1
|
Yang M, Liu X, Jiang M, Hu J, Xiao Z. TAX1BP1/A20 inhibited TLR2-NF-κB activation to induce tolerant expression of IL-6 in endothelial cells. Int Immunopharmacol 2024; 139:112789. [PMID: 39079200 DOI: 10.1016/j.intimp.2024.112789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 09/25/2024]
Abstract
The inflammatory cascadedriven by interleukin-6 (IL-6) plays a crucial role in the initiation and progression of chronic inflammatory conditions such as atherosclerosis. Research has demonstrated that prolonged exposure to inflammatory stimuli leads to the development of "immune tolerance" in specialized immune cells such as monocytes and macrophages, serving as a mechanism to prevent tissue damage and curb the inflammatory cascade. However, our recent investigation revealed that immune tolerance did not effectively regulate the production of IL-6 in human umbilical vein endothelial cells (HUVECs) when stimulated by a Toll-like receptor 2 (TLR2) ligand Pam3CSK4, which is a potent activator of the pro-inflammatory transcription factor NF-κB. Furthermore, the negative regulator of NF-κB signaling, A20, was ineffective in suppressing TLR2-induced IL-6 synthesis in this context. Notably, all A20 auxiliary molecules, with the exception of TAX1BP1, were found to be significantly expressed in HUVECs. DNA methylation in TAX1BP1 was confirmed in GEO database. According to the information provided, it is hypothesized that altered DNA methylation in HUVECs could potentially lead to decreased expression of TAX1BP1, thereby impeding A20's capacity to modulate continuous activation of the TLR2-NF-κB pathway. This may consequently lead to unregulated production of IL-6, evading immune tolerance mechanisms. Subsequent investigations suggested that demethylating TAX1BP1 could enhance its expression, potentially reducing the endogenous IL-6 levels induced by repeated TLR2 stimulation and restoring A20's inhibitory role in NF-κB signaling. Additionally, over-expression of TAX1BP1 coulddecrease the production of atherosclerosis-associated cytokines like IL-6, MCP-1, ICAM-1, and VCAM-1, while increasing NO release following repeated Pam3cks4 stimulation, along with enhanced co-localization of TAX1BP1 and A20. These findings indicate that inducing immune tolerance in endothelial cells may effectively suppress endogenous IL-6 production and halt the IL-6-mediated inflammatory cascade, with TAX1BP1/A20 identified as crucial components in this process.These insights provide novel perspectives and potential targets for therapeutic strategies in inflammatoryimmunological disorders involving the overproduction of IL-6.
Collapse
Affiliation(s)
- Mei Yang
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Zhilin Xiao
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Tabatabaei FS, Shafeghat M, Azimi A, Akrami A, Rezaei N. Endosomal Toll-Like Receptors intermediate negative impacts of viral diseases, autoimmune diseases, and inflammatory immune responses on the cardiovascular system. Expert Rev Clin Immunol 2024:1-13. [PMID: 39137281 DOI: 10.1080/1744666x.2024.2392815] [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: 04/30/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of morbidity globally, with chronic inflammation as a key modifiable risk factor. Toll-like receptors (TLRs), pivotal components of the innate immune system, including TLR-3, -7, -8, and -9 within endosomes, trigger intracellular cascades, leading to inflammatory cytokine production by various cell types, contributing to systemic inflammation and atherosclerosis. Recent research highlights the role of endosomal TLRs in recognizing self-derived nucleic acids during sterile inflammation, implicated in autoimmune conditions like myocarditis. AREAS COVERED This review explores the impact of endosomal TLRs on viral infections, autoimmunity, and inflammatory responses, shedding light on their intricate involvement in cardiovascular health and disease by examining literature on TLR-mediated mechanisms and their roles in CVD pathophysiology. EXPERT OPINION Removal of endosomal TLRs mitigates myocardial damage and immune reactions, applicable in myocardial injury. Targeting TLRs with agonists enhances innate immunity against fatal viruses, lowering viral loads and mortality. Prophylactic TLR agonist administration upregulates TLRs, protecting against fatal viruses and improving survival. TLRs play a complex role in CVDs like atherosclerosis and myocarditis, with therapeutic potential in modulating TLR reactions for cardiovascular health.
Collapse
Affiliation(s)
- Fatemeh Sadat Tabatabaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melika Shafeghat
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Azimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashley Akrami
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
3
|
Molinaro R, Sellar RS, Vromman A, Sausen G, Folco E, Sukhova GK, McConke ME, Corbo C, Ebert BL, Libby P. The clonal hematopoiesis mutation Jak2 V617F aggravates endothelial injury and thrombosis in arteries with erosion-like intimas. Int J Cardiol 2024; 409:132184. [PMID: 38759798 DOI: 10.1016/j.ijcard.2024.132184] [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: 04/07/2023] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Superficial plaque erosion causes many acute coronary syndromes. However, mechanisms of plaque erosion remain poorly understood, and we lack directed therapeutics for thrombotic complication. Human eroded plaques can harbor neutrophil extracellular traps (NETs) that propagate endothelial damage at experimental arterial lesions that recapitulate superficial erosion. Clonal Hematopoiesis of Indeterminate Potential (CHIP) denotes age-related clonal expansion of bone marrow-derived cells harboring somatic mutations in the absence of overt hematological disease. CHIP heightens the risk of cardiovascular disease, with the greatest increase seen in individuals with JAK2V617F. Neutrophils from mice and humans with JAK2V617F undergo NETosis more readily than Jak2WT (wild-type) cells. We hypothesized that JAK2V617F, by increasing propensity to NETosis, exacerbates aspects of superficial erosion. METHODS AND RESULTS We generated Jak2V617F and Jak2WT mice with heterozygous Jak2V617F in myeloid cells. We induced areas of denuded endothelium that recapitulate features of superficial erosion and assessed endothelial integrity, cellular composition of the erosion, thrombosis rates, and response to ruxolitinib, a clinically available JAK1/2 inhibitor, in relation to genotype. Following experimental erosion, Jak2V617F mice have greater impairment of endothelial barrier function and increased rates of arterial thrombosis. Neointimas in Jak2V617F mice exhibit increased apoptosis, NETosis, and platelet recruitment. Jak2V617F mice treated with ruxolitinib show increased endothelial continuity and reduced apoptosis in the neointima comparable to levels in Jak2WT. CONCLUSIONS These observations provide new mechanistic insight into the pathophysiology of superficial erosion, the heightened risk for myocardial infarction in JAK2V617F CHIP, and point the way to personalized therapeutics based on CHIP status.
Collapse
Affiliation(s)
- Roberto Molinaro
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, London, UK; Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Amélie Vromman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Grasiele Sausen
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Eduardo Folco
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Galina K Sukhova
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Marie E McConke
- Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Claudia Corbo
- University of Milano-Bicocca, Department of Medicine and Surgery, NANOMIB Center, Monza 20900, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan 20161, Italy
| | - Benjamin L Ebert
- Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
4
|
Bruoha S, Galli M, Sabouret P, Yosefy C, Taha L, Gragnano F, Savage MP, Shuvy M, Biondi-Zoccai G, Glikson M, Asher E. Atherosclerotic Plaque Erosion: Mechanisms, Clinical Implications, and Potential Therapeutic Strategies-A Review. J Cardiovasc Pharmacol 2024; 83:547-556. [PMID: 38421206 DOI: 10.1097/fjc.0000000000001554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
ABSTRACT Atherosclerosis is an insidious and progressive inflammatory disease characterized by the formation of lipid-laden plaques within the intima of arterial walls with potentially devastating consequences. While rupture of vulnerable plaques has been extensively studied, a distinct mechanism known as plaque erosion (PE) has gained recognition and attention in recent years. PE, characterized by the loss of endothelial cell lining in the presence of intact fibrous cap, contributes to a significant and growing proportion of acute coronary events. However, despite a heterogeneous substrate underlying coronary thrombosis, treatment remains identical. This article provides an overview of atherosclerotic PE characteristics and its underlying mechanisms, highlights its clinical implications, and discusses potential therapeutic strategies.
Collapse
Affiliation(s)
- Sharon Bruoha
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Pierre Sabouret
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- National College of French Cardiologists, 13 rue Niepce, 75014 Paris, France
| | - Chaim Yosefy
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Louay Taha
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Michael P Savage
- Division of Cardiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mony Shuvy
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy ; and
- Mediterranea Cardiocentro, Naples, Italy
| | - Michael Glikson
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Elad Asher
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| |
Collapse
|
5
|
Kidder E, Gangopadhyay S, Francis S, Alfaidi M. "How to Release or Not Release, That Is the Question." A Review of Interleukin-1 Cellular Release Mechanisms in Vascular Inflammation. J Am Heart Assoc 2024; 13:e032987. [PMID: 38390810 PMCID: PMC10944040 DOI: 10.1161/jaha.123.032987] [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: 10/06/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, characterized by atherosclerotic activity within large and medium-sized arteries. Inflammation has been shown to be a primary driver of atherosclerotic plaque formation, with interleukin-1 (IL-1) having a principal role. This review focuses on the current state of knowledge of molecular mechanisms of IL-1 release from cells in atherosclerotic plaques. A more in-depth understanding of the process of IL-1's release into the vascular environment is necessary for the treatment of inflammatory disease processes, as the current selection of medicines being used primarily target IL-1 after it has been released. IL-1 is secreted by several heterogenous mechanisms, some of which are cell type-specific and could provide further specialized targets for therapeutic intervention. A major unmet challenge is to understand the mechanism before and leading to IL-1 release, especially by cells in atherosclerotic plaques, including endothelial cells, vascular smooth muscle cells, and macrophages. Data so far indicate a heterogeneity of IL-1 release mechanisms that vary according to cell type and are stimulus-dependent. Unraveling this complexity may reveal new targets to block excess vascular inflammation.
Collapse
Affiliation(s)
- Evan Kidder
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| | - Siddhartha Gangopadhyay
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| | - Sheila Francis
- School of Medicine and Population HealthUniversity of SheffieldSheffieldUK
| | - Mabruka Alfaidi
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| |
Collapse
|
6
|
Libby P. Inflammation and the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 154:107255. [PMID: 38157682 DOI: 10.1016/j.vph.2023.107255] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
The notion that inflammation contributes to atherosclerosis has now gained considerable currency. Inflammation participates in atherosclerosis inception, progression, and thrombotic complications. Induced expression of endothelial leukocyte adhesion molecules and chemoattractant cytokines recruit blood cells to the arterial intima. Lesions can contain virtually every type of leukocyte. Monocytes mature into macrophages and imbibe lipids becoming foam cells, a hallmark of the atherosclerotic lesion. T lymphocytes can instruct the more numerous macrophages to express genes involved in the progression of the atheroma and its eventual destabilization. Inflammation is becoming clinically actionable to refine risk prediction, allocate treatments, and as a therapeutic target.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
7
|
Zheng S, Liu Z, Liu H, Lim JY, Li DWH, Zhang S, Luo F, Wang X, Sun C, Tang R, Zheng W, Xie Q. Research development on gut microbiota and vulnerable atherosclerotic plaque. Heliyon 2024; 10:e25186. [PMID: 38384514 PMCID: PMC10878880 DOI: 10.1016/j.heliyon.2024.e25186] [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: 05/31/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
The relationship between gut microbiota and its metabolites with cardiovascular disease (CVD) has been proven. In this review, we aim to conclude the potential mechanism of gut microbiota and its metabolites on inducing the formation of vulnerable atherosclerotic plaque, and to discuss the effect of intestinal metabolites, including trimethylamine-N-oxide (TMAO), lipopolysaccharide (LPS), phenylacetylglutamine (PAG), short-chain fatty acids (SCFAs) on plaque stability. Finally, we include the impact of gut microbiota and its metabolites on plaque stability, to propose a new therapeutic direction for coronary heart disease. Gut microbiota regulation intervenes the progress of arteriosclerosis, especially on coronary atherosclerosis, by avoiding or reducing the formation of vulnerable plaque, to lower the morbidity rate of myocardial infarction.
Collapse
Affiliation(s)
- Shujiao Zheng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Zuheng Liu
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic Testing, The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Ying Lim
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Dolly Wong Hui Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shaofeng Zhang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fang Luo
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiujing Wang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changqing Sun
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rong Tang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wuyang Zheng
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qiang Xie
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| |
Collapse
|
8
|
Drysdale A, Blanco-Lopez M, White SJ, Unsworth AJ, Jones S. Differential Proteoglycan Expression in Atherosclerosis Alters Platelet Adhesion and Activation. Int J Mol Sci 2024; 25:950. [PMID: 38256024 PMCID: PMC10815981 DOI: 10.3390/ijms25020950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Proteoglycans are differentially expressed in different atherosclerotic plaque phenotypes, with biglycan and decorin characteristic of ruptured plaques and versican and hyaluronan more prominent in eroded plaques. Following plaque disruption, the exposure of extracellular matrix (ECM) proteins triggers platelet adhesion and thrombus formation. In this study, the impact of differential plaque composition on platelet function and thrombus formation was investigated. Platelet adhesion, activation and thrombus formation under different shear stress conditions were assessed in response to individual proteoglycans and composites representing different plaque phenotypes. The results demonstrated that all the proteoglycans tested mediated platelet adhesion but not platelet activation, and the extent of adhesion observed was significantly lower than that observed with type I and type III collagens. Thrombus formation upon the rupture and erosion ECM composites was significantly reduced (p < 0.05) compared to relevant collagen alone, indicating that proteoglycans negatively regulate platelet collagen responses. This was supported by results demonstrating that the addition of soluble biglycan or decorin to whole blood markedly reduced thrombus formation on type I collagen (p < 0.05). Interestingly, thrombus formation upon the erosion composite displayed aspirin sensitivity, whereas the rupture composite was intensive to aspirin, having implications for current antiplatelet therapy regimes. In conclusion, differential platelet responses and antiplatelet efficacy are observed on ECM composites phenotypic of plaque rupture and erosion. Proteoglycans inhibit thrombus formation and may offer a novel plaque-specific approach to limit arterial thrombosis.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Maria Blanco-Lopez
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Stephen J. White
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK;
| | - Amanda J. Unsworth
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| |
Collapse
|
9
|
Shin J, Hong J, Edwards-Glenn J, Krukovets I, Tkachenko S, Adelus ML, Romanoski CE, Rajagopalan S, Podrez E, Byzova TV, Stenina-Adongravi O, Cherepanova OA. Unraveling the Role of Sex in Endothelial Cell Dysfunction: Evidence From Lineage Tracing Mice and Cultured Cells. Arterioscler Thromb Vasc Biol 2024; 44:238-253. [PMID: 38031841 PMCID: PMC10842863 DOI: 10.1161/atvbaha.123.319833] [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: 07/21/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Biological sex differences play a vital role in cardiovascular diseases, including atherosclerosis. The endothelium is a critical contributor to cardiovascular pathologies since endothelial cells (ECs) regulate vascular tone, redox balance, and inflammatory reactions. Although EC activation and dysfunction play an essential role in the early and late stages of atherosclerosis development, little is known about sex-dependent differences in EC. METHODS We used human and mouse aortic EC as well as EC-lineage tracing (Cdh5-CreERT2 Rosa-YFP [yellow fluorescence protein]) atherosclerotic Apoe-/- mice to investigate the biological sexual dimorphism of the EC functions in vitro and in vivo. Bioinformatics analyses were performed on male and female mouse aortic EC and human lung and aortic EC. RESULTS In vitro, female human and mouse aortic ECs showed more apoptosis and higher cellular reactive oxygen species levels than male EC. In addition, female mouse aortic EC had lower mitochondrial membrane potential (ΔΨm), lower TFAM (mitochondrial transcription factor A) levels, and decreased angiogenic potential (tube formation, cell viability, and proliferation) compared with male mouse aortic EC. In vivo, female mice had significantly higher lipid accumulation within the aortas, impaired glucose tolerance, and lower endothelial-mediated vasorelaxation than males. Using the EC-lineage tracing approach, we found that female lesions had significantly lower rates of intraplaque neovascularization and endothelial-to-mesenchymal transition within advanced atherosclerotic lesions but higher incidents of missing EC lumen coverage and higher levels of oxidative products and apoptosis. RNA-seq analyses revealed that both mouse and human female EC had higher expression of genes associated with inflammation and apoptosis and lower expression of genes related to angiogenesis and oxidative phosphorylation than male EC. CONCLUSIONS Our study delineates critical sex-specific differences in EC relevant to proinflammatory, pro-oxidant, and angiogenic characteristics, which are entirely consistent with a vulnerable phenotype in females. Our results provide a biological basis for sex-specific proatherosclerotic mechanisms.
Collapse
Affiliation(s)
- Junchul Shin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jonnelle Edwards-Glenn
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Irene Krukovets
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Maria L. Adelus
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
- Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, USA
| | - Casey E. Romanoski
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Sanjay Rajagopalan
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Eugene Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tatiana V. Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga Stenina-Adongravi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga A. Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
10
|
Kohs TC, Vu HH, Jordan KR, Parra-Izquierdo I, Hinds MT, Shatzel JJ, Kievit P, Morgan TK, Yunga ST, Ngo TT, Aslan JE, Wallisch M, Lorentz CU, Tucker EI, Gailani D, Lindner JR, Puy C, McCarty OJ. Activation of coagulation FXI promotes endothelial inflammation and amplifies platelet activation in a nonhuman primate model of hyperlipidemia. Res Pract Thromb Haemost 2024; 8:102276. [PMID: 38226339 PMCID: PMC10788631 DOI: 10.1016/j.rpth.2023.102276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 01/17/2024] Open
Abstract
Background Hyperlipidemia is associated with chronic inflammation and thromboinflammation. This is an underlying cause of several cardiovascular diseases, including atherosclerosis. In diseased blood vessels, rampant thrombin generation results in the initiation of the coagulation cascade, activation of platelets, and endothelial cell dysfunction. Coagulation factor (F) XI represents a promising therapeutic target to reduce thromboinflammation, as it is uniquely positioned at an intersection between inflammation and thrombin generation. Objectives This study aimed to investigate the role of FXI in promoting platelet and endothelial cell activation in a model of hyperlipidemia. Methods Nonhuman primates (NHPs) were fed a standard chow diet (lean, n = 6) or a high-fat diet (obese, n = 8) to establish a model of hyperlipidemia. Obese NHPs were intravenously administered a FXI blocking antibody (2 mg/kg) and studied at baseline and at 1, 7, 14, 21, and 28 days after drug administration. Platelet activation and inflammatory markers were measured using fluorescence-activated cell sorting or enzyme-linked immunosorbent assay. Molecular imaging was used to quantify vascular cell adhesion molecule 1 (VCAM-1) expression at the carotid bifurcation. Results Obese NHPs demonstrated increased sensitivity for platelet P-selectin expression and phosphatidylserine exposure in response to platelet GPVI or PAR agonists compared with lean NHPs. Obese NHPs exhibited elevated levels of C-reactive protein, cathepsin D, and myeloperoxidase compared with lean NHPs. Following pharmacological inhibition of FIX activation by FXIa, platelet priming for activation by GPVI or PAR agonists, C-reactive protein levels, and endothelial VCAM-1 levels were reduced in obese NHPs. Conclusion FXI activation promotes the proinflammatory phenotype of hyperlipidemia by priming platelet activation and inciting endothelial cell dysfunction.
Collapse
Affiliation(s)
- Tia C.L. Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Helen H. Vu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Kelley R. Jordan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Iván Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Terry K. Morgan
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Samuel Tassi Yunga
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Thuy T.M. Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E. Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael Wallisch
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - Christina U. Lorentz
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - Erik I. Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan R. Lindner
- Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Institute, University of Virginia, Charlottesville, Virginia, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
11
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Gerhardt T, Seppelt C, Abdelwahed YS, Meteva D, Wolfram C, Stapmanns P, Erbay A, Zanders L, Nelles G, Musfeld J, Sieronski L, Stähli BE, Montone RA, Vergallo R, Haghikia A, Skurk C, Knebel F, Dreger H, Trippel TD, Rai H, Joner M, Klotsche J, Libby P, Crea F, Kränkel N, Landmesser U, Leistner DM. Culprit plaque morphology determines inflammatory risk and clinical outcomes in acute coronary syndrome. Eur Heart J 2023; 44:3911-3925. [PMID: 37381774 DOI: 10.1093/eurheartj/ehad334] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023] Open
Abstract
AIMS Rupture of the fibrous cap (RFC) and erosion of an intact fibrous cap (IFC) are the two predominant mechanisms causing acute coronary syndromes (ACS). It is uncertain whether clinical outcomes are different following RFC-ACS vs. IFC-ACS and whether this is affected by a specific inflammatory response. The prospective, translational OPTIcal-COherence Tomography in Acute Coronary Syndrome study programme investigates the impact of the culprit lesion phenotype on inflammatory profiles and prognosis in ACS patients. METHODS AND RESULTS This analysis included 398 consecutive ACS patients, of which 62% had RFC-ACS and 25% had IFC-ACS. The primary endpoint was a composite of cardiac death, recurrent ACS, hospitalization for unstable angina, and target vessel revascularization at 2 years [major adverse cardiovascular events (MACE+)]. Inflammatory profiling was performed at baseline and after 90 days. Patients with IFC-ACS had lower rates of MACE+ than those with RFC-ACS (14.3% vs. 26.7%, P = 0.02). In 368-plex proteomic analyses, patients with IFC-ACS showed lower inflammatory proteome expression compared with those with RFC-ACS, including interleukin-6 and proteins associated with the response to interleukin-1β. Circulating plasma levels of interleukin-1β decreased from baseline to 3 months following IFC-ACS (P < 0.001) but remained stable following RFC-ACS (P = 0.25). Interleukin-6 levels decreased in patients with RFC-ACS free of MACE+ (P = 0.01) but persisted high in those with MACE+. CONCLUSION This study demonstrates a distinct inflammatory response and a lower risk of MACE+ following IFC-ACS. These findings advance our understanding of inflammatory cascades associated with different mechanisms of plaque disruption and provide hypothesis generating data for personalized anti-inflammatory therapeutic allocation to ACS patients, a strategy that merits evaluation in future clinical trials.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, USA
| | - Claudio Seppelt
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Denitsa Meteva
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Christopher Wolfram
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
| | - Philip Stapmanns
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
| | - Aslihan Erbay
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Lukas Zanders
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Gregor Nelles
- Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Johanna Musfeld
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
| | - Lara Sieronski
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Barbara E Stähli
- Klinik für Kardiologie, Universitäres Herzzentrum, Universitätsspital Zürich, Zurich, Switzerland
| | - Rocco A Montone
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rocco Vergallo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Fabian Knebel
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Cardiology, Charité University Medicine Berlin, Campus Charité Mitte (CCM), Berlin 10117, Germany
- Sana Klinikum Lichtenberg, Innere Medizin II: Schwerpunkt Kardiologie, Berlin, Germany
| | - Henryk Dreger
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Cardiology, Charité University Medicine Berlin, Campus Charité Mitte (CCM), Berlin 10117, Germany
| | - Tobias D Trippel
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Cardiology, Charité University Medicine Berlin, Campus Virchow Clinic (CVK), Berlin 13353, Germany
| | - Himanshu Rai
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, 80636 Munich, Germany
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Michael Joner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Munich, Munich 80636, Germany
| | - Jens Klotsche
- German Rheumatism Research Center Berlin, and Institute for Social Medicine, Epidemiology und Health Economy, Charité University Medicine Berlin, Campus Charité Mitte, Berlin 10117, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Filippo Crea
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Nicolle Kränkel
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - David M Leistner
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, Germany and Berlin Institute of Health (BIH), Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| |
Collapse
|
13
|
Diez Benavente E, Karnewar S, Buono M, Mili E, Hartman RJ, Kapteijn D, Slenders L, Daniels M, Aherrahrou R, Reinberger T, Mol BM, de Borst GJ, de Kleijn DP, Prange KH, Depuydt MA, de Winther MP, Kuiper J, Björkegren JL, Erdmann J, Civelek M, Mokry M, Owens GK, Pasterkamp G, den Ruijter HM. Female Gene Networks Are Expressed in Myofibroblast-Like Smooth Muscle Cells in Vulnerable Atherosclerotic Plaques. Arterioscler Thromb Vasc Biol 2023; 43:1836-1850. [PMID: 37589136 PMCID: PMC10521798 DOI: 10.1161/atvbaha.123.319325] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Women presenting with coronary artery disease more often present with fibrous atherosclerotic plaques, which are currently understudied. Phenotypically modulated smooth muscle cells (SMCs) contribute to atherosclerosis in women. How these phenotypically modulated SMCs shape female versus male plaques is unknown. METHODS Gene regulatory networks were created using RNAseq gene expression data from human carotid atherosclerotic plaques. The networks were prioritized based on sex bias, relevance for smooth muscle biology, and coronary artery disease genetic enrichment. Network expression was linked to histologically determined plaque phenotypes. In addition, their expression in plaque cell types was studied at single-cell resolution using single-cell RNAseq. Finally, their relevance for disease progression was studied in female and male Apoe-/- mice fed a Western diet for 18 and 30 weeks. RESULTS Here, we identify multiple sex-stratified gene regulatory networks from human carotid atherosclerotic plaques. Prioritization of the female networks identified 2 main SMC gene regulatory networks in late-stage atherosclerosis. Single-cell RNA sequencing mapped these female networks to 2 SMC phenotypes: a phenotypically modulated myofibroblast-like SMC network and a contractile SMC network. The myofibroblast-like network was mostly expressed in plaques that were vulnerable in women. Finally, the mice ortholog of key driver gene MFGE8 (milk fat globule EGF and factor V/VIII domain containing) showed retained expression in advanced plaques from female mice but was downregulated in male mice during atherosclerosis progression. CONCLUSIONS Female atherosclerosis is characterized by gene regulatory networks that are active in fibrous vulnerable plaques rich in myofibroblast-like SMCs.
Collapse
Affiliation(s)
- Ernest Diez Benavente
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center (S.K., G.K.O.), University of Virginia, Charlottesville
| | - Michele Buono
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Eloi Mili
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Robin J.G. Hartman
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Daniek Kapteijn
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Lotte Slenders
- Central Diagnostic Laboratory (L.S., M.M., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Mark Daniels
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Redouane Aherrahrou
- Center for Public Health Genomics (R.A., M.C.), University of Virginia, Charlottesville
- Institute for Cardiogenetics, University of Lübeck, Germany (R.A., T.R., J.E.)
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland (R.A.)
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Germany (R.A., T.R., J.E.)
| | - Barend M. Mol
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Dominique P.V. de Kleijn
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Koen H.M. Prange
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers — location AMC, University of Amsterdam, Netherlands (K.H.M.P., M.P.J.d.W.)
| | - Marie A.C. Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.A.C.D., J.K.)
| | - Menno P.J. de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers — location AMC, University of Amsterdam, Netherlands (K.H.M.P., M.P.J.d.W.)
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.A.C.D., J.K.)
| | - Johan L.M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York (J.L.M.B.)
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden (J.L.M.B.)
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Germany (R.A., T.R., J.E.)
| | - Mete Civelek
- Center for Public Health Genomics (R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (M.C.)
- University of Virginia, Charlottesville (M.C.)
| | - Michal Mokry
- Central Diagnostic Laboratory (L.S., M.M., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center (S.K., G.K.O.), University of Virginia, Charlottesville
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory (L.S., M.M., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology (E.D.B., M.B., E.M., R.J.G.H., D.K., M.D., H.M.d.R.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| |
Collapse
|
14
|
O’Riordan C, Savage E, Newell M, Flaherty G, Hartigan MSc I. Cardiovascular Disease Risk Factor Profile of Experienced Male Amateur Marathon Runners: A Systematic Review. Sports Health 2023; 15:661-672. [PMID: 37249222 PMCID: PMC10467474 DOI: 10.1177/19417381231176534] [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] [Indexed: 05/31/2023] Open
Abstract
CONTEXT Male amateur marathon runners represent a unique subset of the population who may be at increased risk of cardiovascular disease (CVD) due to their underlying risk factors and their involvement in vigorous exercise such as marathon running. OBJECTIVE To assess the modifiable risk factors (MRFs) of CVD in experienced male amateur marathon runners and health interventions on CVD risk factors. DATA SOURCES CINAHL, Cochrane Library, Embase, Medline, and SPORTDiscus. STUDY SELECTION Studies selected according to the inclusion criteria. STUDY DESIGN Systematic review. LEVEL OF EVIDENCE Level 3. DATA EXTRACTION The publication dates included were from June 1, 2008 to February 29, 2020.Published primary epidemiological, observational, randomized controlled trial (RCT) and/or non-RCT studies assessing the MRFs of CVD and health interventions on CVD risk factors in male amateur marathon runners aged ≥18 years and written in the English language were included in the review. RESULTS Five studies met the inclusion criteria for analysis. These included male amateur marathon runners (n = 862), aged 42 to 77 years. Hypertension, hyperlipidemia, smoking, and alcohol use were MRFs positively associated with an increased risk of coronary atherosclerosis found in a subset of male marathon runners. No studies examined health interventions on CVD risk factors in any of the included studies. All 5 studies were of good quality from the National Heart, Lung, and Blood Institute quality assessment tools used. The risk of bias was low to moderate. CONCLUSION There is a paucity of observational studies evaluating the CVD MRFs. Negative lifestyle behaviors exist within this population despite their engagement in physical exercise through marathon running. Marathon running does not negate the long-term effects caused by past negative lifestyle behaviors. This systematic review identifies that this population may not be aware of their possible risk of atherosclerosis and, consequently, CVD.
Collapse
Affiliation(s)
- Catherine O’Riordan
- School of Nursing and Midwifery, University College Cork, Ireland
- National Institute for Prevention and Cardiovascular Health, Croí Heart and Stroke Centre, Newcastle, Galway, Ireland
| | - Eileen Savage
- School of Nursing and Midwifery, University College Cork, Ireland
| | - Micheál Newell
- School of Medicine, University of Galway, Ireland
- National Institute for Prevention and Cardiovascular Health, Croí Heart and Stroke Centre, Newcastle, Galway, Ireland
| | - Gerard Flaherty
- School of Medicine, University of Galway, Ireland
- National Institute for Prevention and Cardiovascular Health, Croí Heart and Stroke Centre, Newcastle, Galway, Ireland
| | | |
Collapse
|
15
|
Satta S, Beal R, Smith R, Luo X, Ferris GR, Langford-Smith A, Teasdale J, Ajime TT, Serré J, Hazell G, Newby GS, Johnson JL, Kurinna S, Humphries MJ, Gayan-Ramirez G, Libby P, Degens H, Yu B, Johnson T, Alexander Y, Jia H, Newby AC, White SJ. A Nrf2-OSGIN1&2-HSP70 axis mediates cigarette smoke-induced endothelial detachment: implications for plaque erosion. Cardiovasc Res 2023; 119:1869-1882. [PMID: 36804807 PMCID: PMC10405570 DOI: 10.1093/cvr/cvad022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 01/05/2023] [Indexed: 02/20/2023] Open
Abstract
AIMS Endothelial erosion of plaques is responsible for ∼30% of acute coronary syndromes (ACS). Smoking is a risk factor for plaque erosion, which most frequently occurs on the upstream surface of plaques where the endothelium experiences elevated shear stress. We sought to recreate these conditions in vitro to identify potential pathological mechanisms that might be of relevance to plaque erosion. METHODS AND RESULTS Culturing human coronary artery endothelial cells (HCAECs) under elevated flow (shear stress of 7.5 Pa) and chronically exposing them to cigarette smoke extract (CSE) and tumour necrosis factor-alpha (TNFα) recapitulated a defect in HCAEC adhesion, which corresponded with augmented Nrf2-regulated gene expression. Pharmacological activation or adenoviral overexpression of Nrf2 triggered endothelial detachment, identifying Nrf2 as a mediator of endothelial detachment. Growth/Differentiation Factor-15 (GDF15) expression was elevated in this model, with protein expression elevated in the plasma of patients experiencing plaque erosion compared with plaque rupture. The expression of two Nrf2-regulated genes, OSGIN1 and OSGIN2, was increased by CSE and TNFα under elevated flow and was also elevated in the aortas of mice exposed to cigarette smoke in vivo. Knockdown of OSGIN1&2 inhibited Nrf2-induced cell detachment. Overexpression of OSGIN1&2 induced endothelial detachment and resulted in cell cycle arrest, induction of senescence, loss of focal adhesions and actin stress fibres, and disturbed proteostasis mediated in part by HSP70, restoration of which reduced HCAEC detachment. In ACS patients who smoked, blood concentrations of HSP70 were elevated in plaque erosion compared with plaque rupture. CONCLUSION We identified a novel Nrf2-OSGIN1&2-HSP70 axis that regulates endothelial adhesion, elevated GDF15 and HSP70 as biomarkers for plaque erosion in patients who smoke, and two therapeutic targets that offer the potential for reducing the risk of plaque erosion.
Collapse
Affiliation(s)
- Sandro Satta
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Robert Beal
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Rhys Smith
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Xing Luo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, & The Key Laboratory of Medical Ischemia, Chinese Ministry of Education, Harbin 150086, China
| | - Glenn R Ferris
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Alex Langford-Smith
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Jack Teasdale
- Bristol Medical School, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Tom Tanjeko Ajime
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jef Serré
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Georgina Hazell
- Bristol Medical School, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Graciela Sala Newby
- Bristol Medical School, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Jason L Johnson
- Bristol Medical School, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Svitlana Kurinna
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Peter Libby
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hans Degens
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
- Institute of Sport Science and Innovations, Lithuanian Sports University, Sporto g. 6, LT-44221 Kaunas, Lithuania
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, & The Key Laboratory of Medical Ischemia, Chinese Ministry of Education, Harbin 150086, China
| | - Thomas Johnson
- Department of Cardiology, Bristol Heart Institute, Upper Maudlin St., Bristol BS2 8HW, UK
| | - Yvonne Alexander
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Haibo Jia
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, & The Key Laboratory of Medical Ischemia, Chinese Ministry of Education, Harbin 150086, China
| | - Andrew C Newby
- Bristol Medical School, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Stephen J White
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| |
Collapse
|
16
|
Gou T, Hu M, Xu M, Chen Y, Chen R, Zhou T, Liu J, Guo L, Ao H, Ye Q. Novel wine in an old bottle: Preventive and therapeutic potentials of andrographolide in atherosclerotic cardiovascular diseases. J Pharm Anal 2023; 13:563-589. [PMID: 37440909 PMCID: PMC10334359 DOI: 10.1016/j.jpha.2023.05.010] [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: 11/30/2022] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/15/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) frequently results in sudden death and poses a serious threat to public health worldwide. The drugs approved for the prevention and treatment of ASCVD are usually used in combination but are inefficient owing to their side effects and single therapeutic targets. Therefore, the use of natural products in developing drugs for the prevention and treatment of ASCVD has received great scholarly attention. Andrographolide (AG) is a diterpenoid lactone compound extracted from Andrographis paniculata. In addition to its use in conditions such as sore throat, AG can be used to prevent and treat ASCVD. It is different from drugs that are commonly used in the prevention and treatment of ASCVD and can not only treat obesity, diabetes, hyperlipidaemia and ASCVD but also inhibit the pathological process of atherosclerosis (AS) including lipid accumulation, inflammation, oxidative stress and cellular abnormalities by regulating various targets and pathways. However, the pharmacological mechanisms of AG underlying the prevention and treatment of ASCVD have not been corroborated, which may hinder its clinical development and application. Therefore, this review summarizes the physiological and pathological mechanisms underlying the development of ASCVD and the in vivo and in vitro pharmacological effects of AG on the relative risk factors of AS and ASCVD. The findings support the use of the old pharmacological compound ('old bottle') as a novel drug ('novel wine') for the prevention and treatment of ASCVD. Additionally, this review summarizes studies on the availability as well as pharmaceutical and pharmacokinetic properties of AG, aiming to provide more information regarding the clinical application and further research and development of AG.
Collapse
Affiliation(s)
- Tingting Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Minghao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Min Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuchen Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rong Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junjing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiang Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
17
|
Mahdinia E, Shokri N, Taheri AT, Asgharzadeh S, Elahimanesh M, Najafi M. Cellular crosstalk in atherosclerotic plaque microenvironment. Cell Commun Signal 2023; 21:125. [PMID: 37254185 DOI: 10.1186/s12964-023-01153-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Atherosclerosis is an underlying pathology of many vascular diseases as a result of cellular, structural and molecular dysfunctions within the sub-endothelial space. This review deals with the events involved in the formation, growth and remodeling of plaque, including the cell recruitment, cell polarization, and cell fat droplets. It also describes cross talking between endothelial cells, macrophages, and vascular smooth muscle cells, as well as the cellular pathways involved in plaque development in the plaque microenvironment. Finally, it describes the plaque structural components and the role of factors involved in the rupture and erosion of plaques in the vessel. Video Abstract.
Collapse
Affiliation(s)
- Elmira Mahdinia
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolkarim Talebi Taheri
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Asgharzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ghazvin University of Medical Sciences, Ghazvin, Iran
| | - Mohammad Elahimanesh
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Baaten CCFMJ, Vondenhoff S, Noels H. Endothelial Cell Dysfunction and Increased Cardiovascular Risk in Patients With Chronic Kidney Disease. Circ Res 2023; 132:970-992. [PMID: 37053275 PMCID: PMC10097498 DOI: 10.1161/circresaha.123.321752] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The endothelium is considered to be the gatekeeper of the vessel wall, maintaining and regulating vascular integrity. In patients with chronic kidney disease, protective endothelial cell functions are impaired due to the proinflammatory, prothrombotic and uremic environment caused by the decline in kidney function, adding to the increase in cardiovascular complications in this vulnerable patient population. In this review, we discuss endothelial cell functioning in healthy conditions and the contribution of endothelial cell dysfunction to cardiovascular disease. Further, we summarize the phenotypic changes of the endothelium in chronic kidney disease patients and the relation of endothelial cell dysfunction to cardiovascular risk in chronic kidney disease. We also review the mechanisms that underlie endothelial changes in chronic kidney disease and consider potential pharmacological interventions that can ameliorate endothelial health.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| |
Collapse
|
19
|
Yuan D, Chu J, Qian J, Lin H, Zhu G, Chen F, Liu X. New Concepts on the Pathophysiology of Acute Coronary Syndrome. Rev Cardiovasc Med 2023; 24:112. [PMID: 39076267 PMCID: PMC11273028 DOI: 10.31083/j.rcm2404112] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 07/31/2024] Open
Abstract
Acute coronary syndrome (ACS) is the most severe form of ischemic heart disease. Although it is caused by atherosclerotic plaque thrombosis or nonatherosclerotic causes, its pathophysiological mechanism of ACS is not fully understood, and its concept is constantly updated and developed. At present, the main pathophysiological mechanisms include plaque rupture, plaque erosion, calcified nodules (CN) and non-atherosclerotic causes such as coronary vasospasm and myocardial bridging (MB). These mechanisms may overlap and coexist in some ACS patients. Therefore, the pathophysiological mechanism of ACS is complex, and is of great significance for the diagnosis and treatment of ACS. This review will discuss the pathophysiological mechanisms of ACS to provide new thoughts on the pathogenesis, diagnosis and treatment of ACS.
Collapse
Affiliation(s)
- Deqiang Yuan
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jiapeng Chu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Jun Qian
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Hao Lin
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Guoqi Zhu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Fei Chen
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| |
Collapse
|
20
|
Lin X, Deng Y, Ye L, Chen B, Tong J, Shi W, Wang B, Yu B, Tang J. RNA Sequencing Reveals the Differentially Expressed circRNAs between Stable and Unstable Carotid Atherosclerotic Plaques. Genet Res (Camb) 2023; 2023:7006749. [PMID: 37020895 PMCID: PMC10070021 DOI: 10.1155/2023/7006749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Objective. This study aimed to identify circular RNA profiles (circRNAs) via high-throughput RNA sequencing and distinguish the differentially expressed (DE) circRNAs between stable and unstable plaques. Methods. RNA sequencing was performed on unstable and stable carotid plaque samples obtained from patients with carotid artery stenosis. DE circRNAs were screened, and six DE circRNAs were verified using quantitative real-time PCR (qRT-PCR). Functional evaluation of the DE circRNAs was conducted via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Results. We screened 344 DE circRNAs in unstable plaques, consisting of 342 upregulated and 2 downregulated circRNAs. GO analysis showed that the host genes of the upregulated circRNAs were related to ER to Golgi transport vesicle membrane, endocytic vesicle membrane, and Ran GTPase binding. KEGG analysis revealed that the host genes of the upregulated circRNAs were primarily associated with protein processing in endoplasmic reticulum, lysine degradation, homologous recombination, epithelial cell signaling in Helicobacter pylori infection, and yersinia infection. The results of qRT-PCR verified three upregulated DE circRNAs and two downregulated DE circRNAs in unstable plaques. Conclusion. Hsa-circ-0001523, hsa-circ-0008950, hsa-circ-0000571, hsa-circ-0001946, and hsa-circ-0000745 may be involved in regulating the stability of atherosclerotic plaques and serves as a therapeutic target for unstable plaques.
Collapse
|
21
|
Benavente ED, Karnewar S, Buono M, Mili E, Hartman RJG, Kapteijn D, Slenders L, Daniels M, Aherrahrou R, Reinberger T, Mol BM, de Borst GJ, de Kleijn DPV, Prange KHM, Depuydt MAC, de Winther MPJ, Kuiper J, Björkegren JLM, Erdmann J, Civelek M, Mokry M, Owens GK, Pasterkamp G, den Ruijter HM. Female gene networks are expressed in myofibroblast-like smooth muscle cells in vulnerable atherosclerotic plaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527690. [PMID: 36798294 PMCID: PMC9934638 DOI: 10.1101/2023.02.08.527690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Women presenting with coronary artery disease (CAD) more often present with fibrous atherosclerotic plaques, which are currently understudied. Phenotypically modulated smooth muscle cells (SMCs) contribute to atherosclerosis in women. How these phenotypically modulated SMCs shape female versus male plaques is unknown. Here, we show sex-stratified gene regulatory networks (GRNs) from human carotid atherosclerotic tissue. Prioritization of these networks identified two main SMC GRNs in late-stage atherosclerosis. Single-cell RNA-sequencing mapped these GRNs to two SMC phenotypes: a phenotypically modulated myofibroblast-like SMC network and a contractile SMC network. The myofibroblast-like GRN was mostly expressed in plaques that were vulnerable in females. Finally, mice orthologs of the female myofibroblast-like genes showed retained expression in advanced plaques from female mice but were downregulated in male mice during atherosclerosis progression. Female atherosclerosis is driven by GRNs that promote a fibrous vulnerable plaque rich in myofibroblast-like SMCs.
Collapse
Affiliation(s)
- Ernest Diez Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Michele Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Eloi Mili
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Robin J. G. Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Daniek Kapteijn
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lotte Slenders
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mark Daniels
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Redouane Aherrahrou
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | | | - Barend M. Mol
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Dominique P. V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Koen H. M. Prange
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Marie A. C. Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Menno P. J. de Winther
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Johan L. M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Michal Mokry
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| |
Collapse
|
22
|
Nguyen AB, Iqbal O, Block RC, Mousa SA. Prevention and treatment of atherothrombosis: Potential impact of nanotechnology. Vascul Pharmacol 2023; 148:107127. [PMID: 36375733 DOI: 10.1016/j.vph.2022.107127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Complications with atherosclerosis can often lead to fatal clot formation and blood vessel occlusion - also known as atherothrombosis. A key component to the development of atherosclerosis and atherothrombosis is the endothelium and its ability to regulate the balance between prothrombotic and antithrombotic activities. Endothelial surface glycocalyx has a critical role in maintenance of vascular integrity. The endothelial glycocalyx, nitric oxide, prostacyclins, heparan sulfate, thrombomodulin, and tissue factor pathway inhibitor all prevent thrombosis, while P-selectin, among many other factors, favors thrombosis. However, endothelial dysfunction gives rise to the acceleration of thrombotic development and eventually the requirement of antithrombotic therapy. Most FDA-approved anticoagulant and antiplatelet therapies today carry a side effect profile of major bleed. Within the past five years, several preclinical studies using different endothelial targets and nanotechnology as a drug delivery method have emerged to target the endothelium and to enhance current antithrombosis without increasing bleed risk. While clinical studies are required, this review illustrates the proof-of-concept of nanotechnology in promoting a greater safety and efficacy profile through multiple in vitro and in vivo studies.
Collapse
Affiliation(s)
- Anthony B Nguyen
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York 12144, United States of America
| | - Omer Iqbal
- Stritch School of Medicine, Loyola University, Chicago, IL, United States of America
| | - Robert C Block
- University of Rochester, 601 Elmwood Ave, Rochester, NY 14642, United States of America
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York 12144, United States of America.
| |
Collapse
|
23
|
Loneliness, Social Isolation, and Living Alone Associations With Mortality Risk in Individuals Living With Cardiovascular Disease: A Systematic Review, Meta-Analysis, and Meta-Regression. Psychosom Med 2023; 85:8-17. [PMID: 36441849 DOI: 10.1097/psy.0000000000001151] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE This study aimed to conduct a systematic review and meta-analysis of associations between loneliness, social isolation, and living alone and risk of mortality among individuals with established cardiovascular disease. METHODS Five electronic databases were searched (MEDLINE [Ovid], PsycINFO, EMBASE, PubMed, and SCOPUS) from inception to November 25, 2021. In all, 35 studies were included in a narrative synthesis and, where appropriate, a meta-analytic evaluation using a random-effects model. RESULTS Living alone was associated with increased risk of all-cause mortality ( k = 15, n = 80,243, hazard ratio [HR] = 1.48, 95% confidence interval [CI] = 1.20-1.83, I2 = 83%), and meta-regressions found that the effects were stronger in studies from Europe and with longer follow-up. However, there was evidence of publication bias. Social isolation was associated with increased risk of all-cause mortality, measured both as a dichotomous variable ( k = 3, n = 2648, HR = 1.46, 95% CI = 1.08-2.04, I2 = 31%) and as a continuous variable ( k = 5, n = 2388, HR = 1.11, 95% CI = 1.02-1.24, I2 = 51%). Meta-analysis was not feasible for loneliness as exposure, but narrative synthesis of four studies suggested a tentative association between loneliness and increased risk of all-cause mortality. CONCLUSIONS Supporting public health concerns, our review finds that loneliness, social isolation, and living alone are associated with premature mortality among individuals with established cardiovascular disease. However, evidence of publication bias and large methodological differences across studies point to the need for more rigorous research.
Collapse
|
24
|
Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Sex-specific and hormone-related differences in vascular remodelling in atherosclerosis. Eur J Clin Invest 2023; 53:e13885. [PMID: 36219492 DOI: 10.1111/eci.13885] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022]
Abstract
Atherosclerosis, a lipid-driven inflammatory disease, is the main underlying cause of cardiovascular diseases (CVDs) both in men and women. Sex-related dimorphisms regarding CVDs and atherosclerosis were observed since more than a decade ago. Inflammatory mediators such as cytokines, but also endothelial dysfunction, vascular smooth muscle cell migration and proliferation lead to vascular remodelling but are differentially affected by sex. Each year a greater number of men die of CVDs compared with women and are also affected by CVDs at an earlier age (40-70 years old) while women develop atherosclerosis-related complications mainly after menopause (60+ years). The exact biological reasons behind this discrepancy are still not well-understood. From the numerous animal studies on atherosclerosis, only a few include both sexes and even less investigate and highlight the sex-specific differences that may arise. Endogenous sex hormones such as testosterone and oestrogen modulate the atherosclerotic plaque composition and the frequency of such plaques. In men, testosterone seems to act like a double-edged sword as its decrease with ageing correlates with an increased risk of atherosclerotic CVDs, while testosterone is also reported to promote inflammatory immune cell recruitment into the atherosclerotic plaque. In premenopausal women, oestrogen exerts anti-atherosclerotic effects, which decline together with its level after menopause resulting in increased CVD risk in ageing women. However, the interplay of sex hormones, sex-specific immune responses and other sex-related factors is still incompletely understood. This review highlights reported sex differences in atherosclerotic vascular remodelling and the role of endogenous sex hormones in this process.
Collapse
Affiliation(s)
- Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
25
|
Slowey C, Nyhan D. The Vascular System: Anatomical, Physiological, Pathological, and Aging Considerations. Anesthesiol Clin 2022; 40:557-574. [PMID: 36328615 DOI: 10.1016/j.anclin.2022.08.004] [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] [Indexed: 06/16/2023]
Abstract
The vascular system is one of the earliest recognized anatomical systems. It is composed of 3 parts; arterial, capillary, and venous, each with their own unique anatomy and physiology. Blood flow through this system is compromised in aging, atherosclerosis and peripheral vascular disease, and the practicing anesthesiologist must understand both the physiology and pathophysiology of the vascular tree.
Collapse
Affiliation(s)
- Charlie Slowey
- Johns Hopkins Department of Anesthesiology and Critical Care Medicine, 600 North Wolf Street, Baltimore, MD 21287, USA.
| | - Daniel Nyhan
- Johns Hopkins Department of Anesthesiology and Critical Care Medicine, 600 North Wolf Street, Baltimore, MD 21287, USA
| |
Collapse
|
26
|
Gu SZ, Bennett MR. Plaque Structural Stress: Detection, Determinants and Role in Atherosclerotic Plaque Rupture and Progression. Front Cardiovasc Med 2022; 9:875413. [PMID: 35872913 PMCID: PMC9300846 DOI: 10.3389/fcvm.2022.875413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/10/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis remains a major cause of death worldwide, with most myocardial infarctions being due to rupture or erosion of coronary plaques. Although several imaging modalities can identify features that confer risk, major adverse cardiovascular event (MACE) rates attributable to each plaque are low, such that additional biomarkers are required to improve risk stratification at plaque and patient level. Coronary arteries are exposed to continual mechanical forces, and plaque rupture occurs when plaque structural stress (PSS) exceeds its mechanical strength. Prospective studies have shown that peak PSS is correlated with acute coronary syndrome (ACS) presentation, plaque rupture, and MACE, and provides additional prognostic information to imaging. In addition, PSS incorporates multiple variables, including plaque architecture, plaque material properties, and haemodynamic data into a defined solution, providing a more detailed overview of higher-risk lesions. We review the methods for calculation and determinants of PSS, imaging modalities used for modeling PSS, and idealized models that explore structural and geometric components that affect PSS. We also discuss current experimental and clinical data linking PSS to the natural history of coronary artery disease, and explore potential for refining treatment options and predicting future events.
Collapse
Affiliation(s)
| | - Martin R. Bennett
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Semaglutide treatment attenuates vessel remodelling in ApoE-/- mice following vascular injury and blood flow perturbation. ATHEROSCLEROSIS PLUS 2022; 49:32-41. [PMID: 36644202 PMCID: PMC9833261 DOI: 10.1016/j.athplu.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Randomized clinical studies have shown a reduction in cardiovascular outcomes with glucagon-like peptide 1 receptor agonist (GLP-1RA) treatment with the hypothesized mechanisms being an underlying effect on atherosclerosis. Here, we aimed to assess the pharmacological effects of semaglutide in an atheroprone murine model that recapitulates central mechanisms related to vascular smooth muscle cell (VSMC) phenotypic switching and endothelial dysfunction known to operate within the atherosclerotic plaque. Methods In study A, we employed an electrical current to the carotid artery in ApoE-/- mice to induce severe VSMC injury and death, after which the arteries were allowed to heal for 4 weeks. In study B, a constrictive cuff was added for 6 h at the site of the healed segment to induce a disturbance in blood flow. Results Compared to vehicle, semaglutide treatment reduced the intimal and medial area by ∼66% (p = 0.007) and ∼11% (p = 0.0002), respectively. Following cuff placement, expression of the pro-inflammatory marker osteopontin and macrophage marker Mac-2 was reduced (p < 0.05) in the semaglutide-treated group compared to vehicle. GLP-1R were not expressed in murine carotid artery and human coronary vessels with and without atherosclerotic plaques, and semaglutide treatment did not affect proliferation of cultured primary human VSMCs. Conclusions Semaglutide treatment reduced vessel remodelling following electrical injury and blood flow perturbation in an atheroprone mouse model. This effect appears to be driven by anti-inflammatory and -proliferative mechanisms independent of GLP-1 receptor-mediated signalling in the resident vascular cells. This mechanism of action may be important for cardiovascular protection.
Collapse
|
28
|
Shishikura D, Octavia Y, Hayat U, Thondapu V, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
29
|
Hu ZB, Zhong QQ, Lu ZX, Zhu F. Association of platelet-to-white blood cell ratio and platelet-to-neutrophil ratio with the risk of fatal stroke occurrence in middle-aged to older Chinese. BMC Geriatr 2022; 22:430. [PMID: 35581556 PMCID: PMC9112464 DOI: 10.1186/s12877-022-03134-z] [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: 02/15/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background White blood cell (WBC) and neutrophil (NEUT) counts, which are commonly inflammatory markers, have been related to an increased risk of fatal stroke. However, it is unclear whether platelet-to-white blood cell ratio (PWR) and platelet-to-neutrophil ratio (PNR) are related to the risk of fatal stroke in middle-aged to older populations. Method In total, 27,811 participants without a stroke history at baseline were included and followed up for a mean of 14.3 years (standard deviation = 3.2), and 838 stroke deaths were recorded. The Cox proportional hazards regression was used to assess the relationships between the PWR and the PNR and the risk of fatal strokes. Results Compared to the 1st quartile, an increased risk of fatal all stroke showed among the participants in the highest quartiles of both the WBC (adjusted hazard ratio (aHR) = 1.35, 95% confidence interval (CI) 1.09–1.66) and the NEUT (aHR = 1.45, 95% CI 1.18–1.79). The restricted cubic splines showed decreased trends in associations of the PWR and the PNR with the risk of fatal all stroke. A decreased risk of fatal all stroke showed in those with the highest quartiles for both the PWR (aHR = 0.73, 95% CI 0.53–1.00) and the PNR (aHR = 0.74, 95% CI 0.54–1.01). The participants with the 2nd, the 3rd and the 4th change quartiles for the PWR and the PNR had weak decreasing trends for the risk of fatal all stroke, compared to those in the 1st change quartile, and the significant associations were observed in those with an increase of 20% for the PWR with the risk of fatal haemarragic stroke (aHR = 0.47, 95% CI 0.22–0.95) and a decrease of 20% for the PNR with the risk of fatal all stroke (aHR = 1.33, 95% CI 0.99–1.79), compared to those with stable dynamic changes. Conclusions Higher neutrophil count and platelet-to-neutrophil ratio were associated with a contrary risk of fatal stroke, with an increased for the former and a decreased for the later. A potentially chronic inflammation should be paid close attention to stroke occurrence in relatively healthy middle-aged to older populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-022-03134-z.
Collapse
Affiliation(s)
- Zhi-Bing Hu
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Qiong-Qiong Zhong
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China.,Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Jinan, China
| | - Ze-Xiong Lu
- Department of Internal Medicine, Sanya Central Hospital, Sanya, China
| | - Feng Zhu
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China.
| |
Collapse
|
30
|
Endothelial Dysfunction Induced by Extracellular Neutrophil Traps Plays Important Role in the Occurrence and Treatment of Extracellular Neutrophil Traps-Related Disease. Int J Mol Sci 2022; 23:ijms23105626. [PMID: 35628437 PMCID: PMC9147606 DOI: 10.3390/ijms23105626] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Many articles have demonstrated that extracellular neutrophil traps (NETs) are often described as part of the antibacterial function. However, since the components of NETs are non-specific, excessive NETs usually cause inflammation and tissue damage. Endothelial dysfunction (ED) caused by NETs is the major focus of tissue damage, which is highly related to many inflammatory diseases. Therefore, this review summarizes the latest advances in the primary and secondary mechanisms between NETs and ED regarding inflammation as a mediator. Moreover, the detailed molecular mechanisms with emphasis on the disadvantages from NETs are elaborated: NETs can use its own enzymes, release particles as damage-associated molecular patterns (DAMPs) and activate the complement system to interact with endothelial cells (ECs), drive ECs damage and eventually aggravate inflammation. In view of the role of NETs-induced ED in different diseases, we also discussed possible molecular mechanisms and the treatments of NETs-related diseases.
Collapse
|
31
|
Newman AAC, Cyr Y, Moore KJ. Advancing therapeutic targeting of the vulnerable plaque. Eur Heart J 2022; 43:1878-1880. [PMID: 35567566 DOI: 10.1093/eurheartj/ehac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Alexandra A C Newman
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Yannick Cyr
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
32
|
Khomtchouk BB, Lee YS, Khan ML, Sun P, Mero D, Davidson MH. Targeting the cytoskeleton and extracellular matrix in cardiovascular disease drug discovery. Expert Opin Drug Discov 2022; 17:443-460. [PMID: 35258387 PMCID: PMC9050939 DOI: 10.1080/17460441.2022.2047645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Currently, cardiovascular disease (CVD) drug discovery has focused primarily on addressing the inflammation and immunopathology aspects inherent to various CVD phenotypes such as cardiac fibrosis and coronary artery disease. However, recent findings suggest new biological pathways for cytoskeletal and extracellular matrix (ECM) regulation across diverse CVDs, such as the roles of matricellular proteins (e.g. tenascin-C) in regulating the cellular microenvironment. The success of anti-inflammatory drugs like colchicine, which targets microtubule polymerization, further suggests that the cardiac cytoskeleton and ECM provide prospective therapeutic opportunities. AREAS COVERED Potential therapeutic targets include proteins such as gelsolin and calponin 2, which play pivotal roles in plaque development. This review focuses on the dynamic role that the cytoskeleton and ECM play in CVD pathophysiology, highlighting how novel target discovery in cytoskeletal and ECM-related genes may enable therapeutics development to alter the regulation of cellular architecture in plaque formation and rupture, cardiac contractility, and other molecular mechanisms. EXPERT OPINION Further research into the cardiac cytoskeleton and its associated ECM proteins is an area ripe for novel target discovery. Furthermore, the structural connection between the cytoskeleton and the ECM provides an opportunity to evaluate both entities as sources of potential therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bohdan B. Khomtchouk
- University of Chicago, Department of Medicine, Section of Computational Biomedicine and Biomedical Data Science, Institute for Genomics and Systems Biology, Chicago, IL USA
| | - Yoon Seo Lee
- The College of the University of Chicago, Chicago, IL USA
| | - Maha L. Khan
- The College of the University of Chicago, Chicago, IL USA
| | - Patrick Sun
- The College of the University of Chicago, Chicago, IL USA
| | | | - Michael H. Davidson
- University of Chicago, Department of Medicine, Section of Cardiology, Chicago, IL USA
| |
Collapse
|
33
|
Luan Y, Liu H, Luan Y, Yang Y, Yang J, Ren KD. New Insight in HDACs: Potential Therapeutic Targets for the Treatment of Atherosclerosis. Front Pharmacol 2022; 13:863677. [PMID: 35529430 PMCID: PMC9068932 DOI: 10.3389/fphar.2022.863677] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis (AS) features include progressive hardening and reduced elasticity of arteries. AS is the leading cause of morbidity and mortality. An increasing amount of evidence showed that epigenetic modifications on genes serve are a main cause of several diseases, including AS. Histone deacetylases (HDACs) promote the deacetylation at lysine residues, thereby condensing the chromatin structures and further inhibiting the transcription of downstream genes. HDACs widely affect various physiological and pathological processes through transcriptional regulation or deacetylation of other non-histone proteins. In recent years, the role of HDACs in vascular systems has been revealed, and their effects on atherosclerosis have been widely reported. In this review, we discuss the members of HDACs in vascular systems, determine the diverse roles of HDACs in AS, and reveal the effects of HDAC inhibitors on AS progression. We provide new insights into the potential of HDAC inhibitors as drugs for AS treatment.
Collapse
Affiliation(s)
- Yi Luan
- Research Center for Clinical System Biology, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Ying Luan
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Research Center for Clinical System Biology, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yang Yang, ; Jing Yang, ; Kai-Di Ren,
| | - Jing Yang
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yang Yang, ; Jing Yang, ; Kai-Di Ren,
| | - Kai-Di Ren
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yang Yang, ; Jing Yang, ; Kai-Di Ren,
| |
Collapse
|
34
|
Krohn JB, Nguyen YN, Akhavanpoor M, Erbel C, Domschke G, Linden F, Kleber ME, Delgado G, März W, Katus HA, Gleissner CA. Identification of Specific Coronary Artery Disease Phenotypes Implicating Differential Pathophysiologies. Front Cardiovasc Med 2022; 9:778206. [PMID: 35355960 PMCID: PMC8960070 DOI: 10.3389/fcvm.2022.778206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background and Aims The roles of multiple risk factors of coronary artery disease (CAD) are well established. Commonly, CAD is considered as a single disease entity. We wish to examine whether coronary angiography allows to identify distinct CAD phenotypes associated with major risk factors and differences in prognosis. Methods In a cohort of 4,344 patients undergoing coronary angiography at Heidelberg University Hospital between 2014 and 2016, cluster analysis of angiographic reports identified subgroups with similar patterns of spatial distribution of high-grade stenoses. Clusters were independently confirmed in 3,129 patients from the LURIC study. Results Four clusters were identified: cluster one lacking critical stenoses comprised the highest percentage of women with the lowest cardiovascular risk. Patients in cluster two exhibiting high-grade stenosis of the proximal RCA had a high prevalence of the metabolic syndrome, and showed the highest levels of inflammatory biomarkers. Cluster three with predominant proximal LAD stenosis frequently presented with acute coronary syndrome and elevated troponin levels. Cluster four with high-grade stenoses throughout had the oldest patients with the highest overall cardiovascular risk. All-cause and cardiovascular mortality differed significantly between the clusters. Conclusions We identified four phenotypic subgroups of CAD bearing distinct demographic and biochemical characteristics with differences in prognosis, which may indicate multiple disease entities currently summarized as CAD.
Collapse
Affiliation(s)
- Jona B. Krohn
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Y Nhi Nguyen
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Christian Erbel
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Gabriele Domschke
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Fabian Linden
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Marcus E. Kleber
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Graciela Delgado
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Winfried März
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Hugo A. Katus
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christian A. Gleissner
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Cardiology and Stroke Centre, Rottal-Inn Kliniken, Eggenfelden, Germany
- *Correspondence: Christian A. Gleissner
| |
Collapse
|
35
|
Atherosclerosis in HIV Patients: What Do We Know so Far? Int J Mol Sci 2022; 23:ijms23052504. [PMID: 35269645 PMCID: PMC8910073 DOI: 10.3390/ijms23052504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
For the past several decades, humanity has been dealing with HIV. This disease is one of the biggest global health problems. Fortunately, modern antiretroviral therapy allows patients to manage the disease, improving their quality of life and their life expectancy. In addition, the use of these drugs makes it possible to reduce the risk of transmission of the virus to almost zero. Atherosclerosis is another serious pathology that leads to severe health problems, including disability and, often, the death of the patient. An effective treatment for atherosclerosis has not yet been developed. Both types of immune response, innate and adaptive, are important components of the pathogenesis of this disease. In this regard, the peculiarities of the development of atherosclerosis in HIV carriers are of particular scientific interest. In this review, we have tried to summarize the data on atherosclerosis and its development in HIV carriers. We also looked at the classic therapeutic methods and their features concerning the concomitant diagnosis.
Collapse
|
36
|
Slenders L, Tessels DE, van der Laan SW, Pasterkamp G, Mokry M. The Applications of Single-Cell RNA Sequencing in Atherosclerotic Disease. Front Cardiovasc Med 2022; 9:826103. [PMID: 35211529 PMCID: PMC8860895 DOI: 10.3389/fcvm.2022.826103] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/03/2022] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis still is the primary cause of death worldwide. Our characterization of the atherosclerotic lesion is mainly rooted in definitions based on pathological descriptions. We often speak in absolutes regarding plaque phenotypes: vulnerable vs. stable plaques or plaque rupture vs. plaque erosion. By focusing on these concepts, we may have oversimplified the atherosclerotic disease and its mechanisms. The widely used definitions of pathology-based plaque phenotypes can be fine-tuned with observations made with various -omics techniques. Recent advancements in single-cell transcriptomics provide the opportunity to characterize the cellular composition of the atherosclerotic plaque. This additional layer of information facilitates the in-depth characterization of the atherosclerotic plaque. In this review, we discuss the impact that single-cell transcriptomics may exert on our current understanding of atherosclerosis.
Collapse
Affiliation(s)
- Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Daniëlle E. Tessels
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| |
Collapse
|
37
|
Abstract
Despite effective therapeutic and preventive strategies, atherosclerosis and its complications still represent a substantial health burden. Leukocytes and inflammatory mechanisms are increasingly recognized as drivers of atherosclerosis. Neutrophil granulocytes within the circulation were recently shown to undergo neutrophil extracellular trap (NET) formation, linking innate immunity with acute complications of atherosclerosis. In this chapter, we summarize mechanisms of NET formation, evidence for their involvement in atherosclerosis and thrombosis, and potential therapeutic regimens specifically targeting NET components.
Collapse
|
38
|
Gerhardt T, Haghikia A, Stapmanns P, Leistner DM. Immune Mechanisms of Plaque Instability. Front Cardiovasc Med 2022; 8:797046. [PMID: 35087883 PMCID: PMC8787133 DOI: 10.3389/fcvm.2021.797046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation crucially drives atherosclerosis from disease initiation to the emergence of clinical complications. Targeting pivotal inflammatory pathways without compromising the host defense could compliment therapy with lipid-lowering agents, anti-hypertensive treatment, and lifestyle interventions to address the substantial residual cardiovascular risk that remains beyond classical risk factor control. Detailed understanding of the intricate immune mechanisms that propel plaque instability and disruption is indispensable for the development of novel therapeutic concepts. In this review, we provide an overview on the role of key immune cells in plaque inception and progression, and discuss recently identified maladaptive immune phenomena that contribute to plaque destabilization, including epigenetically programmed trained immunity in myeloid cells, pathogenic conversion of autoreactive regulatory T-cells and expansion of altered leukocytes due to clonal hematopoiesis. From a more global perspective, the article discusses how systemic crises such as acute mental stress or infection abruptly raise plaque vulnerability and summarizes recent advances in understanding the increased cardiovascular risk associated with COVID-19 disease. Stepping outside the box, we highlight the role of gut dysbiosis in atherosclerosis progression and plaque vulnerability. The emerging differential role of the immune system in plaque rupture and plaque erosion as well as the limitations of animal models in studying plaque disruption are reviewed.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philip Stapmanns
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
| | - David Manuel Leistner
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: David Manuel Leistner
| |
Collapse
|
39
|
Hu ZB, Zhong QQ, Lu ZX, Zhu F. Association of Neutrophil-to-Lymphocyte Ratio with the Risk of Fatal Stroke Occurrence in Older Chinese. Clin Appl Thromb Hemost 2022; 28:10760296221098720. [PMID: 35538863 PMCID: PMC9102137 DOI: 10.1177/10760296221098720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Associations of neutrophil-to-lymphocyte ratio (NLR) and its longitudinal change with risk of fatal strokes are unclear in older populations. METHODS In this retrospective analysis, a total of 27,799 participants were included and followed up for a mean of 14.3 years (standard deviation = 3.2). 838 stroke deaths were recorded. Cox proportional hazards regression was used to assess associations of NLR with fatal strokes. RESULTS Compared to those in the first quartile and after adjustment for a series of factors, the participants in the highest neutrophil quartile had an increased risk of fatal all stroke (adjusted hazard ratio (aHR) = 1.45, 95% confidence interval (CI), 1.18-1.79) and fatal ischaemic stroke (aHR = 1.58, 95% CI, 1.17-2.12). Restricted cubic splines showed an increased trend of relationship between the NLR and fatal all stroke. The participants with the highest NLR quartile had an increased risk of fatal all stroke (aHR = 1.52, 95% CI, 1.23-1.88) and fatal ischaemic stroke (aHR = 1.59, 95% CI, 1.13-2.26), respectively; Similar associations repeated after further C-reactive protein adjustment; a 21% and a 32% increased risk of fatal all stroke and fatal ischaemic stroke showed in a continuous variable model. Those in NLR change with 5% increase had a 70% increased risk of fatal all stroke (aHR = 1.70, 95%CI, 1.13-2.57), compared to those in stable (-5%∼5%). CONCLUSIONS Higher NLR was associated with an increased risk of fatal all stroke and fatal ischaemic stroke, and its longitudinal change increase of ≥ 5% was associated with an increased risk of fatal all stroke in a relatively healthy older population.
Collapse
Affiliation(s)
- Zhi-Bing Hu
- Department of Internal Medicine and Central laboratory, 477162Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Qiong-Qiong Zhong
- Department of Internal Medicine and Central laboratory, 477162Guangzhou Twelfth People's Hospital, Guangzhou, China.,Department of Public Health and Preventive Medicine, School of Medicine, 47885Jinan University, Guangzhou, China
| | - Ze-Xiong Lu
- Department of Internal Medicine, 477162Sanya Central Hospital, Sanya, China
| | - Feng Zhu
- Department of Internal Medicine and Central laboratory, 477162Guangzhou Twelfth People's Hospital, Guangzhou, China
| |
Collapse
|
40
|
Fang C, Lu J, Zhang S, Wang J, Wang Y, Li L, Wang Y, Jiang S, Yin Y, Guo J, Lei F, Yu H, Wei G, Yao Y, Chen T, Ren X, Xing L, Tu Y, Hou J, Dai J, Yu B. Morphological Characteristics of Eroded Plaques with Noncritical Coronary Stenosis: An Optical Coherence Tomography Study. J Atheroscler Thromb 2022; 29:126-140. [PMID: 33455996 PMCID: PMC8737071 DOI: 10.5551/jat.60301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/30/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS Recent studies suggested plaque erosion with noncritical stenosis could be treated distinctly from that with critical stenosis, but their morphological features remained largely unknown. The present study aimed to investigate morphological features of eroded plaques with different lumen stenosis using optical coherence tomography (OCT). METHODS A total of 348 ST-segment elevated myocardial infarction patients with culprit OCT-defined plaque erosion (OCT-erosion) were analyzed. Based on the severity of lumen area stenosis, all patients with OCT-erosions were divided into the following three groups: Group A (area stenosis <50%, n=50); Group B (50% ≤ area stenosis <75%, n=146); Group C (area stenosis ≥ 75%, n=152). RESULTS Compared with patients in Groups A and B, patients in Group C were older (p=0.008) and had higher prevalence of hypertension (p=0.029). Angiographic analysis showed that 72.0% of the eroded plaques in Group A were located in the left anterior descending artery, followed by 67.8% in Group B, and 53.9% in Group C (p=0.039). OCT analysis showed that Group A had the highest prevalence of fibrous plaques (p<0.001) and nearby bifurcation (p=0.036), but the lowest prevalence of lipid-rich plaques (p<0.001), macrophage accumulation (p<0.001), microvessels (p=0.009), cholesterol crystals (p<0.001), and calcification (p=0.023). Multivariable regression analysis showed fibrous plaque (odds ratio [OR]: 3.014, 95% confidence interval [CI]: 1.932-4.702, p<0.001) and nearby bifurcation (OR: 1.750, 95% CI: 1.109-2.761, p=0.016) were independently associated with OCT-erosion with an area stenosis of <75%. CONCLUSIONS More than half of OCT-erosions presented with <75% area stenosis, having distinct morphological features from those of OCT-erosions with critical stenosis. Fibrous plaque and nearby bifurcation were independently associated with noncritically stenotic OCT-erosion, suggesting that eroded plaques might need individualized treatment.
Collapse
Affiliation(s)
- Chao Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jia Lu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Shaotao Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jifei Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yidan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Lulu Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yini Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Senqing Jiang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yanwei Yin
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Junchen Guo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Fangmeng Lei
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Huai Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Guo Wei
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yuan Yao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Tao Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xuefeng Ren
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Lei Xing
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yingfeng Tu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jingbo Hou
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jiannan Dai
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| |
Collapse
|
41
|
Libby P. Inflammation during the life cycle of the atherosclerotic plaque. Cardiovasc Res 2021; 117:2525-2536. [PMID: 34550337 PMCID: PMC8783385 DOI: 10.1093/cvr/cvab303] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation orchestrates each stage of the life cycle of atherosclerotic plaques. Indeed, inflammatory mediators likely link many traditional and emerging risk factors with atherogenesis. Atheroma initiation involves endothelial activation with recruitment of leucocytes to the arterial intima, where they interact with lipoproteins or their derivatives that have accumulated in this layer. The prolonged and usually clinically silent progression of atherosclerosis involves periods of smouldering inflammation, punctuated by episodes of acute activation that may arise from inflammatory mediators released from sites of extravascular injury or infection or from subclinical disruptions of the plaque. Smooth muscle cells and infiltrating leucocytes can proliferate but also undergo various forms of cell death that typically lead to formation of a lipid-rich 'necrotic' core within the evolving intimal lesion. Extracellular matrix synthesized by smooth muscle cells can form a fibrous cap that overlies the lesion's core. Thus, during progression of atheroma, cells not only procreate but perish. Inflammatory mediators participate in both processes. The ultimate clinical complication of atherosclerotic plaques involves disruption that provokes thrombosis, either by fracture of the plaque's fibrous cap or superficial erosion. The consequent clots can cause acute ischaemic syndromes if they embarrass perfusion. Incorporation of the thrombi can promote plaque healing and progressive intimal thickening that can aggravate stenosis and further limit downstream blood flow. Inflammatory mediators regulate many aspects of both plaque disruption and healing process. Thus, inflammatory processes contribute to all phases of the life cycle of atherosclerotic plaques, and represent ripe targets for mitigating the disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
42
|
Yang W, Lin J, Zhou J, Zheng Y, Jiang S, He S, Li D. Innate Lymphoid Cells and Myocardial Infarction. Front Immunol 2021; 12:758272. [PMID: 34867998 PMCID: PMC8636005 DOI: 10.3389/fimmu.2021.758272] [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: 08/13/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Myocardial infarction results from obstruction of a coronary artery that causes insufficient blood supply to the myocardium and leads to ischemic necrosis. It is one of the most common diseases threatening human health and is characterized by high morbidity and mortality. Atherosclerosis is the pathological basis of myocardial infarction, and its pathogenesis has not been fully elucidated. Innate lymphoid cells (ILCs) are an important part of the human immune system and participate in many processes, including inflammation, metabolism and tissue remodeling, and play an important role in atherosclerosis. However, their specific roles in myocardial infarction are unclear. This review describes the current understanding of the relationship between innate lymphoid cells and myocardial infarction during the acute phase of myocardial infarction, myocardial ischemia-reperfusion injury, and heart repair and regeneration following myocardial infarction. We suggest that this review may provide new potential intervention targets and ideas for treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
| | | | | | | | | | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dazhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Nuclear Receptors and Clock Components in Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms22189721. [PMID: 34575881 PMCID: PMC8468608 DOI: 10.3390/ijms22189721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases (CVD) are still the first cause of death worldwide. Their main origin is the development of atherosclerotic plaque, which consists in the accumulation of lipids and inflammatory leucocytes within the vascular wall of large vessels. Beyond dyslipidemia, diabetes, obesity, hypertension and smoking, the alteration of circadian rhythms, in shift workers for instance, has recently been recognized as an additional risk factor. Accordingly, targeting a pro-atherogenic pathway at the right time window, namely chronotherapy, has proven its efficiency in reducing plaque progression without affecting healthy tissues in mice, thus providing the rationale of such an approach to treat CVD and to reduce drug side effects. Nuclear receptors are transcriptional factors involved in the control of many physiological processes. Among them, Rev-erbs and RORs control metabolic homeostasis, inflammatory processes and the biological clock. In this review, we discuss the opportunity to dampen atherosclerosis progression by targeting such ligand-activated core clock components in a (chrono-)therapeutic approach in order to treat CVD.
Collapse
|
44
|
Status of biomarkers for the identification of stable or vulnerable plaques in atherosclerosis. Clin Sci (Lond) 2021; 135:1981-1997. [PMID: 34414413 DOI: 10.1042/cs20210417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is a systemic inflammation of the arteries characterized by atherosclerotic plaque due to the accumulation of lipids, inflammatory cells, apoptotic cells, calcium and extracellular matrix (ECM) proteins. Stable plaques present a chronic inflammatory infiltration, whereas vulnerable plaques present an 'active' inflammation involved in the thinning of the fibrous cap that predisposes to plaque rupture. Several complex biological cellular processes lead plaques to evolve from stable to vulnerable predisposing them to rupture and thrombosis. In this review, we analyze some emerging circulating biomarkers related to inflammation, ECM and lipid infiltration, angiogenesis, metalloproteinases and microRNA (miRNA), as possible diagnostic and prognostic indicators of plaque vulnerability.
Collapse
|
45
|
ten Cate H, Guzik TJ, Eikelboom J, Spronk HMH. Pleiotropic actions of factor Xa inhibition in cardiovascular prevention: mechanistic insights and implications for anti-thrombotic treatment. Cardiovasc Res 2021; 117:2030-2044. [PMID: 32931586 PMCID: PMC8318102 DOI: 10.1093/cvr/cvaa263] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/10/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease in which atherothrombotic complications lead to cardiovascular morbidity and mortality. At advanced stages, myocardial infarction, ischaemic stroke, and peripheral artery disease, including major adverse limb events, are caused either by acute occlusive atherothrombosis or by thromboembolism. Endothelial dysfunction, vascular smooth muscle cell activation, and vascular inflammation are essential in the development of acute cardiovascular events. Effects of the coagulation system on vascular biology extend beyond thrombosis. Under physiological conditions, coagulation proteases in blood are pivotal in maintaining haemostasis and vascular integrity. Under pathological conditions, including atherosclerosis, the same coagulation proteases (including factor Xa, factor VIIa, and thrombin) become drivers of atherothrombosis, working in concert with platelets and vessel wall components. While initially atherothrombosis was attributed primarily to platelets, recent advances indicate the critical role of fibrin clot and plasma coagulation factors. Mechanisms of atherothrombosis and hypercoagulability vary depending on plaque erosion or plaque rupture. In addition to contributing to thrombus formation, factor Xa and thrombin can affect endothelial dysfunction, oxidative stress, vascular smooth muscle cell function as well as immune cell activation and vascular inflammation. By these mechanisms, they promote atherosclerosis and contribute to plaque instability. In this review, we first discuss the postulated vasoprotective mechanisms of protease-activated receptor signalling induced by coagulation enzymes under physiological conditions. Next, we discuss preclinical studies linking coagulation with endothelial cell dysfunction, thromboinflammation, and atherogenesis. Understanding these mechanisms is pivotal for the introduction of novel strategies in cardiovascular prevention and therapy. We therefore translate these findings to clinical studies of direct oral anticoagulant drugs and discuss the potential relevance of dual pathway inhibition for atherothrombosis prevention and vascular protection.
Collapse
Affiliation(s)
- Hugo ten Cate
- Department of Internal Medicine, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Biochemistry, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Tomasz J Guzik
- Institute of Cardiovascular & Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow, UK
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - John Eikelboom
- Population Health Research Institute, Hamilton General Hospital and McMaster University, Hamilton, L8L 2x2, ON, Canada
| | - Henri M H Spronk
- Department of Internal Medicine, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Biochemistry, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
46
|
Glucocorticoids: Fuelling the Fire of Atherosclerosis or Therapeutic Extinguishers? Int J Mol Sci 2021; 22:ijms22147622. [PMID: 34299240 PMCID: PMC8303333 DOI: 10.3390/ijms22147622] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/21/2023] Open
Abstract
Glucocorticoids are steroid hormones with key roles in the regulation of many physiological systems including energy homeostasis and immunity. However, chronic glucocorticoid excess, highlighted in Cushing's syndrome, is established as being associated with increased cardiovascular disease (CVD) risk. Atherosclerosis is the major cause of CVD, leading to complications including coronary artery disease, myocardial infarction and heart failure. While the associations between glucocorticoid excess and increased prevalence of these complications are well established, the mechanisms underlying the role of glucocorticoids in development of atheroma are unclear. This review aims to better understand the importance of glucocorticoids in atherosclerosis and to dissect their cell-specific effects on key processes (e.g., contractility, remodelling and lesion development). Clinical and pre-clinical studies have shown both athero-protective and pro-atherogenic responses to glucocorticoids, effects dependent upon their multifactorial actions. Evidence indicates regulation of glucocorticoid bioavailability at the vasculature is complex, with local delivery, pre-receptor metabolism, and receptor expression contributing to responses linked to vascular remodelling and inflammation. Further investigations are required to clarify the mechanisms through which endogenous, local glucocorticoid action and systemic glucocorticoid treatment promote/inhibit atherosclerosis. This will provide greater insights into the potential benefit of glucocorticoid targeted approaches in the treatment of cardiovascular disease.
Collapse
|
47
|
El-Dahan KS, Machtoub D, Massoud G, Nasser SA, Hamam B, Kobeissy F, Zouein FA, Eid AH. Cannabinoids and myocardial ischemia: Novel insights, updated mechanisms, and implications for myocardial infarction. Curr Med Chem 2021; 29:1990-2010. [PMID: 34102966 DOI: 10.2174/0929867328666210608144818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/22/2022]
Abstract
Cannabis is the most widely trafficked and abused illicit drug due to its calming psychoactive properties. It has been increasingly recognized as having potential health benefits and relatively less adverse health effects as compared to other illicit drugs; however, growing evidence clearly indicates that cannabis is associated with considerable adverse cardiovascular events. Recent studies have linked cannabis use to myocardial infarction (MI); yet, very little is known about the underlying mechanisms. A MI is a cardiovascular disease characterized by a mismatch in the oxygen supply and demand of the heart, resulting in ischemia and subsequent necrosis of the myocardium. Since cannabis is increasingly being considered a risk factor for MI, there is a growing need for better appreciating its potential health benefits and consequences. Here, we discuss the cellular mechanisms of cannabis that lead to an increased risk of MI. We provide a thorough and critical analysis of cannabinoids' actions, which include modulation of adipocyte biology, regional fat distribution, and atherosclerosis, as well as precipitation of hemodynamic stressors relevant in the setting of a MI. By critically dissecting the modulation of signaling pathways in multiple cell types, this paper highlights the mechanisms through which cannabis may trigger life-threatening cardiovascular events. This then provides a framework for future pharmacological studies which can identify targets or develop drugs that modulate cannabis' effects on the cardiovascular system as well as other organ systems. Cannabis' impact on the autonomic outflow, vascular smooth muscle cells, myocardium, cortisol levels and other hemodynamic changes are also mechanistically reviewed.
Collapse
Affiliation(s)
- Karim Seif El-Dahan
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Dima Machtoub
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Gaelle Massoud
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Suzanne A Nasser
- Department of Pharmacology and Therapeutics, Beirut Arab University, P.O. Box 11-5020, Beirut, Lebanon
| | - Bassam Hamam
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, P.O. Box 146404, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha. Qatar
| |
Collapse
|
48
|
Leistner DM, Kränkel N, Meteva D, Abdelwahed YS, Seppelt C, Stähli BE, Rai H, Skurk C, Lauten A, Mochmann HC, Fröhlich G, Rauch-Kröhnert U, Flores E, Riedel M, Sieronski L, Kia S, Strässler E, Haghikia A, Dirks F, Steiner JK, Mueller DN, Volk HD, Klotsche J, Joner M, Libby P, Landmesser U. Differential immunological signature at the culprit site distinguishes acute coronary syndrome with intact from acute coronary syndrome with ruptured fibrous cap: results from the prospective translational OPTICO-ACS study. Eur Heart J 2021; 41:3549-3560. [PMID: 33080003 DOI: 10.1093/eurheartj/ehaa703] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/15/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
AIMS Acute coronary syndromes with intact fibrous cap (IFC-ACS), i.e. caused by coronary plaque erosion, account for approximately one-third of ACS. However, the underlying pathophysiological mechanisms as compared with ACS caused by plaque rupture (RFC-ACS) remain largely undefined. The prospective translational OPTICO-ACS study programme investigates for the first time the microenvironment of ACS-causing culprit lesions (CL) with intact fibrous cap by molecular high-resolution intracoronary imaging and simultaneous local immunological phenotyping. METHODS AND RESULTS The CL of 170 consecutive ACS patients were investigated by optical coherence tomography (OCT) and simultaneous immunophenotyping by flow cytometric analysis as well as by effector molecule concentration measurements across the culprit lesion gradient (ratio local/systemic levels). Within the study cohort, IFC caused 24.6% of ACS while RFC-ACS caused 75.4% as determined and validated by two independent OCT core laboratories. The IFC-CL were characterized by lower lipid content, less calcification, a thicker overlying fibrous cap, and largely localized near a coronary bifurcation as compared with RFC-CL. The microenvironment of IFC-ACS lesions demonstrated selective enrichment in both CD4+ and CD8+ T-lymphocytes (+8.1% and +11.2%, respectively, both P < 0.05) as compared with RFC-ACS lesions. T-cell-associated extracellular circulating microvesicles (MV) were more pronounced in IFC-ACS lesions and a significantly higher amount of CD8+ T-lymphocytes was detectable in thrombi aspirated from IFC-culprit sites. Furthermore, IFC-ACS lesions showed increased levels of the T-cell effector molecules granzyme A (+22.4%), perforin (+58.8%), and granulysin (+75.4%) as compared with RFC plaques (P < 0.005). Endothelial cells subjected to culture in disturbed laminar flow conditions, i.e. to simulate coronary flow near a bifurcation, demonstrated an enhanced adhesion of CD8+T cells. Finally, both CD8+T cells and their cytotoxic effector molecules caused endothelial cell death, a key potential pathophysiological mechanism in IFC-ACS. CONCLUSIONS The OPTICO-ACS study emphasizes a novel mechanism in the pathogenesis of IFC-ACS, favouring participation of the adaptive immune system, particularly CD4+ and CD8+ T-cells and their effector molecules. The different immune signatures identified in this study advance the understanding of coronary plaque progression and may provide a basis for future development of personalized therapeutic approaches to ACS with IFC. TRIAL REGISTRATION The study was registered at clinicalTrials.gov (NCT03129503).
Collapse
Affiliation(s)
- David M Leistner
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Nicolle Kränkel
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Denitsa Meteva
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Claudio Seppelt
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Barbara E Stähli
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Himanshu Rai
- DZHK (German Centre for Cardiovascular Research) Partner Site Munch, Munich, 80636, Germany
| | - Carsten Skurk
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Alexander Lauten
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Hans-Christian Mochmann
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany
| | - Georg Fröhlich
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Eduardo Flores
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany
| | - Matthias Riedel
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Lara Sieronski
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Sylvia Kia
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Elisabeth Strässler
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Arash Haghikia
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Fabian Dirks
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Julia K Steiner
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Dominik N Mueller
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany.,Experimental and Clinical Research Centre (ECRC), a cooperation of Charité University Medicine Berlin and Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health (BIH), Berlin 10117, Germany.,Institute for Medical Immunology and BIH Centre for Regenerative Therapies (BCRT), and Berlin Centre for Advanced Therapies (BeCAT), Charité University Medicine Berlin, Berlin 13353, Germany
| | - Jens Klotsche
- German Rheumatism Research Centre Berlin, and Institute for Social Medicine, Epidemiology und Heath Economy, Charité University Medicine Berlin, Campus Charité Mitte, Berlin 10117, Germany
| | - Michael Joner
- Department of Cardiology and ISAR Research Centre, German Heart Centre, Munich, 80636, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Munch, Munich, 80636, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Ave Louis Pasteur, Boston, MA 02115, USA
| | - Ulf Landmesser
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| |
Collapse
|
49
|
Tomaniak M, Katagiri Y, Modolo R, de Silva R, Khamis RY, Bourantas CV, Torii R, Wentzel JJ, Gijsen FJH, van Soest G, Stone PH, West NEJ, Maehara A, Lerman A, van der Steen AFW, Lüscher TF, Virmani R, Koenig W, Stone GW, Muller JE, Wijns W, Serruys PW, Onuma Y. Vulnerable plaques and patients: state-of-the-art. Eur Heart J 2021; 41:2997-3004. [PMID: 32402086 DOI: 10.1093/eurheartj/ehaa227] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/01/2019] [Accepted: 03/26/2020] [Indexed: 01/21/2023] Open
Abstract
Despite advanced understanding of the biology of atherosclerosis, coronary heart disease remains the leading cause of death worldwide. Progress has been challenging as half of the individuals who suffer sudden cardiac death do not experience premonitory symptoms. Furthermore, it is well-recognized that also a plaque that does not cause a haemodynamically significant stenosis can trigger a sudden cardiac event, yet the majority of ruptured or eroded plaques remain clinically silent. In the past 30 years since the term 'vulnerable plaque' was introduced, there have been major advances in the understanding of plaque pathogenesis and pathophysiology, shifting from pursuing features of 'vulnerability' of a specific lesion to the more comprehensive goal of identifying patient 'cardiovascular vulnerability'. It has been also recognized that aside a thin-capped, lipid-rich plaque associated with plaque rupture, acute coronary syndromes (ACS) are also caused by plaque erosion underlying between 25% and 60% of ACS nowadays, by calcified nodule or by functional coronary alterations. While there have been advances in preventive strategies and in pharmacotherapy, with improved agents to reduce cholesterol, thrombosis, and inflammation, events continue to occur in patients receiving optimal medical treatment. Although at present the positive predictive value of imaging precursors of the culprit plaques remains too low for clinical relevance, improving coronary plaque imaging may be instrumental in guiding pharmacotherapy intensity and could facilitate optimal allocation of novel, more aggressive, and costly treatment strategies. Recent technical and diagnostic advances justify continuation of interdisciplinary research efforts to improve cardiovascular prognosis by both systemic and 'local' diagnostics and therapies. The present state-of-the-art document aims to present and critically appraise the latest evidence, developments, and future perspectives in detection, prevention, and treatment of 'high-risk' plaques occurring in 'vulnerable' patients.
Collapse
Affiliation(s)
- Mariusz Tomaniak
- Department of Cardiology, Erasmus Medical Centre, Thorax Centre, Rotterdam, The Netherlands.,First Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Yuki Katagiri
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rodrigo Modolo
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Cardiology Division, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, Brazil
| | - Ranil de Silva
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Institute of Cardiovascular Medicine and Science, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Ramzi Y Khamis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Christos V Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London EC1A 7BE, UK.,William Harvey Research Institute, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK.,Institute of Cardiovascular Sciences, University College London, 62 Huntley St, Fitzrovia, London WC1E 6DD, UK
| | - Ryo Torii
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Jolanda J Wentzel
- Department of Cardiology, Biomedical Engineering, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank J H Gijsen
- Department of Biomedical Engineering, Erasmus Medical Centre, Thorax Centre, Rotterdam, The Netherlands
| | - Gijs van Soest
- Department of Cardiology, Biomedical Engineering, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Peter H Stone
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nick E J West
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Rd, Trumpington, Cambridge CB2 0AY, UK
| | - Akiko Maehara
- Division of Cardiology, New York-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA.,Clinical Trials Centre, Cardiovascular Research Foundation, New York, NY, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Antonius F W van der Steen
- Department of Cardiology, Biomedical Engineering, Erasmus Medical Centre, Rotterdam, The Netherlands.,Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Department of Imaging Physics, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Thomas F Lüscher
- Royal Brompton and Harefield Hospital Trust, Imperial College London, , London, UK.,Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Wolfgang Koenig
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Gregg W Stone
- Division of Cardiology, New York-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA.,Clinical Trials Centre, Cardiovascular Research Foundation, New York, NY, USA
| | - James E Muller
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - William Wijns
- The Lambe Institute for Translational Medicine and Curam, National University of Ireland, Galway, Ireland.,Saolta University Healthcare Group, Galway, Ireland
| | - Patrick W Serruys
- National Heart and Lung Institute, Imperial College London, London, UK.,Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland, Galway, Ireland
| |
Collapse
|
50
|
Shimonaga K, Matsushige T, Takahashi H, Hashimoto Y, Yoshiyama M, Ono C, Sakamoto S. Peptidylarginine Deiminase 4 as a Possible Biomarker of Plaque Instability in Carotid Artery Stenosis. J Stroke Cerebrovasc Dis 2021; 30:105816. [PMID: 33906071 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105816] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil extracellular traps (NETs) exhibit pro-inflammatory and pro-thrombotic properties. However, they have only been reported as important regulators in atherosclerosis, especially in atherothrombosis. We investigated the presence of NETs and plaque instability in patients with carotid artery stenosis. MATERIAL AND METHODS A total of 39 consecutive patients with carotid artery stenosis were evaluated. All patients underwent carotid artery stenting (CAS) with dual protection (simultaneous flow reversal + distal filter) and blood aspiration as a method of distal embolism prevention. Local arterial blood was aspirated at the stent site and peptidylarginine deiminase 4 (PAD4), which is essential for the formation of NETs, was measured. The relationships between PAD4 and the patient profile, blood examination and plaque data were investigated. RESULTS The mean value of PAD4 in local arterial blood in CAS was 0.5 ng/ml. Bivariate analysis demonstrated that PAD4 was associated with the neutrophil to lymphocyte ratio (p = 0.007), high-density lipoprotein (p = 0.02), triglycerides to high-density lipoprotein ratio (p = 0.007), ulceration (p = 0.02) and plaque contrast enhancement on T1 black blood imaging (p = 0.03). In multiple linear regression analyses, PAD4 was correlated with the neutrophil to lymphocyte ratio (p = 0.01) and ulceration (p = 0.01, cut-off value: 0.49 odds ratio: 19.3). CONCLUSIONS PAD4, representative of the presence of NETs, was high in carotid plaques with unstable features. The neutrophil to lymphocyte ratio in peripheral blood was suggested to be a biomarker of vulnerable plaques. Elucidating the role of NETs may aid in clarifying factors that promote the instability of carotid plaques.
Collapse
Affiliation(s)
- Koji Shimonaga
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City Asa Citizens Hospital, 2-1-1 Kabeminami, Asakitaku, 731 0293 Hiroshima, Japan.
| | - Toshinori Matsushige
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City Asa Citizens Hospital, 2-1-1 Kabeminami, Asakitaku, 731 0293 Hiroshima, Japan; Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Hiroki Takahashi
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City Asa Citizens Hospital, 2-1-1 Kabeminami, Asakitaku, 731 0293 Hiroshima, Japan.
| | - Yukishige Hashimoto
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City Asa Citizens Hospital, 2-1-1 Kabeminami, Asakitaku, 731 0293 Hiroshima, Japan.
| | - Michitsura Yoshiyama
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City Asa Citizens Hospital, 2-1-1 Kabeminami, Asakitaku, 731 0293 Hiroshima, Japan.
| | - Chiaki Ono
- Department of Radiology, Hiroshima City Asa Citizens Hospital, Hiroshima, Japan.
| | - Shigeyuki Sakamoto
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|