1
|
Wu Y, Ma Q, Meng X, Sun Q, Wang Z, Zhang W. MicroRNA-9-3p inhibits endothelial pyroptosis in atherosclerosis by targeting the PTEN/AKT axis. Int J Biol Macromol 2025; 315:144289. [PMID: 40393590 DOI: 10.1016/j.ijbiomac.2025.144289] [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: 10/18/2024] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
Inflammation and pyroptosis are key characteristic features of Atherosclerosis (AS), a complex, multifaceted chronic vascular disease. Although microRNAs (miRNAs) have been substantially implicated, as pivotal post-transcriptional regulators, in molecular mechanisms underlying atherosclerotic development, the precise role of miR-9-3p in atherosclerotic progression remains unclear. Herein, we aimed to elucidate the regulatory function of miR-9-3p in Endothelial Cell pyroptosis and AS via integrated bioinformatics analysis, as well as in vitro and in vivo assays. We focused on identifying signaling pathways miR-9-3p modulated and assessing its potential to mitigate atherosclerotic plaque formation. According to the results, miR-9-3p promoted AKT phosphorylation by directly targeting the 3'-Untranslated Region (3'-UTR) of PTEN mRNA, suppressing EC pyroptosis. Notably, the AKT signaling pathway, which was significantly enriched in our analyses, functions upstream of the NLRP3 inflammasome, a principal mediator of pyroptosis. We also identified the SP1 Transcription Factor (TF) binding sites within the promoter region of MIR9-1HG, implying a regulatory mechanism in which SP1 modulates miR-9-3p expression and function. Given the reversibility of epigenetic modifications, the potential of restoring miR-9-3p expression presents a novel therapeutic strategy for AS. Overall, in addition to advancing the understanding of atherosclerotic treatment mechanisms, this study positions miR-9-3p as a potential therapeutic target.
Collapse
Affiliation(s)
- Yan Wu
- Department of Ultrasound, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Ultrasound Molecular Imaging Joint laboratory of Heilongjiang Province (International Cooperation), Harbin, Heilongjiang 150086, China
| | - Qi Ma
- Department of Ultrasound, Harbin Red Cross Central Hospital, Harbin, China
| | - Xiangrong Meng
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Qi Sun
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Heilong Jiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Zhuozhong Wang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, 150086, China
| | - Wenjing Zhang
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
2
|
Peng P, Ding S, Liang M, Zheng W, Kang Y, Liu W, Shi H, Gao C. A self-sacrificing anti-inflammatory coating promotes simultaneous cardiovascular repair and reendothelialization of implanted devices. Bioact Mater 2025; 47:502-512. [PMID: 40026826 PMCID: PMC11872464 DOI: 10.1016/j.bioactmat.2025.01.037] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
During interventional surgeries of implantable cardiovascular devices in addressing cardiovascular diseases (CVD), the inevitable tissue damage will trigger host inflammation and vascular lumen injury, leading to delayed re-endothelization and intimal hyperplasia. Endowing cardiovascular implants with anti-inflammatory and endothelialization functions is conducive to the target site, offering significant tissue repair and regeneration benefits. Herein, inspired by the snake's molting process, a ShedWise device was developed by using the poly(propylene fumarate) polyurethane (PPFU) as the foundational material, which was clicked with hyperbranched polylysine (HBPL) and followed by conjugation with pro-endothelial functional Arg-Glu-Asp-Val peptide (REDV), and finally coated with a "self-sacrificing" layer having reactive oxygen species (ROS)-scavenging ability and degradability. During the acute inflammation in the initial stage of implantation, the ROS-responsive hyperbranched poly(acrylate-capped thioketone-containing ethylene glycol (HBPAK) coating effectively modulated the level of environmental inflammation and resisted initial protein adsorption, showcasing robust tissue protection. As the coating gradually "sacrificed", the exposed hyperbranched HBPL-REDV layer recruited specifically endothelial cells and promoted surface endothelialization. In a rat vascular injury model, the ShedWise demonstrated remarkable efficiency in reducing vascular restenosis, protecting the injured tissue, and fostering re-endothelization of the target site. This innovative design will introduce a novel strategy for surface engineering of cardiovascular implants and other medical devices.
Collapse
Affiliation(s)
- Pai Peng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shili Ding
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Min Liang
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Haifei Shi
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
3
|
Xiong X, An M, Yuan L, Long X, Huang S. Increased monocytes and their derived indicators are associated with clinical severity of acute heart failure following acute myocardial infarction. Front Cardiovasc Med 2025; 12:1566635. [PMID: 40276256 PMCID: PMC12018373 DOI: 10.3389/fcvm.2025.1566635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Objective Monocytes play a significant role in the pathophysiology of acute myocardial infarction (AMI). The relationship between monocytes, their derived indicators, and the severity of acute heart failure following AMI remains unclear. Therefore, this study aims to investigate the association of monocytes and their derived indicators with clinical severity of acute heart failure in the patients with AMI. Methods In total of 173 patients with AMI were enrolled in this retrospective study. The demographic data and relevant medical histories were obtained. Monocytes and lipid levels were measured. All patients were divided into two groups based on killip classification. Killip class III-IV was defined as acute severe heart failure, while killip class I-II was defined as acute non-severe heart failure. Results Monocyte count, monocyte-to-white blood cell ratio, and monocyte-to-lymphocyte ratio were significantly higher in patients with acute severe heart failure compared to those with acute non-severe heart failure (P < 0.05). Multivariate logistic regression analysis showed that monocyte count, monocyte-to-white blood cell ratio, and monocyte-to-lymphocyte ratio were independently associated with acute severe heart failure (P < 0.05). Moreover, monocyte count, monocyte-to-white blood cell ratio, and monocyte-to-lymphocyte ratio were linked to NT-proBNP concentrations (P < 0.05). Receiver-operating characteristic curve analysis showed that monocyte count, monocyte-to-white blood cell ratio, and monocyte-to-lymphocyte ratio could identify acute severe heart failure in patients following AMI to some extent (P < 0.05). Conclusion The elevation of monocyte count, monocyte-to-white blood cell ratio, and monocyte-to-lymphocyte ratio correlated with clinical severity of acute heart failure following AMI, and offered potential discriminating value for cardiogenic pulmonary edema and shock following AMI.
Collapse
Affiliation(s)
- Xinlin Xiong
- Department of Cardiology, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | | | | | | | | |
Collapse
|
4
|
Wang P, Chen W, Zhang J, Pan C, Lv Y, Sun Y, Wang Y, Ma X, Gao C, Chen T, Wu A, Zheng J. Advances in the treatment of atherosclerotic plaque based on nanomaterials. Nanomedicine (Lond) 2025; 20:869-881. [PMID: 40109186 PMCID: PMC11988221 DOI: 10.1080/17435889.2025.2480049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Atherosclerosis is the leading cause of cardiovascular disease worldwide, posing not only a significant threat to cardiovascular health but also impairing the function of multiple organs, with severe cases potentially being life-threatening. Consequently, the effective treatment of atherosclerosis is of paramount importance in reducing the mortality associated with cardiovascular diseases. With the advancement of nanomedicine and a deeper understanding of the pathological mechanisms underlying atherosclerosis, nanomaterials have emerged as promising platforms for precise diagnosis and targeted therapeutic strategies. These materials offer notable advantages, including targeted drug delivery, enhanced bioavailability, improved drug stability, and controlled release. This review provides an overview of the mechanisms underlying atherosclerotic plaque development and examines nanomaterial-based therapeutic approaches for managing atherosclerotic plaques, including therapies targeting cholesterol metabolism, anti-inflammatory strategies, macrophage clearance, and immunotherapy. Additionally, the paper discusses the current technical challenges associated with the clinical transformation of these therapies. Finally, the potential future integration of nanomaterials, smart nanomaterials, and artificial intelligence in the treatment of atherosclerosis is also explored.
Collapse
Affiliation(s)
- Pengyu Wang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Weiwei Chen
- Traditional Chinese Medicine Department, Minglou Street Community Health Service Center, Yingzhou, Ningbo, China
| | - Jingfeng Zhang
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Chunshu Pan
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| | - Yagui Lv
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yanzi Sun
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yanan Wang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Xuehua Ma
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Changyong Gao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Tianxiang Chen
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Jianjun Zheng
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, China
- Department of Radiology, Ningbo No.2 Hospital, Ningbo, China
| |
Collapse
|
5
|
Ramoni D, Carbone F, Kraler S, Di Vece D, Montecucco F, Liberale L. Inflammation-Driven Plaque Erosion in Atherosclerosis: A Focus on Complement System Pathways. Curr Atheroscler Rep 2025; 27:42. [PMID: 40119227 PMCID: PMC11928383 DOI: 10.1007/s11883-025-01279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/24/2025]
Abstract
PURPOSE OF REVIEW Complement system activation is implicated in various stages of atherogenesis, from fatty streak formation to plaque destabilization and thrombus formation, with its dreadful clinical sequelae such as myocardial infarction, stroke and premature death. In this review, we consider these issues and explore recent studies on complement activation in atherosclerotic plaque initiation and progression. RECENT FINDINGS Complement pathways impact plaque stability and healing through the modulation of inflammatory processes. Recent studies indicate that complement components, notably C3 and C5b-9, accelerate atherosclerosis progression through their interactions with endothelial cells, smooth muscle cells, and immune cells. Nonetheless, the beneficial versus deleterious effects of complement activation at different stages of atherogenesis remains a matter of ongoing debates. Research also investigates therapies targeting the complement cascade to mitigate plaque erosion and rupture. This review explores the ongoing debates surrounding complement activation in atherogenesis. We bring forward controversial findings and therapeutic strategies aimed at modulating complement cascade activation with the ultimate goal to reduce the burden of atherosclerotic cardiovascular disease.\.
Collapse
Affiliation(s)
- Davide Ramoni
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Davide Di Vece
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| |
Collapse
|
6
|
Zhao G, Zhao L, Miao Y, Yang L, Huang L, Hu Z. HSPB1 Orchestrates the Inflammation-Associated Transcriptome Profile of Atherosclerosis in HUVECs. FRONT BIOSCI-LANDMRK 2025; 30:36306. [PMID: 40018940 DOI: 10.31083/fbl36306] [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: 08/10/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Atherosclerosis (AS), with a profound inflammatory response, is the basis of cardiovascular diseases. Previous reports showed that heat shock protein family B member 1 (HSPB1) has a protective effect against AS, but the specific mechanism is still unclear. In this study, we aim to explore the functions and downstream targets of HSPB1 in human umbilical vein endothelial cells (HUVECs). METHODS Expression of the HSPB1 gene was knocked down in HUVECs. Cellular phenotype was then assessed and transcriptome data (RNA-seq) was analyzed to identify the potential targets regulated by HSPB1. Moreover, RNA-seq data for human fibroatheroma (GSE104140) from the gene expression omnibus (GEO) database was re-analyzed to verify the targets of HSPB1 in AS. RESULTS Silencing of HSPB1 significantly reduced apoptosis (p < 0.0001) and increased the proliferation (p < 0.05) of HUVECs. The 608 differentially expressed genes (DEGs) were identified after HSPB1 knockdown, including 423 upregulated genes. DEGs, including CXCL1, CXCL8, CXCL2, TRIB3, GAS5, SELE, and TNIP1, were enriched in inflammatory and immune response pathways. HSPB1 was also shown to affect alternative splicing patterns of hundreds of genes, especially those enriched in apoptotic processes, including ACIN1, IFI27, PAK4, UBE2D3, and FIS1. An overlapping gene set was found between the HSPB1-regulated and AS-induced transcriptome. This included 171 DEGs and 250 alternatively spliced genes that were also enriched in inflammatory/immune response- and apoptosis-associated pathways, respectively. CONCLUSION In summary, HSPB1 knockdown modulates the proliferation and apoptosis of HUVECs by regulating RNA levels and alternative splicing patterns. HSPB1 plays an important role in AS pathogenesis by modulating the inflammatory and immune response. This study provides novel insights for the investigation of future AS therapeutic strategies.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Vascular Surgery, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Li Zhao
- Department of Anesthesiology, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Yulin Miao
- Clinical Medical School, Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Lei Yang
- Department of Vascular Surgery, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Lizhen Huang
- Clinical Medical School, Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Zhipeng Hu
- Department of Vascular Surgery, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| |
Collapse
|
7
|
Tariq H, Bukhari SZ, An R, Dong J, Ihsan A, Younis MR. Stem cell-derived exosome delivery systems for treating atherosclerosis: The new frontier of stem cell therapy. Mater Today Bio 2025; 30:101440. [PMID: 39866781 PMCID: PMC11758955 DOI: 10.1016/j.mtbio.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. As a chronic inflammatory disease with a complicated pathophysiology marked by abnormal lipid metabolism and arterial plaque formation, atherosclerosis is a major contributor to CVDs and can induce abrupt cardiac events. The discovery of exosomes' role in intercellular communication has sparked a great deal of interest in them recently. Exosomes are involved in strategic phases of the onset and development of atherosclerosis because they have been identified to control pathophysiologic pathways including inflammation, angiogenesis, or senescence. This review investigates the potential role of stem cell-derived exosomes in atherosclerosis management. We briefly introduced atherosclerosis and stem cell therapy including stem cell-derived exosomes. The biogenesis of exosomes along with their secretion and isolation have been elaborated. The design engineering of exosomes has been summarized to present how drug loading and surface modification with targeting ligands can improve the therapeutic and targeting capacity of exosomes, demonstrating atheroprotective action. Moreover, the mechanism of action (endothelial dysfunction, reduction of dyslipidemia, macrophage polarization, vascular calcification, and angiogenesis) of drug-loaded exosomes to treat atherosclerosis has been discussed in detail. In the end, a comparative and balanced viewpoint has been given regarding the current challenges and potential solutions to advance exosome engineering for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Hassan Tariq
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ruibing An
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Jian Dong
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Rizwan Younis
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
8
|
Yang X, Yu Y, Hu G, Bai S, Wu J, Guo C. Causal effects of circulating immune cells on coronary atherosclerosis: Evidence from Mendelian randomization. Medicine (Baltimore) 2025; 104:e41361. [PMID: 39889170 PMCID: PMC11789883 DOI: 10.1097/md.0000000000041361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 02/02/2025] Open
Abstract
The role of circulating immune cells in coronary atherosclerosis remains unclear. This study aimed to assess the causal effects of various immune cells on coronary atherosclerosis using Mendelian randomization (MR). Circulating immune cell datasets were obtained from genome-wide association studies, and coronary atherosclerosis datasets were obtained from FinnGen. Single-nucleotide polymorphisms satisfying the assumptions of association, independence, and exclusivity were screened in the datasets and analyzed using MR, with inverse-variance weighted as the main method. Horizontal pleiotropy, heterogeneity, and sensitivity analyses were performed using the MR-Egger, Cochran Q, and leave-one-out analyses, respectively. The MR analysis showed that effector memory double negative (DN) (cluster of differentiation [CD]4-CD8-) %DN (odds ratio [OR]: 1.042, 95% confidence interval [CI]: 1.008-1.077, P = .014), CD4 on CD39+ CD4+ (OR: 1.027, 95% CI: 1.001-1.054, P = .040), C-X3-C motif chemokine receptor 1 on CD14+ CD16- monocytes (OR: 1.035, 95% CI: 1.010-1.060, P = .006), C-C chemokine receptor 7 on naive CD4+ (OR: 1.035, 95% CI: 1.006-1.076, P = .023), and immunoglobulin D- CD38- %lymphocytes (OR: 1.098, 95% CI: 1.016-1.187, P = .019) were associated with an increased genetic susceptibility to coronary atherosclerosis, with no horizontal pleiotropy (P ≥ .05). Cochran Q showed no heterogeneity (P ≥ .05), and the sensitivity analysis indicated that the results were robust. The MR analysis revealed various markers and immune cell subsets, including effector memory DN (CD4-CD8-) %DN, CD4 on CD39+ CD4+, C-X3-C motif chemokine receptor on CD14+ CD16- monocytes, C-C chemokine receptor 7 on naive CD4+, and IgD- CD38- %lymphocytes, associated with increased genetic susceptibility to coronary atherosclerosis. This provides a genetic explanation for the role of specific immune cells in inducing and exacerbating coronary artery disease and offers new ideas for the exploration of immune markers and immune-targeted drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yunfeng Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gang Hu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Bai
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingyi Wu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chenlu Guo
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
9
|
Ungvari Z, Fekete M, Varga P, Fekete JT, Buda A, Szappanos Á, Lehoczki A, Mózes N, Grosso G, Menyhart O, Munkácsy G, Tarantini S, Yabluchanskiy A, Ungvari A, Győrffy B. Impact of adherence to the Mediterranean diet on stroke risk. GeroScience 2025:10.1007/s11357-024-01491-8. [PMID: 39777701 DOI: 10.1007/s11357-024-01491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Stroke is a leading cause of morbidity and mortality worldwide, and dietary patterns have emerged as a significant modifiable factor in stroke prevention. The Mediterranean diet, characterized by high intake of fruits, vegetables, whole grains, nuts, olive oil, and fish, has been widely recognized for its cardiovascular benefits. However, its specific impact on stroke risk requires further elucidation. We conducted a comprehensive meta-analysis of 30 studies, including both cohort and case-control designs, to evaluate the relationship between adherence to the Mediterranean diet and the risk of stroke. A systematic search was performed across multiple databases, and a random-effects model was used to estimate pooled hazard ratios (HRs) with 95% confidence intervals (CIs). Heterogeneity was assessed using the I2 statistic, and publication bias was examined through funnel plots and Egger's regression test. Additionally, trial sequential analysis was conducted to determine the adequacy of the sample size. The meta-analysis revealed a significant reduction in stroke risk among individuals adhering to the Mediterranean diet, with a pooled HR of 0.88 (95% CI: 0.84-0.91). Notably, a significant heterogeneity was detected (I2 = 34%). The Z-score plot from trial sequential analysis confirmed that the sample sizes were sufficient to draw definitive conclusions. However, a potential publication bias was identified. The case-control studies confirmed a highly significant effect (HR = 0.54, 95% CI: 0.4-0.73). The funnel plots in both settings hinted at the presence of a potential publication bias, supported by a significant Egger's test. Our findings provide robust evidence supporting the protective effect of the Mediterranean diet against stroke. Despite the presence of some heterogeneity and potential publication bias, the cumulative evidence suggests that promoting the Mediterranean diet could serve as an effective public health strategy for stroke prevention. Further research is recommended to explore the underlying mechanisms and to assess the diet's impact across diverse populations.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Péter Varga
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - János Tibor Fekete
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Annamaria Buda
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Ágnes Szappanos
- Department of Vascular and Endovascular Surgery, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Noémi Mózes
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Otilia Menyhart
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Gyöngyi Munkácsy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, 7624, Pecs, Hungary
| |
Collapse
|
10
|
Li YS, Ren HC, Li H, Xing M, Cao JH. From oxidative stress to metabolic dysfunction: The role of TRPM2. Int J Biol Macromol 2025; 284:138081. [PMID: 39603285 DOI: 10.1016/j.ijbiomac.2024.138081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Metabolic syndromes including atherosclerosis, diabetes, obesity, and hypertension are increasingly prevalent worldwide. The disorders are the primary attributes of oxidative stress and inflammation. The transient receptor potential M2 (TRPM2) channel is a pivotal mediator linking oxidative stress to metabolic dysfunction. TRPM2, a non-selective cation channel activated by reactive oxygen species (ROS) and adenosine diphosphate ribose (ADPR), regulates calcium influx, inflammation, and cell death across various tissues. This review explores the structural and activation mechanisms of TRPM2, emphasizing its significance in metabolic diseases. Elevated levels of TRPM2 play a vital role in the disease progression by influencing physiological and cellular processes such as endothelial dysfunction, immune cell activation, and mitochondrial impairment. In conditions such as atherosclerosis, ischemic stroke, diabetes, obesity, and hypertension; TRPM2 exacerbates oxidative damage, amplifies inflammatory responses, and disrupts metabolic homeostasis. Recent research highlights the potential of TRPM2 as a therapeutic target, developing specified inhibitors. This review underscores the multifaceted role of TRPM2 in metabolic disorders and its promise as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Ying-Shuang Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hua-Cheng Ren
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hui Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Man Xing
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Jian-Hua Cao
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China.
| |
Collapse
|
11
|
Celeski M, Segreti A, Crisci F, Cricco R, Piscione M, Di Gioia G, Nusca A, Fossati C, Pigozzi F, Ussia GP, Solaro RJ, Grigioni F. The Role of Cardiac Troponin and Other Emerging Biomarkers Among Athletes and Beyond: Underlying Mechanisms, Differential Diagnosis, and Guide for Interpretation. Biomolecules 2024; 14:1630. [PMID: 39766337 PMCID: PMC11727179 DOI: 10.3390/biom14121630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Cardiovascular (CV) disease remains the leading cause of morbidity and mortality worldwide, highlighting the necessity of understanding its underlying molecular and pathophysiological pathways. Conversely, physical activity (PA) and exercise are key strategies in reducing CV event risks. Detecting latent CV conditions in apparently healthy individuals, such as athletes, presents a unique challenge. The early identification and treatment of CV disorders are vital for long-term health and patient survival. Cardiac troponin is currently the most commonly used biomarker for assessing CV changes in both athletes and the general population. However, there remains considerable debate surrounding the mechanisms underlying exercise-induced troponin elevations and its release in non-ischemic contexts. Thus, there is a pressing need to identify and implement more sensitive and specific biomarkers for CV disorders in clinical practice. Indeed, research continues to explore reliable biomarkers for evaluating the health of athletes and the effectiveness of physical exercise. It is essential to analyze current evidence on troponin release in non-ischemic conditions, post-strenuous exercise, and the complex biological pathways that influence its detection. Furthermore, this study summarizes current research on cytokines and exosomes, including their physiological roles and their relevance in various CV conditions, especially in athletes. In addition, this paper gives special attention to underlying mechanisms, potential biomarkers, and future perspectives.
Collapse
Affiliation(s)
- Mihail Celeski
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Andrea Segreti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Filippo Crisci
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Riccardo Cricco
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Giuseppe Di Gioia
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
- Institute of Sports Medicine and Science, Italian National Olympic Committee, Largo Piero Gabrielli 1, 00197 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Ross John Solaro
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
12
|
Pacinella G, Ciaccio AM, Tuttolomondo A. Molecular Links and Clinical Effects of Inflammation and Metabolic Background on Ischemic Stroke: An Update Review. J Clin Med 2024; 13:7515. [PMID: 39768436 PMCID: PMC11679813 DOI: 10.3390/jcm13247515] [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: 11/02/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025] Open
Abstract
Stroke is a major global health concern, with 12.2 million new cases and 6.6 million deaths reported in 2019, making it the second leading cause of death and third leading cause of disability worldwide. Ischemic stroke, caused by blood vessel occlusion, accounts for 87% of stroke cases and results in neuronal death due to oxygen and nutrient deprivation. The rising global stroke burden is linked to aging populations and increased metabolic risk factors like high blood pressure, obesity, and elevated glucose levels, which promote chronic inflammation. This article explores the intricate molecular and clinical interplay between inflammation and metabolic disorders, emphasizing their role in ischemic stroke development, progression, and outcomes.
Collapse
Affiliation(s)
| | | | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialized Medicine (PROMISE), University of Palermo, 90127 Palermo, Italy; (G.P.); (A.M.C.)
| |
Collapse
|
13
|
Onnis C, Virmani R, Madra A, Nardi V, Salgado R, Montisci R, Cau R, Boi A, Lerman A, De Cecco CN, Libby P, Saba L. Whys and Wherefores of Coronary Arterial Positive Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:2416-2427. [PMID: 39479766 PMCID: PMC11594009 DOI: 10.1161/atvbaha.124.321504] [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: 11/28/2024]
Abstract
Positive remodeling (PR) is an atherosclerotic plaque feature defined as an increase in arterial caliber at the level of an atheroma, in response to increasing plaque burden. The mechanisms that lead to its formation are incompletely understood, but its role in coronary atherosclerosis has major clinical implications. Indeed, plaques with PR have elevated risk of provoking acute cardiac events. Hence, PR figures among the high-risk plaque features that cardiac imaging studies should report. This review aims to provide an overview of the current literature on coronary PR. It outlines the pathophysiology of PR, the different techniques used to assess its presence, and the imaging findings associated to PR, on both noninvasive and invasive studies. This review also summarizes clinical observations, trials, and studies, focused on the impact of PR on clinical outcome.
Collapse
Affiliation(s)
- Carlotta Onnis
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Anna Madra
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital and Antwerp University Lier, Belgium (R.S.)
| | - Roberta Montisci
- Clinical Cardiology, Department of Medical Science and Public Health, University of Cagliari, Italy (R.M.)
| | - Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Alberto Boi
- Department of Cardiology, Azienda Ospedaliera Brotzu, Cagliari, Italy (A.B.)
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Carlo N. De Cecco
- Division of Cardiothoracic Imaging and Biomedical Informatics, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA (C.N.D.C.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (P.L.)
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| |
Collapse
|
14
|
Abduljabbar MH. PCSK9 Inhibitors: Focus on Evolocumab and Its Impact on Atherosclerosis Progression. Pharmaceuticals (Basel) 2024; 17:1581. [PMID: 39770423 PMCID: PMC11676890 DOI: 10.3390/ph17121581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
This paper investigates the therapeutic use of PCSK9 inhibitors, particularly Evolocumab, as monoclonal antibodies for the treatment of atherosclerosis based on recent literature reviews. PCSK9 is an outstanding example of a breakthrough in medical science, with advancements in understanding its biological function driving substantial progress in atherosclerosis treatment. Atherosclerotic cardiovascular disease (ASCVD) is a leading global cause of mortality, imposing substantial financial burdens on healthcare systems. Elevated low-density lipoprotein cholesterol (LDL-C), a modifiable risk factor, plays a pivotal role in the development of ASCVD. Emerging treatments such as PCSK9 inhibitors are now being introduced to combat this issue, with the goal of reducing ASCVD risk by directly targeting LDL-C levels. This discovery highlighted the potential of monoclonal antibodies to inhibit PCSK9, thereby enhancing LDL-C receptor activity. This breakthrough led to the development of Alirocumab and Evolocumab inhibitors, which typically reduce LDL-C levels by approximately 50%. This research underscores the importance of PCSK9 inhibitors in treating ASCVD, drawing on evidence from various randomized controlled trials such as FOURIER, ODYSSEY OUTCOMES, and VESALIUS-CV. These trials have also shown that PCSK9 inhibitors are effective and safe for the treatment of several cardiovascular disorders. PCSK9 inhibitors are therefore useful in patients who do not reach their target LDL-C levels when on the highest doses of statins or patients with very high cardiovascular risk who cannot tolerate statins at all.
Collapse
Affiliation(s)
- Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
15
|
Kelly FA, de Moraes FCA, Lôbo ADOM, Siebel VM, Leite M, de Almeida AM, Consolim-Colombo FM. Safety and efficacy of moderate-intensity statin plus ezetimibe versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease: A meta-analysis. J Clin Lipidol 2024; 18:e893-e904. [PMID: 39550238 DOI: 10.1016/j.jacl.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ASCVD), affects approximately 18.6 million individuals worldwide and poses a significant healthcare related challenge. Despite the established efficacy of both high-intensity statin monotherapy (HIS) and moderate-intensity statin plus ezetimibe (MIS+EZT) in ASCVD management, the optimal treatment strategy remains unclear. METHODS A thorough literature study was conducted across PubMed, Embase, and the Cochrane databases, focusing on studies that compared the effects of MIS+EZT with HIS monotherapy in ASCVD patients. RESULTS In the 13 included studies, involving 8,592 patients, 4,525 (52.67%) of which received moderate-intensity statin plus ezetimibe treatment. The follow-up period ranged from 4 to 156 weeks, with participant ages varying from 55.2 to 71 years old. Analysis revealed that MIS+EZT was associated with a significantly greater percentage of patients who achieved the goal of low-density lipoprotein cholesterol (LDL-C) <70 mg/dL. (odds ratio [OR] 1.76; 95% CI [1.26; 2.45]; p = 0.001; I² = 73%); LDL-C reduction (mean difference [MD] -5.05 mg/dL; 95% CI [-9.02;-1.07]; p < 0.013; I² = 56%); total cholesterol reduction (MD -7.91 mg/ dL; 95% CI [-14.90; -0.91]; p < 0.027; I² = 60%) and triglyceride reduction (MD -8.20 mg/ dL; 95% CI [-13.05; -3.35]; p < 0.001; I² = 2%). There was no statistical difference between groups in drug adverse reaction (risk ratio [RR] 1.19; 95% CI [0.79; 1.78]; p = 0.404; I² = 0%) and drug intolerance (RR 0.78; 95% CI [0.32; 1.92]; p = 0.584; I² = 35%). CONCLUSIONS This meta-analysis highlights the effectiveness of MIS+EZT in significantly improving lipid profile components for ASCVD patients, as can been seen through the greater percentage of patients achieving the LDL-C < 70 mg/dL target and lower LDL-C, total cholesterol and triglyceride levels. Importantly, there were no significant differences in the occurrence of overall adverse events and adverse drug reactions between the two groups.
Collapse
Affiliation(s)
| | | | | | | | - Marianna Leite
- Santa Marcelina College Medical School, São Paulo 08290-005, Brazil (Leite)
| | | | | |
Collapse
|
16
|
Trimarchi G, Pizzino F, Lilli A, De Caterina AR, Esposito A, Dalmiani S, Mazzone A, Di Bella G, Berti S, Paradossi U. Advanced Lung Cancer Inflammation Index as Predictor of All-Cause Mortality in ST-Elevation Myocardial Infarction Patients Undergoing Primary Percutaneous Coronary Intervention. J Clin Med 2024; 13:6059. [PMID: 39458009 PMCID: PMC11508711 DOI: 10.3390/jcm13206059] [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: 08/25/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background: The advanced lung cancer inflammation index (ALI) is an independent prognostic biomarker used to assess inflammation and nutritional status in various cancers, heart failure, and acute coronary syndromes. This study investigates the prognostic significance of ALI in patients experiencing ST-elevation myocardial infarction (STEMI) treated with primary percutaneous coronary intervention (pPCI), comparing its predictive abilities with the established Neutrophil-Lymphocyte Ratio (NLR). Methods: We conducted a retrospective analysis of 1171 patients from the Matrix Registry, encompassing demographic and clinical data for STEMI cases treated with pPCI, and ALI was determined using the formula [serum albumin (g/dL) × body mass index (kg/m2)]/NLR at the time of hospital admission. The primary outcome was all-cause mortality. Results: Of the 1171 patients, 86 died during the follow-up period. Univariate analysis identified age, female gender, smoking, hypertension, diabetes, prior myocardial infarction (PMI), lower left ventricular ejection fraction (LVEF), and reduced ALI as factors associated with mortality. Multivariate analysis confirmed age (HR: 1.1, 95% CI: 1.05-1.11, p < 0.001) and PMI (HR: 2.4, 95% CI: 1.4-4.3, p = 0.001) as prominent independent predictors, alongside ALI (HR: 0.95, 95% CI: 0.92-0.97, p < 0.001) and LVEF (HR: 0.98, 95% CI: 0.97-0.99, p = 0.04). An ALI cut-off of ≤10 indicated a higher mortality risk (HR: 2.3, 95% CI: 1.5-3.7, p < 0.001). The area under the curve for ALI (0.732) surpassed that for NLR (0.685), demonstrating ALI's superior predictive capability. Conclusions: ALI is an independent prognostic factor for all-cause mortality in STEMI patients undergoing pPCI, showing greater discriminatory power than NLR, particularly in patients with ALI values ≤ 10, who face a 2.3-fold higher mortality risk.
Collapse
Affiliation(s)
- Giancarlo Trimarchi
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.D.B.)
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Fausto Pizzino
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Alessio Lilli
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Alberto Ranieri De Caterina
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Augusto Esposito
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Stefano Dalmiani
- Fondazione Toscana Gabriele Monasterio, Via Moruzzi 1, 56100 Pisa, Italy;
| | - Annamaria Mazzone
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.D.B.)
| | - Sergio Berti
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| | - Umberto Paradossi
- Cardiology Unit, Heart Centre, Fondazione Gabriele Monasterio—Regione Toscana, 54100 Massa, Italy; (A.L.); (A.R.D.C.); (A.E.); (A.M.); (S.B.); (U.P.)
| |
Collapse
|
17
|
Guo Z, Zhang W, Gao H, Li Y, Li X, Yang X, Fan L. High expression levels of haem oxygenase-1 promote ferroptosis in macrophage-derived foam cells and exacerbate plaque instability. Redox Biol 2024; 76:103345. [PMID: 39255694 PMCID: PMC11414708 DOI: 10.1016/j.redox.2024.103345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Plaque rupture with consequent thrombosis is the leading cause of acute cardiovascular events, during which macrophage death is a hallmark. Ferroptosis is a pivotal intermediate link between early and advanced atherosclerosis. Existing evidence indicates the involvement of macrophage ferroptosis in plaque vulnerability; however, the exact mechanism remains elusive. The aim of this study was to explore key ferroptosis-related genes (FRGs) involved in plaque progression and the underlying molecular mechanisms involved. The expression landscape of FRGs was obtained from atherosclerosis-related GEO datasets. Molecular mechanism studies of ferroptosis were performed using bone marrow-derived macrophages (BMDMs) and macrophage-derived foam cells (MDFCs). Bioinformatics analysis and immunohistochemistry revealed that macrophage haem oxygenase-1 (HMOX1) is the key FRG involved in plaque destabilization. Hypoxic conditions induced a significant increase in Hmox1 expression in MDFCs but not in macrophages. In addition, the beneficial or deleterious effects of Hmox1 were dependent on the degree of Hmox1 induction. Hmox1 overexpression drove inflammatory responses and ferroptotic oxidative stress in MDFCs and aggravated the plaque burden in atherosclerotic model mice. Further mechanistic investigations demonstrated that hypoxia-mediated degradation of egl-9 family hypoxia-inducible factor 3 (Egln3) stabilized Hif1a, which subsequently promoted Hmox1 transcription. Our findings suggest that high Hmox1 expression under hypoxia is deleterious to MDFC viability and plaque stability, providing a reference for the management of acute cardiovascular events.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wan Zhang
- Department of Vascular Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Hongxia Gao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Yang Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Xiaohu Yang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Longhua Fan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China.
| |
Collapse
|
18
|
Buonpane A, Trimarchi G, Ciardetti M, Coceani MA, Alagna G, Benedetti G, Berti S, Andò G, Burzotta F, De Caterina AR. Optical Coherence Tomography in Myocardial Infarction Management: Enhancing Precision in Percutaneous Coronary Intervention. J Clin Med 2024; 13:5791. [PMID: 39407851 PMCID: PMC11477163 DOI: 10.3390/jcm13195791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
In acute myocardial infarction (AMI), the urgency of coronary revascularization through percutaneous coronary intervention (PCI) is paramount, offering notable advantages over pharmacologic treatment. However, the persistent risk of adverse events, including recurrent AMI and heart failure post-revascularization, underscores the necessity for enhanced strategies in managing coronary artery disease. Traditional angiography, while widely employed, presents significant limitations by providing only two-dimensional representations of complex three-dimensional vascular structures, hampering the accurate assessment of plaque characteristics and stenosis severity. Intravascular imaging, specifically optical coherence tomography (OCT), significantly addresses these limitations with superior spatial resolution compared to intravascular ultrasound (IVUS). Within the context of AMI, OCT serves dual purposes: as a diagnostic tool to accurately identify culprit lesions in ambiguous cases and as a guide for optimizing PCI procedures. Its capacity to differentiate between various mechanisms of acute coronary syndrome, such as plaque rupture and spontaneous coronary dissection, enhances its diagnostic potential. Furthermore, OCT facilitates precise lesion preparation, optimal stent sizing, and confirms stent deployment efficacy. Recent meta-analyses indicate that OCT-guided PCI markedly improves safety and efficacy in revascularization, subsequently decreasing the risks of mortality and complications. This review emphasizes the critical role of OCT in refining patient-specific therapeutic approaches, aligning with the principles of precision medicine to enhance clinical outcomes for individuals experiencing AMI.
Collapse
Affiliation(s)
- Angela Buonpane
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Largo Agostino Gemelli, 1, 00168 Roma, Italy; (A.B.); (F.B.)
| | - Giancarlo Trimarchi
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Marco Ciardetti
- Cardiology and Pneumology Division, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy; (M.C.); (M.A.C.)
| | - Michele Alessandro Coceani
- Cardiology and Pneumology Division, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy; (M.C.); (M.A.C.)
| | - Giulia Alagna
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
| | - Giovanni Benedetti
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| | - Sergio Berti
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| | - Giuseppe Andò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
| | - Francesco Burzotta
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Largo Agostino Gemelli, 1, 00168 Roma, Italy; (A.B.); (F.B.)
| | - Alberto Ranieri De Caterina
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| |
Collapse
|
19
|
Diz-Küçükkaya R, İyigün T, Albayrak Ö, Eker C, Günel T. JAK2V617F Mutation in Endothelial Cells of Patients with Atherosclerotic Carotid Disease. Turk J Haematol 2024; 41:167-174. [PMID: 38801025 PMCID: PMC11589362 DOI: 10.4274/tjh.galenos.2024.2024.0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 05/29/2024] Open
Abstract
Objective It has been shown that clonal mutations occur in hematopoietic stem cells with advancing age and increase the risk of death due to atherosclerotic vascular diseases, similarly to myeloproliferative neoplasms. Endothelial cells (ECs) and hematopoietic stem cells develop from common stem cells called hemangioblasts in the early embryonic period. However, the presence of hemangioblasts in the postnatal period is controversial. In this study, JAK2 gene variants were examined in patients with atherosclerotic carotid disease and without any hematological malignancies. Materials and Methods Ten consecutive patients (8 men and 2 women) with symptomatic atherosclerotic carotid stenosis were included in this study. ECs (CD31+CD45-) were separated from tissue samples taken by carotid endarterectomy. JAK2 variants were examined in ECs, peripheral blood mononuclear cells, and oral epithelial cells of the patients with next-generation sequencing. Results The median age of the patients was 74 (range: 58-80) years and the median body mass index value was 24.44 (range: 18.42-30.85) kg/m2. Smoking history was present in 50%, hypertension in 80%, diabetes in 70%, and ischemic heart disease in 70% of the cases. The JAK2V617F mutation was detected in the peripheral blood mononuclear cells of 3 of the 10 patients, and 2 patients also had the JAK2V617F mutation in their ECs. The JAK2V617F mutation was not found in the oral epithelial cells of any of the patients. Conclusion In this study, for the first time in the literature, we showed that the JAK2V617F mutation was found somatically in both peripheral blood cells and ECs in patients with atherosclerosis. This finding may support that ECs and hematopoietic cells originate from a common clone or that somatic mutations can be transmitted to ECs by other mechanisms. Examining the molecular and functional changes caused by the JAK2V617F mutation in ECs may help open a new avenue for treating atherosclerosis.
Collapse
Affiliation(s)
- Reyhan Diz-Küçükkaya
- İstanbul University, Institute of Graduate Studies in Science, Department of Molecular Biology and Genetics, İstanbul, Türkiye
| | - Taner İyigün
- Turkish Ministry of Health, Mehmet Akif Ersoy Chest and Cardiovascular Surgery Education and Research Hospital, Clinic of Cardiovascular Surgery, İstanbul, Türkiye
| | - Özgür Albayrak
- Koç University Hospital Research Center for Translational Medicine, Flow Cytometry Core Facility, İstanbul, Türkiye
| | - Candan Eker
- İstanbul Bilgi University Faculty of Engineering and Natural Sciences, Department of Genetics and Bioengineering, İstanbul, Türkiye
| | - Tuba Günel
- İstanbul University, Institute of Graduate Studies in Science, Department of Molecular Biology and Genetics, İstanbul, Türkiye
| |
Collapse
|
20
|
Wu X, Pan X, Zhou Y, Pan J, Kang J, Yu JJJ, Cao Y, Quan C, Gong L, Li Y. Identification of key genes for atherosclerosis in different arterial beds. Sci Rep 2024; 14:6543. [PMID: 38503760 PMCID: PMC10951242 DOI: 10.1038/s41598-024-55575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/25/2024] [Indexed: 03/21/2024] Open
Abstract
Atherosclerosis (AS) is the pathologic basis of various cardiovascular and cerebrovascular events, with a high degree of heterogeneity among different arterial beds. However, mechanistic differences between arterial beds remain unexplored. The aim of this study was to explore key genes and potential mechanistic differences between AS in different arterial beds through bioinformatics analysis. Carotid atherosclerosis (CAS), femoral atherosclerosis (FAS), infrapopliteal atherosclerosis (IPAS), abdominal aortic atherosclerosis (AAS), and AS-specific differentially expressed genes (DEGs) were screened from the GSE100927 and GSE57691 datasets. Immune infiltration analysis was used to identify AS immune cell infiltration differences. Unsupervised cluster analysis of AS samples from different regions based on macrophage polarization gene expression profiles. Weighted gene co-expression network analysis (WGCNA) was performed to identify the most relevant module genes with AS. Hub genes were then screened by LASSO regression, SVM-REF, and single-gene differential analysis, and a nomogram was constructed to predict the risk of AS development. The results showed that differential expression analysis identified 5, 4, 121, and 62 CAS, FAS, IPAS, AAS-specific DEGs, and 42 AS-common DEGs, respectively. Immune infiltration analysis demonstrated that the degree of macrophage and mast cell enrichment differed significantly in different regions of AS. The CAS, FAS, IPAS, and AAS could be distinguished into two different biologically functional and stable molecular clusters based on macrophage polarization gene expression profiles, especially for cardiomyopathy and glycolipid metabolic processes. Hub genes for 6 AS (ADAP2, CSF3R, FABP5, ITGAX, MYOC, and SPP1), 4 IPAS (CLECL1, DIO2, F2RL2, and GUCY1A2), and 3 AAS (RPL21, RPL26, and RPL10A) were obtained based on module gene, gender stratification, machine learning algorithms, and single-gene difference analysis, respectively, and these genes were effective in differentiating between different regions of AS. This study demonstrates that there are similarities and heterogeneities in the pathogenesis of AS between different arterial beds.
Collapse
Affiliation(s)
- Xize Wu
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No. 41 Jianshe Road, Chongchuan District, Nantong, 226000, Jiangsu, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Xue Pan
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
- Dazhou Vocational College of Chinese Medicine, Dazhou, 635000, Sichuan, China
| | - Yi Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Jiaxiang Pan
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jian Kang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - J J Jiajia Yu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Yingyue Cao
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Chao Quan
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No. 41 Jianshe Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| | - Lihong Gong
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China.
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| | - Yue Li
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| |
Collapse
|
21
|
Laurino S, Russi S, Omer LC, D’Angelo A, Bozza G, Gallucci G, Falco G, Roviello G, Bochicchio AM. The Conundrum of Cancer-Associated Thrombosis: Lesson Learned from Two Intriguing Cases and Literature Review. Diseases 2024; 12:47. [PMID: 38534971 PMCID: PMC10969593 DOI: 10.3390/diseases12030047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 01/12/2025] Open
Abstract
The correlation between cancer and venous thromboembolism (VTE) is solid, whereas the knowledge about cancer-related arterial thromboembolism (ATE) still needs a deeper investigation to clarify its pathogenesis. We describe two cases that represent useful hints for a comprehensive review of the thrombotic issue. A 75-year-old man with advanced rectal cancer treated with fluoropyrimidines suffered two catheter-related VTE events managed according to current guidelines. There was no indication for "extended" anticoagulant therapy for him, but during antithrombotic wash-out and fluoropyrimidines plus panitumumab regimen, he suffered a massive right coronary artery (RCA) thrombosis. Another patient with no cardiovascular (CV) risk factors and affected by advanced bladder cancer was treated with a platinum-containing regimen and suffered an acute inferior myocardial infarction 2 days after chemotherapy administration. He was successfully treated with primary Percutaneous Transluminal Coronary Angioplasty of RCA, discontinuing platinum-based therapy. Our observations raise the issue of cancer-associated thrombosis (CAT) complexity and the potential correlation between arterial and venous thrombotic events. Moreover, physicians should be aware of the thrombotic risk associated with anticancer therapies, suggesting that an appropriate prophylaxis should be considered.
Collapse
Affiliation(s)
- Simona Laurino
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| | - Sabino Russi
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| | - Ludmila Carmen Omer
- Trial Office, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| | - Alberto D’Angelo
- Department of Oncology, Royal United Hospital, Bath BA1 3NG, UK;
| | - Giovanni Bozza
- Medical Oncology Unit, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| | - Giuseppina Gallucci
- Cardiology Unit, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| | - Geppino Falco
- Department of Biology, Università degli Studi di Napoli Federico II, 80138 Naples, Italy;
| | - Giandomenico Roviello
- Clinical Oncologic Unit, Careggi Hospital, University of Florence, 50121 Florence, Italy;
| | - Anna Maria Bochicchio
- Multispecialty Tumor Board, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, Italy;
| |
Collapse
|
22
|
Kounatidis D, Vallianou NG, Poulaki A, Evangelopoulos A, Panagopoulos F, Stratigou T, Geladari E, Karampela I, Dalamaga M. ApoB100 and Atherosclerosis: What's New in the 21st Century? Metabolites 2024; 14:123. [PMID: 38393015 PMCID: PMC10890411 DOI: 10.3390/metabo14020123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
ApoB is the main protein of triglyceride-rich lipoproteins and is further divided into ApoB48 in the intestine and ApoB100 in the liver. Very low-density lipoprotein (VLDL) is produced by the liver, contains ApoB100, and is metabolized into its remnants, intermediate-density lipoprotein (IDL) and low-density lipoprotein (LDL). ApoB100 has been suggested to play a crucial role in the formation of the atherogenic plaque. Apart from being a biomarker of atherosclerosis, ApoB100 seems to be implicated in the inflammatory process of atherosclerosis per se. In this review, we will focus on the structure, the metabolism, and the function of ApoB100, as well as its role as a predictor biomarker of cardiovascular risk. Moreover, we will elaborate upon the molecular mechanisms regarding the pathophysiology of atherosclerosis, and we will discuss the disorders associated with the APOB gene mutations, and the potential role of various drugs as therapeutic targets.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Second Department of Internal Medicine, Hippokration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Aikaterini Poulaki
- Hematology Unit, Second Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | | | - Fotis Panagopoulos
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|