1
|
Yin X, Chen X, Wang T, Yang J, Yu J, Yang J. LncRNA SCARNA8 promotes atherosclerotic plaque instability by inhibiting macrophage efferocytosis. Epigenetics 2025; 20:2487317. [PMID: 40356342 PMCID: PMC12077458 DOI: 10.1080/15592294.2025.2487317] [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: 09/20/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
In recent years, findings suggest that long noncoding RNAs (lncRNAs) are closely related to the development of atherosclerosis (AS), but there is a lack of studies on the involvement of lncRNA-regulated cytosolic burial in the regulation of AS. In this study, we investigated the mechanism by which lncRNA SCARNA8 affects macrophage cell burial to regulate AS. The cytosolic burial-associated target gene regulated by lncRNA SCARNA8 was PPARG. LncRNA SCARNA8 was increased in the carotid unstable plaque group, whereas PPARG was decreased. Ox-LDL led to the up-regulation of lncRNA SCARNA8 expression and apoptosis in Raw264.7 cells in a time-, concentration-dependent manner. Knockdown of lncRNA SCARNA8 upregulated PPARG and reduced apoptosis in Raw264.7 cells. In addition, knockdown of lncRNA SCARNA8 improved the stability of atherosclerotic plaques by promoting cellular burial of Raw264.7 cells. LncRNA SCARNA8 is a key regulator of plaque vulnerability, and targeting lncRNA SCARNA8 May provide a novel means for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Xiaoliang Yin
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Xiaodong Chen
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Jianling Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Jiahui Yu
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
- Center for Precision Neurosurgery and Oncology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
2
|
Xiao Y, Bi X, Zhang R, Li Y, Sun W, Hao Y. Restoration of vascular dysfunction resulting from maternal high-fat diet via modulation of the NLRP3/IL-1β axis. Clin Exp Hypertens 2025; 47:2440342. [PMID: 39722596 DOI: 10.1080/10641963.2024.2440342] [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: 10/28/2024] [Revised: 11/24/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
This study investigated the impact of maternal high-fat diet on vascular function and endothelial homeostasis in offspring. We found that offspring exposed to maternal high-fat diet exhibited elevated blood pressure, impaired abdominal aortic vascular function, and endothelial homeostasis imbalance. These changes were accompanied by increased levels of reactive oxygen species (ROS) and upregulation of pro-inflammatory cytokines (including IL-1β, TNF-α, IL-6, and IL-10). Treatment with NLRP3 or IL-1β inhibitors prevented the deterioration in vascular function, reduced endothelial NO production, and inflammation induced by maternal high-fat diet exposure compared to the control group. The findings suggest that during pregnancy, mitigating the vascular impairments in offspring induced by maternal high-fat diet can be achieved by inhibiting the NLRP3/IL-1β pathway.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xianru Bi
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Rongjie Zhang
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yu Li
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Wei Sun
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yingxue Hao
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Hua S, Sun L, Zhang H, Shiu C, Zhang S, Zhu Y, Yan X, Gu P, Huang Z, Jiang W. Yiqi Wenyang decoction protects against the development of atherosclerosis by inhibiting vascular inflammation. PHARMACEUTICAL BIOLOGY 2025; 63:264-274. [PMID: 40254717 PMCID: PMC12010649 DOI: 10.1080/13880209.2025.2492650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
CONTEXT Vascular inflammation is a key process in the pathogenesis of atherosclerosis, which is regulated by NF-κB pathway. Yiqi Wenyang decoction (YQWY), a Traditional Chinese medicine (TCM) formula, has anti-inflammatory properties and may inhibit this pathway, potentially offering anti-atherosclerotic effects. OBJECTIVE The purpose of this study is to investigate the effects of YQWY on atherosclerosis and the underlying mechanism. Materials and methods: ApoE-/- mice were fed a Western diet and administered with YQWY (low or high dose), atorvastatin, or vehicle for 13 weeks. The size of atherosclerotic plaques was assessed using ORO staining. Vascular inflammation was evaluated with IF or IHC staining. The mechanisms and signaling pathways underlying the effect of YQWY on vasculature were studied using transcriptomic analysis and were validated in vitro in endothelial cells and macrophages. RESULTS YQWY attenuated atherosclerotic plaque development which was associated with reduced vascular inflammation as demonstrated by transcriptomic analysis of aorta. This was verified by reduced expression of proinflammatory chemokines, adhesion molecules, and inflammatory cytokines in aortas from YQWY-treated mice at both mRNA and protein levels. Mechanistically, YQWY suppressed NF-κB activation in endothelial cells and, to a lesser extent, macrophages possibly. DISCUSSION AND CONCLUSIONS YQWY protects against vascular inflammation and atherosclerosis by suppressing NF-κB pathway, suggesting the potential of YQWY and its active ingredients as novel anti-atherosclerotic therapeutics.
Collapse
Affiliation(s)
- Shuang Hua
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Lingling Sun
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chiwen Shiu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujie Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingqun Yan
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Gu
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Endocrinology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Zhe Huang
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Department of Cardiology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Li J, Gao Q, Liu H, Liu S, Wang Y, Sun X, Zheng J, Yang H, Hu B. Integrating 16S rDNA sequencing analysis and targeted metabolomics to explore the mechanism of Xiexin Tang in treating atherosclerosis mice induced by high-fat diet. J Pharm Biomed Anal 2025; 259:116760. [PMID: 40014894 DOI: 10.1016/j.jpba.2025.116760] [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: 12/11/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Xiexin Tang (XXT) is a classic Chinese medicine formula that can be used to treat Atherosclerosis (AS). This study aimed to investigate the mechanism by which XXT regulated AS lipid levels. Firstly, the mixture components of XXT were analyzed by High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Then, the AS model based on Apolipoprotein E knockout (ApoE-/-) mice was established. Cytokines related to lipid metabolism and bile acid metabolism were detected by Quantitative Real-time PCR (qRT-PCR). 16S rDNA gene sequencing was performed to analyze differential bacterial populations, and the mechanism of XXT regulation of bile acids affecting lipid metabolism was further explored by targeted metabolomics. Further, antibiotic-treated mice were used to investigate the role of gut microbiota in the anti-AS effect of XXT. The results showed that XXT attenuated the lipid levels and reversed the abnormal elevation of cytokines, such as hepatic lipid metabolism and inflammatory reaction in AS mice. XXT also repaired the gut barrier damage and reversed gut microbiota disorders in AS mice. Furthermore, the metabolic levels of bile acids were reshaped by XXT. Whereas, in the absence of gut microbiota, XXT failed to attenuate lipid levels and inhibit the expression of cytokines related to inflammation and bile acid metabolism in AS mice and failed to play a role in ultimately treating AS. In conclusion, XXT could effectively inhibit the inflammatory reaction and lipid accumulation in AS mice, and this effect was closely related to its remodeling of gut microbiota to regulate bile acid metabolism.
Collapse
MESH Headings
- Animals
- Drugs, Chinese Herbal/pharmacology
- Atherosclerosis/drug therapy
- Atherosclerosis/metabolism
- Mice
- Gastrointestinal Microbiome/drug effects
- Metabolomics/methods
- Lipid Metabolism/drug effects
- Diet, High-Fat/adverse effects
- Male
- Bile Acids and Salts/metabolism
- Mice, Inbred C57BL
- Chromatography, High Pressure Liquid/methods
- Tandem Mass Spectrometry/methods
- RNA, Ribosomal, 16S/genetics
- Mice, Knockout, ApoE
- Disease Models, Animal
- DNA, Ribosomal/genetics
- Cytokines/metabolism
- Mice, Knockout
- Liver/metabolism
- Liver/drug effects
Collapse
Affiliation(s)
- Junling Li
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Qianru Gao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Songlin Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanchun Wang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiongjie Sun
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China.
| |
Collapse
|
5
|
Adamopoulou E, Dimitriadis K, Kyriakoulis K, Pyrpyris N, Beneki E, Fragkoulis C, Konstantinidis D, Aznaouridis K, Tsioufis K. Defining "Vulnerable" in coronary artery disease: predisposing factors and preventive measures. Cardiovasc Pathol 2025; 77:107736. [PMID: 40228760 DOI: 10.1016/j.carpath.2025.107736] [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: 01/28/2025] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
The likelihood of a plaque to cause an acute coronary syndrome (ACS) depends on several factors, both lesion- and patient-related. One of the most investigated and established contributing factors is the presence of high-risk or "vulnerable plaque" characteristics, which have been correlated with increased incidence of major adverse cardiovascular events (MACE). The recognition, however, that a significant percentage of vulnerable plaques do not result in causing clinical events has led the scientific community towards the more multifaceted concept of "vulnerable patients". Incorporating the morphological features of an atherosclerotic plaque into its hemodynamic surroundings can better predict the chance of its disruption, as altered fluid dynamics play a significant role in plaque destabilization. The advances in coronary imaging and the field of computational fluid dynamics (CFD) can contribute to develop more accurate lesion- and patient-related ACS prediction models that take into account both the morphology of a plaque and the forces applied upon it. The aim of this review is to provide the latest data regarding the aforementioned predictive factors as well as relevant preventive measures.
Collapse
Affiliation(s)
- Eleni Adamopoulou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece.
| | - Konstantinos Kyriakoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Dimitris Konstantinidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Aznaouridis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| |
Collapse
|
6
|
Rahimi F, Westermann D, Breitbart P. Editorial: Age-related ten-year outcomes after percutaneous coronary intervention of in-stent restenosis. Int J Cardiol 2025; 430:133183. [PMID: 40127824 DOI: 10.1016/j.ijcard.2025.133183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Affiliation(s)
- Faridun Rahimi
- Department of Cardiology and Angiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Südring 15, 79189 Bad Krozingen, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Südring 15, 79189 Bad Krozingen, Germany
| | - Philipp Breitbart
- Department of Cardiology and Angiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Südring 15, 79189 Bad Krozingen, Germany.
| |
Collapse
|
7
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [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: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
8
|
Farias HR, Ramos JMO, Griesang CT, Santos L, Junior OVR, Souza DG, Ferreira FS, Somacal S, Martins LAM, de Souza DOG, Moreira JCF, Wyse ATS, Guma FTCR, de Oliveira J. LDL Exposure Disrupts Mitochondrial Function and Dynamics in a Hippocampal Neuronal Cell Line. Mol Neurobiol 2025; 62:6939-6950. [PMID: 39302616 DOI: 10.1007/s12035-024-04476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Hypercholesterolemia has been associated with cognitive dysfunction and neurodegenerative diseases. Moreover, this metabolic condition disrupts the blood-brain barrier, allowing low-density lipoprotein (LDL) to enter the central nervous system. Thus, we investigated the effects of LDL exposure on mitochondrial function in a mouse hippocampal neuronal cell line (HT-22). HT-22 cells were exposed to human LDL (50 and 300 μg/mL) for 24 h. After this, intracellular lipid droplet (LD) content, cell viability, cell death, and mitochondrial parameters were assessed. We found that the higher LDL concentration increases LD content compared with control. Both concentrations increased the number of Annexin V-positive cells, indicating apoptosis. Moreover, in mitochondrial parameters, the LDL exposure on hippocampal neuronal cell line leads to a decrease in mitochondrial complexes I and II activities in both concentrations tested and a reduction in Mitotracker™ Red fluorescence and Mitotracker™ Red and Mitotracker™ Green ratio in the higher concentration, indicating mitochondrial impairment. The LDL incubation induces mitochondrial superoxide production and decreases superoxide dismutase activity in the lower concentration in HT-22 cells. Finally, LDL exposure increases the expression of genes associated with mitochondrial fusion (OPA1 and mitofusin 2) in the lower concentration. In conclusion, our findings suggest that LDL exposure induces mitochondrial dysfunction and modulates mitochondrial dynamics in the hippocampal neuronal cells.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jessica Marques Obelar Ramos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Caroline Tainá Griesang
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Lucas Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Osmar Vieira Ramires Junior
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Debora Guerini Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Silva Ferreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Sabrina Somacal
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Leo Anderson Meira Martins
- Programa de Pós-Graduação em Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
9
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025; 22:399-413. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [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] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Englisch C, Ay C. Clonal Hematopoiesis and Thrombosis. Am J Hematol 2025; 100:1049-1060. [PMID: 40192106 PMCID: PMC12067151 DOI: 10.1002/ajh.27682] [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: 02/18/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 05/13/2025]
Abstract
Clonal hematopoiesis (CH) has been the focus of many research efforts in the last years and has emerged as a risk modifier for cardiovascular disease morbidity and mortality. While substantial evidence has accumulated regarding its impact on arterial system diseases, the connection with venous thrombosis has only recently been explored. Both clinical and preclinical evidence suggest that the magnitude and mechanism underlying the association of CH with cardiovascular events vary depending on the specific mutated gene involved, indicating a causal link between CH and thrombosis development, not only in the arterial system, particularly in the context of atherosclerosis, but also in venous thrombosis. Although this growing body of knowledge has driven translational research and provided insights for improving clinical management, several questions remain unanswered. This review aims to summarize the available evidence on the link between CH and thrombosis, while highlighting the gaps that need to be addressed in future research.
Collapse
Affiliation(s)
- Cornelia Englisch
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| | - Cihan Ay
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| |
Collapse
|
11
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025; 22:431-442. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [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] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
12
|
Anghelache M, Voicu G, Anton R, Safciuc F, Boteanu D, Deleanu M, Turtoi M, Simionescu M, Manduteanu I, Calin M. Inflammation resolution-based treatment of atherosclerosis using biomimetic nanocarriers loaded with specialized pro-resolving lipid mediators. Mater Today Bio 2025; 32:101733. [PMID: 40255582 PMCID: PMC12008601 DOI: 10.1016/j.mtbio.2025.101733] [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: 01/23/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Recent studies have shown that chronic inflammation in atherosclerotic (ATH) lesions is due to an inability to resolve the inflammatory response. We evaluated the therapeutic potential of specialized pro-resolving mediators (SPMs) incorporated into biomimetic lipid nanoemulsions covered with macrophage membranes (Bio-LN/SPMs) to enhance their stability, targeting, and bioactivity in resolving atherosclerotic plaque inflammation. We utilized both in vitro and in vivo experimental models to test this hypothesis. In vitro, we found that Bio-LN/SPMs significantly reduced the inflammatory markers VCAM-1, MCP-1 in TNF-α-activated endothelial and smooth muscle cells, and iNOS, and NLRP3 in LPS-activated macrophages. In contrast, free SPMs exhibited a more modest effect. In vivo, the i.v. administration of Bio-LN/SPMs in ApoE-deficient mice with progressive atherosclerotic lesions developed after administration for 4 and 8 weeks of a high-fat diet, reduced plasma triglycerides, improved renal function, and decreased plasma proteins associated with complement activation and inflammation (i.e. C4d, C5b-9, IL-6, and MCP-1) to a greater extent than other treatment groups. Bio-LN/SPMs also affected circulated monocyte subpopulations by increasing the percentage of anti-inflammatory Ly6Clow monocytes and reducing that of pro-inflammatory Ly6Chigh monocytes. Additionally, they promoted the transition of macrophages in atherosclerotic plaques to a reparative M2 phenotype. They decreased the production of TNF-α, IL-1β, and IL-6 cytokines, along with lipid deposits in the aorta of ApoE-deficient mice. These findings demonstrate the improved therapeutic efficacy of Bio-LN/SPMs compared to unincorporated SPMs and standard nanoemulsions (LN/SPMs), emphasizing their potential as a novel approach for treating atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Maria Anghelache
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Geanina Voicu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Ruxandra Anton
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Florentina Safciuc
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Delia Boteanu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Mariana Deleanu
- “Liquid and Gas Chromatography” Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Mihaela Turtoi
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Maya Simionescu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Ileana Manduteanu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Manuela Calin
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| |
Collapse
|
13
|
Zhang N, Tian X, Sun D, Tse G, Xie B, Zhao Z, Liu T. Clonal hematopoiesis, cardiovascular disease and cancer treatment-induced cardiotoxicity. Semin Cancer Biol 2025; 111:89-114. [PMID: 40023267 DOI: 10.1016/j.semcancer.2025.02.007] [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: 04/23/2024] [Revised: 01/05/2025] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
Clonal hematopoiesis (CH) arises when a substantial proportion of mature blood cells is derived from a single hematopoietic stem cell lineage. It is considered to be a premalignant state that predisposes individuals to an increased risk of cancers. Recently, emerging evidence has demonstrated a strong association between CH and both the incidence and mortality of cardiovascular diseases (CVD), with the relative risks being comparable to those attributed to traditional cardiovascular risk factors. In addition, CH has been suggested to play a role in CVD and anti-cancer treatment-related cardiotoxicity amongst cancer survivors. Moreover, certain forms of chemotherapy and radiation therapy have been shown to promote the clonal expansion of specific CH-related mutations. Consequently, CH may play a substantial role in the realm of cardio-oncology. In this review, we discuss the association between CH with cancer and CVD, with a special focus on anti-cancer treatment-related cardiotoxicity, discuss possible future research avenues and propose a systematic approach for clinical practice.
Collapse
Affiliation(s)
- Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xu Tian
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dongkun Sun
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
14
|
Sanghvi MM, Young WJ, Naderi H, Burns R, Ramírez J, Bell CG, Munroe PB. Using Genomics to Develop Personalized Cardiovascular Treatments. Arterioscler Thromb Vasc Biol 2025; 45:866-881. [PMID: 40244646 DOI: 10.1161/atvbaha.125.319221] [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: 02/04/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Advances in genomic technologies have significantly enhanced our understanding of both monogenic and polygenic etiologies of cardiovascular disease. In this review, we explore how the utilization of genomic information is bringing personalized medicine approaches to the forefront of cardiovascular disease management. We describe how genomic data can resolve diagnostic uncertainty, support cascade screening, and inform treatment strategies. We discuss how genome-wide association studies have identified thousands of genetic variants associated with polygenic cardiovascular diseases, and how integrating these insights into polygenic risk scores can enhance personalized risk prediction beyond traditional clinical algorithms. We detail how pharmacogenomics approaches leverage genotype information to guide drug selection and mitigate adverse events. Finally, we present the paradigm-shifting approach of gene therapy, which holds the promise of being a curative intervention for cardiovascular conditions.
Collapse
Affiliation(s)
- Mihir M Sanghvi
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - William J Young
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - Hafiz Naderi
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - Richard Burns
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| | - Julia Ramírez
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Aragon Institute of Engineering Research, University of Zaragoza, Spain (J.R.)
- Centro de Investigación Biomédica en Red, Biomedicina, Bioingeniería y Nanomedicina, Zaragoza, Spain (J.R.)
| | - Christopher G Bell
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| | - Patricia B Munroe
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| |
Collapse
|
15
|
Barbui T, Stefano VD, Rossi E, Ghirardi A, Carobbio A, Loscocco GG, Condorelli A, Guglielmelli P. Thrombosis-Driven Disease Progression in JAK2-Mutant Polycythemia Vera and Essential Thrombocythemia: Reassessing Risk-Based Management. Am J Hematol 2025; 100 Suppl 4:66-73. [PMID: 40062566 DOI: 10.1002/ajh.27657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 05/13/2025]
Abstract
This paper explores emerging therapies in polycythemia vera and essential thrombocythemia, focusing on thrombosis as a driver of disease progression leading to myelofibrosis, blast phase, second cancers, and mortality. While the thrombosis rate in high-risk patients has declined, it remains persistently high in low-risk individuals, with most events being arterial. Inflammation driven by JAK2 V617F mutation plays a primary role in pathogenesis, and mounting evidence suggests arterial thrombosis itself can fuel a self-sustaining cycle of inflammation, thereby accelerating hematologic and systemic complications. Early intervention with cytoreductive and anti-inflammatory drugs may not only prevent incidental thrombosis but also disrupt this inflammatory circuit.
Collapse
Affiliation(s)
- Tiziano Barbui
- FROM, Fondazione per la Ricerca Ospedale di Bergamo ETS, Bergamo, Italy
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Elena Rossi
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Arianna Ghirardi
- FROM, Fondazione per la Ricerca Ospedale di Bergamo ETS, Bergamo, Italy
| | - Alessandra Carobbio
- Dipartimento di Scienze Mediche e Chirurgiche, Materno-Infantili e dell'Adulto, Università di Modena-Reggio Emilia, Modena, Italy
| | - Giuseppe Gaetano Loscocco
- CRIMM, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | | | - Paola Guglielmelli
- CRIMM, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| |
Collapse
|
16
|
Caller T, Moore KJ, Lehmann LH, Wu SM, Leor J. Insights Into Heart-Tumor Interactions in Heart Failure. Circ Res 2025; 136:1262-1285. [PMID: 40403117 DOI: 10.1161/circresaha.124.325490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure (HF) often coexists with cancer. Beyond the known cardiotoxicity of some cancer treatments, HF itself has been associated with increased cancer incidence. The 2 conditions share common risk factors, mechanisms, and interactions that can worsen patient outcomes. The bidirectional relationship between HF and cancer presents a complex interplay of factors that are not fully understood. Recent preclinical evidence suggests that HF may promote tumor growth via the release of protumorigenic factors from the injured heart, revealing HF as a potentially protumorigenic condition. Our review discusses the biological crosstalk between HF and cancer, emphasizing the impact of HF on tumor growth, with inflammation, and modulating the immune system as central mechanisms. We further explore the clinical implications of this connection and propose future research directions. Understanding the mechanistic overlap and interactions between HF and cancer could lead to new biomarkers and therapies, addressing the growing prevalence of both conditions and enhancing approaches to diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Tal Caller
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| | - Kathryn J Moore
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine (K.J.M.)
| | - Lorenz H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Germany (L.H.L.)
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg/Mannheim, Germany (L.H.L.)
- German Cancer Research Center (DKFZ), Heidelberg, Germany (L.H.L.)
| | - Sean M Wu
- Stanford Cardiovascular Institute (S.M.W.), Stanford University School of Medicine, CA
- Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| |
Collapse
|
17
|
Avery EG, Haag LM, McParland V, Kedziora SM, Zigra GJ, Valdes DS, Kirchner M, Popp O, Geisberger S, Nonn O, Karlsen TV, N’Diaye G, Yarritu A, Bartolomaeus H, Bartolomaeus TUP, Tagiyeva NA, Wimmer MI, Haase N, Zhang YD, Wilhelm A, Grütz G, Tenstad O, Wilck N, Forslund SK, Klopfleisch R, Kühl AA, Atreya R, Kempa S, Mertins P, Siegmund B, Wiig H, Müller DN. Intestinal interstitial fluid isolation provides novel insight into the human host-microbiome interface. Cardiovasc Res 2025; 121:803-816. [PMID: 39804196 PMCID: PMC12101326 DOI: 10.1093/cvr/cvae267] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 03/28/2025] Open
Abstract
AIMS The gastrointestinal (GI) tract is composed of distinct sub-regions, which exhibit segment-specific differences in microbial colonization and (patho)physiological characteristics. Gut microbes can be collectively considered as an active endocrine organ. Microbes produce metabolites, which can be taken up by the host and can actively communicate with the immune cells in the gut lamina propria with consequences for cardiovascular health. Variation in bacterial load and composition along the GI tract may influence the mucosal microenvironment and thus be reflected its interstitial fluid (IF). Characterization of the segment-specific microenvironment is challenging and largely unexplored because of lack of available tools. METHODS AND RESULTS Here, we developed methods, namely tissue centrifugation and elution, to collect IF from the mucosa of different intestinal segments. These methods were first validated in rats and mice, and the tissue elution method was subsequently translated for use in humans. These new methods allowed us to quantify microbiota-derived metabolites, mucosa-derived cytokines, and proteins at their site-of-action. Quantification of short-chain fatty acids showed enrichment in the colonic IF. Metabolite and cytokine analyses revealed differential abundances within segments, often significantly increased compared to plasma, and proteomics revealed that proteins annotated to the extracellular phase were site-specifically identifiable in IF. Lipopolysaccharide injections in rats showed significantly higher ileal IL-1β levels in IF compared to the systemic circulation, suggesting the potential of local as well as systemic effect. CONCLUSION Collection of IF from defined segments and the direct measurement of mediators at the site-of-action in rodents and humans bypasses the limitations of indirect analysis of faecal samples or serum, providing direct insight into this understudied compartment.
Collapse
Affiliation(s)
- Ellen G Avery
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Lea-Maxie Haag
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, Berlin 10117, Germany
| | - Victoria McParland
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sarah M Kedziora
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Gabriel J Zigra
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Daniela S Valdes
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charite—Universitätsmedizin Berlin, Berlin, Germany
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sabrina Geisberger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Olivia Nonn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Gabriele N’Diaye
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alex Yarritu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Theda U P Bartolomaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nurana A Tagiyeva
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Moritz I Wimmer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Faculty of Medicine, Universität Tübingen, Tübingen, Germany
| | - Nadine Haase
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Yiming D Zhang
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Andreas Wilhelm
- CheckImmune GmbH, BerlinBioCube, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Gerald Grütz
- CheckImmune GmbH, BerlinBioCube, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Nicola Wilck
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Charité—Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Sofia K Forslund
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Univeristät Berlin and Humboldt Universität zu Berlin, iPATH, Berlin, Berlin, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander University, Erlangen, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charite—Universitätsmedizin Berlin, Berlin, Germany
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Britta Siegmund
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Dominik N Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
18
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
19
|
Kallikourdis M, Cochran JD, Walsh K, Condorelli G. Contributions of Noncardiac Organ-Heart Immune Crosstalk and Somatic Mosaicism to Heart Failure: Current Knowledge and Perspectives. Circ Res 2025; 136:1208-1232. [PMID: 40403105 DOI: 10.1161/circresaha.125.325489] [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: 02/12/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is the final outcome of most cardiovascular diseases and shares risk factors with other cardiovascular pathologies. Among these, inflammation plays a central role in disease progression and myocardial remodeling. Over the past 2 decades, numerous studies have explored immune-related mechanisms in cardiovascular disease, highlighting the importance of immune cross-talk between the heart and extra-cardiac organs, including bone marrow, spleen, liver, gut, and adipose tissue. This review examines how immune interactions among these organs contribute to heart failure pathogenesis, with a focus on clonal hematopoiesis, an age-related alteration of hematopoietic stem cells that fosters pathological bone marrow-heart communication. Additionally, we explore recent advances in the understanding of clonal hematopoiesis and its role in heart failure, emphasizing its implications for prognosis and potential therapeutic interventions. By integrating insights from immunology, metabolism, and aging, we provide a comprehensive perspective on the immunologic determinants of heart failure, paving the way for precision medicine approaches aimed at mitigating cardiovascular risk.
Collapse
Affiliation(s)
- Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (M.K., G.C.)
- Department of Cardiovascular Medicine, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (M.K., G.C.)
| | - Jesse D Cochran
- Hematovascular Biology Center, Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Center (J.D.C., K.W.), University of Virginia School of Medicine, Charlottesville, VA
- Medical Scientist Training Program (J.D.C.), University of Virginia School of Medicine, Charlottesville, VA
| | - Kenneth Walsh
- Hematovascular Biology Center, Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Center (J.D.C., K.W.), University of Virginia School of Medicine, Charlottesville, VA
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (M.K., G.C.)
- Department of Cardiovascular Medicine, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (M.K., G.C.)
| |
Collapse
|
20
|
Charchar FJ, Marques FZ. A paradigm shift in cardiovascular research: new method isolates intestinal interstitial fluid to understand gut microbiome and host cross-talk. Cardiovasc Res 2025; 121:697-698. [PMID: 40145177 DOI: 10.1093/cvr/cvaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Affiliation(s)
- Fadi J Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Victoria 3350, Australia
- Deparment of Medicine, Austin Health, University of Melbourne, Melbourne, Victoria 3350, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute, Monash University, Melbourne, Australia
- Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
21
|
Janssen M, Müller-Tidow C. Inflammation conjoins differentiation and resistance in AML. Blood 2025; 145:2405-2407. [PMID: 40402530 DOI: 10.1182/blood.2025028651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Affiliation(s)
- Maike Janssen
- University Hospital Heidelberg
- University of Heidelberg
- European Molecular Biology Laboratory
| | - Carsten Müller-Tidow
- University Hospital Heidelberg
- University of Heidelberg
- European Molecular Biology Laboratory
| |
Collapse
|
22
|
Lindkvist M, Göthlin Eremo A, Paramel GV, Anisul Haque S, Rydberg Millrud C, Rattik S, Grönberg C, Liberg D, Sirsjö A, Fransén K. IL1RAP Expression in Human Atherosclerosis: A Target of Novel Antibodies to Reduce Vascular Inflammation and Adhesion. J Am Heart Assoc 2025; 14:e039557. [PMID: 40371594 DOI: 10.1161/jaha.124.039557] [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: 10/21/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Blockade of IL1RAP (interleukin 1 receptor associated protein) was recently shown to reduce atherosclerosis in mice, but the effect on human vascular cells is largely unknown. Targeting the IL1RAP coreceptor represents a novel strategy to block the IL1RAP-dependent cytokines IL (interleukin)-1, IL-33, and IL-36. In the present study, we aimed to evaluate the role of novel antibodies targeting IL1RAP to reduce the effects of IL-1β, IL-33, or IL-36γ in human vascular cells. METHODS Expression of IL1RAP was observed in human atherosclerotic plaques by immunohistochemistry and microarray and in endothelial cells by flow cytometry. Endothelial cells were cultured with IL-1β, IL-33, or IL-36γ cytokines with or without IL1RAP antibodies and analyzed with Olink proteomics, ELISA, Western blot, and real-time quantitative polymerase chain reaction. The functional effect of IL1RAP antibodies on endothelial cells were analyzed with adhesion and permeability assays. RESULTS Olink proteomics showed inhibition of the inflammatory proteins LIF (leukemia inhibitory factor), OPG (osteoprotegerin), CCL4 (C-C motif chemokine ligand 4), and MCP-3 (monocyte chemoattractant protein 3) by IL1RAP-blockade in endothelial cells after IL-1β stimulation. In addition, the IL1RAP antibodies inhibited IL-1β, and IL-33 induced IL-6 and IL-8 secretion. Secretion of MCP-1 (monocyte chemoattractant protein 1) was induced by IL-1β, IL-33, and IL-36γ, and subsequently was inhibited by IL1RAP antibodies. Similar effects were found on mRNA expression level. Endothelial expression of the adhesion markers ICAM1, VCAM1, and SELE were significantly reduced by IL1RAP antibodies, and neutrophil adhesion to endothelial cells induced by IL-1β and IL-33 was reduced by IL1RAP blockade. In human atherosclerotic lesions, IL1RAP expression correlated with markers of inflammation like IL6, IL8, and MCP1. CONCLUSIONS IL1RAP-targeting antibodies can reduce the expression of inflammatory cytokines and markers of adhesion in endothelial cells, which may be of importance for future putative targeted treatments against cardiovascular disease.
Collapse
Affiliation(s)
- Madelene Lindkvist
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Anna Göthlin Eremo
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- Department of Clinical Research Laboratory, School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Geena Varghese Paramel
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Sheikh Anisul Haque
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | | | | | | | | | - Allan Sirsjö
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| | - Karin Fransén
- Cardiovascular Research Centre, Faculty of Medicine and Health Örebro University Örebro Sweden
- School of Medical Sciences, Faculty of Medicine and Health Örebro University Örebro Sweden
| |
Collapse
|
23
|
Zimerman A, Kunzler ALF, Weber BN, Ran X, Murphy SA, Wang H, Honarpour N, Keech AC, Sever PS, Sabatine MS, Giugliano RP. Intensive Lowering of LDL Cholesterol Levels With Evolocumab in Autoimmune or Inflammatory Diseases: An Analysis of the FOURIER Trial. Circulation 2025; 151:1467-1476. [PMID: 40255182 DOI: 10.1161/circulationaha.124.072756] [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: 10/15/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Patients with an autoimmune or inflammatory disease (AIID) are at increased cardiovascular risk and may benefit more from statin therapy. In the FOURIER trial (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk), the PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor evolocumab lowered low-density lipoprotein cholesterol levels, but not hsCRP (high-sensitivity C-reactive protein) levels, and reduced the risk of cardiovascular events. METHODS FOURIER was a randomized trial of evolocumab versus placebo in 27 564 patients with stable atherosclerosis who were taking statins. This analysis focused on the effect of evolocumab in patients with or without an AIID, defined as any autoimmune or chronic inflammatory condition. The primary end point was a composite of cardiovascular death, myocardial infarction, stroke, unstable angina, or coronary revascularization. RESULTS At baseline, 889 patients (3.2%) had an AIID, most commonly rheumatoid arthritis (33.7%) or psoriasis (15.6%). Median (interquartile range) low-density lipoprotein cholesterol levels were 90.0 mg/dL (79.5-105.5) and 91.5 mg/dL (79.5-108.5) in patients with or without an AIID, respectively (P=0.025), and the placebo-adjusted percent reduction with evolocumab was consistent (60.2% versus 59.0%; P=0.57). Baseline hsCRP was higher in patients with an AIID (median 2.1 versus 1.7 mg/L; P<0.001) and did not significantly change with evolocumab in either group. Compared with placebo, evolocumab reduced the rate of the primary end point by 14% in patients without an AIID (hazard ratio, 0.86 [95% CI, 0.80-0.93]) and by 42% in patients with an AIID (hazard ratio, 0.58 [95% CI, 0.38-0.89]; Pinteraction=0.066). Likewise, evolocumab reduced the key secondary end point of cardiovascular death, myocardial infarction, or stroke by 19% in patients without an AIID (hazard ratio, 0.81 [95% CI, 0.74-0.89]) and 58% in those with an AIID (hazard ratio, 0.42 [95% CI, 0.24-0.74]; Pinteraction=0.022). CONCLUSIONS Intensive lowering of low-density lipoprotein cholesterol levels with evolocumab may lead to greater relative reduction in cardiovascular events in patients with an AIID. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT01764633.
Collapse
Affiliation(s)
- Andre Zimerman
- Hospital Moinhos de Vento, Moinhos de Vento College of Health Sciences, Porto Alegre, Brazil (A.Z., A.L.F.K.)
| | - Ana Laura F Kunzler
- Hospital Moinhos de Vento, Moinhos de Vento College of Health Sciences, Porto Alegre, Brazil (A.Z., A.L.F.K.)
| | - Brittany N Weber
- Division of Cardiovascular Medicine (B.N.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Xinhui Ran
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sabina A Murphy
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Huei Wang
- Biostatistics (H.W.), Amgen, Thousand Oaks, CA
| | | | - Anthony C Keech
- Sydney Medical School, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Australia (A.C.K.)
| | - Peter S Sever
- National Heart and Lung Institute, Imperial College London, UK (P.S.S.)
| | - Marc S Sabatine
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Robert P Giugliano
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Zhao C, Xu S, Yang Y, Shen X, Wang J, Xing S, Yu Z. Intersection of Cardio-Oncology: An Overview of Radiation-Induced Heart Disease in the Context of Tumors. J Am Heart Assoc 2025; 14:e040937. [PMID: 40357679 DOI: 10.1161/jaha.124.040937] [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] [Indexed: 05/15/2025]
Abstract
Radiation-induced heart disease (RIHD) is a prevalent cardiovascular complication of radiation therapy, with coronary heart disease being the most common manifestation. Clinical presentations of RIHD vary and may include conduction abnormalities, ischemic heart disease, cardiomyopathy, heart failure, and valvular damage. Even low doses of radiation significantly increase the risk of cardiovascular disease, often associated with severe stenosis detected via angiography. Radiation-induced damage to the cardiac endothelium triggers inflammatory responses and oxidative stress, which contribute to the progression of atherosclerosis. This study explores how radiation activates multiple signaling pathways through the generation of reactive oxygen species, resulting in vascular endothelial damage, cellular senescence, inflammatory responses, and DNA damage. It further examines the impact of radiation on vascular integrity and tight junction proteins, leading to increased vascular permeability and infiltration by inflammatory cells. From a clinical perspective, we emphasize the challenges posed by the coexistence of tumors in many patients with RIHD, as tumors complicate the microenvironment and may have mutually reinforcing interactions with radiation-induced damage. We also discuss various therapeutic strategies, including novel approaches targeting cellular senescence and immune responses, with a focus on the potential use of navitoclax and IL-6 (interleukin-6) inhibitors to prevent irreversible cardiomyocyte fibrosis and ongoing vascular damage. In conclusion, RIHD is a multifaceted disease involving complex biological processes and signaling pathways. Early intervention and targeted therapies are crucial for improving patient outcomes. Future research should prioritize uncovering the molecular mechanisms of RIHD and developing more effective therapeutic strategies.
Collapse
Affiliation(s)
- Chunan Zhao
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology Beijing Institute of Radiation Medicine Beijing China
| | - Shuai Xu
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Yanru Yang
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Xing Shen
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology Beijing Institute of Radiation Medicine Beijing China
| | - Jingjing Wang
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Shuang Xing
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology Beijing Institute of Radiation Medicine Beijing China
| | - Zuyin Yu
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology Beijing Institute of Radiation Medicine Beijing China
| |
Collapse
|
25
|
Mohammadnia N, Wesselink BE, Bax WA, Nidorf SM, Mosterd A, Fiolet ATL, Cetinyurek-Yavuz A, Thompson PL, Bangdiwala SI, Eikelboom JW, Cornel JH, El Messaoudi S. Cardiovascular Benefit of Colchicine in Relation to Baseline Risk: A Secondary Analysis of the LoDoCo2 Trial. J Am Heart Assoc 2025; 14:e038687. [PMID: 40371626 DOI: 10.1161/jaha.124.038687] [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: 09/04/2024] [Accepted: 04/04/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND The LoDoCo2 (Low-Dose Colchicine 2) trial showed that colchicine reduced the risk for cardiovascular events in patients with chronic coronary syndrome. Current guidelines recommend colchicine use in selected high-risk patients. The aim of this secondary analysis was to explore the relative and absolute benefits of colchicine according to baseline risk. METHODS The LoDoCo2 trial randomized 5522 patients to colchicine 0.5 mg or placebo. The primary end point was a composite of cardiovascular death, spontaneous myocardial infarction, ischemic stroke, or ischemia-driven coronary revascularization. First, a LoDoCo2 risk score was developed by Cox regression to identify high-risk features for the primary end point. Second, the Thrombolysis in Myocardial Infarction Risk Score for Secondary Prevention was applied to explore robustness of findings. RESULTS In the LoDoCo2 risk score, high-risk features were age ≥75, diabetes, and current smoker. In high-risk (≥1 high-risk feature), compared with low-risk (0 high-risk features) patients, colchicine was associated with consistent relative (high risk: hazard ratio [HR], 0.72 [95% CI, 0.56-0.94] versus low risk: HR, 0.67 [95% CI, 0.52-0.88]; P for interaction=0.73) and absolute benefits (high risk: HR, -1.33 [95% CI, -2.38 to -0.27] versus low risk: HR, -0.93 [95% CI -1.57 to -0.30] events per 100 person-years). Using the Thrombolysis in Myocardial Infarction Risk Score for Secondary Prevention, consistent relative and absolute benefits were found in high-, intermediate-, and low-risk patients. CONCLUSIONS In patients with chronic coronary syndrome, the relative and absolute benefits of colchicine were consistent in those at high, intermediate, and low risk for cardiovascular events. These findings support the use of colchicine across the spectrum of baseline risk. REGISTRATION URL: https://www.anzctr.org.au; Unique identifier: 12614000093684.
Collapse
Affiliation(s)
| | - Britta E Wesselink
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
| | - Willem A Bax
- Department of Internal Medicine Northwest Clinics Alkmaar Netherlands
| | - Stefan M Nidorf
- Heart and Vascular Research Institute of Western Australia Perth WA Australia
| | - Arend Mosterd
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology Meander Medical Center Amersfoort The Netherlands
| | - Aernoud T L Fiolet
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology University Medical Center Utrecht Utrecht The Netherlands
| | | | - Peter L Thompson
- Heart and Vascular Research Institute of Western Australia Perth WA Australia
- School of Medicine University of Western Australia Perth WA Australia
- Sir Charles Gairdner Hospital Perth WA Australia
| | - Shrikant I Bangdiwala
- Department of Health Research Methods, Evidence and Impact McMaster University Hamilton ON Canada
- Statistics Department Population Health Research Institute, McMaster University Hamilton ON Canada
| | | | - Jan H Cornel
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology Northwest Clinics Alkmaar Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
26
|
Fiolet ATL, Lin A, Kwiecinski J, Tutein Nolthenius J, McElhinney P, Grodecki K, Kietselaer B, Opstal TS, Cornel JH, Knol RJ, Schaap J, Aarts RAHM, Tutein Nolthenius AMFA, Nidorf SM, Velthuis BK, Dey D, Mosterd A. Effect of low-dose colchicine on pericoronary inflammation and coronary plaque composition in chronic coronary disease: a subanalysis of the LoDoCo2 trial. Heart 2025:heartjnl-2024-325527. [PMID: 40393691 DOI: 10.1136/heartjnl-2024-325527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/11/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Low-dose colchicine (0.5 mg once daily) reduces the risk of major cardiovascular events in coronary disease, but its mechanism of action is not yet fully understood. We investigated whether low-dose colchicine is associated with changes in pericoronary inflammation and plaque composition in patients with chronic coronary disease. METHODS We performed a cross-sectional, nationwide, subanalysis of the Low-Dose Colchicine 2 Trial (LoDoCo2, n=5522). CT angiography studies were performed in 151 participants randomised to colchicine or placebo coronary after a median treatment duration of 28.2 months. Pericoronary adipose tissue (PCAT) attenuation measurements around proximal coronary artery segments and quantitative plaque analysis for the entire coronary tree were performed using artificial intelligence-enabled plaque analysis software. RESULTS Median PCAT attenuation was not significantly different between the two groups (-79.5 Hounsfield units (HU) for colchicine versus -78.7 HU for placebo, p=0.236). Participants assigned to colchicine had a higher volume (169.6 mm3 vs 113.1 mm3, p=0.041) and burden (9.6% vs 7.0%, p=0.035) of calcified plaque, and higher volume of dense calcified plaque (192.8 mm3 vs 144.3 mm3, p=0.048) compared with placebo, independent of statin therapy. Colchicine treatment was associated with a lower burden of low-attenuation plaque in participants on a low-intensity statin, but not in those on a high-intensity statin (pinteraction=0.037). CONCLUSIONS Pericoronary inflammation did not differ among participants who received low-dose colchicine compared with placebo. Low-dose colchicine was associated with a higher volume of calcified plaque, particularly dense calcified plaque, which is considered a feature of plaque stability.
Collapse
Affiliation(s)
- Aernoud T L Fiolet
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
| | - Andrew Lin
- Monash Victorian Heart Institute and Monash Health Heart, Victorian Heart Hospital, Clayton, Victoria, Australia
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| | - Jacek Kwiecinski
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California, USA
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Julie Tutein Nolthenius
- Faculty of Medicine, Amsterdam University Medical Centre Amsterdam, Amsterdam, The Netherlands
| | - Priscilla McElhinney
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| | - Kajetan Grodecki
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| | - Bas Kietselaer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiology, Zuyderland Medical Centre, Heerlen, The Netherlands
| | - Tjerk S Opstal
- Department of Cardiology, Amsterdam University Medical Centre Amsterdam, Amsterdam, The Netherlands
| | - Jan Hein Cornel
- Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Remco Jj Knol
- Cardiac Imaging Division, Department of Nuclear Medicine, Noordwest Ziekenhuisgroep, Alkmaar, Noord-Holland, The Netherlands
| | - Jeroen Schaap
- Department of Cardiology, Amphia Hospital, Breda, The Netherlands
| | - Ruud A H M Aarts
- Department of Radiology, Amphia Hospital, Breda, The Netherlands
| | | | - Stefan M Nidorf
- Heart and Vascular Research Institute of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Birgitta K Velthuis
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| | - Arend Mosterd
- Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
- Department of Cardiology, Meander MC, Amersfoort, The Netherlands
| |
Collapse
|
27
|
Obare LM, Stephens VR, Wanjalla CN. Understanding residual risk of cardiovascular disease in people with HIV. Curr Opin HIV AIDS 2025:01222929-990000000-00164. [PMID: 40397567 DOI: 10.1097/coh.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
PURPOSE OF REVIEW Traditional cardiovascular risk factors, combined with persistent systemic inflammation, contribute to the increased prevalence of atherosclerotic cardiovascular disease (ASCVD) in people with HIV (PWH). This review highlights key findings from the REPRIEVE trial on statin-based primary prevention of major adverse cardiovascular events in PWH. It explores HIV-specific immune mechanisms contributing to residual cardiovascular risk. RECENT FINDINGS In REPRIEVE, statin therapy used for primary prevention of major adverse cardiovascular events in PWH decreased the plasma lipoprotein-associated phospholipase A2, oxidized low-density lipoprotein, and high-sensitivity C-reactive protein (hs-CRP). However, several inflammatory markers including soluble CD14 (sCD14), sCD163, interleukin (IL)-1β, interleukin (IL)-6, IL-10, and caspase 1 did not change. The HIV reservoir, dysfunctional CD4+ T cells, immunoglobulin G N-glycans, antiapolipoprotein A1 autoantibodies, trained immunity, and clonal hematopoiesis of indeterminate potential may contribute to residual inflammation. SUMMARY Despite antiretroviral and statin therapy, residual ASCVD risk in PWH underscores the need for targeted interventions. Anti-inflammatory therapies, including IL-6 and IL-1β inhibitors, CCR5 antagonists (e.g., maraviroc, cenicriviroc mesylate), and immunomodulatory agents like methotrexate and colchicine, are being explored. Understanding HIV-driven immune dysregulation may lead to novel strategies to mitigate cardiovascular risk in this population.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center
| | | | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center
- The Center for AIDS Health Disparities Research
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
28
|
Farias HR, Costa-Beber LC, Costa Rodrigues Guma FT, de Oliveira J. Hypercholesterolemia, oxidative stress, and low-grade inflammation: a potentially dangerous scenario to blood-brain barrier. Metab Brain Dis 2025; 40:205. [PMID: 40380979 DOI: 10.1007/s11011-025-01620-y] [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: 01/31/2024] [Accepted: 04/23/2025] [Indexed: 05/19/2025]
Abstract
For more than a century, hypercholesterolemia has been linked to atherosclerotic cardiovascular disease. Notably, this metabolic condition has also been pointed out as a risk factor for neurodegenerative diseases, such as Alzheimer's disease (AD). Oxidative stress seems to be the connective factor between hypercholesterolemia and cardio and neurological disorders. By disturbing redox homeostasis, hypercholesterolemia impairs nitric oxide (NO) availability, an essential vasoprotective element, and jeopardizes endothelial function and selective permeability. The central nervous system (CNS) is partially protected from peripheral insults due to an arrangement between endothelial cells, astrocytes, microglia, and pericytes that form the blood-brain barrier (BBB). The endothelial dysfunction related to hypercholesterolemia increases the risk of developing cardiovascular diseases and also initiates BBB breakdown, which is a cause of brain damage characterized by neuroinflammation, oxidative stress, mitochondrial dysfunction, and, ultimately, neuronal and synaptic impairment. In this regard, we reviewed the mechanisms by which hypercholesterolemia-induced oxidative stress affects peripheral vessels, BBB, and leads to memory deficits. Finally, we suggest oxidative stress as the missing link between hypercholesterolemia and dementia.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Lílian Corrêa Costa-Beber
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
29
|
Zheng Q, Lin Y, Zeng L, Chen S, Chen L, Lin X, Zhu J, Lin J, Weng X, Chai D. ITE-mediated AhR activation attenuates atherosclerosis by promoting macrophage M2-like polarization through NF-κB/LCN2 pathway suppression. Life Sci 2025; 375:123715. [PMID: 40389023 DOI: 10.1016/j.lfs.2025.123715] [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: 03/16/2025] [Revised: 05/04/2025] [Accepted: 05/10/2025] [Indexed: 05/21/2025]
Abstract
AIMS Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid accumulation and inflammation. Macrophage phenotypic transformation plays a critical role in AS progression. Aryl hydrocarbon receptor (AhR) has been proved to regulate the phenotype of macrophages. This study investigates the role and molecular mechanism of AhR activation by its endogenous ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) attenuates AS. MATERIALS AND METHODS We employed Western blotting to analyze the expression of AhR, NF-κB, and lipocalin-2 (LCN2). Flow cytometry and immunofluorescence staining were used to assess the phenotype of macrophages. Plaque progression was evaluated using pathological staining. Transcriptome sequencing was utilized to explore the potential mechanism by which AhR promotes macrophage phenotypic transformation. CUT&Tag-qPCR and lentivirus infection confirmed that the AhR/NF-κB/LCN2 pathway regulates macrophage polarization. KEY FINDINGS Activation of AhR by ITE reduced plaque area and inhibited lipid deposition. ITE significantly increased the number of M2-like macrophages both in vivo and in vitro. Transcriptome sequencing identified LCN2 as a key target for AhR-mediated macrophage M2-like polarization. Furthermore, AhR activation suppressed the NF-κB/LCN2 pathway. SIGNIFICANCE Our findings reveal that AhR promotes the macrophages to exhibit M2-like characteristics to attenuate AS by inhibiting the NF-κB/LCN2 pathway. These results suggest that AhR may serve as a novel therapeutic target for AS.
Collapse
Affiliation(s)
- Qiaowen Zheng
- Cardiovascular Department, Fuqing City Hospital, Fuzhou 350005, China
| | - Yifei Lin
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Lishan Zeng
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Shuaijie Chen
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Longqing Chen
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiaoyan Lin
- Echocardiological Department, the First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Jiang Zhu
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jinxiu Lin
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiuzhu Weng
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dajun Chai
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Key Laboratory of Metabolic Heart Disease in Fujian Province, Clinical Research Centre of Metabolic Cardiovascular Disease in Fujian Province, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
30
|
Gao A, Peng B, Gao Y, Yang Z, Li Z, Guo T, Qiu H, Gao R. Evaluation and comparison of inflammatory and insulin resistance indicators on recurrent cardiovascular events in patients undergoing percutaneous coronary intervention: a single center retrospective observational study. Diabetol Metab Syndr 2025; 17:157. [PMID: 40380257 PMCID: PMC12082960 DOI: 10.1186/s13098-025-01687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/30/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND The aim of this study was to evaluate and compare the usefulness of the C-reactive protein (CRP)-triglyceride glucose (TyG) index (CTI) and other insulin resistance (IR) or inflammatory indexes for predicting recurrent cardiovascular events in percutaneous coronary intervention (PCI)-treated patients. In addition, the mediating effects of systemic inflammation, represented by high-sensitive CRP (hs-CRP), on TyG index-associated adverse cardiovascular events across different subgroups were also evaluated. METHODS The formula for calculating the CTI was 0.412 × ln [high-sensitivity CRP (mg/L)] + ln [triglyceride (mg/dl) × fasting glucose (mg/dl)/2]. The primary endpoint was defined as the incidence of major adverse cerebrovascular and cardiovascular events (MACCEs), including cardiovascular death, nonfatal acute myocardial infarction (AMI), nonfatal ischemic stroke and repeat coronary revascularization. RESULTS Among the 2383 PCI-treated patients, 413 experienced MACCEs during a median of 34 months follow-up. Correlation analysis showed CTI was significantly associated with cardiometabolic factors. The CTI was the strongest predictor for MACCEs (adjusted HR 1.85, 95% CI 1.44-2.38) among the inflammatory and IR indicators. CTI had an incremental effect on the predictive ability of the prognostic model for MACCEs (NRI: 0.220, p < 0.001; IDI: 0.009, p < 0.001). Subgroup analysis revealed that the prognostic value of the CTI remained significant across all subgroups (all p < 0.05) whereas the predictive abilities of other IR or inflammatory indicators were more or less influenced by the metabolic abnormalities. Finally, mediation analysis revealed that the effects of systemic inflammation on TyG index-associated MACCEs were more prominent in patients with metabolic disorders. CONCLUSIONS CTI was a practical indicator for evaluating cardiometabolic diseases. Among the IR and inflammatory indicators, CTI was the most promising index for predicting recurrent cardiovascular risks in PCI-treated patients. TyG index-associated cardiovascular risks were partially mediated by systemic inflammation in patients with metabolic abnormalities.
Collapse
Affiliation(s)
- Ang Gao
- Department of Cardiology, Cardio-Metabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Peng
- Department of Cardiology, 3 Ward of Structural Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanan Gao
- Department of Cardiology, Cardio-Metabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Yang
- Department of Cardiology, Cardio-Metabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifan Li
- Department of Cardiology, Cardio-Metabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingting Guo
- Intensive Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- People's Hospital of Xizang Autonomous Region, Lhasa, China.
| | - Hong Qiu
- Department of Cardiology, Cardio-Metabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China.
| | - Runlin Gao
- Department of Cardiology, Coronary Artery Disease Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Huntermann R, Peres de Oliveira J, Barbosa LM, Queiroz I, Nunes Cavalcante D, de Oliveira Fischer Bacca C. Colchicine in acute coronary syndromes: a systematic review and meta-analysis of randomised controlled trials. Heart 2025:heartjnl-2025-325826. [PMID: 40379469 DOI: 10.1136/heartjnl-2025-325826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/27/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Acute coronary syndrome (ACS) is a global leading cause of morbidity, with residual inflammation contributing to recurrent events. Colchicine has been proposed as an adjunct therapy, but its efficacy remains uncertain. METHODS We performed a systematic review and meta-analysis. PubMed, Embase and Cochrane databases were searched for randomised controlled trials (RCTs) data comparing colchicine versus placebo in ACS. Risk ratio (RR) and mean difference with 95% CIs were computed for binary and continuous outcomes, respectively. Primary outcomes were adverse cardiovascular events (ACEs), mortality and safety. Random-effects models were used for pooled estimates. RESULTS Seventeen RCTs comprising 14 794 patients were included, of whom 7390 (50%) were randomised to colchicine. The mean patient age across the studies ranged from 54 to 63 years, in a follow-up period ranging from 5 days to 12 months. Colchicine reduced the incidence of recurrent ACS (RR 0.41, 95% CI 0.19 to 0.92; p=0.03; I²=55%) and unstable angina (RR 0.27, 95% CI 0.11 to 0.63; p<0.01; I²=0%). No meaningful differences were observed in all-cause mortality (RR 0.95, 95% CI 0.79 to 1.14; I²=12%), cardiovascular death (RR 1.03, 95% CI 0.82 to 1.30; I²=0%) or ACE (RR 0.77, 95% CI 0.59 to 1.01; p=0.05; I²=58%). Subgroup analyses suggested a dose-dependent effect, with 0.5 mg/day potentially reducing ACE (RR 0.63, 95% CI 0.45 to 0.88; I²=41%), but higher doses increasing gastrointestinal symptoms. CONCLUSION Low-dose colchicine may reduce recurrent ischaemic events in ACS, but evidence remains uncertain due to heterogeneity and limited long-term data. Safety and efficacy in women and optimal dosing require further investigation. TRIAL REGISTRATION NUMBER CRD42024627348.
Collapse
Affiliation(s)
- Ramon Huntermann
- Medical Sciences Research Center, University Center for the Development of Alto Vale, UNIDAVI, Rio do Sul, Brazil
| | - Juan Peres de Oliveira
- Medical Sciences Research Center, University Center for the Development of Alto Vale, UNIDAVI, Rio do Sul, Brazil
| | - Lucas M Barbosa
- Department of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ivo Queiroz
- Radiology Department, University of Wisconsin-Madison, Madison, Winsconsin, USA
| | | | | |
Collapse
|
32
|
Antonello J, Roy P. Damage-Associated Molecular Patterns (DAMPs) In Vascular Diseases. J Biol Chem 2025:110241. [PMID: 40381697 DOI: 10.1016/j.jbc.2025.110241] [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: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Research into the role of chronic sterile inflammation (i.e. a prolonged inflammatory state not caused by an infectious agent), in vascular disease progression has continued to grow over the last few decades. DAMPs have a critical role in this research due to their ability to link stress-causing cardiovascular risk factors to inflammatory phenotypes seen in vascular disease. In this mini-review, we will briefly summarize the DAMPs and receptor signaling pathways that have been extensively studied in the context of vascular disease, including TLRs, RAGE, cGAS-STING, and the NLRP3 inflammasome. In particular, we will discuss how these pathways can promote the release of pro-inflammatory cytokines and chemokines as well as vascular remodeling. Next, we will summarize the results of studies which have linked the various pro-inflammatory effects of DAMPs with the phenotypes in the context of vascular diseases including atherosclerosis, fibrosis, aneurysm, ischemia, and hypertension. Finally, we will discuss some pre-clinical and clinical trials that have targeted DAMPs, their receptors, or the products of their signaling pathways, and discuss the outlook and future directions for the field at large.
Collapse
Affiliation(s)
| | - Partha Roy
- Bioengineering, University of Pittsburgh; Pathology, University of Pittsburgh.
| |
Collapse
|
33
|
Mury P, Dagher O, Fortier A, Diaz A, Lamarche Y, Noly PE, Ibrahim M, Pagé P, Demers P, Bouchard D, Bernier PL, Poirier N, Moss E, Durrleman N, Jeanmart H, Pellerin M, Lettre G, Thorin-Trescases N, Carrier M, Thorin E. Quercetin Reduces Vascular Senescence and Inflammation in Symptomatic Male but Not Female Coronary Artery Disease Patients. Aging Cell 2025:e70108. [PMID: 40375481 DOI: 10.1111/acel.70108] [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: 01/24/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
Recent studies suggest that vascular senescence and its associated inflammation fuel the inflammaging to favor atherogenesis; whether these pathways can be therapeutically targeted in coronary artery disease (CAD) patients remains unknown. In a randomized, double-blind trial, 97 patients (78 men) undergoing coronary artery bypass graft surgery were treated with either quercetin (500 mg twice daily, 47 patients) or placebo (50 patients) for two days pre-surgery through hospital discharge. Primary outcomes were reduced inflammation and improved endothelial function ex vivo. Exploratory analyses included plasma proteomics and single-nuclei RNA sequencing of internal thoracic artery (ITA) samples. Quercetin treatment showed a trend toward reduced C-reactive protein at discharge (p = 0.073) and differentially modulated circulating inflammatory protein expression between men and women, with a pro-inflammatory effect of quercetin in females. Endothelial acetylcholine-induced relaxation improved significantly with quercetin (p = 0.049), with effects in men (p = 0.043) but not in women (p = 0.852). ITA transcriptomics revealed the overexpression of senescence and inflammaging pathways in male vascular cells, which quercetin reversed. In female cells, quercetin had minimal endothelial benefit and increased inflammaging in fibroblasts. In male cells, a candidate target of quercetin involves interactions between the receptor PLAUR and its ligands PLAU and SERPINE1. Post-operative atrial fibrillation incidence was significantly lower with quercetin, representing 4% of the patients compared to 18% in the placebo group (p = 0.033). In conclusion, short-term quercetin treatment effectively targeted vascular senescence in male CAD patients, improving inflammatory and functional outcomes. However, these benefits were not observed in female patients. Trial Registration: https://clinicaltrials.gov, NCT04907253.
Collapse
Affiliation(s)
- Pauline Mury
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Olina Dagher
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Annik Fortier
- Department of Biostatistics, Montreal Health Innovations Coordinating Centre (MHICC), Montréal, Québec, Canada
| | - Ariel Diaz
- CIUSSS-MCQ, Université de Montréal, Campus Mauricie, Trois-Rivières, Québec, Canada
| | - Yoan Lamarche
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre-Emmanuel Noly
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Marina Ibrahim
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Pagé
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Philippe Demers
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Denis Bouchard
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre-Luc Bernier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Nancy Poirier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Emmanuel Moss
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Nicolas Durrleman
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Hughes Jeanmart
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Michel Pellerin
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | - Michel Carrier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Eric Thorin
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
34
|
Furtado GE, de Barros MP, Rodrigues RN, Bachi ALL, Chupel MU, Rocha SV, Vieira RP, Hogervorst E, Teixeira AM, Ferreira JP. Examining the impact of 28-week multicomponent and strength exercises on brain health, salivary stress, and mental well-being in frail older women: A controlled trial analysis. Physiol Behav 2025; 294:114868. [PMID: 40024357 DOI: 10.1016/j.physbeh.2025.114868] [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: 03/25/2024] [Revised: 02/11/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND In recent years, the efficacy of various physical exercise programs in enhancing functional fitness among frail older adults has gained recognition. However, limited research has concurrently explored the long-term effects of exercise on brain health, stress biomarkers, and mental well-being. This study aimed to investigate the impact of two distinct chair-based exercise programs on salivary stress hormones and psychological well-being in frail older women over a 28-week period. METHODS A total of 140 individuals participated in the enrollment phase, with 84 eligible participants randomly assigned to three groups. Following the intervention, data from 60 participants were analyzed across the multicomponent exercise (MCE, n = 23), elastic band muscle-strength exercise (ESE, n=19), and non-exercise control (CG n=18) groups. Salivary biomarkers of alpha-amylase (α-AMY) Cortisol (COR), alpha-amylase/cortisol ratio, psychological indicators and physical frailty (PF) and functional fitness were assessed pre- and post-intervention. RESULTS Salivary COR levels exhibited a significant time × group interaction, with a moderate increase in MCE, a small decrease in ESE, and a substantial increase in CGne. Salivary α-AMY levels varied significantly over time and by group, with a small decrease in both exercise groups and a moderate increase in CGne. The α-AMY /COR ratio also displayed a significant interaction effect. Additionally, significant improvements were observed in PF compound scores, general self-efficacy, attitudes toward aging, and reductions in perceived stress and depressive symptoms (p < 0.05). CONCLUSIONS Notably, the MCE program demonstrated greater benefits than ESE. The observed associations between changes in α-AMY levels, mental well-being, and functional fitness indicators contribute novel evidence on the psychophysiological adaptations to long-term exercise. Importantly, reductions in PF scores correlated with improvements in self-efficacy, attitudes toward aging, and handgrip strength, reinforcing the link between functional fitness, stress regulation, and psychological well-being. These findings emphasize the need for tailored exercise interventions to enhance both physiological resilience and mental health in frail older populations.
Collapse
Affiliation(s)
- Guilherme Eustáquio Furtado
- Polytechnic University of Coimbra, Lagar dos Cortiços - S. Martinho do Bispo, Coimbra 3045-093, Portugal; Center for Studies on Natural Resources, Environment, and Society (CERNAS), Polytechnic University of Coimbra, Bencanta, Coimbra 3045-601, Portugal; SPRINT - Sport Physical activity and health Research & INnovation cenTer, Polytechnic University of Coimbra, Portugal.
| | - Marcelo Paes de Barros
- MSc/PhD Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo 01506-000, Brazil
| | - Rafael N Rodrigues
- Research Unit for Sport and Physical Activity (UID/PTD/04213/2020) at Faculty of Sport Science and Physical Education, University of Coimbra (FCDEF-UC), Portugal
| | - André Luís Lacerda Bachi
- Post-Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo 04829-300, Brazil
| | - Matheus Uba Chupel
- Research Unit for Sport and Physical Activity (UID/PTD/04213/2020) at Faculty of Sport Science and Physical Education, University of Coimbra (FCDEF-UC), Portugal; Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Saulo Vasconcelos Rocha
- Transnordestina Avenue, State University of Feira de Santana, s/n - Novo Horizonte, CEP 44036-900 - Feira de Santana, Bahia, Brazil
| | - Rodolfo P Vieira
- Postgraduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos-SP 11060-001, Brazil; Postgraduate Programs in Humam Movement and Rehabilitation and in Pharmaceutical Sciences, Evangelical University of Goias (UniEvangélica), Avenida Universitária Km 3,5, Anápolis-GO 75083-515, Brazil
| | - Eef Hogervorst
- Applied Cognitive Research NCSEM, Loughborough University, Loughborough, United Kingdom
| | - Ana Maria Teixeira
- Research Unit for Sport and Physical Activity (UID/PTD/04213/2020) at Faculty of Sport Science and Physical Education, University of Coimbra (FCDEF-UC), Portugal
| | - José Pedro Ferreira
- Research Unit for Sport and Physical Activity (UID/PTD/04213/2020) at Faculty of Sport Science and Physical Education, University of Coimbra (FCDEF-UC), Portugal
| |
Collapse
|
35
|
Medamana JL, Gelfand JM, Weber BN, Garshick MS. Cardiovascular disease risk in psoriatic disease: mechanisms and implications for clinical practice. Curr Opin Rheumatol 2025:00002281-990000000-00176. [PMID: 40357683 DOI: 10.1097/bor.0000000000001092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW Psoriasis is an immune-mediated pro-inflammatory skin condition that is associated with an increase in risk factors for cardiovascular disease, risk of ischemic heart disease, and cardiovascular death. Despite this, traditional modifiable atherosclerotic cardiovascular disease (ASCVD) risk factors are underdiagnosed and undertreated in patients with psoriasis. RECENT FINDINGS At a cellular level, psoriasis and atherosclerosis are driven by a host of shared inflammatory pathways, such as pro-inflammatory cytokines (TNF, IL-6), immune cells, and platelets which act synergistically to drive endothelial damage and atherosclerosis progression. SUMMARY Optimal prevention of cardiovascular disease in psoriasis centers around modifying known risk factors for the development of ASCVD and emerging data highlight the promise of treating inflammation to further decrease the risk of ASCVD.
Collapse
Affiliation(s)
- John L Medamana
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Joel M Gelfand
- Department of Dermatology and Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Sciences in Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Brittany N Weber
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael S Garshick
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
36
|
Cabral JE, Wu A, Zhou H, Pham MA, Lin S, McNulty R. Targeting the NLRP3 inflammasome for inflammatory disease therapy. Trends Pharmacol Sci 2025:S0165-6147(25)00073-2. [PMID: 40374417 DOI: 10.1016/j.tips.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/17/2025]
Abstract
The NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome is a megadalton complex implicated in numerous inflammation-driven diseases including COVID-19, Alzheimer's disease, and gout. Although past efforts have focused on inhibiting IL-1β downstream of NLRP3 activation using drugs such as canakinumab, no FDA-approved NLRP3-targeted inhibitors are currently available. MCC950, a direct NLRP3 inhibitor, showed promise but exhibited off-target effects. Recent research has focused on optimizing the sulfonylurea-based MCC950 scaffold by leveraging recent structural and medicinal chemistry insights into the NLRP3 nucleotide-binding and oligomerization (NACHT) domain to improve solubility and clinical efficacy. In addition, oxidized DNA (oxDNA) has emerged as a key inflammasome trigger, and molecules targeting the pyrin domain have shown promise in inhibiting NLRP3 activation. This review discusses the role of NLRP3 in inflammation-related diseases, the status of ongoing clinical trials, and emerging small-molecule therapeutics targeting NLRP3.
Collapse
Affiliation(s)
- Julia Elise Cabral
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Anna Wu
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Haitian Zhou
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Minh Anh Pham
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Sophia Lin
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Reginald McNulty
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA.
| |
Collapse
|
37
|
Roman-Pepine D, Serban AM, Capras RD, Cismaru CM, Filip AG. A Comprehensive Review: Unraveling the Role of Inflammation in the Etiology of Heart Failure. Heart Fail Rev 2025:10.1007/s10741-025-10519-w. [PMID: 40360833 DOI: 10.1007/s10741-025-10519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) remains a leading cause of morbidity and mortality worldwide, with inflammation playing a pivotal role in its pathogenesis. This comprehensive review aims to elucidate the intricate mechanisms by which inflammation contributes to the development and progression of HF. The review synthesizes current research on the involvement of both innate and adaptive immune responses in HF, highlighting the roles of cytokines, chemokines, and other inflammatory mediators. Recent studies have demonstrated that chronic inflammation, driven by factors such as oxidative stress, neurohormonal activation, and metabolic disturbances, leads to adverse cardiac remodeling and impaired myocardial function. The review explores how systemic inflammation, characterized by elevated levels of inflammatory biomarkers like C-reactive protein (CRP) and interleukin-6 (IL-6), correlates with HF severity and outcomes. Additionally, it discusses the impact of comorbid conditions such as diabetes, obesity, and hypertension on inflammatory pathways and HF risk. The review also delves into the therapeutic implications of targeting inflammation in HF. Despite mixed results from early clinical trials, emerging evidence suggests that anti-inflammatory therapies offer benefits in specific HF phenotypes. The potential of novel therapeutic strategies, including the use of biologics and small molecule inhibitors, is examined in the context of their ability to modulate inflammatory responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Diana Roman-Pepine
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania.
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania.
| | - Adela Mihaela Serban
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania
- 5 Th Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Roxana-Denisa Capras
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Cristina Mihaela Cismaru
- Department of Infectious Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Adriana Gabriela Filip
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| |
Collapse
|
38
|
Poredoš P, Mangaroska AS, Poredoš P. Atherosclerotic plaque stabilization and regression. VASA 2025. [PMID: 40356553 DOI: 10.1024/0301-1526/a001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Atherosclerotic plaques represent a typical deterioration of arterial wall in atherosclerotic process and are the source of cardiovascular events. Plaque progression and composition represent a major risk for cardiovascular events. Therefore, recently many studies have assessed changes in plaque characteristics and their response to various treatment modalities. In the last two decades, improvement in plaque imaging modalities that can assess plaque volumes and composition enable to follow plaque characteristics in a non-invasive way. Clinical trials utilizing arterial imaging modalities have shown that reducing LDL cholesterol to low levels can reduce atherosclerotic plaque burden and favourably modify plaque composition. These outcomes have been achieved with statin therapy and newer lipid-lowering strategies such as protein convertase subtilisin/kexin type 9 inhibitors. Also, some anti-inflammatory drugs and other anti-atherosclerotic medications can lead to significant reduction in plaque burden. However, the data assessing association of plaque regression to reduction of cardiovascular events are limited. Therefore, the aim of this narrative review is to elucidate the possibilities and the role of plaque assessment and if it might offer the potential to guide personalized management of patients at risk for cardiovascular events in the future.
Collapse
Affiliation(s)
- Pavel Poredoš
- Department of Vascular Disease, University Medical Centre Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Peter Poredoš
- Faculty of Medicine, University of Ljubljana, Slovenia
- Department of Anaesthesiology and Surgical Intensive Care, University Medical Centre Ljubljana, Slovenia
| |
Collapse
|
39
|
Kraler S, Mueller C, Libby P, Bhatt DL. Acute coronary syndromes: mechanisms, challenges, and new opportunities. Eur Heart J 2025:ehaf289. [PMID: 40358623 DOI: 10.1093/eurheartj/ehaf289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/03/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Despite advances in research and patient management, atherosclerosis and its dreaded acute and chronic sequelae continue to account for one out of three deaths globally. The vast majority of acute coronary syndromes (ACS) arise from either plaque rupture or erosion, but other mechanisms, including calcific nodules, embolism, spontaneous coronary artery dissection, coronary spasm, and microvascular dysfunction, can also cause ACS. This ACS heterogeneity necessitates a paradigm shift in its management that extends beyond the binary interpretation of electrocardiographic and biomarker data. Indeed, given the evolution in the global risk factor profile, the increasing importance of previously underappreciated mechanisms, the evolving appreciation of sex-specific disease characteristics, and the advent of rapidly evolving technologies, a precision medicine approach is warranted. This review provides an update of the mechanisms of ACS, delineates the role of previously underappreciated contributors, discusses sex-specific differences, and explores novel tools for contemporary and personalized management of patients with ACS. Beyond mechanistic insights, it examines evolving imaging techniques, biomarkers, and regression- and machine learning-based approaches for the diagnosis (e.g. CoDE-ACS, MI3) and prognosis (e.g. PRAISE, GRACE, SEX-SHOCK scores) of ACS, along with their implications for future ACS management. A more individualized approach to patients with ACS is advocated, emphasizing the need for innovative studies on emerging technologies, including artificial intelligence, which may collectively facilitate clinical decision-making within a more mechanistic framework, thereby personalizing patient care and potentially improving long-term outcomes.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave Levy Place, Box 1030, New York, NY 10029, USA
| |
Collapse
|
40
|
von Ehr A, Steenbuck ID, Häfele C, Remmersmann F, Vico TA, Ehlert C, Lindner D, Wolf D, Tholen S, Schilling O, Czerny M, Westermann D, Hilgendorf I. Experimental evidence on colchicine's mode of action in human carotid artery plaques. Atherosclerosis 2025; 406:119239. [PMID: 40381496 DOI: 10.1016/j.atherosclerosis.2025.119239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND AIMS Atherosclerosis, driven by inflammation, is a leading cause of cardiovascular events. Recent clinical trials have highlighted the therapeutic potential of anti-inflammatory treatments. Consequently, colchicine is being recommended for secondary prevention in current guidelines, although the drug's mechanistic actions are not fully understood. METHODS To this end, we conducted a multiomic investigation of colchicine's effect on human carotid plaques. Sections from endarterectomy specimens were exposed to colchicine at concentrations of 2 ng/ml and 10 ng/ml ex vivo for 24 h and compared to untreated segments of the same plaque. Gene expression changes were analyzed by bulk RNA sequencing, and plaque secretomes underwent mass spectrometry for proteomic analysis. In situ cell proliferation was assessed by histology. RESULTS Our data indicate, that colchicine suppresses neutrophil and platelet degranulation and activation, collagen degradation and atheromatous plaque macrophage proliferation in a dose-dependent manner in human plaques, while stimulating myofibroblast activation. Unexpectedly, interleukine (IL)-1beta release from colchicine treated plaques was not reduced. These results indicate that the inflammasome may not be the predominant target of low-dose colchicine in human carotid artery plaques. CONCLUSION Our study identifies multifactorial pathways through which colchicine, the first cardiovascular guideline-recommended anti-inflammatory drug, predominantly acts on human atherosclerotic lesions beyond the inflammasome. Targeting neutrophil and platelet degranulation, collagen degradation and macrophage proliferation, selectively, may provide substantial therapeutic benefit in atherosclerotic cardiovascular disease without colchicine's undesired side effects.
Collapse
Affiliation(s)
- Alexander von Ehr
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Ines Derya Steenbuck
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Häfele
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Remmersmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamara A Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolin Ehlert
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
41
|
Zannas AS. Cell Type-Level Epigenetics at the Frontier of Atherosclerosis Research. Circulation 2025; 151:1409-1411. [PMID: 40354452 PMCID: PMC12074577 DOI: 10.1161/circulationaha.125.074275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Anthony S Zannas
- Departments of Psychiatry and Genetics, University of North Carolina at Chapel Hill
| |
Collapse
|
42
|
Cheng L, Zhang Y. Cell death, IL-1 cytokines, and tumor progression. Cancer Cell 2025; 43:817-819. [PMID: 40250443 DOI: 10.1016/j.ccell.2025.03.030] [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: 03/18/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/20/2025]
Abstract
Tumor cell death shapes tumorigenesis and antitumor immunity in complex ways. Recently, Hänggi et al. revealed that necrotic-like death releases interleukin-1α (IL-1α), driving myeloid-mediated immunosuppression. Lamorte et al. demonstrated that medullary sinus macrophage (MSM) efferocytosis of apoptotic tumor cells activates the IL-33-regulatory T cell (Treg) axis, accelerating tumor growth. Blocking these pathways enhances the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Long Cheng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Ying Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
43
|
Ghafoury R, Malek M, Ismail-Beigi F, Khamseh ME. Role of Residual Inflammation as a Risk Factor Across Cardiovascular-Kidney-Metabolic (CKM) Syndrome: Unpacking the Burden in People with Type 2 Diabetes. Diabetes Ther 2025:10.1007/s13300-025-01743-6. [PMID: 40343683 DOI: 10.1007/s13300-025-01743-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a global health crisis, with cardiovascular disease (CVD) accounting for 75% of mortality in this population. Despite advances in managing traditional risk factors, such as low-density lipoprotein cholesterol (LDL) cholesterol reduction (IMPROVE-IT, FOURIER), antithrombotic therapies (PEGASUS, COMPASS), and triglyceride-lowering agents (REDUCE-IT), a substantial residual cardiovascular risk persists, driven in part by chronic low-grade systemic inflammation. Chronic low-grade inflammation is a central driver of cardiovascular-kidney-metabolic (CKM) syndrome in T2DM, perpetuating residual cardiovascular risk despite optimal management of traditional risk factors. This narrative review synthesizes evidence on how inflammation accelerates coronary heart disease (CHD), heart failure (HF), stroke, diabetic kidney disease (DKD), and peripheral artery disease (PAD). We evaluate the anti-inflammatory mechanisms of current therapies such as statins, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and glucagon-like peptide 1 (GLP-1) receptor agonists, as well as emerging agents like colchicine and interleukin (IL)-1β/IL-6 inhibitors, emphasizing their differential efficacy across CKM traits. By integrating pathophysiological insights with clinical trial data, we propose biomarker-guided strategies to target inflammation as a modifiable risk factor, offering a roadmap to bridge the gap in diabetes-related cardiovascular care.
Collapse
Affiliation(s)
- Roya Ghafoury
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | - Mojtaba Malek
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | | | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran.
| |
Collapse
|
44
|
Pasut A, Lama E, Van Craenenbroeck AH, Kroon J, Carmeliet P. Endothelial cell metabolism in cardiovascular physiology and disease. Nat Rev Cardiol 2025:10.1038/s41569-025-01162-x. [PMID: 40346347 DOI: 10.1038/s41569-025-01162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 05/11/2025]
Abstract
Endothelial cells are multifunctional cells that form the inner layer of blood vessels and have a crucial role in vasoreactivity, angiogenesis, immunomodulation, nutrient uptake and coagulation. Endothelial cells have unique metabolism and are metabolically heterogeneous. The microenvironment and metabolism of endothelial cells contribute to endothelial cell heterogeneity and metabolic specialization. Endothelial cell dysfunction is an early event in the development of several cardiovascular diseases and has been shown, at least to some extent, to be driven by metabolic changes preceding the manifestation of clinical symptoms. Diabetes mellitus, hypertension, obesity and chronic kidney disease are all risk factors for cardiovascular disease. Changes in endothelial cell metabolism induced by these cardiometabolic stressors accelerate the accumulation of dysfunctional endothelial cells in tissues and the development of cardiovascular disease. In this Review, we discuss the diversity of metabolic programmes that control endothelial cell function in the cardiovascular system and how these metabolic programmes are perturbed in different cardiovascular diseases in a disease-specific manner. Finally, we discuss the potential and challenges of targeting endothelial cell metabolism for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Alessandra Pasut
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Eleonora Lama
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Jeffrey Kroon
- Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, The Netherlands.
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium.
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
45
|
Taylor EB, Hall JE, Mouton AJ. Current anti-inflammatory strategies for treatment of heart failure: From innate to adaptive immunity. Pharmacol Res 2025; 216:107761. [PMID: 40348101 DOI: 10.1016/j.phrs.2025.107761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in developed countries worldwide, often manifesting in the form of heart failure (HF). Recent successful clinical outcomes of anti-inflammatory therapies in HF patients have greatly boosted interest in basic and translational research on the role of inflammation in development of HF. In this review, we discuss recent and ongoing therapies targeting inflammation in CVD/HF, including broad-spectrum anti-inflammatory drugs, supplements, and biologicals such as canakinumab and anakinra. We also discuss the growing body of literature supporting off-target/anti-inflammatory actions of mainline CVD/HF agents, including guideline-directed medical therapy (GDMT) drugs that target the neurohormonal axis, and statins. We discuss therapeutics that target autoimmune mechanisms, and their implications for treating patients with autoimmune diseases with HF or at-risk of developing HF. We also discuss recent evidence for vaccines in modulating the immune response during HF. We conclude that despite the wealth of knowledge gained in the past decade, the therapeutic efficacy of anti-inflammatory therapy is driven by many biological and logistical factors that vary from patient to patient. Furthermore, more studies are needed to understand the adaptive/autoimmune component of HF, particularly in women and patients with pre-existing autoimmune disease. As the number of patients with HF patients who suffer from obesity, diabetes, or autoimmune disease continues to grow, our understanding of inflammation must continue to evolve to reflect these underlying co-morbidities.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - John E Hall
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Alan J Mouton
- Department of Physiology and Biophysics and Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| |
Collapse
|
46
|
Yan Y, Zhang T, He X, Du T, Dai G, Xu X, Chen Z, Wu J, Zhou H, Peng Y, Li Y, Liu C, Liao X, Dong Y, Ou JS, Huang ZP. A cardiac fibroblast-enriched micropeptide regulates inflammation in ischemia/reperfusion injury. JCI Insight 2025; 10:e187848. [PMID: 40111415 DOI: 10.1172/jci.insight.187848] [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: 10/08/2024] [Accepted: 03/19/2025] [Indexed: 03/22/2025] Open
Abstract
Inflammation is a critical pathological process in myocardial infarction. Although immunosuppressive therapies can mitigate inflammatory responses and improve outcomes in myocardial infarction, they also increase the risk of infections. Identifying novel regulators of local cardiac inflammation could provide safer therapeutic targets for myocardial ischemia/reperfusion injury. In this study, we identified a previously uncharacterized micropeptide, which we named Inflammation Associated MicroPeptide (IAMP). IAMP is predominantly expressed in cardiac fibroblasts, and its expression is closely associated with cardiac inflammation. Downregulation of IAMP promotes, whereas its overexpression prevents, the transformation of cardiac fibroblasts into a more inflammatory phenotype under stressed/stimulated conditions, as evidenced by changes in the expression and secretion of proinflammatory cytokines. Consequently, loss of IAMP function leads to uncontrolled inflammation and worsens cardiac injury following ischemia/reperfusion surgery. Mechanistically, IAMP promotes the degradation of HIF-1α by interacting with its stabilizing partner HSP90 and, thus, suppresses the transcription of proinflammatory genes downstream of HIF-1α. This study underscores the significance of fibroblast-mediated inflammation in cardiac ischemia/reperfusion injury and highlights the therapeutic potential of targeting micropeptides for myocardial infarction.
Collapse
Affiliation(s)
- Youchen Yan
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Tingting Zhang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Xin He
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Tailai Du
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Gang Dai
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Xingfeng Xu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Zhuohui Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Jialing Wu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Huimin Zhou
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yazhi Peng
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Chen Liu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Jing-Song Ou
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
47
|
Kim S, Nam HS, Kang DO, Han J, Kim H, Song JW, Park EJ, Kim RH, Kim HJ, Kim JH, Lee S, Kim YS, Park P, Baik MJ, Yoo H, Kim JW. Intracoronary Structural-Molecular Imaging for Multitargeted Characterization of High-Risk Plaque: First-in-Human OCT-FLIm. JAMA Cardiol 2025:2833590. [PMID: 40332864 DOI: 10.1001/jamacardio.2025.0928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Importance Fluorescence lifetime imaging (FLIm) is a molecular imaging technique used to visualize the biochemical composition of atherosclerosis. Novel dual-modal imaging using optical coherence tomography (OCT)-FLIm has the potential to provide both microstructural and biocompositional information on coronary plaques; however, it needs validation for clinical application. Objective To investigate the clinical feasibility and safety of OCT-FLIm for characterizing plaque compositions in patients with coronary artery disease (CAD) undergoing revascularization therapy. Design, Setting, and Participants A prospective, open-label, single-center diagnostic feasibility study involving 40 patients with significant CAD requiring coronary revascularization. This first-in-human clinical study of the novel intracoronary OCT-FLIm imaging was conducted between February and August 2022. The analyses were performed from August 2022 to July 2023. Interventions An OCT-FLIm system with 2.6-F catheters was constructed. All patients underwent OCT-FLIm for target/culprit and nontarget/nonculprit lesions during coronary revascularization. Intravascular ultrasound imaging was performed for comparison. Main Outcomes and Measures The primary outcome was to assess the FLIm-derived molecular readouts of prespecified plaque compositions. The secondary outcome was the feasibility of OCT-FLIm in determining target/culprit plaque compositions across different subsets of atherosclerotic disease activity: (1) acute coronary syndrome (ACS) vs chronic stable angina (CSA) and (2) angiographic rapid disease progression vs nonprogressive controls. Results We prospectively enrolled 40 patients (mean [SD] age, 63.1 [8.1] years; 32 men [80.0%]), of whom 20 presented with ACS and 20 with CSA. OCT provided the structural features of plaques, and FLIm characterized the molecular signatures of atheroma compositions, including macrophages, healed plaques, superficial calcification, and fibrosis, in a reproducible manner. Fluorescence lifetime (FL) values of the plaque compositions correlated with findings from prior autopsy studies. Plaque inflammation was significantly greater in patients with ACS than those with CSA. The mean (SD) of inflammation-FL was 7.59 (0.96) nanoseconds for patients with ACS vs 6.46 (0.87) nanoseconds for patients with CSA (P < .001). The healed plaque phenotype was more prominently distributed in the segments of rapid disease progression than in nonprogressive controls. The mean (SD) healed plaque-FL was 5.31 (0.20) nanoseconds for the rapidly progressive lesions vs 4.81 (0.30) nanoseconds for the rapidly nonprogressive lesions (P < .001). All patients underwent OCT-FLIm safely without adverse clinical events. Conclusions and Relevance This diagnostic feasibility study found that an OCT-FLIm structural-molecular intracoronary imaging is clinically feasible and safe for the comprehensive characterization of human atheromas, supporting its potential role in the diagnosis and biological understanding of high-risk plaques.
Collapse
Affiliation(s)
- Sunwon Kim
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
- Department of Cardiology, Korea University Ansan Hospital, Ansan, South Korea
| | - Hyeong Soo Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Dong Oh Kang
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Jeongmoo Han
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hyokee Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Seoul, South Korea
| | - Joon Woo Song
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Eun Jin Park
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Ryeong Hyun Kim
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Jin Hyuk Kim
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Sunki Lee
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Young Su Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Seoul, South Korea
| | - Pyoungjae Park
- Division of Transplantation and Vascular Surgery, Korea University Guro Hospital, Seoul, South Korea
| | - Man-Jong Baik
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Seoul, South Korea
| | - Hongki Yoo
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jin Won Kim
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| |
Collapse
|
48
|
Romeo S, Vidal-Puig A, Husain M, Ahima R, Arca M, Bhatt DL, Diehl AM, Fontana L, Foo R, Frühbeck G, Kozlitina J, Lonn E, Pattou F, Plat J, Quaggin SE, Ridker PM, Rydén M, Segata N, Tuttle KR, Verma S, Roeters van Lennep J, Benn M, Binder CJ, Jamialahmadi O, Perkins R, Catapano AL, Tokgözoğlu L, Ray KK. Clinical staging to guide management of metabolic disorders and their sequelae: a European Atherosclerosis Society consensus statement. Eur Heart J 2025:ehaf314. [PMID: 40331343 DOI: 10.1093/eurheartj/ehaf314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Obesity rates have surged since 1990 worldwide. This rise is paralleled by increases in pathological processes affecting organs such as the heart, liver, and kidneys, here termed systemic metabolic disorders (SMDs). For clinical management of SMD, the European Atherosclerosis Society proposes a pathophysiology-based system comprising three stages: Stage 1, where metabolic abnormalities such as dysfunctional adiposity and dyslipidaemia occur without detectable organ damage; Stage 2, which involves early organ damage manifested as Type 2 diabetes, asymptomatic diastolic dysfunction, metabolic-associated steatohepatitis (MASH), and chronic kidney disease (CKD); and Stage 3, characterized by more advanced organ damage affecting multiple organs. Various forms of high-risk obesity, driven by maintained positive energy balance, are the most common cause of SMD, leading to ectopic lipid accumulation and insulin resistance. This progression affects various organs, promoting comorbidities such as hypertension and atherogenic dyslipidaemia. Genetic factors influence SMD susceptibility, and ethnic disparities in SMD are attributable to genetic and socioeconomic factors. Key SMD features include insulin resistance, inflammation, pre-diabetes, Type 2 diabetes, MASH, hypertension, CKD, atherogenic dyslipidaemia, and heart failure. Management strategies involve lifestyle changes, pharmacotherapy, and metabolic surgery in severe cases, with emerging treatments focusing on genetic approaches. The staging system provides a structured approach to understanding and addressing the multi-faceted nature of SMD, which is crucial for improving health outcomes. Categorization of SMD abnormalities by presence and progression is aimed to improve awareness of a multi-system trait and encourage a tailored and global approach to treatment, ultimately aiming to reduce the burden of obesity-related comorbidities.
Collapse
Affiliation(s)
- Stefano Romeo
- Department of Medicine, H7 Medicin, Huddinge, H7 Endokrinologi och Diabetes Romeo, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital Huddinge, 141 57 Huddinge, Stockholm, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Viale Europa, 88100 Catanzaro, Italy
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, C/ d'Eduardo Primo Yufera, 3, 46012 Valencia, Spain
- Cambridge University Nanjing Centre of Technology and Innovation, No. 23, Rongyue Road, Jiangbei New Area, Nanjing, Jiangsu, China
| | - Mansoor Husain
- Ted Rogers Centre for Heart Research, Department of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, Canada M5G 1M1
| | - Rexford Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Unit of Internal Medicine and Metabolic Diseases, Hospital Policlinico Umberto I, Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC, USA
| | - Luigi Fontana
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Metabolic Research Laboratory, CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eva Lonn
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Francois Pattou
- Department of Endocrine and Metabolic Surgery, CHU Lille, University of Lille, Inserm, Institut Pasteur Lille, Lille, France
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Susan E Quaggin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Katherine R Tuttle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, WA, USA
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Centre of Diagnostic Investigation, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Alberico L Catapano
- Center for the Study of Atherosclerosis, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK
| |
Collapse
|
49
|
Tejedor-Santamaria L, Marquez-Exposito L, Villacampa A, Marchant V, Battaglia-Vieni A, Rayego-Mateos S, Rodrigues-Diez RR, Santos FM, Valentijn FA, Knoppert SN, Broekhuizen R, Ruiz-Torres MP, Goldschmeding R, Ortiz A, Peiró C, Nguyen TQ, Ramos AM, Ruiz-Ortega M. CCN2 Activates Cellular Senescence Leading to Kidney Fibrosis in Folic Acid-Induced Experimental Nephropathy. Int J Mol Sci 2025; 26:4401. [PMID: 40362638 PMCID: PMC12072722 DOI: 10.3390/ijms26094401] [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] [Received: 03/20/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Cellular communication network factor 2 (CCN2, also known as CTGF) is a complex protein that regulates numerous cellular functions. This biomolecule exhibits dual functions, depending on the context, and can act as a matricellular protein or as a growth factor. CCN2 is an established marker of fibrosis and a well-known mediator of kidney damage, involved in the regulation of inflammation, extracellular matrix remodeling, cell death, and activation of tubular epithelial cell (TECs) senescence. In response to kidney damage, cellular senescence mechanisms are activated, linked to regeneration failure and progression to fibrosis. Our preclinical studies using a total conditional CCN2 knockout mouse demonstrate that CCN2 plays a significant role in the development of a senescence phenotype after exposure to a nephrotoxic agent. CCN2 induces cell growth arrest in TECs, both in the early phase and in the chronic phase of folic acid nephropathy (FAN), associated with cell-death/necroinflammation and fibrosis, respectively. Renal CCN2 overexpression was found to be linked to excessive collagen accumulation in tubulointerstitial areas, microvascular rarefaction, and a decline in renal function, which were observed three weeks following the initial injury. All these findings were markedly diminished in conditional CCN2 knockout mice. In the FAN model, injured senescent TECs are associated with microvascular rarefaction, and both were modulated by CCN2. In primary cultured endothelial cells, as previously described in TECs, CCN2 directly induced senescence. The findings collectively demonstrate the complexity of CCN2, highlight the pivotal role of cellular senescence as an important mechanism in renal injury, and underscore the critical function of this biomolecule in kidney damage progression.
Collapse
Affiliation(s)
- Lucia Tejedor-Santamaria
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Laura Marquez-Exposito
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Alicia Villacampa
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.V.); (C.P.)
- Vascular Pharmacology and Metabolism (FARMAVASM) Group, IdiPAZ, 28029 Madrid, Spain
| | - Vanessa Marchant
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Antonio Battaglia-Vieni
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
| | - Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Raul R. Rodrigues-Diez
- Department of Cell Biology, School of Medicine, Complutense University, 28040 Madrid, Spain;
| | - Fatima Milhano Santos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Floris A. Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Sebastian N. Knoppert
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | | | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Alberto Ortiz
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28049 Madrid, Spain
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.V.); (C.P.)
- Vascular Pharmacology and Metabolism (FARMAVASM) Group, IdiPAZ, 28029 Madrid, Spain
| | - Tri Q. Nguyen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Adrián M. Ramos
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28049 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| |
Collapse
|
50
|
Dong Z, Jin Y, Shen Y, Huang J, Tan J, Feng Q, Gong Z, Zhu S, Chen H, Yu F, Li W, Jia Y, Kong W, Fu Y. Methyltransferase-like 3-catalysed N6-methyladenosine methylation facilitates the contribution of vascular smooth muscle cells to atherosclerosis. Cardiovasc Res 2025; 121:568-584. [PMID: 39977233 DOI: 10.1093/cvr/cvaf029] [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: 04/20/2024] [Revised: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 02/22/2025] Open
Abstract
AIMS Vascular smooth muscle cells (VSMCs) are involved in the aetiology of atherosclerosis, but whether methyltransferase-like 3 (METTL3)-catalysed N6-methyladenosine (m6A) modulates the contribution of VSMCs to atherosclerosis remains elusive. METHODS AND RESULTS We generated tamoxifen-inducible VSMC-specific METTL3 knockout mice with VSMC lineage tracing and found that VSMC-specific METTL3 deficiency substantially attenuated atherosclerosis and reduced the proportion of VSMCs in plaques, due to the inhibition of VSMC atheroprone phenotype as characterized by macrophage-like and inflammatory features as well as high migratory and proliferative capacity. m6A-methylated RNA immunoprecipitation sequencing (MeRIP-Seq) combined with polysome profiling analysis mechanistically displayed METTL3-catalysed m6A methylation of myocardin-related transcription factor A (MRTFA) mRNA and further enhanced YTH N6-methyladenosine RNA-binding protein F3 (YTHDF3)-dependent MRTFA mRNA translation. Conversely, adenovirus or adeno-associated virus-mediated VSMC-specific MRTFA overexpression abolished METTL3 deficiency-mediated alleviation of VSMC atheroprone phenotypic switching and atherosclerotic progression both in vitro and in vivo. CONCLUSION METTL3 facilitated the contribution of VSMCs to atherosclerosis through the m6A-YTHDF3-dependent MRTFA mRNA translation enhancement.
Collapse
MESH Headings
- Animals
- Methyltransferases/genetics
- Methyltransferases/metabolism
- Methyltransferases/deficiency
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Atherosclerosis/enzymology
- Atherosclerosis/prevention & control
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Methylation
- Mice, Knockout
- Disease Models, Animal
- Cells, Cultured
- Plaque, Atherosclerotic
- Phenotype
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Mice, Inbred C57BL
- Aortic Diseases/pathology
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Cell Proliferation
- Male
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Cell Movement
- Signal Transduction
- Humans
- Mice
Collapse
Affiliation(s)
- Zhigang Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yourong Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- Department of Pathophysiology, School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Jiaai Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- Hwamei College of Life and Health Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Shirong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Huiyue Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wei Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- Department of Vascular Surgery, Peking University People's Hospital, Peking University, Beijing 100191, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| |
Collapse
|