 9:809955. [PMID: 35096837 PMCID: PMC8793783 DOI: 10.3389/fcell.2021.809955] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual buildup of plaques within the vessel wall of middle-sized and large arteries. The occurrence and development of atherosclerosis and the rupture of plaques are related to the injury of vascular cells, including endothelial cells, smooth muscle cells, and macrophages. Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles, and the autophagy disorder of vascular cells is closely related to atherosclerosis. Pyroptosis is a proinflammatory form of regulated cell death, while ferroptosis is a form of regulated nonapoptotic cell death involving overwhelming iron-dependent lipid peroxidation. Both of them exhibit distinct features from apoptosis, necrosis, and autophagy in morphology, biochemistry, and genetics. However, a growing body of evidence suggests that pyroptosis and ferroptosis interact with autophagy and participate in the development of cancers, degenerative brain diseases and cardiovascular diseases. This review updated the current understanding of autophagy, pyroptosis, and ferroptosis, finding potential links and their effects on atherogenesis and plaque stability, thus providing ways to develop new pharmacological strategies to address atherosclerosis and stabilize vulnerable, ruptured plaques.
Collapse
Affiliation(s)
- Lin Lin
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mu-Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Zhang
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun-Lun Li
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Liu C, Jiang Z, Pan Z, Yang L. The Function, Regulation and Mechanism of Programmed Cell Death of Macrophages in Atherosclerosis. Front Cell Dev Biol 2022; 9:809516. [PMID: 35087837 PMCID: PMC8789260 DOI: 10.3389/fcell.2021.809516] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory vascular disease, which is an important pathological basis for inducing a variety of cardio-cerebrovascular diseases. As a kind of inflammatory cells, macrophages are the most abundant immune cells in atherosclerotic plaques and participate in the whole process of atherosclerosis and are the most abundant immune cells in atherosclerotic plaques. Recent studies have shown that programmed cell death plays a critical role in the progression of many diseases. At present, it is generally believed that the programmed death of macrophages can affect the development and stability of atherosclerotic vulnerable plaques, and the intervention of macrophage death may become the target of atherosclerotic therapy. This article reviews the role of macrophage programmed cell death in the progression of atherosclerosis and the latest therapeutic strategies targeting macrophage death within plaques.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Zecheng Jiang
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | | | - Liang Yang
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
30
|
Sakamoto A, Cornelissen A, Sato Y, Mori M, Kawakami R, Kawai K, Ghosh SKB, Xu W, Abebe BG, Dikongue A, Kolodgie FD, Virmani R, Finn AV. Vulnerable Plaque in Patients with Acute Coronary Syndrome: Identification, Importance, and Management. US CARDIOLOGY REVIEW 2022; 16:e01. [PMID: 39600843 PMCID: PMC11588187 DOI: 10.15420/usc.2021.22] [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: 07/07/2021] [Accepted: 10/07/2021] [Indexed: 11/04/2022] Open
Abstract
MI is a leading cause of morbidity and mortality worldwide. Coronary artery thrombosis is the final pathologic feature of the most cases of acute MI primarily caused by atherosclerotic coronary artery disease. The concept of vulnerable plaque has evolved over the years but originated from early pioneering work unveiling the crucial role of plaque rupture and subsequent coronary thrombosis as the dominant cause of MI. Along with systemic cardiovascular risk factors, developments of intravascular and non-invasive imaging modalities have allowed us to identify coronary plaques thought to be at high risk for rupture. However, morphological features alone may only be one of many factors which promote plaque progression. The current vulnerable-plaque-oriented approaches to accomplish personalized risk assessment and treatment have significant room for improvement. In this review, the authors discuss recent advances in the understanding of vulnerable plaque and its management strategy from pathology and clinical perspectives.
Collapse
Affiliation(s)
| | | | - Yu Sato
- CVPath InstituteGaithersburg, MD
| | | | | | | | | | - Weili Xu
- CVPath InstituteGaithersburg, MD
| | | | | | | | | | - Aloke V Finn
- CVPath InstituteGaithersburg, MD
- University of Maryland, School of MedicineBaltimore, MD
| |
Collapse
|
31
|
Abstract
Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.
Collapse
Affiliation(s)
- Dawn M Fernandez
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- New York University Cardiovascular Research Center, New York University Langone Health, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
32
|
Zhang J, Xu Y, Ding W, Zhao M, Liu J, Ye J, Wang Z, Ye D, Wang M, Wan J. Increased expression of IL-20 is associated with ischemic cardiomyopathy and acute myocardial infarction. Biomark Med 2021; 15:1641-1650. [PMID: 34704818 DOI: 10.2217/bmm-2020-0529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/01/2021] [Indexed: 11/21/2022] Open
Abstract
Background: The expression and clinical significance of IL-20 in coronary artery diseases needs to be analyzed. Methods: IL-20 and its receptors were analyzed in coronary artery tissues. In a separate study, plasma IL-20 was also evaluated. Results: IL-20 and its receptors were significantly higher in coronary artery stenosis tissues from ischemic cardiomyopathy patients than that from controls. T lymphocytes and macrophages were the main source of IL-20 and expressed its receptors abundantly. Plasma IL-20 was significantly higher in acute myocardial infarction patients than that in controls. Conclusion: IL-20 was closely associated with the presence of acute myocardial infarction. IL-20 may participate in the progression of coronary artery stenosis and plaque vulnerability via regulating T lymphocytes and macrophages.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| |
Collapse
|
33
|
Molecular Imaging of Vulnerable Coronary Plaque with Radiolabeled Somatostatin Receptors (SSTR). J Clin Med 2021; 10:jcm10235515. [PMID: 34884218 PMCID: PMC8658082 DOI: 10.3390/jcm10235515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis is responsible for the majority of heart attacks and is characterized by several modifications of the arterial wall including an inflammatory reaction. The silent course of atherosclerosis has made it necessary to develop predictors of disease complications before symptomatic lesions occur. Vulnerable to rupture atherosclerotic plaques are the target for molecular imaging. To this aim, different radiopharmaceuticals for PET/CT have emerged for the identification of high-risk plaques, with high specificity for the identification of the cellular components and pathophysiological status of plaques. By targeting specific receptors on activated macrophages in high-risk plaques, radiolabelled somatostatin analogues such as 68Ga-DOTA-TOC, TATE,0 or NOC have shown high relevance to detect vulnerable, atherosclerotic plaques. This PET radiopharmaceutical has been tested in several pre-clinical and clinical studies, as reviewed here, showing an important correlation with other risk factors.
Collapse
|
34
|
Sun J, Singh P, Österlund J, Orho-Melander M, Melander O, Engström G, Edsfeldt A. Hyperglycaemia-associated Caspase-3 predicts diabetes and coronary artery disease events. J Intern Med 2021; 290:855-865. [PMID: 34309093 PMCID: PMC7612448 DOI: 10.1111/joim.13327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/05/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Apoptosis is central in both diabetes and atherosclerosis, linked to pancreatic beta cell death and plaque progression. Circulating Caspase-3 has also been associated with diabetes and coronary calcium score. Here, we explored if soluble Caspase-3 (sCaspase-3) is associated with cardio-metabolic risk factors and predicts incidence of diabetes and coronary artery disease (CAD). METHODS Clinical data and plasma from 4637 individuals from the Malmö Diet and Cancer cohort were studied. Plasma sCaspase-3 was measured by a Proximity Extension Assay. National registers were used to identify diabetes and CAD events during follow-up. Type 2 diabetes risk variants and expression quantitative trait loci (eQTL) for sCaspase-3 were retrieved from the DIAGRAM consortium and the Genotype-Tissue Expression project. RESULTS HbA1c was the factor with the strongest association with sCaspase-3 (r = 0.18, P = 1.3x10-36 ). During follow-up 666 individuals developed diabetes and 648 individuals suffered from CAD. Increasing sCaspase-3 was associated with a higher risk of developing diabetes (hazard ratio (HR) 1.18 per 1unit; P = 7 × 10-5 ) and CAD (HR 1.2 per 1 unit, P = 1 × 10-4 ) during follow-up. A genetic variant rs60780116, located upstream of CASP3, showed strong association with type 2 diabetes (OR 1.06, 95%CI 1.04-1.07, P = 8.4 × 10-11 ). An eQTL was identified between this variant and gene expression of CASP3, where the allele positively correlated with type 2 diabetes was associated with increased CASP3 expression in blood. CONCLUSIONS The present study provides evidence for plasma sCaspase-3 as a marker of cardio-metabolic risk factors and as a predictor of future diabetes and CAD in a cohort without cardiovascular disease or diabetes at baseline.
Collapse
Affiliation(s)
- Jiangming Sun
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Pratibha Singh
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Johan Österlund
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Marju Orho-Melander
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olle Melander
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gunnar Engström
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Andreas Edsfeldt
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Blackburn CMR, Schilke RM, Vozenilek AE, Chandran S, Bamgbose TT, Finck BN, Woolard MD. Myeloid-associated lipin-1 transcriptional co-regulatory activity is atheroprotective. Atherosclerosis 2021; 330:76-84. [PMID: 34256308 DOI: 10.1016/j.atherosclerosis.2021.06.927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/27/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is the most prominent underlying cause of cardiovascular disease (CVD). It is initiated by cholesterol deposition in the arterial intima, which causes macrophage recruitment and proinflammatory responses that promote plaque growth, necrotic core formation, and plaque rupture. Lipin-1 is a phosphatidic acid phosphohydrolase for glycerolipid synthesis. We have shown that lipin-1 phosphatase activity promotes macrophage pro-inflammatory responses when stimulated with modified low-density lipoprotein (modLDL) and accelerates atherosclerosis. Lipin-1 also independently acts as a transcriptional co-regulator where it enhances the expression of genes involved in β-oxidation. In hepatocytes and adipocytes, lipin-1 augments the activity of transcription factors such as peroxisome proliferator-activated receptor (PPARs). PPARs control the expression of anti-inflammatory genes in macrophages and slow or reduce atherosclerotic progression. Therefore, we hypothesize myeloid-derived lipin-1 transcriptional co-regulatory activity reduces atherosclerosis. METHODS We used myeloid-derived lipin-1 knockout (lipin-1mKO) and littermate control mice and AAV8-PCSK9 along with high-fat diet to elicit atherosclerosis. RESULTS Lipin-1mKO mice had larger aortic root plaques than littermate control mice after 8 and 12 weeks of a high-fat diet. Lipin-1mKO mice also had increased serum proinflammatory cytokine concentrations, reduced apoptosis in plaques, and larger necrotic cores in the plaques compared to control mice. CONCLUSIONS Combined, the data suggest lipin-1 transcriptional co-regulatory activity in myeloid cells is atheroprotective.
Collapse
Affiliation(s)
- Cassidy M R Blackburn
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Aimee E Vozenilek
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sunitha Chandran
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Temitayo T Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Brian N Finck
- Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
36
|
Bajaj R, Garcia-Garcia HM, Courtney BK, Ramasamy A, Tufaro V, Erdogan E, Khan AH, Alves N, Rathod KS, Onuma Y, Serruys PW, Mathur A, Baumbach A, Bourantas C. Multi-modality intravascular imaging for guiding coronary intervention and assessing coronary atheroma: the Novasight Hybrid IVUS-OCT system. Minerva Cardiol Angiol 2021; 69:655-670. [PMID: 33703857 DOI: 10.23736/s2724-5683.21.05532-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intravascular imaging has evolved alongside interventional cardiology as an adjunctive tool for assessing plaque pathology and for guiding and optimising percutaneous coronary intervention (PCI) in challenging lesions. The two modalities which have dominated the field are intravascular ultrasound (IVUS), which relies on sound waves and optical coherence tomography (OCT), relying on light waves. These approaches however have limited efficacy in assessing plaque morphology and vulnerability that are essential for guiding PCI in complex lesions and identifying patient at risk that will benefit from emerging therapies targeting plaque evolution. These limitations are complementary and, in this context, it has been recognised and demonstrated in multi-modality studies that the concurrent use of IVUS and OCT can help overcome these deficits enabling a more complete and accurate plaque assessment. The Conavi Novasight Hybrid IVUS-OCT catheter is the first commercially available device that is capable of invasive clinical coronary assessment with simultaneously acquired and co-registered IVUS and OCT imaging. It represents a significant evolution in the field and is expected to have broad application in clinical practice and research. In this review article we present the limitations of standalone intravascular imaging techniques, summarise the data supporting the value of multimodality imaging in clinical practice and research, describe the Novasight Hybrid IVUS-OCT system and highlight the potential utility of this technology in coronary intervention and in the study of atherosclerosis.
Collapse
Affiliation(s)
- Retesh Bajaj
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Brian K Courtney
- Sunnybrook Research Institute, Schulich Heart Program, University of Toronto, Toronto, ON, Canada.,Conavi Medical, North York, ON, Canada
| | - Anantharaman Ramasamy
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Vincenzo Tufaro
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Emrah Erdogan
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Ameer H Khan
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Natasha Alves
- Sunnybrook Research Institute, Schulich Heart Program, University of Toronto, Toronto, ON, Canada
| | - Krishnaraj S Rathod
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yoshinobu Onuma
- School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Patrick W Serruys
- School of Medicine, National University of Ireland Galway, Galway, Ireland.,National Heart & Lung Institute, Imperial College London, London, UK
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andreas Baumbach
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Christos Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK - .,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
37
|
Anandan V, Thankayyan Retnabai SK, Jaleel A, Thulaseedharan T, Mullasari A, Pillai MR, Kartha CC, Ramachandran S. Cyclophilin A induces macrophage apoptosis and enhances atherosclerotic lesions in high-fat diet-fed hyperglycemic rabbits. FASEB Bioadv 2021; 3:305-322. [PMID: 33977232 PMCID: PMC8103716 DOI: 10.1096/fba.2020-00135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 11/11/2022] Open
Abstract
Macrophage apoptosis is a key contributor to the progression of atherosclerosis. Cyclophilin A, a monocyte secretory protein associated with the initiation of atherosclerosis has an inherent nuclease activity. This study reports the mechanism by which cyclophilin A causes apoptosis of macrophages and accelerates the progression of atherosclerosis. Aortic lesion formation and apoptosis were studied in New Zealand White rabbits (NZW) which were fed high-fat diet (HFD) for 12 weeks. Using monocytes and HFD-fed rabbits we demonstrate that cyclophilin A induces mitochondrial membrane potential loss and mitochondrial pore transition protein opening through caspase 3 activation. En face staining revealed a significant increase in the lesion area in HFD-fed rabbits. Levels of glucose, cholesterol and proinflammatory cytokines were higher in these animals compared to rabbits fed with a normal diet. In the aorta of HFD-fed rabbits, medial vascular smooth muscle cells were disorganized and there was a loss of integrity of the endothelium. An 8-fold increase was seen in the number of apoptotic cells in the lesion area of HFD-fed NZW rabbits which were associated with an elevation in plasma cyclophilin A levels. siRNA knockdown of cyclophilin A gene reduced activation of caspase 3 in macrophages. Treatment with cyclosporine A, an inhibitor of cyclophilin A, significantly attenuated apoptosis in macrophages. Our study indicates that inhibitors of proinflammatory cytokines such as cyclophilin A may arrest macrophage apoptosis and result in a regression of advanced atherosclerotic lesions.
Collapse
Affiliation(s)
- Vinitha Anandan
- Cardiovascular Diseases and Diabetes Biology Rajiv Gandhi Centre for Biotechnology Trivandrum India.,Manipal Academy of Higher Education Manipal India
| | | | - Abdul Jaleel
- Cardiovascular Diseases and Diabetes Biology Rajiv Gandhi Centre for Biotechnology Trivandrum India
| | - Thushara Thulaseedharan
- Cardiovascular Diseases and Diabetes Biology Rajiv Gandhi Centre for Biotechnology Trivandrum India
| | | | | | | | - Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology Rajiv Gandhi Centre for Biotechnology Trivandrum India
| |
Collapse
|
38
|
Chaudhry F, Kawai H, Johnson KW, Narula N, Shekhar A, Chaudhry F, Nakahara T, Tanimoto T, Kim D, Adapoe MKMY, Blankenberg FG, Mattis JA, Pak KY, Levy PD, Ozaki Y, Arbustini E, Strauss HW, Petrov A, Fuster V, Narula J. Molecular Imaging of Apoptosis in Atherosclerosis by Targeting Cell Membrane Phospholipid Asymmetry. J Am Coll Cardiol 2021; 76:1862-1874. [PMID: 33059832 DOI: 10.1016/j.jacc.2020.08.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Apoptosis in atherosclerotic lesions contributes to plaque vulnerability by lipid core enlargement and fibrous cap attenuation. Apoptosis is associated with exteriorization of phosphatidylserine (PS) and phosphatidylethanolamine (PE) on the cell membrane. Although PS-avid radiolabeled annexin-V has been employed for molecular imaging of high-risk plaques, PE-targeted imaging in atherosclerosis has not been studied. OBJECTIVES This study sought to evaluate the feasibility of molecular imaging with PE-avid radiolabeled duramycin in experimental atherosclerotic lesions in a rabbit model and compare duramycin targeting with radiolabeled annexin-V. METHODS Of the 27 rabbits, 21 were fed high-cholesterol, high-fat diet for 16 weeks. Nine of the 21 rabbits received 99mTc-duramycin (test group), 6 received 99mTc-linear duramycin (duramycin without PE-binding capability, negative radiotracer control group), and 6 received 99mTc-annexin-V for radionuclide imaging. The remaining normal chow-fed 6 animals (disease control group) received 99mTc-duramycin. In vivo microSPECT/microCT imaging was performed, and the aortas were explanted for ex vivo imaging and for histological characterization of atherosclerosis. RESULTS A significantly higher duramycin uptake was observed in the test group compared with that of disease control and negative radiotracer control animals; duramycin uptake was also significantly higher than the annexin-V uptake. Quantitative duramycin uptake, represented as the square root of percent injected dose per cm (√ID/cm) of abdominal aorta was >2-fold higher in atherosclerotic lesions in test group (0.08 ± 0.01%) than in comparable regions of disease control animals (0.039 ± 0.0061%, p = 3.70·10-8). Mean annexin uptake (0.060 ± 0.010%) was significantly lower than duramycin (p = 0.001). Duramycin uptake corresponded to the lesion severity and macrophage burden. The radiation burden to the kidneys was substantially lower with duramycin (0.49% ID/g) than annexin (5.48% ID/g; p = 4.00·10-4). CONCLUSIONS Radiolabeled duramycin localizes in lipid-rich areas with high concentration of apoptotic macrophages in the experimental atherosclerosis model. Duramycin uptake in atherosclerotic lesions was significantly greater than annexin-V uptake and produced significantly lower radiation burden to nontarget organs.
Collapse
Affiliation(s)
- Farhan Chaudhry
- Icahn School of Medicine at Mount Sinai, New York, New York; Wayne State University School of Medicine, Detroit, Michigan
| | - Hideki Kawai
- Icahn School of Medicine at Mount Sinai, New York, New York; Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Navneet Narula
- New York University Langone Medical Center, New York, New York
| | - Aditya Shekhar
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Dongbin Kim
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Jeffrey A Mattis
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Phillip D Levy
- Wayne State University School of Medicine, Detroit, Michigan
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | | | - H William Strauss
- Icahn School of Medicine at Mount Sinai, New York, New York; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Artiom Petrov
- Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Valentin Fuster
- Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
39
|
Paszek E, Zajdel W, Rajs T, Żmudka K, Legutko J, Kleczyński P. Profilin 1 and Mitochondria-Partners in the Pathogenesis of Coronary Artery Disease? Int J Mol Sci 2021; 22:1100. [PMID: 33499277 PMCID: PMC7865810 DOI: 10.3390/ijms22031100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis remains a large health and economic burden. Even though it has been studied for more than a century, its complex pathophysiology has not been elucidated. The relatively well-established contributors include: chronic inflammation in response to oxidized cholesterol, reactive oxygen species-induced damage and apoptosis. Recently, profilin 1, a regulator of actin dynamics emerged as a potential new player in the field. Profilin is abundant in stable atherosclerotic plaques and in thrombi extracted from infarct-related arteries in patients with acute myocardial infarction. The exact role of profilin in atherosclerosis and its complications, as well as its mechanisms of action, remain unknown. Here, we summarize several pathways in which profilin may act through mitochondria in a number of processes implicated in atherosclerosis.
Collapse
Affiliation(s)
- Elżbieta Paszek
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
| | - Wojciech Zajdel
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
| | - Tomasz Rajs
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
| | - Krzysztof Żmudka
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Krakow, Poland
| | - Jacek Legutko
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Krakow, Poland
| | - Paweł Kleczyński
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Krakow, Poland; (E.P.); (W.Z.); (T.R.); (K.Ż.); (J.L.)
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Krakow, Poland
| |
Collapse
|
40
|
Tosun H, Kamışlı S, Tecellioğlu M, Alan S, Tecellioğlu FS, Öztanır MN, Kablan Y. Red and White Thrombus Characteristics in Patients Undergoing Carotid Endarterectomy. J Stroke Cerebrovasc Dis 2020; 30:105451. [PMID: 33278805 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/12/2020] [Accepted: 10/31/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The study aimed to compare the characteristics of red and white thrombi in patients undergoing carotid endarterectomy. MATERIAL AND METHODS The study was conducted in 81 patients with ischemic stroke who underwent carotid endarterectomy for carotid artery stenosis. Carotid plaques were graded by two pathologists. Thrombus materials were divided into two groups: white and red. The parameters of assessment were plaque rupture, lipid core, fibrous cap thickness, inflammation, intraplaque hemorrhage, calcification, necrotic core, and neovascularization. Normally distributed data were evaluated using Mann-Whitney U and Chi-squared tests. RESULTS The ratio of white and red thrombus was 19.8% and 80.2%, respectively. Lipid core, plaque rupture, necrotic core, neovascularization, intraplaque hemorrhage, obstruction, and inflammation were observed more in red thrombus, which were statistically significant. Calcification and fibrous cap thickness were not statistically significant in the two groups. Moreover, intimal smooth muscle cells were present in all thrombus types. CONCLUSION In our study, we found that red thrombi had more unstable characteristics than white thrombi. Thus, the risk for ischemic cerebrovascular events is more in red thrombi. However, this finding cannot be generalized due to the small number of patients in this study. Therefore, studies involving more patients are needed.
Collapse
Affiliation(s)
| | | | | | - Saadet Alan
- Turgut Özal Medical Center, Malatya, Turkey.
| | | | | | | |
Collapse
|
41
|
Sun SW, Tong WJ, Zheng GQ, Tuo QH, Lei XY, Liao DF. Pyroptotic cell-derived microparticle: An atherogenic factor in infectious diseases. Med Hypotheses 2020; 146:110370. [PMID: 33308934 DOI: 10.1016/j.mehy.2020.110370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/21/2020] [Accepted: 10/26/2020] [Indexed: 11/25/2022]
Abstract
Chronic infection is considered a risk factor for atherosclerosis. The link between infectious agents and atherosclerosis is manifested by the presence of infection-induced pyroptotic cells in atherosclerotic lesions. Pyroptosis is an inflammatory form of programmed cell death that occurs most frequently upon infection. However, inflammation is not the only cause by which pyroptosis involved in atherosclerosis. During pyroptosis, a large amount of microparticles are released from pyroptotic cells, which not only transfer inflammatory mediators to arterial vessel, but also mediate the interaction between a variety of cells, leading to endothelial injury, macrophage infiltration, vascular smooth muscle cell migration and proliferation, thereby accelerating atherosclerosis. Thus, we proposed hypothesis that pyroptotic cell-derived microparticle is an atherogenic factor in infectious diseases.
Collapse
Affiliation(s)
- Shao-Wei Sun
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, Hunan, China
| | - Wen-Juan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Gui-Qiong Zheng
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, Hunan, China
| | - Qin-Hui Tuo
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Xiao-Yong Lei
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, Hunan, China
| | - Duan-Fang Liao
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| |
Collapse
|
42
|
Targeted multiplex proteomics for prediction of all-cause mortality during long-term follow-up in outpatients with peripheral arterial disease. Atherosclerosis 2020; 311:143-149. [PMID: 32711845 DOI: 10.1016/j.atherosclerosis.2020.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 04/20/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Patients with peripheral arterial disease (PAD) are at high risk for fatal events. We aimed to investigate the ability among several serum proteins to predict all-cause mortality in outpatients with PAD. METHODS Consecutive outpatients with carotid and/or lower extremity PAD were included in the discovery cohort (n = 436), and subjects with PAD from a population-based sample in the validation cohort (n = 129). Blood samples were analyzed for 81 proteins by a proximity extension assay. The proteins best predicting incident all-cause mortality were identified using L1-regularized Cox regression. The added value of the identified proteins to clinical risk markers was evaluated by Cox regression models and presented by the area under the receiver operator characteristics curves (AUC). RESULTS In the discovery cohort (mean age 70 years; 59% men), 195 died (4.8 events per 100 person-years) during a 10.3 years median follow-up. The clinical risk markers generated an AUC of 0.70 (95% confidence interval [95%CI] 0.65-0.76). The two serum protein biomarkers with best prediction of all-cause mortality were growth differentiation factor 15 and tumor necrosis factor-related apoptosis-inducing ligand receptor 2. Adding these proteins to the clinical risk markers significantly improved prediction (p < 0.001) and yielded an AUC of 0.76 (95%CI 0.71-0.80). A higher discriminatory performance was observed in the validation cohort (AUC 0.84; 95% CI 0.76-0.92). CONCLUSIONS In a large-sample targeted proteomics assay, we identified two proteins that improved risk prediction beyond the COPART risk score. The use of high-throughput proteomics assays may identify potential biomarkers for improved risk prediction in patients with PAD.
Collapse
|
43
|
Liang X, Wang L, Wang M, Liu Z, Liu X, Zhang B, Liu E, Li G. MicroRNA-124 inhibits macrophage cell apoptosis via targeting p38/MAPK signaling pathway in atherosclerosis development. Aging (Albany NY) 2020; 12:13005-13022. [PMID: 32611832 PMCID: PMC7377888 DOI: 10.18632/aging.103387] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
The objective of this study is to characterize the function of microRNA (miR)-124 in the process of coronary artery disease (CAD). Eighty patients, including 40 CAD patients and 40 non-CAD control patients were enrolled in this study. Atherosclerosis model was established in vivo in ApoE-/- mice and in vitro in RAW264.7 cells. Expression of miR-124 and p38 in patients, animal models and cell models were measured by qRT-PCR, western blot and immunohistochemistry assay. Overexpression or suppression of miR-124 was introduced in vitro and in vivo and the expression levels of p38, miR-124, pro- and anti-inflammatory cytokines, and pro- and anti-apoptotic factors were examined. Results showed that miR-124 was decreased, while p38 was increased in CAD patients and atherosclerosis models compared with control group. MiR-124 could target p38 by binding its 3’ untranslated region and negatively regulated the protein expression of p38. Overexpression of miR-124 increased the expression of anti-inflammatory cytokines, reduced the expression of pro- inflammatory cytokines, and inhibited macrophage apoptosis. MiR-124 overexpression may be a promising treatment for atherosclerosis and CAD via inhibiting p38.
Collapse
Affiliation(s)
- Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Lijun Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Manman Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhaohong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xing Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Baoshuai Zhang
- Department of Scientific Research, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Enzhao Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
44
|
Correlation between SHP-1 and carotid plaque vulnerability in humans. Cardiovasc Pathol 2020; 49:107258. [PMID: 32674045 DOI: 10.1016/j.carpath.2020.107258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Advanced atherosclerotic plaques tend to indicate an increased risk of cerebral ischemic events. SH2 domain-containing protein tyrosine phosphatase 1 (SHP-1) is a class I classical nonreceptor protein tyrosine phosphatase associated with plaque stability, as shown by analysis of a Gene Expression Omnibus (GEO) dataset showing differences in mRNA levels. However, the correlation between SHP-1 and human carotid plaque stability at the protein level remains unclear. METHODS AND RESULTS Thirty-nine carotid plaque tissue samples were acquired from 39 carotid artery stenosis patients after carotid endarterectomy. Hematoxylin and eosin, Masson trichrome, and CD68 staining was performed for pathological characterization, and immunohistochemical staining for SHP-1 was carried out. Within stable and unstable plaques, SHP-1 mainly accumulated in the necrotic area, plaque shoulder, and fibrous cap, similar to the distribution of CD68. A quantitative analysis of SHP-1 was carried out. The relative SHP-1-positive cell area was higher in the vulnerable group than in the stable group (P < .001). The number of symptomatic patients in the vulnerable group was no greater than that in the stable group (P = .098). Moreover, the integrated optical density (IOD)/area of SHP-1 was significantly higher in the vulnerable group than in the stable group (P < .001). Besides, SHP-1 colocalized with CD68 and vascular cell adhesion protein 1(VCAM-1). CONCLUSIONS We demonstrate that SHP-1 expression increases during carotid atherosclerotic plaque progression. The protein expression of SHP-1 was related to an increase in plaque instability in not only symptomatic but also asymptomatic patients with carotid artery stenosis. SHP-1 may play a role in atherosclerosis progression by macrophage polarization-mediated efferocytosis. Furthermore, SHP-1 may become a promising biomarker for plaque vulnerability in the future.
Collapse
|
45
|
Jinnouchi H, Sato Y, Sakamoto A, Cornelissen A, Mori M, Kawakami R, Gadhoke NV, Kolodgie FD, Virmani R, Finn AV. Calcium deposition within coronary atherosclerotic lesion: Implications for plaque stability. Atherosclerosis 2020; 306:85-95. [PMID: 32654790 DOI: 10.1016/j.atherosclerosis.2020.05.017] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerotic lesion progression is associated with intimal calcification. The earliest lesion that shows calcification is pathologic intimal thickening in which calcifications appear as microcalcifications that vary in size from <0.5 to 15 μm. The calcifications become larger as plaques progress, becoming punctate (>15 μm to 1 mm in diameter), fragmented (>1 mm), and eventually sheet-like calcification (>3 mm). When stratified by plaque type, maximum calcifications are observed in fibrocalcific plaques, followed by healed plaque ruptures. Lesions of acute thrombi, i.e., plaque rupture and erosions, which are the most frequent causes of acute coronary syndromes, show much less calcification than stable fibrocalcific plaques. Conversely, a calcified nodule, the least common lesion of acute thrombosis, occurs in highly calcified lesions. Pro-inflammatory cytokines observed in unstable plaques may provoke an early phase of osteogenic differentiation of smooth muscle cells (SMCs), a release of calcifying extracellular matrix vesicles, and/or induce apoptosis of macrophages and SMCs, which also calcify. Recent pathologic and imaging based studies indicate that lesions with dense calcifications are more likely to be stable plaques (fibrocalcific plaques), while micro, punctate, or fragmented calcifications are associated with either early stage plaques or unstable lesions (plaque rupture or erosion). Clinical non-invasive computed tomography (CT) studies show that the greater the calcium score, the higher the likelihood of patients developing future acute coronary events. This appears contradictory with the findings from pathologic autopsy studies. However, CT analysis of calcium subtypes is limited by resolution and blooming artifacts. Thus, areas of heavy calcification may not be the cause of future events as pathologic studies suggest. Rather, calcium may be an overall marker for the extent of disease. These types of discrepancies can perhaps be resolved by invasive or non-invasive high resolution imaging studies carried out at intervals in patients who present with acute coronary syndromes versus stable angina patients. Coronary calcium burden is greater in stable plaques than unstable plaques and there is a negative correlation between necrotic core area and area of calcification. Recent clinical studies have demonstrated that statins can reduce plaque burden by demonstrating a reduction in percent and total atheroma volume. However, calcification volume increases. In summary, pathologic studies show that sheet calcification is highly prevalent in stable plaques, while microcalcifications, punctate, and fragmented calcifications are more frequent in unstable lesions. Both pathologic and detailed analysis of imaging studies in living patients can resolve some of the controversies in our understanding of coronary calcification.
Collapse
Affiliation(s)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
The role of optical coherence tomography and other intravascular imaging modalities in cardiac allograft vasculopathy. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2020; 16:19-29. [PMID: 32368233 PMCID: PMC7189132 DOI: 10.5114/aic.2020.93909] [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] [Received: 10/30/2019] [Accepted: 02/13/2020] [Indexed: 11/17/2022] Open
Abstract
Orthotopic heart transplantation (OHT) is the standard-of-care for end-stage heart disease. Although a significant improvement in the prognosis of patients after OHT has been observed in recent years, their overall mortality remains relatively high, with a median survival of approximately 10 years after transplantation. One of the primary causes of death in patients after OHT is cardiac allograft vasculopathy (CAV), the condition developing specifically in the coronary vasculature after OHT, the pathophysiology of which is still inadequately known. It is estimated that CAV development and progression is responsible for approximately 30% of deaths within five years post-OHT. According to the International Society for Heart and Lung Transplantation (ISHLT) Nomenclature for CAV, its presence should be assessed primarily by the coronary angiography performed routinely after OHT, mostly due to its wide availability, reproducibility, and low complication rate. However, the analysis of CAV in coronary angiography has limitations, mostly concerning its - sometimes inadequate - sensitivity and specificity. Hence, there is a growing need for the introduction of more accurate methods of CAV assessment, such as intravascular imaging, which through a thorough evaluation of the arterial wall structure and thickness allows the drawbacks of routine angiography to be minimised. The aim of the article was to critically summarise the current findings derived from the analysis of CAV by optical coherence tomography, the other intravascular imaging modalities, such as intravascular ultrasound (IVUS) and IVUS-derived virtual histology, along with physiological assessment with the use of the fractional flow reserve.
Collapse
|
47
|
Jinnouchi H, Guo L, Sakamoto A, Torii S, Sato Y, Cornelissen A, Kuntz S, Paek KH, Fernandez R, Fuller D, Gadhoke N, Surve D, Romero M, Kolodgie FD, Virmani R, Finn AV. Diversity of macrophage phenotypes and responses in atherosclerosis. Cell Mol Life Sci 2020; 77:1919-1932. [PMID: 31720740 PMCID: PMC11104939 DOI: 10.1007/s00018-019-03371-3] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
The presence of macrophages within the plaque is a defining hallmark of atherosclerosis. Macrophages are exposed to various microenvironments such as oxidized lipids and cytokines which effect their phenotypic differentiation and activation. Classically, macrophages have been divided into two groups: M1 and M2 macrophages induced by T-helper 1 and T-helper 2 cytokines, respectively. However, for a decade, greater phenotypic heterogeneity and plasticity of these cells have since been reported in various models. In addition to M1 and M2 macrophage phenotypes, the concept of additional macrophage phenotypes such as M (Hb), Mox, and M4 has emerged. Understanding the mechanisms and functions of distinct phenotype of macrophages can lead to determination of their potential role in atherosclerotic plaque pathogenesis. However, there are still many unresolved controversies regarding their phenotype and function with respect to atherosclerosis. Here, we summarize and focus on the differential subtypes of macrophages in atherosclerotic plaques and their differing functional roles based upon microenvironments such as lipid, intraplaque hemorrhage, and plaque regression.
Collapse
Affiliation(s)
| | - Liang Guo
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Atsushi Sakamoto
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Sho Torii
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Yu Sato
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Anne Cornelissen
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Salome Kuntz
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Ka Hyun Paek
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Raquel Fernandez
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Daniela Fuller
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Neel Gadhoke
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Dipti Surve
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Maria Romero
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Frank D Kolodgie
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Renu Virmani
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Aloke V Finn
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
48
|
Abstract
Atherosclerosis is a chronic and most often progressive disease with a long clinically apparently silent period, and can become unstable at any time, due to a plaque rupture or erosion, leading to an acute atherothrombotic event. Atherosclerosis has a progression rate that is highly variable among patients and in the same patient. The progression of atherosclerotic plaque from asymptomatic to symptomatic phase depends on its structure and composition in which inflammation plays an essential role. Prototype of the ruptured plaque contains a large, soft, lipid-rich necrotic core with intraplaque hemorrhage that accounts for more than half of the volume of the plaque covered by a thin and inflamed fibrous cap with few smooth muscle cells, and a heavy infiltrate of inflammatory cells. Noninvasive imaging modalities might provide an assessment of the atherosclerotic disease process through the exploration of these plaque features. Computed tomography angiography and magnetic resonance imaging can characterize plaque morphology, whereas molecular imaging, owing to the high sensitivity of nuclear medicine for the detection of radiopharmaceuticals in tissues, allows to explore plaque biology. During the last 2 decades, FDG-PET imaging has also emerged as a powerful tool to explore noninvasively inflammatory activities in atherosclerotic plaques providing new insights on the evolution of metabolic activities in the vascular wall over time. This review highlights the role of PET imaging for the exploration of metabolic activities in atherosclerotic plaques. It will resume the evidence that have been gathered from clinical studies using FDG-PET and will discuss the perspectives of new radiopharmaceuticals for vulnerable plaque imaging.
Collapse
Affiliation(s)
- Olivier Lairez
- Cardiac Imaging Centre, Rangueil University Hospital, Toulouse, France
| | - Fabien Hyafil
- Department of Nuclear Medicine, Bichat University Hospital, Hôpitaux de Paris, Université René Diderot, Paris, France.
| |
Collapse
|
49
|
Vigne J, Hyafil F. Inflammation imaging to define vulnerable plaque or vulnerable patient. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2020; 64:21-34. [PMID: 32077668 DOI: 10.23736/s1824-4785.20.03231-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The role of nuclear imaging in the characterization of high-risk atherosclerotic plaque is increasing thanks to its high sensitivity to detect radiopharmaceuticals signal in tissues. Currently, 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) is the most studied and widely used radiopharmaceutical for the molecular imaging of atherosclerotic plaques with positron emission tomography (PET). [18F]FDG PET is a valuable tool to non-invasively detect, monitor and quantify inflammatory processes occurring in atherosclerotic plaques. The aim of this review is to gather insights provided by [18F]FDG PET to better understand the role of inflammation in the definitions of the vulnerable plaque and the vulnerable patient. Alternatives radiopharmaceuticals targeting inflammation and other potential high-risk plaque related processed are also discussed.
Collapse
Affiliation(s)
- Jonathan Vigne
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie University (UNICAEN), Caen, France -
- INSERM U1148, Laboratory for Vascular Translational Science (LVTS), DHU FIRE, University of Paris, Paris, France -
- Department of Pharmacy, CHU de Caen Normandie, Normandie University (UNICAEN), Caen, France -
| | - Fabien Hyafil
- INSERM U1148, Laboratory for Vascular Translational Science (LVTS), DHU FIRE, University of Paris, Paris, France
- Department of Nuclear Medicine, Bichat University Hospital, Paris, France
| |
Collapse
|
50
|
Effects of thrombospondin-4 on pro-inflammatory phenotype differentiation and apoptosis in macrophages. Cell Death Dis 2020; 11:53. [PMID: 31974349 PMCID: PMC6978349 DOI: 10.1038/s41419-020-2237-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/25/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
Thrombospondin-4 (TSP-4) attracted renewed attention recently as a result of assignment of new functions to this matricellular protein in cardiovascular, muscular, and nervous systems. We have previously reported that TSP-4 promotes local vascular inflammation in a mouse atherosclerosis model. A common variant of TSP-4, P387-TSP-4, was associated with increased cardiovascular disease risk in human population studies. In a mouse atherosclerosis model, TSP-4 had profound effect on accumulation of macrophages in lesions, which prompted us to examine its effects on macrophages in more detail. We examined the effects of A387-TSP-4 and P387-TSP-4 on mouse macrophages in cell culture and in vivo in the model of LPS-induced peritonitis. In tissues and in cell culture, TSP-4 expression was associated with inflammation: TSP-4 expression was upregulated in peritoneal tissues in LPS-induced peritonitis, and pro-inflammatory signals, INFγ, GM-CSF, and LPS, induced TSP-4 expression in macrophages in vivo and in cell culture. Deficiency in TSP-4 in macrophages from Thbs4−/− mice reduced the expression of pro-inflammatory macrophage markers, suggesting that TSP-4 facilitates macrophage differentiation into a pro-inflammatory phenotype. Expression of TSP-4, especially more active P387-TSP-4, was associated with higher cellular apoptosis. Cultured macrophages displayed increased adhesion to TSP-4 and reduced migration in presence of TSP-4, and these responses were further increased with P387 variant. We concluded that TSP-4 expression in macrophages increases their accumulation in tissues during the acute inflammatory process and supports macrophage differentiation into a pro-inflammatory phenotype. In a model of acute inflammation, TSP-4 supports pro-inflammatory macrophage apoptosis, a response that is closely related to their pro-inflammatory activity and release of pro-inflammatory signals. P387-TSP-4 was found to be the more active form of TSP-4 in all examined functions.
Collapse
|