1
|
Guo B, Shi S, Xiong J, Guo Y, Wang B, Bai L, Qiu Y, Li S, Gao D, Dong Z, Tu Y. Identification of potential biomarkers in cardiovascular calcification based on bioinformatics combined with single-cell RNA-seq and multiple machine learning analysis. Cell Signal 2025; 131:111705. [PMID: 40024421 DOI: 10.1016/j.cellsig.2025.111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The molecular and genetic mechanisms underlying vascular calcification remain unclear. This study aimed to determine the differences in calcification marker-related gene expression in macrophages. METHODS The expression profiling datasets GSE104140 and GSE235995 were analysed to identify differentially expressed genes (DEGs) between fibroatheroma with calcification and diffuse intimal thickening. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, Weighted Gene Co-expression Network Analysis (WGCNA), and Gene Set Enrichment Analysis (GSEA) were performed to assess functional characteristics. Hub genes were identified through a protein-protein interaction (PPI) network and machine learning approaches. Single-cell RNA sequencing data (GSE159677) validated the expression of calcification-related genes in macrophages, while Mendelian randomization analysis explored their potential causal relationship with coronary calcification. Further validation was conducted using enzyme-linked immunosorbent assay (ELISA) on coronary calcification samples and immunohistochemistry in ApoE-/- mice. Intravascular ultrasound was performed to assess coronary calcification severity. RESULTS AND CONCLUSIONS Two key biomarkers, ITGAX and MYD88, were identified as diagnostic indicators of cardiovascular calcification. Both biomarkers were significantly upregulated in calcified samples and were strongly associated with immune processes. Single-cell RNA sequencing confirmed their high expression in multiple immune cell types. Additionally, molecular docking analysis revealed that retinoic acid interacted with both biomarkers, suggesting potential therapeutic relevance. Immunohistochemical and ELISA analyses further validated their elevated expression in calcified samples. These findings provide novel insights into the molecular mechanisms of vascular calcification and highlight potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Bingchen Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| | - Si Shi
- Harbin Medical University, Harbin, China; Department of Respirology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Jie Xiong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yutong Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Bo Wang
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Liyan Bai
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yi Qiu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Shucheng Li
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Dianyu Gao
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Zengxiang Dong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yingfeng Tu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China; Department of Cardiology, The Shanxi Provincial People's Hospital, Taiyuan 030000, China.
| |
Collapse
|
2
|
Lau C, Primus CP, Shabbir A, Chhetri I, Ono M, Masucci M, Bin Noorany Aubdool MA, Amarin J, Hamers AJ, Khan Z, Kumar NA, Montalvo Moreira SA, Nuredini G, Osman M, Whitear C, Godec T, Kapil V, Massimo G, Khambata RS, Rathod KS, Ahluwalia A. Accelerating inflammatory resolution in humans to improve endothelial function and vascular health: Targeting the non-canonical pathway for NO. Redox Biol 2025; 82:103592. [PMID: 40209616 PMCID: PMC12005330 DOI: 10.1016/j.redox.2025.103592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chronic cardiovascular diseases (CVD) are characterised by low-grade systemic inflammation in part due to reduced nitric oxide (NO) bioavailability associated with endothelial dysfunction. Bioavailability of NO can be enhanced by activation of the non-canonical pathway, through increased dietary inorganic nitrate consumption with the potential to attenuate inflammation. METHODS We sought to determine whether dietary inorganic nitrate influences the inflammatory response in models of localised (cantharidin-induced blisters) and systemic inflammation (typhoid vaccine), in healthy male volunteers and conducted two clinical trials; Blister-NITRATE and Typhoid-NITRATE respectively. RESULTS We show that dietary nitrate attenuates endothelial dysfunction following typhoid vaccine administration and accelerates resolution of cantharidin-induced blisters. Both phenomena were associated with an increased level of pro-resolving mediators consequent to a reduction in the expression and activity of pro-inflammatory monocytes. Moreover, we show that leukocytes of the monocyte lineage express the nitrite reductase XOR, that may drive localised nitrite reduction to elevate NO (and cGMP) to drive the protective phenotype. CONCLUSIONS Inorganic nitrate improves endothelial function in the setting of systemic inflammation. Whilst the immediate inflammatory response appeared unaffected by inorganic nitrate treatment, during the resolution phase of the acute inflammatory response lower levels of pro-inflammatory classical inflammatory and intermediate monocytes and attenuated levels of inflammatory cytokines and chemokines were evident. We propose that this reflects a pro-resolution phenotype that may be of potential therapeutic benefit in patients with established CVD. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; unique identifiers NCT02715635, NCT03183830.
Collapse
Affiliation(s)
- Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christopher P Primus
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Masucci
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad Aadil Bin Noorany Aubdool
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Amarin
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jp Hamers
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zara Khan
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nitin Ajit Kumar
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Gani Nuredini
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Miski Osman
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Charlotte Whitear
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tom Godec
- Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vikas Kapil
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Cardiology, Barts Heart Centre, 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
3
|
Bay B, Tanner R, Gao M, Oliva A, Sartori S, Vogel B, Gitto M, Smith KF, Di Muro FM, Hooda A, Sweeny J, Krishnamoorthy P, Moreno P, Krishnan P, Dangas G, Kini A, Sharma SK, Mehran R. Residual cholesterol and inflammatory risk in statin-treated patients undergoing percutaneous coronary intervention†. Eur Heart J 2025:ehaf196. [PMID: 40208236 DOI: 10.1093/eurheartj/ehaf196] [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: 07/21/2024] [Revised: 12/03/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND AIMS Elevated LDL-cholesterol levels and inflammation, as assessed by high-sensitivity C-reactive protein, correlate with cardiovascular risk. However, data on the relative impact of residual LDL-cholesterol and inflammatory risk among statin-treated patients undergoing percutaneous coronary intervention (PCI) is lacking. Hence, this study aimed to investigate the impact of residual cholesterol/inflammatory risk in patients on statin therapy undergoing PCI. METHODS From 2012 to 2022, patients at a tertiary centre undergoing PCI were analysed. Patients were stratified according to LDL-cholesterol (≥70 vs <70 mg/dL) and high-sensitivity C-reactive protein (≥2 vs <2 mg/L) levels: no residual cholesterol or inflammatory risk, residual cholesterol risk, residual inflammatory risk, and combined residual cholesterol and inflammatory risk. Patients presenting with acute myocardial infarction, cancer, no statin treatment at admission, or high-sensitivity C-reactive protein levels >10 mg/L were excluded. The primary endpoint was major adverse cardiovascular events (MACEs), defined as the composite of all-cause mortality, spontaneous myocardial infarction, and stroke 1 year after the index PCI. RESULTS A total of 15 494 patients were included. After 1-year follow-up, individuals with isolated residual inflammatory risk had the highest MACE rate (5.1%), followed by patients with combined cholesterol and inflammatory risk, no residual risk, and isolated residual cholesterol risk. After multivariable Cox regression analysis, patients with residual inflammatory risk had a 1.8-fold higher risk for MACE (adjusted hazard ratio: 1.78, 95% confidence interval 1.36-2.33, P < .001) compared with those with no residual cholesterol or inflammatory risk. This was similar in patients with combined residual cholesterol and inflammatory risk (adjusted hazard ratio: 1.56, 95% confidence interval 1.19-2.04, P = 0.001). Of note, no independent association of isolated residual cholesterol risk (adjusted hazard ratio: 1.01, 95% confidence interval .76-1.35, P-value = .920) with MACE was noted (P-trend across all groups <.001). CONCLUSIONS Among statin-treated patients undergoing PCI, residual inflammation but not cholesterol risk was associated with an increased risk of MACE during follow-up.
Collapse
Affiliation(s)
- Benjamin Bay
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard Tanner
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Cardiology, Cork University Hospital, Cork, Ireland
| | - Michael Gao
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Angelo Oliva
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Samantha Sartori
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Birgit Vogel
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Mauro Gitto
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Kenneth F Smith
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Francesca Maria Di Muro
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Structural Interventional Cardiology Division, Department of Cardiac Thoracic and Vascular Medicine, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Amit Hooda
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Joseph Sweeny
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Parasuram Krishnamoorthy
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Pedro Moreno
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Prakash Krishnan
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - George Dangas
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Annapoorna Kini
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Samin K Sharma
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| |
Collapse
|
4
|
Dastjerdi P, Mahalleh M, Shayesteh H, Najafi MS, Narimani-Javid R, Dashtkoohi M, Mofidi SA, Hosseini K, Tajdini M. Liver biomarkers as predictors of prognosis in heart failure with preserved ejection fraction: a systematic review and meta-analysis. BMC Cardiovasc Disord 2025; 25:244. [PMID: 40175926 PMCID: PMC11963275 DOI: 10.1186/s12872-025-04647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) accounts for 50% of heart failure cases, with increasing prevalence due to aging and risk factors such as hypertension and obesity. Liver dysfunction is common in HFpEF and may impact prognosis. This systematic review and meta-analysis aimed to evaluate the prognostic value of liver function markers (albumin, bilirubin, AST, ALT, ALP) in HFpEF patients. METHODS A systematic search of PubMed, Embase, Web of Science, and Scopus was conducted for studies assessing the association of liver markers with adverse outcomes in HFpEF. The primary outcome was a composite of heart failure-related hospitalization or death. Hazard ratios (HR) were pooled using a random-effects model, and heterogeneity was assessed using the I² statistic. RESULTS Twenty studies involving 30,623 patients were included. Serum albumin, the main marker of our study, was significantly associated with a reduced risk of adverse outcomes in a meta-analysis of 16 studies (HR 0.71, 95% CI: 0.61-0.83; I² = 87%). After excluding outliers, heterogeneity decreased (I² = 23%), and the association remained significant (HR 0.75, 95% CI: 0.69-0.82). Although no significant associations were found for AST, ALT, ALP, or bilirubin with adverse outcomes, the limited number of studies for these markers may have contributed to the lack of statistical significance. CONCLUSION Higher serum albumin levels predict better outcomes in HFpEF, while other liver function markers showed limited prognostic utility. Serum albumin may serve as a valuable marker for risk stratification in HFpEF.
Collapse
Affiliation(s)
- Parham Dastjerdi
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Mahalleh
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hedieh Shayesteh
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadeq Najafi
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Roozbeh Narimani-Javid
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadese Dashtkoohi
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mofidi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Hosseini
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cardiac Primary Prevention Research Center, Cardiovascular Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masih Tajdini
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Wang Y, Aivalioti E, Stamatelopoulos K, Zervas G, Mortensen MB, Zeller M, Liberale L, Di Vece D, Schweiger V, Camici GG, Lüscher TF, Kraler S. Machine learning in cardiovascular risk assessment: Towards a precision medicine approach. Eur J Clin Invest 2025; 55 Suppl 1:e70017. [PMID: 40191920 DOI: 10.1111/eci.70017] [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: 11/28/2024] [Accepted: 02/22/2025] [Indexed: 04/24/2025]
Abstract
Cardiovascular diseases remain the leading cause of global morbidity and mortality. Validated risk scores are the basis of guideline-recommended care, but most scores lack the capacity to integrate complex and multidimensional data. Limitations inherent to traditional risk prediction models and the growing burden of residual cardiovascular risk highlight the need for refined strategies that go beyond conventional paradigms. Artificial intelligence and machine learning (ML) provide unique opportunities to refine cardiovascular risk assessment and surveillance through the integration of diverse data types and sources, including clinical, electrocardiographic, imaging and multi-omics derived data. In fact, ML models, such as deep neural networks, can handle high-dimensional data through which phenotyping and cardiovascular risk assessment across diverse patient populations become much more precise, fostering a paradigm shift towards more personalized care. Here, we review the role of ML in advancing cardiovascular risk assessment and discuss its potential to identify novel therapeutic targets and to improve prevention strategies. We also discuss key challenges inherent to ML, such as data quality, standardized reporting, model transparency and validation, and discuss barriers in its clinical translation. We highlight the transformative potential of ML in precision cardiology and advocate for more personalized cardiovascular prevention strategies that go beyond previous notions.
Collapse
Affiliation(s)
- Yifan Wang
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Georgios Zervas
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Martin Bødtker Mortensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marianne Zeller
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
- Physiolopathologie et Epidémiologie Cérébro-Cardiovasculaire (PEC2), EA 7460, Univ Bourgogne, Dijon, France
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Davide Di Vece
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Victor Schweiger
- Deutsches Herzzentrum der Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals GSTT and Cardiovascular Academic Group, King's College, London, UK
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine and Cardiology, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
6
|
Güven B, Deniz MF, Geylan NA, Kültürsay B, Dönmez A, Bulat Z, Gül ÖB, Kaya M, Oktay V. A novel indicator of all-cause mortality in acute coronary syndrome: the CALLY index. Biomark Med 2025; 19:287-294. [PMID: 40125936 PMCID: PMC11980495 DOI: 10.1080/17520363.2025.2483159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
AIMS This study aimed to identify the prognostic significance of the C-reactive protein-albumin-lymphocyte (CALLY) index for predicting all-cause mortality in acute coronary syndrome (ACS) patients who have undergone primary percutaneous coronary intervention (pPCI) for revascularization. MATERIALS AND METHODS 505 patients who presented with ACS and underwent pPCI were retrospectively included in this single center study. CALLY index and other five prognostic scores were calculated. The median follow-up was 40 months. All-cause mortality was defined as the primary endpoint. RESULTS The median age of the patients was 59 years, 23.4% were female. The CALLY index was categorized into low (<0.7) and high (≥0.7). Age (p = 0.038), concomitant atrial fibrillation (p = 0.023), previous CABG (p = 0.001), ACE-I/ARB/ARNI use (p = 0.015), diuretic use (p = 0.021), and a low-CALLY index (p < 0.001) were identified as independent predictors of all-cause mortality in multivariate cox regression analysis. When compared to other prognostic scores according to AUC in ROC analysis, the CALLY index demonstrated the best ability to predict all-cause mortality. Additionally, patients with a high-CALLY index exhibited significantly better survival outcomes compared to those with a low-CALLY index (log-rank:p < 0.001). CONCLUSIONS CALLY index can be utilized as a novel prognostic score for predicting all-cause mortality in ACS patients who have undergone pPCI.
Collapse
Affiliation(s)
- Barış Güven
- Department of Cardiology, Idil State Hospital, Sirnak, Turkey
- Department of Cardiology, Sirnak State Hospital, Sirnak, Turkey
| | - Muhammed Furkan Deniz
- Department of Cardiology, Bagcilar Research and Education Hospital, Istanbul, Turkey
| | - Neziha Aybüke Geylan
- Department of Cardiology, Istanbul University Cerrahpasa Institute of Cardiology, Istanbul, Turkey
| | - Barkın Kültürsay
- Department of Cardiology, Tunceli State Hospital, Tunceli, Turkey
| | - Ayça Dönmez
- Department of Cardiology, Istanbul University Cerrahpasa Institute of Cardiology, Istanbul, Turkey
| | - Zübeyir Bulat
- Department of Cardiology, Sirnak State Hospital, Sirnak, Turkey
| | - Ömer Burak Gül
- Department of Cardiology, Istanbul University Cerrahpasa Institute of Cardiology, Istanbul, Turkey
| | - Melike Kaya
- Department of Cardiology, Istanbul University Cerrahpasa Institute of Cardiology, Istanbul, Turkey
| | - Veysel Oktay
- Department of Cardiology, Istanbul University Cerrahpasa Institute of Cardiology, Istanbul, Turkey
| |
Collapse
|
7
|
Jin M, Mamute M, Shapaermaimaiti H, Ji H, Cao Z, Luo S, Abudula M, Aigaixi A, Fu Z. Serum ferritin associated with atherogenic lipid profiles in a high-altitude living general population. PeerJ 2025; 13:e19104. [PMID: 40151449 PMCID: PMC11949108 DOI: 10.7717/peerj.19104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/12/2025] [Indexed: 03/29/2025] Open
Abstract
Background Serum ferritin (SF) levels are associated with metabolic syndrome and dyslipidemia. However, the association between SF and atherogenic lipid profiles in high-altitude living populations remains unclear. Methods In 2021, a cross-sectional study was conducted on adult Tajik individuals residing in Tashkurgan Tajik Autonomous County (average altitude 3,100 meters). Demographic information and anthropometric measurements were collected in local clinics. Fasting blood samples were analyzed using a Beckman AU-680 Automatic Biochemical analyzer at the biochemical laboratory of Fuwai Hospital. Univariate linear regression analyses were used to explore the association between SF and atherogenic lipid levels. Subgroup analysis was used based on gender and different high-sensitivity C-reactive protein (hs-CRP) and serum amyloid A (SAA) levels. The association between higher SF quartiles and different kinds of dyslipidemia were analyzed by logistic regression. Results There were 1,703 participants in total, among which 866 (50.9%) being men. The mean ages of male and female participants were similar (41.50 vs. 42.38 years; P = 0.224). SF levels were significantly correlated with total cholesterol (TC) (Beta = 0.225, P < 0.001), low-density lipoprotein cholesterol (LDL-C) (Beta = 0.197, P < 0.001), high-density lipoprotein cholesterol (HDL-C) (Beta = -0.218, P < 0.001), triglycerides (TG) (Beta = 0.332, P < 0.001), and small dense LDL-C (sdLDL-C) (Beta = 0.316, P < 0.001), with the exception of lipoprotein (a) (Lp(a)) (Beta = 0.018, P = 0.475). SF was significantly correlated with LDL-C and HDL-C in women, and correlated with TC, TG, and sdLDL-C levels in both men and women in different inflammatory conditions. Elevated SF levels was significantly correlated with high TC (OR: 1.413, 95% CI [1.010-1.978]), high TG (OR: 1.602, 95% CI [1.299-1.976]), and high sdLDL-C (OR: 1.631, 95% CI [1.370-1.942]) in men and high TC (OR: 1.461, 95% CI [1.061-2.014]), high LDL-C (OR: 2.104, 95% CI [1.481-2.990]), low HDL-C (OR: 1.447, 95% CI [1.195-1.752]), high TG (OR: 2.106, 95% CI [1.454-3.050]), and high sdLDL-C (OR: 2.000, 95% CI [1.589-2.516]) in women. After adjusting for potential confounders, elevated SF levels continue to be correlated with high TG in male (OR: 1.382, 95% CI [1.100-1.737]) and female (OR: 1.677, 95% CI [1.070-2.628]) participants. In both young and middle-aged subgroups, the associations between SF and TG, TC, HDL-C, LDL-C, and sdLDL-C were still significant. Conclusions SF was closely related to atherogenic lipid profiles, especially with regard to TG in high-altitude populations. This association cannot be attributed to its role as an inflammation marker.
Collapse
Affiliation(s)
- Menglong Jin
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mawusumu Mamute
- Department of Urology, First People’s Hospital of Kashgar District, Kashgar, Xinjiang, China
| | - Hebali Shapaermaimaiti
- Disease Control and Prevention Center of Tashkurgan Tajik Autonomous County, Tashkurgan, Xinjiang, China
| | - Hongyu Ji
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zichen Cao
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sifu Luo
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mayire Abudula
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Abuduhalike Aigaixi
- Health Commission of Tashkurgan Tajik Autonomous County, Tashkurgan, Xinjiang, China
| | - Zhenyan Fu
- Department of Cardiology, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
8
|
Luo Y, Yang L, Cheng X, Bai Y, Xiao Z. The association between blood count based inflammatory markers and the risk of atrial fibrillation heart failure and cardiovascular mortality. Sci Rep 2025; 15:10056. [PMID: 40128300 PMCID: PMC11933413 DOI: 10.1038/s41598-025-94507-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: 08/01/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
The prevalence and progression of cardiovascular disease (CVD) are significantly influenced by low-grade chronic inflammation. Our aim was to investigate the association of blood-count-based inflammatory markers with atrial fibrillation (AF), heart failure (HF), and cardiovascular mortality. We utilized prospective follow-up data from the UK Biobank, including 334,674 individuals (aged between 39 and 70 years) free of HF and AF at baseline. The exposures were blood-count-based inflammatory markers, including neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), systemic immune-inflammation index (SII), aggregate index of systemic inflammation (AISI), and systemic inflammatory response index (SIRI), which were computed using leukocyte counts (lymphocytes, monocytes, and neutrophils). The primary outcomes were the occurrence of AF, HF and cardiovascular-related deaths. Cox proportional hazards models were employed to determine hazard ratios (HRs) and their 95% confidence intervals (CIs) for assessing the longitudinal association between inflammatory markers and the development of cardiovascular events. Survival analysis was illustrated using Kaplan-Meier curves with the occurrence of AF, HF and CVD mortality as the endpoint. There were 16,994 incidences of AF and 7342 incidences of HF over a median follow-up of 10.4 years (range, 2.4-12.1). Additionally, there were 3434 deaths from CVD causes. A higher risk of cardiovascular mortality was significantly associated with NLR, MLR, SIRI, AISI, and SII in multivariable adjusted models (HR 1.44, 95% CI 1.30-1.58; HR 1.46, 95% CI 1.31-1.61; HR 1.57, 95% CI 1.41-1.75; HR 1.57, 95% CI 1.41-1.73; and HR 1.36, 95% CI 1.24-1.49, respectively). Furthermore, our analysis revealed positive associations between NLR, MLR, SIRI, and AISI and the risk for AF (HR 1.18, 95% CI 1.13-1.24; HR 1.19, 95% CI 1.14-1.25; HR 1.22, 95% CI 1.16-1.27; and HR 1.08, 95% CI 1.03-1.13, respectively), and HF (HR 1.42, 95% CI 1.33-1.52; HR 1.28, 95% CI 1.19-1.37; HR 1.45, 95% CI 1.35-1.56; and HR 1.32, 95% CI 1.23-1.41, respectively). Furthermore, the SII demonstrated a positive association with HF (HR 1.24, 95% CI 1.16-1.32). However, the association with AF was not statistically significant (HR 1.01, 95% CI 0.97-1.06). In summary, our research provides preliminary evidence that blood cell indices associated with the systemic inflammatory response, may serve as potential biomarkers for the risk of developing AF, HF and CVD mortality. Additionally, it is likely that a systemic inflammatory-immune response existed before the clinical diagnosis. Identifying high-risk populations based on the levels of inflammatory markers could help reduce the risk of death and prevent the onset of AF and HF. Further investigation is warranted to determine whether reducing inflammatory markers causally improves cardiovascular outcomes.
Collapse
Affiliation(s)
- Yi Luo
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Liu Yang
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - XunJie Cheng
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - YongPing Bai
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - ZhiLin Xiao
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China.
| |
Collapse
|
9
|
Di Muro FM, Vogel B, Sartori S, Bay B, Oliva A, Feng Y, Krishnan P, Sweeny J, Gitto M, Smith K, Moreno P, Nicolas J, Krishnamoorthy P, Leone PP, Bhatt DL, Dangas G, Kini A, Sharma SK, Mehran R. Prognostic impact of residual inflammatory and triglyceride risk in statin-treated patients with well-controlled LDL cholesterol and atherosclerotic cardiovascular disease. Eur J Prev Cardiol 2025:zwaf112. [PMID: 40112036 DOI: 10.1093/eurjpc/zwaf112] [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: 10/14/2024] [Revised: 12/10/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
AIMS Identifying alternative contributors to the residual risk of atherosclerotic cardiovascular disease (ASCVD) beyond LDL cholesterol (LDL-C) levels is crucial. We investigated the relative impact of triglycerides (TGs) and high-sensitivity C-reactive protein (hs-CRP) on outcomes in statin-treated patients with well-controlled LDL-C undergoing percutaneous coronary intervention (PCI) for established ASCVD. METHODS AND RESULTS We included 9446 statin-treated patients with LDL-C < 70 mg/dL undergoing PCI between 2012 and 2022, stratified into four groups: (i) no residual risk (TG <150 mg/dL + hs-CRP <2 mg/L); (ii) residual TG risk (TG ≥150 mg/dL + hs-CRP <2 mg/L); (iii) residual inflammatory risk (TG <150 mg/dL + hs-CRP ≥2 mg/L); and (iv) residual TG and inflammatory risk (TG ≥150 mg/dL + hs-CRP ≥2 mg/L). The primary endpoint was major adverse cardiovascular events (MACE) at 1 year, consisting of all-cause mortality, myocardial infarction, or stroke. Cox regression analysis was performed, using the no residual risk group as a reference. Of the total population, 5339 (56.5%) had no residual risk, 555 (5.9%) presented residual TG risk, 3009 (31.9%) had residual inflammatory risk, and 543 (5.7%) exhibited residual combined risk. After multivariable adjustment, patients with residual inflammatory or combined risk showed a significantly higher hazard of MACE, mainly driven by all-cause mortality. No significant difference was observed between patients with residual TG risk and those with no residual risk. CONCLUSION In statin-treated patients with well-controlled LDL-C undergoing PCI, residual inflammatory risk-alone or in combination with residual TG risk-was associated with a higher incidence of MACE, highlighting the need for targeted preventive strategies beyond LDL-C lowering.
Collapse
Affiliation(s)
- Francesca Maria Di Muro
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
- Department of Experimental and Clinical Medicine, School of Human Health Sciences, Careggi University Hospital, University of Florence, Florence, Italy
| | - Birgit Vogel
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Samantha Sartori
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Benjamin Bay
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Angelo Oliva
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Yihan Feng
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Prakash Krishnan
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Joseph Sweeny
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Mauro Gitto
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Kenneth Smith
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Pedro Moreno
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Johny Nicolas
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Parasuram Krishnamoorthy
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Pier Pasquale Leone
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Deepak L Bhatt
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - George Dangas
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Annapoorna Kini
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Samin K Sharma
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Roxana Mehran
- Icahn School of Medicine at Mount Sinai, Mount Sinai Fuster Heart Hospital, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| |
Collapse
|
10
|
Hu Q, Ma X, Cai T, Li Y. Flavonoid intake, inflammation, and atherosclerotic cardiovascular disease risk in U.S. adults: a cross-sectional study. Nutr Metab (Lond) 2025; 22:24. [PMID: 40102925 PMCID: PMC11917028 DOI: 10.1186/s12986-025-00913-4] [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: 06/25/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND This study aimed to investigate the relationship between the dietary intake of total flavonoids and their six subclasses and the risk of atherosclerotic cardiovascular disease (ASCVD) in adults, and to evaluate the potential mediating effect of inflammation in this association. METHODS Cross-sectional data from 3841 individuals participating in the National Health and Nutrition Examination Survey 2017-2018 were included in the analysis. Flavonoid intake was assessed using a 2-day dietary recall method, and ASCVD status was determined by extracting relevant information from the medical condition questionnaire. To determine the relationship between flavonoid intake and ASCVD risk, we employed logistic regression, subgroup, mediation, and restricted cubic spline analyses. RESULTS Intake of flavan-3-ols, flavones, flavonols, and total flavonoids was negatively correlated with ASCVD risk. Subgroup analysis revealed that the association between flavonoid intake and ASCVD risk exhibits sex-specific differences, with the relationship being more pronounced among women. The significant associations between increased flavonoid intake and reduced ASCVD risk were observed in smokers, non-alcohol consumers, physically inactive individuals, those with hypertension. A nonlinear relationship was observed between the intake of total flavonoids, flavan-3-ols and flavonols and ASCVD risk. Additionally, high-sensitivity C-reactive protein (hs-CRP) and the neutrophil-to-lymphocyte ratio (NLR), inflammatory markers relevant to ASCVD, were found to mediate the association between flavonoid intake and ASCVD risk. Flavonoids demonstrated a dose‒response relationship with reductions in the levels of hs-CRP and the NLR. CONCLUSIONS This study indicates the inverse association between flavonoid intake, particularly flavan-3-ols, flavones, and flavonols, and the risk of ASCVD. It highlights the mediating role of CRP and NLR in this relationship. Furthermore, the study emphasizes the importance of considering lifestyle factors and sex when evaluating the cardiovascular benefits of flavonoids.
Collapse
Affiliation(s)
- Qin Hu
- Department of Epidemiology, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Xiangyu Ma
- Department of Epidemiology, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Tongjian Cai
- Department of Epidemiology, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yafei Li
- Department of Epidemiology, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
11
|
Wang T, Zhang M, Shi W, Li Y, Zhang T, Shi W. Atherogenic index of plasma, high sensitivity C-reactive protein and incident diabetes among middle-aged and elderly adults in China: a national cohort study. Cardiovasc Diabetol 2025; 24:103. [PMID: 40045300 PMCID: PMC11883954 DOI: 10.1186/s12933-025-02653-4] [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: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The atherogenic index of plasma (AIP) and systematic inflammation, as measured by high-sensitivity C-reactive protein (hsCRP), are predictors of diabetes, but their combined impacts on incident diabetes are poorly understood. Using a nationally representative cohort in China, we aimed to investigate the association of AIP and hsCRP with incident diabetes among middle-aged and elderly adults. METHODS This cohort comprised 9,112 participants aged at least 45 years from 125 cities in the China Health and Retirement Longitudinal Study who were free of diabetes at baseline in 2011. Of these, 5,048 participants were followed up until 2015. The AIP was calculated as Log10[TG (mg/dL)/HDL-C(mg/dL)]. Multivariate logistic regression and linear mixed-effect (LME) models were performed to evaluate the associations of AIP, hsCRP, and incident diabetes as well as glycemic biomarkers. Receiver operating characteristic (ROC) curves were used to evaluate their diagnostic values. We conducted a mediation analysis to assess the direct and indirect associations between AIP and hsCRP with diabetes. RESULTS 489 (9.7%) cases developed diabetes during four years. Higher levels of AIP and hsCRP were independently associated with diabetes. Compared to the lowest quartile of AIP or hsCRP, the highest quartile of AIP (adjusted odds ratio, aOR 2.53, 95% CI: 1.90-3.38) and hsCRP (aOR 2.38, 1.79-3.16) was significantly associated with incident diabetes. The joint effects showed that participants with higher levels of AIP and hsCRP had significantly higher aOR of 2.76 (2.13-3.57). The LME models showed AIP and hsCRP were related to an increased level of fasting blood glucose and glycated hemoglobin. The combination of AIP and hsCRP has better predictive efficacy (area under the curve, AUC: 0.628, 0.601-0.654) for incident diabetes than alone. Mediation analyses showed that high AIP significantly mediated 25.4% of the association between hsCRP and diabetes, and hsCRP simultaneously mediated 5.7% of the association between AIP and diabetes. CONCLUSIONS This cohort suggests combined effects and mutual mediation between the AIP and hsCRP on incident diabetes in China. Our findings provide clinical implications for monitoring and managing AIP and hsCRP levels to mitigate the development of diabetes.
Collapse
Affiliation(s)
- Tongshuai Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Clinical Research Unit Office, Tongren Hospital Shanghai Jiao Tong University School of Medicine, 200336, Shanghai, China
| | - Mengru Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Wenxing Shi
- Department of Pharmaceutical and Biomedical Engineering, Clinical College of Anhui Medical University, Anhui, 230031, China
| | - Yongzhen Li
- Clinical Nutrition Department, Starkids Children's Hospital, Shanghai, New Hong Qiao Campus for Children's Hospital of Fudan University, Shanghai, 201106, China
- School of Public Health, Peking University, Beijing, 100191, China
| | - Tiantian Zhang
- School of Public Health, Fudan University, Shanghai, 200032, China
| | - Wenming Shi
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gaoke Road, Pudong New District, Shanghai, 201204, China.
| |
Collapse
|
12
|
Pesce G, Gusto G, Johansen P, Khachatryan A, Lopez-Ledesma B, Vukmirica J, Cases A. Systemic inflammation prevalence in patients with atherosclerotic cardiovascular disease and chronic kidney disease: a population-based study using a nationwide primary care database in Spain. Front Cardiovasc Med 2025; 12:1538466. [PMID: 40104140 PMCID: PMC11913838 DOI: 10.3389/fcvm.2025.1538466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction Systemic inflammation is recognised as a critical driver of atherosclerotic cardiovascular disease (ASCVD), especially in patients with comorbid chronic kidney disease (CKD). This study aims to assess the prevalence of systemic inflammation in the ASCVD population in Spain. Methods Outpatient electronic medical records from The Health Improvement Network (THIN®) database were used to identify patients with ASCVD and a C-reactive protein (CRP) measurement ≥1 between January 2014 and July 2023 in Spain. The proportion of patients with systemic inflammation (defined as CRP ≥ 2 mg/L) was estimated at the first CRP measurement (index date) and at the end of the study. The patients' characteristics, comorbidities, and drug dispensation in the prior 12 months were reported by systemic inflammation status at the index date. Results Overall, 15,798 patients with ASCVD were included in the study (mean age: 71.1 years; 57% men), of whom 34% had CKD. The proportion of patients with systemic inflammation at the index date was 58% (65% among CKD patients) and 56% (62% among CKD patients) at the end of the study. Patients with systemic inflammation were more frequently smokers, obese, with comorbidities, and had higher low-density lipoprotein cholesterol and triglycerides levels than patients without systemic inflammation. Overall, patients with ASCVD and systemic inflammation used statins and aspirin less frequently compared to patients without systemic inflammation, while they used antibiotics, anticoagulants, and antihypertensives more frequently. Conclusion Systemic inflammation prevalence is high among patients with ASCVD in Spain, especially among patients with comorbid CKD. Therapeutic strategies focused on targeting systemic inflammation may have beneficial effects in reducing the burden of ASCVD.
Collapse
Affiliation(s)
- Giancarlo Pesce
- Real-World Evidence & Modeling Solutions, Certara, Milan, Italy
| | - Gaelle Gusto
- Real-World Evidence & Modeling Solutions, Certara, Paris, France
| | | | - Artak Khachatryan
- Real-World Evidence & Modeling Solutions, Certara, London, United Kingdom
| | | | | | - Aleix Cases
- Servicio de Nefrologia, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
13
|
Hou A, Shi D, Huang H, Liu Y, Zhang Y. Inflammation pathways as therapeutic targets in angiotensin II induced atrial fibrillation. Front Pharmacol 2025; 16:1515864. [PMID: 40098617 PMCID: PMC11911380 DOI: 10.3389/fphar.2025.1515864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/30/2025] [Indexed: 03/19/2025] Open
Abstract
Atrial fibrillation (AF), a common cardiac arrhythmia, is associated with severe complications such as stroke and heart failure. Although the precise mechanisms underlying AF remain elusive, inflammation is acknowledged as a pivotal factor in its progression. Angiotensin II (AngII) is implicated in promoting atrial remodeling and inflammation. However, the exact pathways through which AngII exacerbates AF are still not fully defined. This study explores the key molecular mechanisms involved, including dysregulation of calcium ions, altered connexin expression, and activation of signaling pathways such as TGF-β, PI3K/AKT, MAPK, NF-κB/NLRP3, and Rac1/JAK/STAT3. These pathways are instrumental in contributing to atrial fibrosis, electrical remodeling, and increased susceptibility to AF. Ang II-induced inflammation disrupts ion channel function, resulting in structural and electrical remodeling of the atria and significantly elevating the risk of AF. Anti-inflammatory treatments such as RAAS inhibitors, colchicine, and statins have demonstrated potential in reducing the incidence of AF, although clinical outcomes are inconsistent. This manuscript underscores the link between AngII-induced inflammation and the development of AF, proposing the importance of targeting inflammation in the management of AF.
Collapse
Affiliation(s)
- Ailin Hou
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Xiyuan Hospital, Beijing, China
| | - Dazhuo Shi
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongbo Huang
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxuan Liu
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Soussi S, Price S, Proudfoot A, Mebazaa A, Lawler PR. Why Molecular Subphenotyping Is Needed in Cardiogenic Shock and How to Accomplish This. Am J Respir Crit Care Med 2025; 211:319-322. [PMID: 39805090 PMCID: PMC11936121 DOI: 10.1164/rccm.202407-1475vp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Affiliation(s)
- Sabri Soussi
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia and Pain Management, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Cardiovascular Markers in Stress Conditions (MASCOT), Inserm UMR-S 942, Paris, France
| | - Susanna Price
- Division of Heart, Lung and Critical Care, Royal Brompton Hospital, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Alastair Proudfoot
- Critical Care and Perioperative Medicine Group, School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
- Department of Perioperative Medicine, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Alexandre Mebazaa
- Cardiovascular Markers in Stress Conditions (MASCOT), Inserm UMR-S 942, Paris, France
- Department of Anaesthesiology, Critical Care, Lariboisière – Saint-Louis Hospitals, DMU Parabol, Assistance Publique-Hôpitaux de Paris Nord, University of Paris Cité, Paris, France; and
| | - Patrick R. Lawler
- McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Reilly NA, Dekkers KF, Molenaar J, Arumugam S, Kuipers TB, Ariyurek Y, Hoeksema MA, Jukema JW, Heijmans BT. EPA Induces an Anti-Inflammatory Transcriptome in T Cells, Implicating a Triglyceride-Independent Pathway in Cardiovascular Risk Reduction. JACC Basic Transl Sci 2025; 10:383-395. [PMID: 40139879 PMCID: PMC12013851 DOI: 10.1016/j.jacbts.2024.09.002] [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: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 03/29/2025]
Abstract
Twice-daily intake of purified eicosapentaenoic acid (EPA) reduces atherosclerotic cardiovascular disease risk in patients with high triglycerides, but its exact mechanism remains unclear. We exposed non-activated CD4+ T cells to 100μM EPA, oleic acid, palmitic acid, or control, and conducted RNA and ATAC-sequencing after 48 hours. EPA exposure downregulated immune response-related genes like HLA-DRA, CD69, and IL2RA, and upregulated oxidative stress prevention genes like NQO1. Transcription factor footprinting showed decreased GATA3 and PU.1, and increased REV-ERB. These effects were specific to EPA, suggesting it induces an anti-inflammatory transcriptomic landscape in CD4+ T cells, contributing to its observed cardiovascular benefits.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen Molenaar
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sinthuja Arumugam
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas B Kuipers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Yavuz Ariyurek
- Leiden Genome Technology Center, Department of Human Genetics, Leiden University Medical Center, the Netherlands
| | - Marten A Hoeksema
- Department of Medical Biochemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
16
|
Zheng L, Ye J, Liao X, Li J, Wang Q, Wang F. Frailty, high-sensitivity C-reactive protein and cardiovascular disease: a nationwide prospective cohort study. Aging Clin Exp Res 2025; 37:58. [PMID: 40021565 PMCID: PMC11870887 DOI: 10.1007/s40520-025-02928-6] [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: 10/07/2024] [Accepted: 01/07/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND This study aimed to investigate the complex associations of frailty and high-sensitivity C-reactive protein (hsCRP) with cardiovascular disease (CVD) through a nationwide prospective cohort, while also assessing the mediating associations. METHODS According to critical criteria, a total of 5239 participants from the China Health and Retirement Longitudinal Study (CHARLS) in 2011 were ultimately enrolled in this study. Frailty was evaluated by the frailty index with 40 items, and CVD was defined as the presence of physician-diagnosed heart disease and/or stroke. A restricted cubic spline model, receiver operating characteristic curves, adjusted Cox proportional hazards regression, interaction analyses and mediation analyses were performed for association exploration. RESULTS During a maximum follow-up of 7.0 years, 1204 (23.67%) people developed CVD. Both elevated hsCRP and frailty were significantly associated with CVD incidence. Compared with participants with a healthy status and low hsCRP (< 1.015 mg/L), those with a frailty status and elevated hsCRP had the highest risk of CVD (adjusted HR, 2.97; 95% CI 2.29-3.84), heart disease (adjusted HR, 2.93; 95% CI 2.16-3.96), and stroke (adjusted HR, 4.26; 95% CI 2.81-6.44), which were still robust in the subgroup analysis. Moreover, frailty significantly mediated 19.60% of the associations between hsCRP and CVD. CONCLUSIONS Combined assessment of frailty and hsCRP levels helps to better stratify the individual risk of CVD. Frailty could partly mediate the associations between hsCRP and CVD incidence.
Collapse
Affiliation(s)
- Lei Zheng
- Department of Urology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianjun Ye
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyang Liao
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Li
- Department of Cardiology, People's Hospital of Tibet Autonomous Region, Lhasa, China
| | - Qihao Wang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Wang
- Department of Urology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, China.
| |
Collapse
|
17
|
Yan C, Chen G, Jing Y, Ruan Q, Liu P. Association between air pollution and cardiovascular disease risk in middle-aged and elderly individuals with diabetes: inflammatory lipid ratio accelerate this progression. Diabetol Metab Syndr 2025; 17:65. [PMID: 39980049 PMCID: PMC11844067 DOI: 10.1186/s13098-025-01638-3] [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: 11/15/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Long-term exposure to air pollution significantly increases the risk of cardiovascular disease (CVD); however, the association and underlying mechanisms in individuals with diabetes remain unconfirmed. METHODS We used data from the China Health and Retirement Longitudinal Study (CHARLS) to follow 5,430 adults over a four-year period. Baseline CVD and diabetes status were determined, and high-resolution data were used to assess air pollution exposure to PM1, PM2.5, PM10, and O3. The inflammatory lipid ratio (ILR) was calculated to reflect inflammatory and lipid metabolic states. A generalized linear model (GLM) was employed to analyze the effects of air pollution and ILR on diabetes-related CVD risk. RESULTS The prevalence of CVD was 8.5% in the healthy population and 13.8% in the diabetic population. Air pollution exposure was significantly associated with an increased risk of CVD among diabetic individuals. For each interquartile range (IQR) increase in concentrations of pollutants O3, PM1, PM10, and PM2.5, CVD risk in the diabetic group rose by 21%, 19%, 28%, and 19%, respectively. Higher ILR values were positively associated with CVD incidence (OR = 1.019, 95% CI: 1.001-1.037, P < 0.05), with a nonlinear relationship observed between ILR levels and CVD risk (PNonlinear = 0.0381), indicating that higher ILR values exacerbate the impact of air pollution on diabetic individuals. CONCLUSION Among middle-aged and older adults with diabetes, exposure to air pollution is associated with an increased risk of CVD, and ILR intensifies this process. Therefore, implementing effective public health interventions to reduce air pollution exposure in diabetic populations is essential.
Collapse
Affiliation(s)
- Chunyu Yan
- Department of Endocrinology and Metabolism, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China
| | - Guang Chen
- Department of Endocrinology and Metabolism, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China
| | - Yingyu Jing
- Department of Endocrinology and Metabolism, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China
| | - Qi Ruan
- Department of Endocrinology and Metabolism, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China
| | - Ping Liu
- Department of Endocrinology and Metabolism, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China.
- Xi'an Key Laboratory of Metabolic Disease Imaging, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an, Shanxi Province, 710021, China.
| |
Collapse
|
18
|
Loscalzo J. Timing the Taming of Vascular Inflammation. N Engl J Med 2025; 392:712-714. [PMID: 39938099 DOI: 10.1056/nejme2416329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Affiliation(s)
- Joseph Loscalzo
- Brigham and Women's Hospital, Boston
- Harvard Medical School, Boston
| |
Collapse
|
19
|
Rusu CC, Kacso I, Moldovan D, Potra A, Tirinescu D, Ticala M, Maslyennikov Y, Urs A, Bondor CI. Exploring the Associations Between Inflammatory Biomarkers, Survival, and Cardiovascular Events in Hemodialysis Patients and the Interrelationship with Nutritional Parameters-The Experience of a Single Transylvanian Dialysis Center. J Clin Med 2025; 14:1139. [PMID: 40004669 PMCID: PMC11855970 DOI: 10.3390/jcm14041139] [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: 12/25/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: In hemodialysis (HD), inflammatory biomarkers are discussed as prognostic markers for survival and cardiovascular events (CVEs). The results of the studies are not uniform and there are particularities related to population groups and comorbidities. In addition, it is known that inflammation determines protein malnutrition and less about the effect of adipose tissue on inflammation in HD. This study investigates the relationship between inflammatory molecules and nutritional biomarkers, and CVE and survival in HD patients. Methods: We included, in an observational, longitudinal study, 65 patients with chronic HD (53 without diabetes and 22 smokers), with a mean age of 60.1 ± 12.4 years. High-sensitivity C-reactive protein (hs-CRP), interleukin 1 beta, tumor necrosis factor alpha (TNF-alpha), interleukin 6, soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK), soluble CD163 (sCD163), and fibroblast growth factor 21 were determined. We recorded survival and cardiovascular events for 60 months. Univariate and multivariate analyses were performed. Results: Hs-CRP was significantly associated with survival (p = 0.014) in the total group. In smokers and former smokers, TNF-α lower than 368.34 pg/mL was associated with better survival. In multivariate analysis, hs-CRP was correlated with adipose tissue biomarkers (p = 0.006), and sCD163 was correlated with total and LDL cholesterol (p = 0.002). In addition, in univariate analysis, sTWEAK was correlated with serum albumin (p = 0.026, r = -0.30). In conclusion, in HD patients, hs-CRP was significantly associated with survival, and low TNF-alpha values in smokers and former smokers were linked to better survival. Hs-CRP was also correlated with adipose tissue biomarkers, CD163 was correlated with total and LDL cholesterol, and albumin was inversely associated with sTWEAK. The relation between inflammatory molecules and adipose tissue biomarkers was less identified in HD patients until now.
Collapse
Affiliation(s)
- Crina Claudia Rusu
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Ina Kacso
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Diana Moldovan
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alina Potra
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Dacian Tirinescu
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Maria Ticala
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Yuriy Maslyennikov
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alexandra Urs
- Department of Nephrology, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Cosmina Ioana Bondor
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| |
Collapse
|
20
|
Du Z, Lu Y, Ma Y, Yang Y, Luo W, Liu S, Zhang M, Wang Y, Li L, Li C, Wang W, Gao H. The prognostic and therapeutic significance of polyunsaturated fatty acid-derived oxylipins in ST-segment elevation myocardial infarction. IMETA 2025; 4:e266. [PMID: 40027487 PMCID: PMC11865345 DOI: 10.1002/imt2.266] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 03/05/2025]
Abstract
Polyunsaturated fatty acid-derived oxylipins regulate systemic inflammation and exert cardiovascular effects, yet their role in ST-segment elevation myocardial infarction (STEMI) remains unclear. Herein, we used targeted metabolomics and machine learning algorithms to develop an oxylipin-based risk model to accurately predict recurrent major adverse cardiovascular events (MACE) after STEMI in two independent prospective cohorts with 2 years of follow-up. The in vivo effects of significant oxylipin predictors were explored via a murine myocardial ischemia‒reperfusion model and functional metabolomics. Among the 130 plasma oxylipins detected in discovery cohort (n = 645), patients with and without recurrent MACE exhibited significant differences in a variety of oxylipin subclasses. We constructed an oxylipin-based prediction model that showed powerful performance in predicting recurrent MACE in the discovery cohort (predictive accuracy: 91.5%). The predictive value of the oxylipin marker panel was confirmed in an independent external validation cohort (predictive accuracy: 89.9%; n = 401). Furthermore, we found that the anti-inflammatory/pro-resolving oxylipin (ARO) predictor panel showed better prognostic performance than the pro-inflammatory oxylipin predictor panel in both cohorts. Compared with the treatment of pro-inflammatory oxylipin predictor panel, combined treatment of six ARO predictors, including 14,15 epoxy-eicosatrienoic acid, 14(15)-epoxy-eicosatetraenoic acid, 12,13-epoxy-octadecenoic acid, lipoxin A4, resolving D1, and 6 keto-prostaglandin F1 showed significant cardiac activities and synergistic metabolic actions in myocardial infarction‒reperfusion model mice. We also mechanistically identified an important role of ARO predictors in restraining ceramide/lysophosphatidylcholine synthesis and inhibiting inflammatory responses. Overall, the present study depicted the landscape of oxylipin profiles in the largest panel of STEMI patients worldwide. Our results also highlight the great potential of bioactive oxylipins in prognostic prediction and therapeutics after STEMI.
Collapse
Affiliation(s)
- Zhiyong Du
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yingyuan Lu
- School of Pharmaceutical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityPeking UniversityBeijingChina
| | - Ying Ma
- The State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, National Resource Center for Chinese Materia Medica, China, Academy of Chinese Medical SciencesBeijingChina
| | - Yunxiao Yang
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Wei Luo
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Sheng Liu
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ming Zhang
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yong Wang
- Dongzhimen Hospital of Beijing University of Chinese MedicineBeijingChina
| | - Lei Li
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Chun Li
- Institute of Traditional Chinese MedicineBeijing University of Chinese MedicineBeijingChina
- Chinese Medicine Guangdong LaboratoryGuangdong HengqinChina
- State Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Wei Wang
- Institute of Traditional Chinese MedicineBeijing University of Chinese MedicineBeijingChina
- Chinese Medicine Guangdong LaboratoryGuangdong HengqinChina
- State Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Hai Gao
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
21
|
Qin X, Ren J, Xing C, Chen L, Wang R, Tian S. Linear association between high-sensitivity C-reactive protein and postoperative delirium after general anesthesia: a cross-sectional study. Front Neurol 2025; 16:1516800. [PMID: 39958614 PMCID: PMC11827214 DOI: 10.3389/fneur.2025.1516800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Objective To investigate the association between high-sensitivity C-reactive protein (Hs-CRP) levels and the risk of postoperative delirium (POD) following general anesthesia. Methods This retrospective cross-sectional study included 644 patients who underwent general anesthesia. Univariate and multivariate logistic regression analyses were performed to evaluate the relationship between Hs-CRP and POD, with subgroup analyses used to assess stratified associations. Receiver operator characteristic (ROC) curve analysis was employed to assess the predictive efficacy of Hs-CRP for POD. Restricted cubic spline (RCS) analysis was conducted to explore the linear relationship between the log-transformed Hs-CRP (Log10Hs-CRP) and POD risk. Results The total population consisted of 644 individuals with a mean age of 64.02 ± 13.20 years, 506 (78.60%) of whom were male, and 114 patients (17.7%) had POD. Compared to the lower Hs-CRP group, patients in the higher Hs-CRP group exhibited higher age, heart rate, white blood cell count, blood urea nitrogen, creatinine, uric acid, fasting glucose, hemoglobin A1c, fibrinogen, D-dimer, and a higher prevalence of CKD, but lower hemoglobin, high-density lipoprotein cholesterol, albumin and estimated glomerular filtration rate. Additionally, the prevalence of POD was higher in the higher Hs-CRP group (24.7% vs. 9.5%, p < 0.001). Multivariate logistic regression confirmed that elevated Hs-CRP and its forms (Log10Hs-CRP, standardized Hs-CRP, and higher Hs-CRP group) consistently increased the risk of POD across all adjusted models (p < 0.05). Stratified analyses further highlighted significant associations between Hs-CRP and POD in specific subgroups, notably in patients aged ≥65 years, female patients, and those with or without hypertension, diabetes, or stroke history, and without chronic kidney disease (p < 0.05). ROC curve analysis demonstrated that Hs-CRP had a significant predictive ability for POD in the overall population (AUC = 0.646), as well as in male (AUC = 0.644) and female patients (AUC = 0.654). Additionally, RCS analysis indicated a linear positive association between Log10Hs-CRP and POD risk (p = 0.003, nonlinear p = 0.896). Conclusion Elevated Hs-CRP levels are significantly associated with an increased risk of POD following general anesthesia.
Collapse
Affiliation(s)
- Xiao Qin
- Department of Anesthesiology, Sixth Hospital of Shanxi Medical University, General Hospital of Tisco, Taiyuan, China
| | - Junming Ren
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Chunping Xing
- Department of Anesthesiology, Sixth Hospital of Shanxi Medical University, General Hospital of Tisco, Taiyuan, China
| | - Lijiao Chen
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renjie Wang
- Department of Urology, General Hospital of Tisco, Sixth Hospital of Shanxi Medical University, Taiyuan, China
| | - Shouyuan Tian
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
22
|
Zhihong G, Yuqiang Z, Linyi J, Yuling Y, Xu Y, Lei X, Zengfang H. Correlation analysis between epicardial adipose tissue and acute coronary syndrome. Sci Rep 2025; 15:3015. [PMID: 39849033 PMCID: PMC11758384 DOI: 10.1038/s41598-025-87594-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025] Open
Abstract
To investigate the correlation between the density and volume of epicardial adipose tissue(EAT)and acute coronary syndrome (ACS). This study included 355 subjects (mean age: 60.65 ± 9.67 years; 54.65% male), comprising 175 patients with ACS and 180 without ACS. Propensity score matching was applied to balance the variables between the two groups, resulting in 96 successfully matched pairs. Clinical data, epicardial adipose tissue volume (EATV), and epicardial adipose tissue density (EATD) were compared. Independent factors influencing ACS were identified using logistic regression analysis, and the predictive ability of each variable was evaluated using receiver operating characteristic (ROC) curves. Systolic blood pressure, EATV, EATD, fasting blood glucose, triglycerides, and high-sensitivity C-reactive protein were significantly elevated in the ACS group compared with the non-ACS group (all p < 0.05). Spearman correlation analysis revealed a moderate positive correlation between EATV and BMI (r = 0.444, p < 0.001), while EATD showed a weak negative correlation with age (r = -0.177, p = 0.014) and a weak positive correlation with EATV (r = 0.239, p = 0.001). Univariable regression analysis demonstrated that both EATV (OR: 2.018, 95% CI: 1.334-3.052) and EATD (OR: 5.341, 95% CI: 3.293-8.663) were associated with ACS. After adjusting for other risk factors, logistic regression model confirmed that EATV (adjusted OR: 1.892, 95%CI: 1.211-2.955) and EATD (adjusted OR: 6.942, 95%CI: 3.875-12.437) were independent predictors of ACS (both p < 0.001), with EATD showing the highest predictive value (AUC = 0.859). This study identifies a close relationship between EAT and ACS, highlighting EATD and EATV as independent influencing factors for ACS. Among them, EATD demonstrated a stronger predictive value for ACS than both traditional risk factors and EATV.
Collapse
Affiliation(s)
- Gao Zhihong
- Health Examination Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, People's Republic of China
| | - Zuo Yuqiang
- Health Examination Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, People's Republic of China
| | - Jia Linyi
- Radiology Department, Xingtai People's Hospital, Xingtai, 054031, Hebei, People's Republic of China
| | - Yin Yuling
- Health Examination Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, People's Republic of China
| | - Yang Xu
- Health Examination Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, People's Republic of China
| | - Xu Lei
- Cardiology Department, Shijiazhuang People's Hospital, Shijiazhuang, 050011, Hebei, People's Republic of China
| | - Hao Zengfang
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
23
|
Goonewardena SN, Murthy VL. The complete blood count and cardiovascular disease: analyses across six cohorts of 23,370 adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.10.17.24315694. [PMID: 39484272 PMCID: PMC11527051 DOI: 10.1101/2024.10.17.24315694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background The complete blood count (CBC) is one of the most performed laboratory studies. However, the CBC and its components are not commonly used to understand and quantify cardiovascular disease (CVD) risk. Objective We sought to define the relationships between the CBC, traditional CVD risk factors, and common CVD biomarkers and their joint association with all-cause mortality and CVD. Methods We examined the relationships between the CBC, traditional CVD risk factors, and mortality in NHANES (n=7843). We validated and extended these findings to more refined CVD endpoints in five additional cohorts (n=15,527). Results We first examined the variance accounted for by common laboratory studies (lipid panel, HbA1c, hs-CRP, and basic metabolic panel) by traditional risk factors in NHANES. Except for hemoglobin (Hb) components, we found that traditional risk factors accounted for less than 20% of the variance in the CBC, suggesting that the CBC provides unique information beyond traditional risk factors and CVD biomarkers. Additionally, the CBC was strongly associated with all-cause mortality (p<0.0001), even more than traditional CVD biomarkers (lipid panel, HbA1c, and CRP). We validated and extended these findings across five additional cohorts with a mean follow-up of 16 years and more refined CVD endpoints. In the fully adjusted analyses, several CBC components, including the white blood cell (WBC) count, neutrophil (PMN) count, Hb level, and an integrated immune cell score, were associated with individual CVD endpoints (incident stroke, MI, or revascularization) and a composite CV endpoint (MACE3) with standardized hazard ratios of 1.13 (p=0.002), 1.15 (p=0.0006), 0.82 (p<0.0001), and 2.16 (p<0.0001) respectively. Conclusion This study represents the first systematic examination of the relationship between the CBC, all-cause mortality, and CVD in a diverse cohort of 23,370 adults. These findings underscore the added value of the CBC over traditional risk factors and common CVD biomarkers for CVD risk assessment. Future studies should explore the integration of CBC parameters into predictive models to enhance our understanding, early identification, and prevention strategies for CVD.
Collapse
Affiliation(s)
- Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Cardiovascular Medicine, VA Ann Arbor Health System, Ann Arbor, Michigan, USA
| | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [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: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
25
|
Siverio-Morales O, Mora-Fernández C, Hernández-Carballo C, Martín-Núñez E, González-Luis A, Martín-Olivera A, Navarro-González JF, Donate-Correa J. Predictive value of triglyceride-glucose index for the evaluation of coronary artery disease severity and occurrence of major adverse cardiovascular events. Am J Physiol Heart Circ Physiol 2025; 328:H14-H20. [PMID: 39560964 DOI: 10.1152/ajpheart.00684.2024] [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: 10/04/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024]
Abstract
The triglyceride-glucose (TyG) index has been proposed as an independent predictor of coronary artery disease (CAD). In this retrospective study, we further examine this association and its utility as a predictor for major adverse cardiovascular events (MACE). A total of 870 patients who underwent coronary angiography between May 2008 and June 2009 were included in this retrospective study. The TyG index was calculated using the formula Ln [fasting TG (mg/dL) × FBG (mg/dL)/2]. The association of the TyG index with the presence and severity of CAD, cardiovascular risk factors, and inflammatory markers was evaluated at baseline. In the longitudinal study, the multivariate-adjusted Cox hazard model was used to investigate the associations of the TyG index with the occurrence of MACE during a 5-yr follow-up, which was defined as the endpoint. The TyG index was significantly associated with the presence and severity of CAD. Multiple linear regression analysis showed that a high TyG index, together with inflammatory markers and dyslipidemia, was independently associated with greater stenotic occlusion of coronary arteries (adjusted R2 = 0.031, P < 0.001). Kaplan-Meier survival curve (free of MACE) by tertiles of the TyG index showed a higher incidence of MACE in the upper tertile (log-rank test, P = 0.02). Multivariate Cox analysis demonstrated that the risk of incident MACE during the follow-up was associated with higher levels of the TyG index, even after adjusting for inflammatory parameters and cardiovascular risk factors: hazard ratio = 1.54 (95% confidence interval: 1.18-2.13; P < 0.01). We conclude that an elevated TyG index is independently associated with a higher risk of CAD and a poor prognosis for MACE.NEW & NOTEWORTHY This retrospective study demonstrates significant associations between the TyG index and the occurrence and severity of CAD, as well as indicates the clinical value of the TyG index as a potential predictor for MACE.
Collapse
Affiliation(s)
- Orlando Siverio-Morales
- Nephrology Service, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Hernández-Carballo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Internal Medicine Service, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Ernesto Martín-Núñez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ainhoa González-Luis
- Doctoral and Graduate School, University of La Laguna, Santa Cruz de Tenerife, Spain
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Alberto Martín-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Juan F Navarro-González
- Nephrology Service, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
26
|
Escobar C, Aldeguer X, Vivas D, Manzano Fernández S, Gonzalez Caballero E, Garcia Martín A, Barrios V, Freixa-Pamias R. The gut microbiota and its role in the development of cardiovascular disease. Expert Rev Cardiovasc Ther 2025; 23:23-34. [PMID: 39915986 DOI: 10.1080/14779072.2025.2463366] [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: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The pathophysiology of cardiovascular diseases encompasses a complex interplay of genetic and environmental risk factors. Even if traditional risk factors are treated to target, there remains a residual risk. AREAS COVERED This manuscript reviews the potential role of gut microbiota in the development of cardiovascular disease, and as potential target. A systematic search was conducted until 30 October 2024 on PubMed (MEDLINE), using the MeSH terms [Gut microbiota] + [Dysbiosis] + [Cardiovascular] + [TMAO] + [bile acids] + [short-chain fatty acids]. EXPERT OPINION The term dysbiosis implies changes in equilibrium, with modifications in the composition and functionality of microbiota and a series of additional factors: reduced diversity and uniformity of microorganisms; reduced short-chain fatty acid-producing bacteria; increased gut permeability; release of metabolites, such as trimethylamine N-oxide, betaine, phenylalanine, tryptophan-kynurenine, phenylacetylglutamine, and lipopolysaccharides; and reduced secondary bile acid excretion, leading to inflammation, oxidative stress, and endothelial dysfunction and facilitating the onset of pathological conditions, including obesity, hypertension, diabetes, atherosclerosis, and heart failure. Attempts to restore gut microbiota balance through different interventions, mainly changes in diet, have been shown to positively affect individual components and metabolites and reduce the risk of cardiovascular disease. In addition, probiotics and prebiotics are potentially useful. Fecal microbiota transplantation is a promising therapy.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Department, University Hospital La Paz, Madrid, Spain
| | - Xavier Aldeguer
- Gastroenterology Department, Hospital Doctor Josep Trueta i Santa Caterina, Institut d'investigació Biomèdica de Girona IDIBGI, Girona/Salt, Spain
| | - David Vivas
- Cardiovascular Institute, San Carlos University Hospital, Madrid, Spain
- Cardiology Department, Cardiovascular Institute Vithas Milagrosa and Aravaca, Madrid, Spain
| | | | | | - Ana Garcia Martín
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Vivencio Barrios
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Román Freixa-Pamias
- Cardiology Department, Complex Hospitalari Moisès Broggi, Sant Joan Despí, Barcelona, Spain
| |
Collapse
|
27
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [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: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
28
|
Bawamia B, Gupta A, Omari M, Farag M, Spyridopoulos I, Alkhalil M. Eosinopenia in patients with acute myocardial infarction- longitudinal imaging insights from the CAPRI study. J Thromb Thrombolysis 2025; 58:136-144. [PMID: 39306654 DOI: 10.1007/s11239-024-03042-6] [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] [Accepted: 09/04/2024] [Indexed: 01/27/2025]
Abstract
Eosinophils are recruited to the heart during acute myocardial infarction (MI) and are considered part of the inflammatory response associated with adverse clinical outcomes. We assessed the impact of eosinopenia on cardiac imaging biomarkers in patients presenting with ST-segment elevation MI. This is a post-hoc analysis of the Evaluating the effectiveness of intravenous Ciclosporin on reducing reperfusion injury in pAtients undergoing PRImary percutaneous coronary intervention (CAPRI) trial. Patients underwent cardiac MRI within 1 week and 12 weeks and low eosinophil was defined as less than 40 cells/ml. The study included 52 patients and 38% had low eosinophil. Ciclosporin administration was comparable between patients with low versus normal eosinophils. The ischaemia time was significantly longer in low eosinophil patients [262 (205-325) vs. 138 (102-195) minutes, P < 0.001]. At 12 weeks, patients with eosinopenia had larger infarct size [9.8% (5.7-18.4) vs. 7.4% (1.9-10.2), P = 0.045], larger left ventricle (LV) end systolic volume (89 ± 28 vs. 68 ± 23, P = 0.02), and lower LV ejection fraction (EF) (49 ± 9 vs. 58 ± 7, P < 0.001). After adjustments for significant predictors, including ischaemia time, low eosinophil count was an independent predictor of worse LVEF at 12 weeks [-5.78, 95% CI (-11.22 to -0.34), P = 0.038] but not infarct size [1.83, 95% CI (-2.77 to 6.43), P = 0.43]. Patients with low eosinophil count had larger infarct size and LV volumes and worse adverse remodeling compared to those with normal eosinophil count. At 12 weeks, eosinopenia was an independent predictor of worse LVEF but not infarct size.
Collapse
Affiliation(s)
- Bilal Bawamia
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Ashish Gupta
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Muntaser Omari
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Mohamed Farag
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Ioakim Spyridopoulos
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Mohammad Alkhalil
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
29
|
Csengo E, Lorincz H, Csosz E, Guba A, Karai B, Toth J, Csiha S, Paragh G, Harangi M, Nagy GG. Newly Initiated Statin Treatment Is Associated with Decreased Plasma Coenzyme Q10 Level After Acute ST-Elevation Myocardial Infarction. Int J Mol Sci 2024; 26:106. [PMID: 39795963 PMCID: PMC11720258 DOI: 10.3390/ijms26010106] [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: 11/07/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Coenzyme Q10 (CoQ10) plays a crucial role in facilitating electron transport during oxidative phosphorylation, thus contributing to cellular energy production. Statin treatment causes a decrease in CoQ10 levels in muscle tissue as well as in serum, which may contribute to the musculoskeletal side effects. Therefore, we aimed to assess the effect of newly initiated statin treatment on serum CoQ10 levels after acute ST-elevation myocardial infarction (STEMI) and the correlation of CoQ10 levels with key biomarkers of subclinical or clinically overt myopathy. In this study, we enrolled 67 non-diabetic, statin-naïve early-onset STEMI patients with preserved renal function. Plasma CoQ10 level was determined by ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC/MS-MS), while the myopathy marker serum fatty acid-binding protein 3 (FABP3) level was measured with enzyme-linked immunosorbent assay (ELISA) at hospital admission and after 3 months of statin treatment. The treatment significantly decreased the plasma CoQ10 (by 43%) and FABP3 levels (by 79%) as well as total cholesterol, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B100 (ApoB100), and oxidized LDL (oxLDL) levels. The change in CoQ10 level showed significant positive correlations with the changes in total cholesterol, LDL-C, ApoB100, and oxLDL levels, while it did not correlate with the change in FABP3 level. Our results prove the CoQ10-reducing effect of statin treatment and demonstrate its lipid-lowering efficacy but contradict the role of CoQ10 reduction in statin-induced myopathy.
Collapse
Affiliation(s)
- Erika Csengo
- Centre of Cardiovascular Diseases and Internal Medicine, Borsod-Abauj-Zemplen County Central Hospital and University Teaching Hospital, Szentpéteri kapu 72-76, 3526 Miskolc, Hungary
| | - Hajnalka Lorincz
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eva Csosz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Andrea Guba
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Bettina Karai
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Judit Toth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Sara Csiha
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | - Gyorgy Paragh
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mariann Harangi
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Institute of Health Studies, Faculty of Health Sciences, University of Debrecen, Kassai út 26, 4032 Debrecen, Hungary
| | - Gergely Gyorgy Nagy
- Centre of Cardiovascular Diseases and Internal Medicine, Borsod-Abauj-Zemplen County Central Hospital and University Teaching Hospital, Szentpéteri kapu 72-76, 3526 Miskolc, Hungary
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| |
Collapse
|
30
|
Comi L, Giglione C, Klinaku FT, Pialorsi F, Tollemeto V, Zurlo M, Seneci A, Magni P. Valorizing Agro‐Food Waste for Nutraceutical Development: Sustainable Approaches for Managing Metabolic Dysfunction‐Associated Steatotic Liver Disease and Related Co‐Morbidities. FOOD FRONTIERS 2024. [DOI: 10.1002/fft2.535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
ABSTRACTThis comprehensive investigation delves into the interconnectedness of different features of cardiometabolic syndrome, such as metabolic dysfunction‐associated steatotic liver disease (MASLD), atherosclerotic cardiovascular disease (ASCVD), and gut dysbiosis, highlighting the crucial role of nutraceuticals in their management and prevention. Given the significant overlap in the pathophysiology of these conditions, the treatment with nutraceuticals, especially those derived from agro‐food waste, offers a promising, sustainable, and innovative approach to healthcare. The 2030 Agenda for Sustainable Development and the One Health concept are key frameworks for selecting the most interesting supply chain for the production of nutraceuticals from agro‐food waste, ensuring environmental sustainability, and innovative agricultural practices. In this review, the therapeutic potential of kiwifruit and apples has been explored, detailing how their bioactive compounds, like polyphenols, fiber, pectin, kaempferol, phloretin, and phlorizin, may contribute to the management of MASLD, ASCVD, and gut dysbiosis. Various extraction methods for active ingredients, including chemical, water, and enzyme extractions, are analyzed for their respective benefits and drawbacks. By integrating scientific research, sustainable agricultural practices, and innovative extraction methods, we can develop effective strategies to combat these pervasive health issues. This holistic approach not only enhances individual health outcomes but also supports broader environmental and societal goals, promoting a healthier future for all.
Collapse
Affiliation(s)
- Laura Comi
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Claudia Giglione
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Fationa Tolaj Klinaku
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | | | | | | | | | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
- IRCCS MultiMedica, Sesto San Giovanni Milan Italy
| |
Collapse
|
31
|
Zeng G, Zhang C, Song Y, Zhang Z, Xu J, Liu Z, Tang X, Wang X, Chen Y, Zhang Y, Zhu P, Guo X, Jiang L, Wang Z, Liu R, Wang Q, Yao Y, Feng Y, Han Y, Yuan J. The potential impact of inflammation on the lipid paradox in patients with acute myocardial infarction: a multicenter study. BMC Med 2024; 22:599. [PMID: 39710711 PMCID: PMC11664818 DOI: 10.1186/s12916-024-03823-z] [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: 06/06/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) is a well-recognized risk factor for cardiovascular diseases. However, several clinical studies demonstrated an inverse association between LDL-C and mortality risk in patients with acute myocardial infarction (AMI), known as the lipid paradox. This study aims to investigate the potential impact of inflammation on the association between LDL-C levels and mortality risks. METHODS A total of 5244 patients with AMI from a large nationwide prospective cohort were included in our analysis. Patients were stratified according to LDL-C quartiles. The primary outcome was all-cause mortality, and the secondary endpoint was cardiac mortality. High-sensitive C-reactive protein (hsCRP) > 3 mg/L was defined as high inflammatory risk. RESULTS During a median follow-up of 2.07 years, 297 mortality events (5.5%) and 227 cardiac mortality events (4.2%) occurred. Patients in the lowest LDL-C quartile had the highest incidence of all-cause mortality (7.3%) and cardiac mortality (5.8%). A U-shaped association between LDL-C levels and mortality risk was observed after multivariable adjustment, which persisted only in patients with high hsCRP levels. In contrast, a linear association between LDL-C and mortality risk was shown in patients with low hsCRP levels. CONCLUSIONS AMI patients with lower LDL-C levels had a higher risk of mortality. However, this association was only observed in those with high inflammatory risk. In contrast, the relationship between LDL-C and mortality risk was linear in patients with low inflammatory risk. This suggests the importance of considering inflammation when managing LDL-C levels in AMI patients.
Collapse
Affiliation(s)
- Guyu Zeng
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Ce Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Ying Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Xiaozeng Wang
- Department of Cardiology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110016, China
| | - Yan Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Jiang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, China
| | - Ru Liu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Qingsheng Wang
- Department of Cardiology, The First Hospital of QinHuangDao, Qinhuangdao, China
| | - Yi Yao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110016, China.
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China.
| |
Collapse
|
32
|
Wang X, Zheng Q, Zha L, Zhang L, Huang M, Zhang S, Zhang X, Li Q, Chen X, Xia N, Zhang M, Lv B, Jiao J, Lu Y, Gu M, Yang F, Li J, Li N, Cheng X, Zhou Z, Tang T. Thymic stromal lymphopoietin modulates T cell response and improves cardiac repair post-myocardial infarction. Front Immunol 2024; 15:1467095. [PMID: 39703503 PMCID: PMC11655303 DOI: 10.3389/fimmu.2024.1467095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Background The inflammatory response is associated with cardiac repair and ventricular remodeling after myocardial infarction (MI). The key inflammation regulatory factor thymic stromal lymphopoietin (TSLP) plays a critical role in various diseases. However, its role in cardiac repair after MI remains uncertain. In this study, we elucidated the biological function and mechanism of action of TSLP in cardiac repair and ventricular remodeling following MI. Method and Result Wild-type and TSLP receptor (TSLPR)-knockout (Crlf2-/-) mice underwent MI induction via ligation of the left anterior descending artery. TSLP expression was upregulated in the infarcted heart, with a peak observed on day 7 post-MI. TSLP expression was enriched in the cardiomyocytes of infarcted hearts and the highest expression of TSLPR was observed in dendritic cells. Crlf2-/- mice exhibited reduced survival and worsened cardiac function, increased interstitial fibrosis and cardiomyocyte cross-sectional area, and reduced CD31+ staining, with no change in the proportion of apoptotic cardiomyocytes within the border zone. Mechanistically, reduced Treg cell counts but increased myeloid cell infiltration and an increased ratio of Ly6Chigh/Ly6Clow monocyte were observed in the ani hearts of Crlf2-/- mice. Further, TSLP regulated CD4+ T cell activation and proliferation at baseline and after MI, with a greater impact on Treg cells than on conventional T cells. RNA-seq analysis revealed significant downregulation of genes involved in T cell activation and TCR signaling in the infarcted heart of Crlf2-/- mice compared with their WT counterparts. Conclusion Collectively, our data indicate a critical role for TSLP in facilitating cardiac repair and conferring protection against MI, primarily through regulating CD4+ T cell responses, which may provide a potential novel therapeutic approach for managing heart failure after MI.
Collapse
Affiliation(s)
- Xuhong Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingxue Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingkai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuzhe Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinlin Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinglin Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Jiao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhi Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Muyang Gu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyong Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nana Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
van der Stouwe JG, Godly K, Kraler S, Godly J, Matter CM, Wenzl FA, von Eckardstein A, Räber L, Mach F, Obeid S, Templin C, Lüscher TF, Niederseer D. Body temperature, systemic inflammation and risk of adverse events in patients with acute coronary syndromes. Eur J Clin Invest 2024; 54:e14314. [PMID: 39350322 DOI: 10.1111/eci.14314] [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: 05/26/2024] [Accepted: 08/18/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Inflammatory processes can trigger acute coronary syndromes (ACS) which may increase core body temperature (BT), a widely available low-cost marker of systemic inflammation. Herein, we aimed to delineate baseline characteristics of ST-segment elevation myocardial infarction (STEMI) and non-ST-segment elevation ACS (NSTE-ACS) patients stratified by initial BT and to assess its predictive utility towards major adverse cardiovascular events (MACE) after the index ACS. METHODS From 2012 until 2017, a total of 1044 ACS patients, 517 with STEMI and 527 with NSTE-ACS, were prospectively recruited at the University Hospital Zurich. BT was measured by digital tympanic thermometer along with high-sensitivity C-reactive protein (hs-CRP) and cardiac troponin-T (hs-cTnT) levels prior to coronary angiography. Patients were stratified according to initial BT and uni- and multivariable regression models were fit to assess associations of BT with future MACE risk. RESULTS Among patients with STEMI, BT was not predictive of 1-year MACE, but a U-shaped relationship between BT and MACE risk was noted in those with NSTE-ACS (p = .029), translating into a 2.4-fold (HR, 2.44, 95% CI, 1.16-5.16) increased 1-year MACE risk in those with BT >36.8°C (reference: 36.6-36.8°C). Results remained robust in multivariable-adjusted analyses accounting for sex, age, diabetes, renal function and hs-cTnT. However, when introducing hs-CRP, the BT-MACE association did not prevail. CONCLUSIONS In prospectively recruited patients with ACS, initial BT shows a U-shaped relationship with 1-year MACE risk among those with NSTE-ACS, but not in those with STEMI. BT is a broadly available low-cost marker to identify ACS patients with high inflammatory burden, at high risk for recurrent ischaemic events, and thus potentially suitable for an anti-inflammatory intervention. REGISTRATION ClinicalTrials.gov Identifier: NCT01000701.
Collapse
Affiliation(s)
- Jan Gerrit van der Stouwe
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Cardiology, Cardiovascular Research Institute Basel, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Konstantin Godly
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Julia Godly
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Christian M Matter
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- National Disease Registration and Analysis Service, NHS, London, UK
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Clinical Sciences, Karolinska Institutet, Stockholm, Sweden
| | | | - Lorenz Räber
- Department of Cardiology, Inselspital Bern, Bern, Switzerland
| | - François Mach
- Department of Cardiology, University Hospital Geneva, Geneva, Switzerland
| | - Slayman Obeid
- Department of Cardiology, Kantonsspital Liestal, Liestal, Switzerland
| | - Christian Templin
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, UK
- Cardiovascular Academic Group, Kings College London, London, UK
| | - David Niederseer
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Hochgebirgsklinik, Medicine Campus Davos, Davos, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Medicine Campus Davos, Davos, Switzerland
| |
Collapse
|
34
|
Bartoli-Leonard F, Pennel T, Caputo M. Immunotherapy in the Context of Aortic Valve Diseases. Cardiovasc Drugs Ther 2024; 38:1173-1185. [PMID: 39017904 PMCID: PMC11680629 DOI: 10.1007/s10557-024-07608-7] [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] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Aortic valve disease (AVD) affects millions of people around the world, with no pharmacological intervention available. Widely considered a multi-faceted disease comprising both regurgitative pathogenesis, in which retrograde blood flows back through to the left ventricle, and aortic valve stenosis, which is characterized by the thickening, fibrosis, and subsequent mineralization of the aortic valve leaflets, limiting the anterograde flow through the valve, surgical intervention is still the main treatment, which incurs considerable risk to the patient. RESULTS Though originally thought of as a passive degeneration of the valve or a congenital malformation that has occurred before birth, the paradigm of AVD is shifting, and research into the inflammatory drivers of valve disease as a potential mechanism to modulate the pathobiology of this life-limiting pathology is taking center stage. Following limited success in mainstay therapeutics such as statins and mineralisation inhibitors, immunomodulatory strategies are being developed. Immune cell therapy has begun to be adopted in the cancer field, in which T cells (chimeric antigen receptor (CAR) T cells) are isolated from the patient, programmed to attack the cancer, and then re-administered to the patient. Within cardiac research, a novel T cell-based therapeutic approach has been developed to target lipid nanoparticles responsible for increasing cardiac fibrosis in a failing heart. With clonally expanded T-cell populations recently identified within the diseased valve, their unique epitope presentation may serve to identify novel targets for the treatment of valve disease. CONCLUSION Taken together, targeted T-cell therapy may hold promise as a therapeutic platform to target a multitude of diseases with an autoimmune aspect, and this review aims to frame this in the context of cardiovascular disease, delineating what is currently known in the field, both clinically and translationally.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK.
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK.
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa.
| | - Tim Pennel
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
35
|
Potere N, Bonaventura A, Abbate A. Novel Therapeutics and Upcoming Clinical Trials Targeting Inflammation in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2024; 44:2371-2395. [PMID: 39387118 PMCID: PMC11602387 DOI: 10.1161/atvbaha.124.319980] [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] [Indexed: 10/12/2024]
Abstract
Cardiovascular disease (CVD) remains a major health burden despite significant therapeutic advances accomplished over the last decades. It is widely and increasingly recognized that systemic inflammation not only represents a major cardiovascular risk and prognostic factor but also plays key pathogenic roles in CVD development and progression. Despite compelling preclinical evidence suggesting large potential of anti-inflammatory pharmacological interventions across numerous CVDs, clinical translation remains incomplete, mainly due to (1) yet undefined molecular signaling; (2) challenges of safety and efficacy profile of anti-inflammatory drugs; and (3) difficulties in identifying optimal patient candidates and responders to anti-inflammatory therapeutics, as well as optimal therapeutic windows. Randomized controlled trials demonstrated the safety/efficacy of canakinumab and colchicine in secondary cardiovascular prevention, providing confirmation for the involvement of a specific inflammatory pathway (NLRP3 [NACHT, LRR, and PYD domain-containing protein 3] inflammasome/IL [interleukin]-1β) in atherosclerotic CVD. Colchicine was recently approved by the US Food and Drug Administration for this indication. Diverse anti-inflammatory drugs targeting distinct inflammatory pathways are widely used for the management of other CVDs including myocarditis and pericarditis. Ongoing research efforts are directed to implementing anti-inflammatory therapeutic strategies across a growing number of CVDs, through repurposing of available anti-inflammatory drugs and development of novel anti-inflammatory compounds, which are herein concisely discussed. This review also summarizes the main characteristics and findings of completed and upcoming randomized controlled trials directly targeting inflammation in CVDs, and discusses major challenges and future perspectives in the exciting and constantly expanding landscape of cardioimmunology.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Italy
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
36
|
Tengbom J, Humoud R, Kontidou E, Jiao T, Yang J, Hedin U, Zhou Z, Jurga J, Collado A, Mahdi A, Pernow J. Red blood cells from patients with ST-elevation myocardial infarction and elevated C-reactive protein levels induce endothelial dysfunction. Am J Physiol Heart Circ Physiol 2024; 327:H1431-H1441. [PMID: 39392478 DOI: 10.1152/ajpheart.00443.2024] [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: 07/01/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Endothelial dysfunction is an early consequence of vascular inflammation and a driver of coronary atherosclerotic disease leading to myocardial infarction. The red blood cells (RBCs) mediate endothelial dysfunction in patients at cardiovascular risk, but their role in patients with acute myocardial infarction is unknown. This study aimed to investigate if RBCs from patients with ST-elevation myocardial infarction (STEMI) induced endothelial dysfunction and the role of systemic inflammation in this effect. RBCs from patients with STEMI and aged-matched healthy controls were coincubated with rat aortic segments for 18 h followed by evaluation of endothelium-dependent (EDR) and endothelium-independent relaxation (EIDR). RBCs and aortic segments were also analyzed for arginase and oxidative stress. The patients were divided into groups depending on C-reactive protein (CRP) levels at admission. RBCs from patients with STEMI and CRP levels ≥2 mg/L induced impairment of EDR, but not EIDR, compared with RBCs from STEMI and CRP <2 mg/L and healthy controls. Aortic expression of arginase 1 was increased following incubation with RBCs from patients with STEMI and CRP ≥2, and arginase inhibition prevented the RBC-induced endothelial dysfunction. RBCs from patients with STEMI and CRP ≥2 had increased reactive oxygen species compared with RBCs from patients with CRP <2 and healthy controls. Vascular inhibition of NADPH oxidases and increased dismutation of superoxide improved EDR. RBCs from patients with STEMI and low-grade inflammation induce endothelial dysfunction through a mechanism involving arginase 1 as well as increased RBC and vascular superoxide by NADPH oxidases.NEW & NOTEWORTHY Red blood cells from patients with STEMI and systemic inflammation induce endothelial dysfunction ex vivo. The RBC-induced endothelial dysfunction is mediated through increased arginase 1 and a shift in the redox balance toward oxidative stress. Inhibition of arginase or free radicals attenuates the impairment of endothelial function. The study suggests that red blood cells deserve attention as a key player in systemic inflammation and STEMI.
Collapse
Affiliation(s)
- John Tengbom
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rawan Humoud
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eftychia Kontidou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tong Jiao
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Juliane Jurga
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Aida Collado
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ali Mahdi
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
37
|
Gavilán-Carrera B, Aguilera-Fernández V, Amaro-Gahete FJ, Rosales-Castillo A, Soriano-Maldonado A, Vargas-Hitos JA. Association of the Mediterranean diet with arterial stiffness, inflammation, and medication use in women with systemic lupus erythematosus: An exploratory study. J Nutr Biochem 2024; 134:109759. [PMID: 39276943 DOI: 10.1016/j.jnutbio.2024.109759] [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: 12/01/2023] [Revised: 07/23/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Patients with systemic lupus erythematosus (SLE) face increased cardiovascular risk not fully explained by traditional cardiovascular risk factors. Arterial stiffness, inflammation and disease-related therapies may be contributors to augmented cardiovascular risk, whereas healthy dietary habits could help in their management. The aim of the present study was to analyze the association of the adherence to the Mediterranean Diet with arterial stiffness, inflammation, and disease-related medication in women with SLE. A total of 76 women with SLE were included in this cross-sectional exploratory study. The adherence to the Mediterranean Diet was assessed using the Mediterranean Diet Score. Arterial stiffness was measured through pulse wave velocity (PWV). Inflammatory profile was evaluated through high-sensitivity C-reactive protein (hsCRP). The use (yes / no) and doses (mg /day and cumulative dose over the last 3 years) of corticosteroids and immunosuppressants were also registered. No association of the overall adherence to the Mediterranean Diet with PWV, hsCRP or medication use was found (all P>.05). Lower intake of full dairy products was related to greater odds of corticosteroids use (odds=1.72; P=.004), and both higher current (β=0.29; P=.024) and cumulative (β=0.21; P=.040) doses. Lower intake of red wine was associated with lower odds of immunosuppressants use (odds=0.63; P=.008). No association of the adherence to the Mediterranean Diet with arterial stiffness, inflammation or disease-related medication was observed in women with SLE with mild disease activity. However, higher dairy products and lower red wine consumption were related to lower use of disease-related medication. Future intervention studies are needed to better understand how nutritional education promoting Mediterranean Diet food groups can complement conventional SLE treatments.
Collapse
Affiliation(s)
- Blanca Gavilán-Carrera
- Department of Internal Medicine, Hospital Universitario Virgen de las Nieves, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; PA-HELP ``Physical Activity for HEaLth Promotion'' Research Group, University of Granada, Granada, Spain; Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain.
| | | | - Francisco J Amaro-Gahete
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Granada, Spain
| | | | - Alberto Soriano-Maldonado
- Department of Education, Faculty of Education Sciences, University of Almería, Almería, Spain; SPORT Research Group (CTS-1024), CIBIS (Centro de Investigación para el Bienestar y la Inclusión Social) Research Center, University of Almería, Almería, Spain
| | - José Antonio Vargas-Hitos
- Department of Internal Medicine, Hospital Universitario Virgen de las Nieves, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
38
|
France-Ratcliffe M, Harrison SL, Verma LA, Abdul-Rahim AH, McCallum L, Young CA, McDowell G, Buckley BJ. Vitamin D and cardiovascular outcomes in multiple sclerosis. Mult Scler Relat Disord 2024; 92:106155. [PMID: 39522463 DOI: 10.1016/j.msard.2024.106155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Vitamin D (25(OH)D) deficiency is linked to increased cardiovascular disease (CVD) risk in the general population, but its implications for people with multiple sclerosis (pwMS) remain unexplored. This study aimed to evaluate the association of 25(OH)D with long-term CVD outcomes in pwMS and the impact of vitamin D supplementation. METHODS This observational cohort study analysed anonymised medical records from 70 healthcare organisations following pwMS for 5-years (2019-2024). PwMS and deficient or inadequate 25(OH)D levels were 1:1 propensity-score matched with pwMS and adequate 25(OH)D levels, for demographics, comorbidities, and cardiovascular care. Cox proportional hazard models analysed the incidence of all-cause mortality, stroke, acute myocardial infarction, heart failure, angina, atrial fibrillation/flutter, and a composite measure of major adverse cardiovascular events (MACE). Propensity-matched pwMS who had deficient or inadequate 25(OH)D levels taking cholecalciferol were compared to pwMS and adequate 25(OH)D levels (not taking supplementation). RESULTS Amongst 74,372 pwMS, 9 % had deficient 25(OH)D levels, 18 % inadequate, and 73 % adequate. Deficient, or inadequate 25(OH)D levels were associated with an increased rate of MACE (HR, 1.32 [95 % CI: 1.19, 1.46], HR, 1.29 [95 % CI: 1.20, 1.40], respectively) compared to those with adequate levels. Cholecalciferol supplementation in pwMS and deficient or inadequate 25(OH)D levels did not alleviate the higher CVD rate (HR, 1.39 [95 % CI: 1.21,1.60], HR, 1.31 [95 % CI: 1.17, 1.47], respectively) in comparison to those with adequate 25(OH)D levels taking no vitamin D supplementation. CONCLUSIONS Deficient or inadequate 25(OH)D levels in pwMS were associated with an increased rate of MACE, which may not be mitigated by vitamin D supplementation.
Collapse
Affiliation(s)
- Madeleine France-Ratcliffe
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Cardiovascular Health Sciences, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5UX, United Kingdom.
| | - Stephanie L Harrison
- Registry of Senior Australians, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Leona A Verma
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences, Liverpool, L3 3AF, United Kingdom; Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Azmil H Abdul-Rahim
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK; Stroke Division, Department of Medicine for Older People, Whiston Hospital, Mersey and West Lancashire Teaching Hospitals NHS Trust, Prescot, UK
| | - Linsay McCallum
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Carolyn A Young
- Molecular and Integrative Biology, Institute of Systems, University of Liverpool, Liverpool, United Kingdom; Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Garry McDowell
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences, Liverpool, L3 3AF, United Kingdom
| | - Benjamin Jr Buckley
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Cardiovascular Health Sciences, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5UX, United Kingdom
| |
Collapse
|
39
|
Deng Q, Zhang Y, Guan X, Wang C, Guo H. Association of healthy lifestyles with risk of all-cause and cause-specific mortality among individuals with metabolic dysfunction-associated steatotic liver disease: results from the DFTJ cohort. Ann Med 2024; 56:2398724. [PMID: 39247937 PMCID: PMC11385647 DOI: 10.1080/07853890.2024.2398724] [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: 10/08/2023] [Revised: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 09/10/2024] Open
Abstract
AIM To examine the associations of healthy lifestyles with risk of all-cause and cause-specific mortality among adults with metabolic dysfunction-associated steatotic liver disease (MASLD), and whether the association was mediated by systemic immune-inflammatory biomarkers (SIIBs). METHODS The study included 10,347 subjects with MASLD, who were enrolled in the Dongfeng-Tongji cohort study. The healthy lifestyles referred to non-smoking, being physically active (≥7.5 metabolic equivalents-hours/week), low-risk alcohol consumption (1-14 g/day for women and 1-28 g/day for men), and optimal sleep duration (≥6 to ≤8 h/day). Cox proportional hazard models were used to examine the relationship between each lifestyle and SIIBs with the risk of all-cause and cause-specific mortality. A mediation analysis was conducted to investigate the role of SIIBs on the association between healthy lifestyles and mortality. RESULTS There were 418 MASLD subjects dead till the follow-up of 2018, including 259 deaths from cardiovascular disease (CVD). Compared to MASLD participants with 0-1 healthy lifestyle score (HLS), those with 3-4 HLS had the lowest risk of all-cause mortality [hazard ratio (HR), 0.46; 95% CI, (0.36-0.60)], and CVD mortality [HR (95%CI), 0.41 (0.29-0.58)]. Mediation analyses indicated that SIIBs mediated the association between healthy lifestyles and mortality, with proportions ranging from 2.5% to 6.1%. CONCLUSIONS These findings suggest that adherence to healthy lifestyles can significantly reduce mortality for MASLD patients, and the decreased SIIBs may partially explain the protection mechanism of healthy lifestyles.
Collapse
Affiliation(s)
- Qilin Deng
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingchen Zhang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Guan
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenming Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Mazhar F, Faucon AL, Fu EL, Szummer KE, Mathisen J, Gerward S, Reuter SB, Marx N, Mehran R, Carrero JJ. Systemic inflammation and health outcomes in patients receiving treatment for atherosclerotic cardiovascular disease. Eur Heart J 2024; 45:4719-4730. [PMID: 39211962 PMCID: PMC11578643 DOI: 10.1093/eurheartj/ehae557] [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: 03/22/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The burden and outcomes of inflammation in patients with atherosclerotic cardiovascular disease (ASCVD) are not well defined beyond the controlled settings of trials and research cohorts. METHODS This was an observational study of ASCVD adults undergoing C-reactive protein testing in Stockholm's healthcare (2007-21). After excluding C-reactive protein tests associated with acute illness or medications/conditions that bias C-reactive protein interpretation, systemic inflammation was evaluated over a 3-month ascertainment window. Determinants of C-reactive protein ≥ 2 mg/L were explored with logistic regression. C-reactive protein categories were compared via negative-binomial/Cox regression for subsequent healthcare resource utilization and occurrence of major adverse cardiovascular events, heart failure hospitalization, and death. RESULTS A total of 84 399 ASCVD adults were included (46% female, mean age 71 years, 59% with C-reactive protein ≥ 2 mg/L). Female sex, older age, lower kidney function, albuminuria, diabetes, hypertension, and recent anaemia were associated with higher odds of C-reactive protein ≥ 2 mg/L. The use of renin-angiotensin system inhibitors, antiplatelets, and lipid-lowering therapy was associated with lower odds. Over a median of 6.4 years, compared with C-reactive protein < 2 mg/L, patients with C-reactive protein ≥ 2 mg/L had higher rates of hospitalizations, days spent in hospital, outpatient consultations, and dispensed medications (P < .05 for all). They also had a higher rate of major adverse cardiovascular events [hazard ratio (HR) 1.30; 95% confidence interval (CI) 1.27-1.33], heart failure (HR 1.24; 95% CI 1.20-1.30), and death (HR 1.35; 95% CI 1.31-1.39). Results were consistent across subgroups and granular C-reactive protein categories and robust to the exclusion of extreme C-reactive protein values or early events. CONCLUSIONS Three in five adults with ASCVD have systemic inflammation, which is associated with excess healthcare resource utilization and increased rates of cardiovascular events and death.
Collapse
Affiliation(s)
- Faizan Mazhar
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Anne-Laure Faucon
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Edouard L Fu
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karolina E Szummer
- Department of Cardiology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | | | | | - Nikolaus Marx
- Department of Internal Medicine I, RWTH Aachen University, Aachen, Germany
| | - Roxana Mehran
- Mount Sinai School of Medicine, Mount Sinai Health System, New York City, NY, USA
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Danderyd, Sweden
| |
Collapse
|
41
|
Sallam M, Hassan H, Connolly D, Rahman MS. Commencement of Atorvastatin and Ezetimibe Immediately in Patients Presenting with Acute Coronary Syndrome. Eur Cardiol 2024; 19:e22. [PMID: 39588251 PMCID: PMC11588103 DOI: 10.15420/ecr.2024.06] [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: 01/30/2024] [Accepted: 08/12/2024] [Indexed: 11/27/2024] Open
Abstract
Lipids are implicated in the development of coronary atherosclerosis. Achieving a significant reduction in lipid levels remains a crucial aspect of secondary prevention following an acute coronary syndrome event. Novel lipid-lowering therapies now provide clinicians with a variety of therapeutic strategies to choose from and tailor to individual patient needs. This review focuses on evidence supporting the importance of early and intensive lipid-lowering therapy use in patients presenting with acute coronary syndrome, specifically addressing data relating to atorvastatin and ezetimibe use in this high-risk cohort of patients.
Collapse
Affiliation(s)
- Mohammed Sallam
- Birmingham City Hospital, Sandwell and West Birmingham Hospitals NHS Trust Birmingham, West Midlands, UK
| | - Hossameldin Hassan
- Birmingham City Hospital, Sandwell and West Birmingham Hospitals NHS Trust Birmingham, West Midlands, UK
| | - Derek Connolly
- Birmingham City Hospital, Sandwell and West Birmingham Hospitals NHS Trust Birmingham, West Midlands, UK
| | - Mohammed Shamim Rahman
- Birmingham City Hospital, Sandwell and West Birmingham Hospitals NHS Trust Birmingham, West Midlands, UK
| |
Collapse
|
42
|
Chen T, Yang Y. Immunologic and inflammatory pathogenesis of chronic coronary syndromes: A review. Medicine (Baltimore) 2024; 103:e40354. [PMID: 39496055 PMCID: PMC11537619 DOI: 10.1097/md.0000000000040354] [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: 09/17/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
Chronic coronary syndrome (CCS) is a major cause of progression to acute coronary syndrome. Due to its insidious onset and complex etiology, this condition is often underestimated and insufficiently recognized, and traditional interventions for risk factors do not effectively control the disease progression. Current research suggests that immune and inflammatory pathways contribute to atherosclerosis and its clinical complications, thereby triggering the progression of CCS to acute coronary syndrome. This article primarily reviews the possible mechanisms of immune and inflammatory responses in CCS, with the aim of providing references for the diagnosis, treatment, and prevention of CCS.
Collapse
Affiliation(s)
- Tingting Chen
- Dali University School of Clinical Medicine, Yunnan, China
| | - Ying Yang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Yunnan, China
| |
Collapse
|
43
|
Yang Q, Liu J, Zhang T, Zhu T, Yao S, Wang R, Wang W, Dilimulati H, Ge J, An S. Exploring shared biomarkers and shared pathways in insomnia and atherosclerosis using integrated bioinformatics analysis. Front Mol Neurosci 2024; 17:1477903. [PMID: 39439987 PMCID: PMC11493776 DOI: 10.3389/fnmol.2024.1477903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Insomnia (ISM) is one of the non-traditional drivers of atherosclerosis (AS) and an important risk factor for AS-related cardiovascular disease. Our study aimed to explore the shared pathways and diagnostic biomarkers of ISM-related AS using integrated bioinformatics analysis. Methods We download the datasets from the Gene Expression Omnibus database and the GeneCards database. Weighted gene co-expression network analysis and gene differential expression analysis were applied to screen the AS-related gene set. The shared genes of ISM and AS were obtained by intersecting with ISM-related genes. Subsequently, candidate diagnostic biomarkers were identified by constructing protein-protein interaction networks and machine learning algorithms, and a nomogram was constructed. Moreover, to explore potential mechanisms, a comprehensive analysis of shared genes was carried out, including enrichment analysis, protein interactions, immune cell infiltration, and single-cell sequencing analysis. Results We successfully screened 61 genes shared by ISM and AS, of which 3 genes (IL10RA, CCR1, and SPI1) were identified as diagnostic biomarkers. A nomogram with excellent predictive value was constructed (the area under curve of the model constructed by the biomarkers was 0.931, and the validation set was 0.745). In addition, the shared genes were mainly enriched in immune and inflammatory response regulation pathways. The biomarkers were associated with a variety of immune cells, especially myeloid immune cells. Conclusion We constructed a diagnostic nomogram based on IL10RA, CCR1, and SPI1 and explored the inflammatory-immune mechanisms, which indicated new insights for early diagnosis and treatment of ISM-related AS.
Collapse
Affiliation(s)
- Qichong Yang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Juncheng Liu
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
- Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhang
- Center for Clinical Single-Cell Biomedicine, Henan Province People’s Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhu
- Department of Cardiopulmonary Functions Test, Henan Province People’s Hospital, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Siyu Yao
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Rongzi Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Wenjuan Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Haliminai Dilimulati
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Songtao An
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| |
Collapse
|
44
|
Pellegrini V, La Grotta R, Carreras F, Giuliani A, Sabbatinelli J, Olivieri F, Berra CC, Ceriello A, Prattichizzo F. Inflammatory Trajectory of Type 2 Diabetes: Novel Opportunities for Early and Late Treatment. Cells 2024; 13:1662. [PMID: 39404426 PMCID: PMC11476093 DOI: 10.3390/cells13191662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Low-grade inflammation (LGI) represents a key driver of type 2 diabetes (T2D) and its associated cardiovascular diseases (CVDs). Indeed, inflammatory markers such as hs-CRP and IL-6 predict the development of T2D and its complications, suggesting that LGI already increases before T2D diagnosis and remains elevated even after treatment. Overnutrition, unhealthy diets, physical inactivity, obesity, and aging are all recognized triggers of LGI, promoting insulin resistance and sustaining the pathogenesis of T2D. Once developed, and even before frank appearance, people with T2D undergo a pathological metabolic remodeling, with an alteration of multiple CVD risk factors, i.e., glycemia, lipids, blood pressure, and renal function. In turn, such variables foster a range of inflammatory pathways and mechanisms, e.g., immune cell stimulation, the accrual of senescent cells, long-lasting epigenetic changes, and trained immunity, which are held to chronically fuel LGI at the systemic and tissue levels. Targeting of CVD risk factors partially ameliorates LGI. However, some long-lasting inflammatory pathways are unaffected by common therapies, and LGI burden is still increased in many T2D patients, a phenomenon possibly underlying the residual inflammatory risk (i.e., having hs-CRP > 2 mg/dL despite optimal LDL cholesterol control). On the other hand, selected disease-modifying drugs, e.g., GLP-1RA, seem to also act on the pathogenesis of T2D, curbing the inflammatory trajectory of the disease and possibly preventing it if introduced early. In addition, selected trials demonstrated the potential of canonical anti-inflammatory therapies in reducing the rate of CVDs in patients with this condition or at high risk for it, many of whom had T2D. Since colchicine, an inhibitor of immune cell activation, is now approved for the prevention of CVDs, it might be worth exploring a possible therapeutic paradigm to identify subjects with T2D and an increased LGI burden to treat them with this drug. Upcoming studies will reveal whether disease-modifying drugs reverse early T2D by suppressing sources of LGI and whether colchicine has a broad benefit in people with this condition.
Collapse
Affiliation(s)
- Valeria Pellegrini
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Rosalba La Grotta
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Francesca Carreras
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Angelica Giuliani
- Cardiac Rehabilitation Unit of Bari Institute, Istituti Clinici Scientifici Maugeri IRCCS, 70124 Bari, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60127 Ancona, Italy; (J.S.); (F.O.)
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60127 Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60127 Ancona, Italy; (J.S.); (F.O.)
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60127 Ancona, Italy
| | | | - Antonio Ceriello
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | | |
Collapse
|
45
|
Mang G, Chen J, Sun P, Ma R, Du J, Wang X, Cui J, Yang M, Tong Z, Yan X, Wang D, Xie H, Chen Y, Yang Q, Kong Y, Jin J, Wu J, Zhang M, Yu B. Von Willebrand factor exacerbates heart failure through formation of neutrophil extracellular traps. Eur Heart J 2024; 45:3853-3867. [PMID: 39165142 DOI: 10.1093/eurheartj/ehae517] [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: 05/22/2023] [Revised: 10/14/2023] [Accepted: 07/28/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND AND AIMS Heart failure (HF) is a leading cause of mortality worldwide and characterized by significant co-morbidities and dismal prognosis. Neutrophil extracellular traps (NETs) aggravate inflammation in various cardiovascular diseases; however, their function and mechanism of action in HF pathogenesis remain underexplored. This study aimed to investigate the involvement of a novel VWF-SLC44A2-NET axis in HF progression. METHODS NET levels were examined in patients with HF and mouse models of transverse aortic constriction (TAC) HF. PAD4 knockout mice and NET inhibitors (GSK-484, DNase I, NEi) were used to evaluate the role of NETs in HF. RNA sequencing was used to investigate the downstream mechanisms. Recombinant human ADAMTS13 (rhADAMTS13), ADAMTS13, and SLC44A2 knockouts were used to identify novel upstream factors of NETs. RESULTS Elevated NET levels were observed in patients with HF and TAC mouse models of HF. PAD4 knockout and NET inhibitors improved the cardiac function. Mechanistically, NETs induced mitochondrial dysfunction in cardiomyocytes, inhibiting mitochondrial biogenesis via the NE-TLR4-mediated suppression of PGC-1α. Furthermore, VWF/ADAMTS13 regulated NET formation via SLC44A2. Additionally, sacubitril/valsartan amplifies the cardioprotective effects of the VWF-SLC44A2-NET axis blockade. CONCLUSIONS This study established the role of a novel VWF-SLC44A2-NET axis in regulating mitochondrial homeostasis and function, leading to cardiac apoptosis and contributing to HF pathogenesis. Targeting this axis may offer a potential therapeutic approach for HF treatment.
Collapse
Affiliation(s)
- Ge Mang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100013, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Jianfeng Chen
- Experimental Animal Centre, Harbin Medical University, Ministry of Education, No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Ping Sun
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Ruishuang Ma
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Jingwen Du
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Xiaoqi Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Jingxuan Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Mian Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Zhonghua Tong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Xiangyu Yan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Dongni Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Huiqi Xie
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Yujia Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Qiannan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Yingjin Kong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Jiaqi Jin
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St., Beijing 100053, China
| | - Jian Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin 150086, Heilongjiang, China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin 150086, Heilongjiang, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), No. 246 Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin 150086, Heilongjiang, China
| |
Collapse
|
46
|
Li J, Yan K, Zhu P, Tang X, Yang Y, Gao R, Yuan J, Zhao X. Prognostic value of Glasgow Prognostic Score and its modified scores on 5-year outcome in patients with coronary heart disease undergoing percutaneous coronary intervention. Heliyon 2024; 10:e37317. [PMID: 39309905 PMCID: PMC11415646 DOI: 10.1016/j.heliyon.2024.e37317] [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: 03/07/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/25/2024] Open
Abstract
Background Glasgow Prognostic Score (GPS) and its modified counterparts, including the modified GPS (mGPS) and hsCRP-modified GPS (hs-mGPS), are widely used inflammatory indices in clinical settings. Inflammation has gained increased attention in the context of coronary heart disease (CHD); however, its long-term predictive value in patients with CHD remains uncertain. Objective This study aimed to assess the predictive values of GPS, mGPS, and hs-mGPS for long-term survival in patients following percutaneous coronary intervention (PCI). Methods Consecutive 10,724 PCI patients were enrolled in 2013. The primary endpoint was 5-year all-cause death. Results This study included 8,909 patients. Individuals with high GPS, mGPS, and hs-mGPS scores exhibited a significantly higher risk of all-cause death compared to those with low scores (all P < 0.05). All three indices (GPS, mGPS, and hs-mGPS) demonstrated predictive values for all-cause death, albeit with relatively low area under the curve values of 0.534, 0.522, and 0.545, respectively. Furthermore, we refined the hs-mGPS using cutoffs (hsCRP at 2 mg/L and albumin at 40 g/L) which are better suited for these patients, to establish the CHD-hs-mGPS. This modification significantly improved the prediction of all-cause death, outperformed the mGPS and demonstrated numerical superiority over both the GPS and hs-mGPS. Notably, only CHD-hs-mGPS exhibited a predictive value for both the ACS and non-ACS subgroups. Conclusion In patients with CHD who underwent PCI, GPS, mGPS, and hs-mGPS demonstrated significant long-term predictive values for all-cause death. Our parameter-adjusted score, the CHD-hs-mGPS, is applicable to a broad population and moderately enhances the predictive accuracy, facilitating the early identification of patients at high risk of long-term death.
Collapse
Affiliation(s)
- Jiawen Li
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kailun Yan
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinqing Yuan
- Corresponding author. National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xueyan Zhao
- Corresponding author. National Clinical Research Center for Cardiovascular Diseases and State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
47
|
Zhang J, Liu J, Ye M, Zhang M, Yao F, Cheng Y. Incidence and risk factors associated with atrioventricular block in the general population: the atherosclerosis risk in communities study and Cardiovascular Health Study. BMC Cardiovasc Disord 2024; 24:509. [PMID: 39327574 PMCID: PMC11428875 DOI: 10.1186/s12872-024-04163-9] [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: 11/13/2023] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
OBJECTIVES To identify risk factors correlated with atrioventricular block (AVB) in the general population. METHODS Participants in the Atherosclerosis Risk in Communities study (ARIC) and the Cardiovascular Health study (CHS) were enrolled. The presence of AVB was confirmed at an electrocardiogram (ECG) reading center using Minnesota ECG Classification. Cox proportional hazards models were performed to investigate potential risk factors of AVB, after adjustment for age, sex, race and traditional cardiovascular risk factors. RESULTS During the 17 years of follow-up, a total of 731 high-degree AVB cases were identified. Age and sex-standardized rate of AVB was 2.79 and 2.35 per 1000 person-years in the white and the black population, respectively. With the increase of the geriatric population, the incidence of high-degree AVB will increase from 378,816 in 2020 to 535,076 in 2060, and most increment would occur among the elderly. Older age, male sex, the white race, overweight, comorbidities, declined forced vital capacity (FVC), elevated inflammation biomarkers, left bundle branch block and bifascicular block were independently associated with the incidence of high-degree AVB. CONCLUSION To conclude, older age, male sex, white population, overweight, combined diabetes or chronic kidney disease, impaired FVC, elevated inflammation biomarkers, left bundle branch block and bifascicular block were independent predictors for high-degree AVB. The next 40 years would witness a dramatic increase in the incidence of high-degree AVB.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Jia Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Min Ye
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fengjuan Yao
- Department of Medical Ultrasonics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- Key Laboratory of Assisted Circulation, NHC, Guangzhou, China.
| | - Yunjiu Cheng
- Department of Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Yu M, Yang Y, Dong SL, Zhao C, Yang F, Yuan YF, Liao YH, He SL, Liu K, Wei F, Jia HB, Yu B, Cheng X. Effect of Colchicine on Coronary Plaque Stability in Acute Coronary Syndrome as Assessed by Optical Coherence Tomography: The COLOCT Randomized Clinical Trial. Circulation 2024; 150:981-993. [PMID: 39166327 DOI: 10.1161/circulationaha.124.069808] [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: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Colchicine has been approved to reduce cardiovascular risk in patients with coronary heart disease on the basis of its potential benefits demonstrated in the COLCOT (Colchicine Cardiovascular Outcomes Trial) and LoDoCo2 (Low-Dose Colchicine 2) studies. Nevertheless, there are limited data available about the specific impact of colchicine on coronary plaques. METHODS This was a prospective, single-center, randomized, double-blind clinical trial. From May 3, 2021, until August 31, 2022, a total of 128 patients with acute coronary syndrome aged 18 to 80 years with lipid-rich plaque (lipid pool arc >90°) detected by optical coherence tomography were included. The subjects were randomly assigned in a 1:1 ratio to receive either colchicine (0.5 mg once daily) or placebo for 12 months. The primary end point was the change in the minimal fibrous cap thickness from baseline to the 12-month follow-up. RESULTS Among 128 patients, 52 in the colchicine group and 52 in the placebo group completed the study. The mean age of the 128 patients was 58.0±9.8 years, and 25.0% were female. Compared with placebo, colchicine therapy significantly increased the minimal fibrous cap thickness (51.9 [95% CI, 32.8 to 71.0] μm versus 87.2 [95% CI, 69.9 to 104.5] μm; difference, 34.2 [95% CI, 9.7 to 58.6] μm; P=0.006), and reduced average lipid arc (-25.2° [95% CI, -30.6° to -19.9°] versus -35.7° [95% CI, -40.5° to -30.8°]; difference, -10.5° [95% CI, -17.7° to -3.4°]; P=0.004), mean angular extension of macrophages (-8.9° [95% CI, -13.3° to -4.6°] versus -14.0° [95% CI, -18.0° to -10.0°]; difference, -6.0° [95% CI, -11.8° to -0.2°]; P=0.044), high-sensitivity C-reactive protein level (geometric mean ratio, 0.6 [95% CI, 0.4 to 1.0] versus 0.3 [95% CI, 0.2 to 0.5]; difference, 0.5 [95% CI, 0.3 to 1.0]; P=0.046), interleukin-6 level (geometric mean ratio, 0.8 [95% CI, 0.6 to 1.1] versus 0.5 [95% CI, 0.4 to 0.7]; difference, 0.6 [95% CI, 0.4 to 0.9]; P=0.025), and myeloperoxidase level (geometric mean ratio, 1.0 [95% CI, 0.8 to 1.2] versus 0.8 [95% CI, 0.7 to 0.9]; difference, 0.8 [95% CI, 0.6 to 1.0]; P=0.047). CONCLUSIONS Our findings suggested that colchicine resulted in favorable effects on coronary plaque stabilization at optical coherence tomography in patients with acute coronary syndrome. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04848857.
Collapse
Affiliation(s)
- Miao Yu
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Lai Dong
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhao
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Fen Yang
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Fan Yuan
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Lin He
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Wei
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Bo Jia
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Bo Yu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Xiang Cheng
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [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: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
50
|
Chen X, Zhang Z, Qiao G, Sun Z, Lu W. Immune and inflammatory insights in atherosclerosis: development of a risk prediction model through single-cell and bulk transcriptomic analyses. Front Immunol 2024; 15:1448662. [PMID: 39364414 PMCID: PMC11446800 DOI: 10.3389/fimmu.2024.1448662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/29/2024] [Indexed: 10/05/2024] Open
Abstract
Background Investigation into the immune heterogeneity linked with atherosclerosis remains understudied. This knowledge gap hinders the creation of a robust theoretical framework essential for devising personalized immunotherapies aimed at combating this disease. Methods Single-cell RNA sequencing (scRNA-seq) analysis was employed to delineate the immune cell-type landscape within atherosclerotic plaques, followed by assessments of cell-cell interactions and phenotype characteristics using scRNA-seq datasets. Subsequently, pseudotime trajectory analysis was utilized to elucidate the heterogeneity in cell fate and differentiation among macrophages. Through integrated approaches, including single-cell sequencing, Weighted Gene Co-expression Network Analysis (WGCNA), and machine learning techniques, we identified hallmark genes. A risk score model and a corresponding nomogram were developed and validated using these genes, confirmed through Receiver Operating Characteristic (ROC) curve analysis. Additionally, enrichment and immune characteristic analyses were conducted based on the risk score model. The model's applicability was further corroborated by in vitro and in vivo validation of specific genes implicated in atherosclerosis. Result This comprehensive scRNA-seq analysis has shed new light on the intricate immune landscape and the role of macrophages in atherosclerotic plaques. The presence of diverse immune cell populations, with a particularly enriched macrophage population, was highlighted by the results. Macrophage heterogeneity was intricately characterized, revealing four distinct subtypes with varying functional attributes that underscore their complex roles in atherosclerotic pathology. Intercellular communication analysis revealed robust macrophage interactions with multiple cell types and detailed pathways differing between proximal adjacent and atherosclerotic core groups. Furthermore, pseudotime trajectories charted the developmental course of macrophage subpopulations, offering insights into their differentiation fates within the plaque microenvironment. The use of machine learning identified potential diagnostic markers, culminating in the identification of RNASE1 and CD14. The risk score model based on these biomarkers exhibited high accuracy in diagnosing atherosclerosis. Immune characteristic analysis validated the risk score model's efficacy in defining patient profiles, distinguishing high-risk individuals with pronounced immune cell activities. Finally, experimental validation affirmed RNASE1's involvement in atherosclerotic progression, suggesting its potential as a therapeutic target. Conclusion Our findings have advanced our understanding of atherosclerosis immunopathology and paved the way for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xiaosan Chen
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai
Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | | | | | | | | |
Collapse
|