51
|
Naveed MA, Neppala S, Rehan MO, Azeem B, Chigurupati HD, Ali A, Iqbal R, Mubeen M, Ahmed M, Rana J, Dani SS. Longitudinal Trends in Heart Failure Mortality Linked to Coronary Artery Disease Among Adults 65 years and older. Am J Med Sci 2025:S0002-9629(25)00991-7. [PMID: 40254220 DOI: 10.1016/j.amjms.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Heart failure (HF) in patients with coronary artery disease (CAD) is a leading cause of mortality among older adults in the United States. This study examines trends in HF with CAD-related mortality among adults aged 65 and older. METHODS A retrospective analysis was performed using the CDC WONDER database death certificates from 1999 to 2020. Age-adjusted mortality rates (AAMRs), annual percent change (APC), and average annual percentage change (AAPC) were calculated per 100,000 persons, stratified by year, sex, race/ethnicity, and geographical region. RESULTS HF associated with CAD led to 1,597,451 deaths among adults > 65, primarily occurring in medical facilities (37.1%). The AAMR for HF with CAD decreased from 241.7 in 1999 to 156.2 in 2020 (AAPC: -2.23, p < 0.001), which was significant from 1999 to 2014. Men had higher AAMRs than women (227.4 vs. 137.1), with women's rates declining more significantly (AAPC: -3.23, p < 0.001). White adults had the highest AAMRs (183.0), while Asians/Pacific Islanders (81.6) recorded the lowest. Geographically, AAMRs varied, from 92.1 in Hawaii to 257.3 in West Virginia, with the Midwest showing the highest mortality (191.0). Nonmetropolitan areas exhibited higher AAMRs than metropolitan areas (202.6 vs. 166.1) CONCLUSION: Our study reveals striking disparities in HF-related mortality among adults aged 65 years and older in the United States. While AAMRs decreased overall from 1999 to 2014, they have reached an inflection point since 2019, indicating rising mortality rates. Persistent inequalities underscore the critical need for targeted public health interventions to address these issues.
Collapse
Affiliation(s)
- Muhammad Abdullah Naveed
- Department of Cardiology, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Sivaram Neppala
- Department of Cardiology, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Muhammad Omer Rehan
- Department of Cardiology, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Bazil Azeem
- Department of Cardiology, Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, Pakistan
| | | | - Ahila Ali
- Department of Cardiology, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Rabia Iqbal
- Department of Cardiology, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Manahil Mubeen
- Department of Cardiology, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Mushood Ahmed
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Jamal Rana
- Department of Cardiology, The Permanente Medical Group, Oakland, California, USA
| | - Sourbha S Dani
- Department of Cardiology, Lahey Hospital and Medical Center, Burlington, MA
| |
Collapse
|
52
|
Generali M, Kehl D, Meier D, Zorndt D, Atrott K, Saito H, Emmert MY, Hoerstrup SP. Generation and purification of iPSC-derived cardiomyocytes for clinical applications. Stem Cell Res Ther 2025; 16:189. [PMID: 40251664 PMCID: PMC12008852 DOI: 10.1186/s13287-025-04319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Over the past decade, the field of cell therapy has rapidly expanded with the aim to replace and repair damaged cells and/or tissue. Depending on the disease many different cell types can be used as part of such a therapy. Here we focused on the potential treatment of myocardial infarction, where currently available treatment options are not able to regenerate the loss of healthy heart tissue. METHOD We generated good manufacturing practice (GMP)-compatible cardiomyocytes (iCMs) from transgene- and xenofree induced pluripotent stem cells (iPSCs) that can be seamless adapted for clinical applications. Further protocols were established for replating and freezing/thawing iCMs under xenofree conditions. RESULTS iCMs showed a cardiac phenotype, with the expression of specific cardiac markers and absence of pluripotency markers at RNA and protein level. To ensure a pure iCMs population for in vivo applications, we minimized risks of iPSC contamination using RNA-switch technology to ensure safety. CONCLUSION We describe the generation and further processing of xeno- and transgene-free iCMs. The use of GMP-compliant differentiation protocols ab initio facilitates the clinical translation of this project in later stages.
Collapse
Affiliation(s)
- M Generali
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
| | - D Kehl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Meier
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - D Zorndt
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Atrott
- Center for Surgical Research, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - H Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - M Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - S P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
53
|
Song Y, Wang L, Peng T, Shan L, Wan B, Tang M, Luan Y, Jiang Y, He W. Brain-targeting biomimetic nanozyme enhances neuroprotection in ischemic stroke by remodeling the neurovascular unit. J Control Release 2025:113750. [PMID: 40254137 DOI: 10.1016/j.jconrel.2025.113750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Dysfunction of the neurovascular unit significantly impacts the prognostic outcomes of ischemic stroke. However, effective strategies to comprehensively modulate the neurovascular unit have yet to be developed. In this work, we introduce a brain-targeting biomimetic nanozyme, A@HPB@THSA, designed to mitigate neurovascular unit dysfunction following ischemia/reperfusion. Specifically, aspirin is encapsulated within hollow Prussian blue nanozyme, which is subsequently modified with brain-targeting T7 peptide-conjugated human serum albumin, ultimately forming the composite A@HPB@THSA. The overexpression of transferrin receptors on cerebral vascular endothelial cells, along with compromised blood-brain barrier (BBB) permeability, facilitates the accumulation of A@HPB@THSA at cerebral ischemic lesions. The hollow Prussian blue nanozyme component effectively scavenges reactive oxygen species in ischemia/reperfusion-affected brain cells. While the aspirin component inhibits platelets aggregation and neutrophils infiltration, thereby preventing microvascular "no-reflow" and preserving the integrity of the BBB. In rat models of transient middle cerebral artery occlusion, A@HPB@THSA demonstrated comprehensive modulation of the neurovascular unit, including reduced BBB permeability, promotion of microglia polarization toward an anti-inflammatory phenotype, and enhanced neuronal survival. This work provides a promising platform to reverse dysfunctional neurovascular unit for ischemic stroke treatment.
Collapse
Affiliation(s)
- Yan Song
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Luyao Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tingting Peng
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Lingling Shan
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Bo Wan
- Departent of Pharmacy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Mingtan Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuxia Luan
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yue Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Wenxiu He
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
54
|
Zhang W, Yu X, Lin Y, Wu C, Zhu R, Jiang X, Tao J, Chen Z, He J, Zhang X, Xu J, Zhang M. Acetyl-CoA synthetase 2 alleviates brain injury following cardiac arrest by promoting autophagy in brain microvascular endothelial cells. Cell Mol Life Sci 2025; 82:160. [PMID: 40244361 PMCID: PMC12006639 DOI: 10.1007/s00018-025-05689-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025]
Abstract
INTRODUCTION Brain injury is a common sequela following cardiac arrest (CA), with up to 70% of hospitalized patients dying from it. Brain microvascular endothelial cells (BMVECs) play a crucial role in post-cardiac arrest brain injury (PCABI). However, the effects and mechanisms of targeting BMVEC energy metabolism to mitigate brain injury remain unclear. METHODS We established a mouse model of cardiac arrest by injecting potassium chloride into the right internal jugular vein. Mass spectrometry detected targeted changes in short-chain fatty acids and energy metabolism metabolites in the CA/CPR group compared to the sham group. Mice with overexpressed ACSS2 in BMVECs were created using an AAV-BR1 vector, and ACSS2 knockout mice were generated using the CRE-LOXP system. The oxygen glucose deprivation/re-oxygenation (OGD/R) model was established to investigate the role and mechanisms of ACSS2 in endothelial cells in vitro. RESULTS Metabolomics analysis revealed disrupted cerebral energy metabolism post-CA/CPR, with decreased acetyl-CoA and amino acids. Overexpression of ACSS2 in BMVECs increased acetyl-CoA levels and improved neurological function. Vascular endothelial cell-specific ACSS2 knockout mice exhibited reduced aortic sprouting in vitro. Overexpression of ACSS2 improved endothelial dysfunction following oxygen glucose deprivation/re-oxygenation (OGD/R) and influenced autophagy by interacting with transcription factor EB (TFEB) and modulating the AMP-activated protein kinase α (AMPKα) pathway. CONCLUSION Our study shows that ACSS2 modulates the biological functions of BMVECs by promoting autophagy. Enhancing energy metabolism via ACSS2 may target PCABI treatment development.
Collapse
Affiliation(s)
- Wenbin Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Xin Yu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Yao Lin
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Chenghao Wu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Ruojie Zhu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Xiangkang Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Jiawei Tao
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Ziwei Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Jiantao He
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Xiaodan Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Jiefeng Xu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China.
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| | - Mao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, Hangzhou, China.
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| |
Collapse
|
55
|
Coons JC, Wang A, Latremouille-Viau D, Russ C, Cheng D, Stellhorn R, Dai F, Steffen DR, Zion A, Deeba S, Hines DM. Oral anticoagulant use among Medicare patients newly diagnosed with venous thromboembolism (VTE): Factors associated with treatment status. PLoS One 2025; 20:e0321106. [PMID: 40245061 PMCID: PMC12005561 DOI: 10.1371/journal.pone.0321106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/01/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE This study aimed to describe oral anticoagulant (OAC) use among patients with venous thromboembolism (VTE). MATERIALS AND METHODS This study included Medicare fee-for-service beneficiaries (data from 1/1/2014-12/31/2019) newly diagnosed with VTE. Factors associated with being untreated with OACs in the first month from VTE (vs. OAC-treated), with receiving direct-acting OACs ([DOACs] vs. warfarin), and with extended OAC treatment (>3 months) were assessed using multivariable logistic regressions. RESULTS Overall, 169,928 patients with VTE (50.3% OAC-untreated) were included. Among the 49.7% OAC-treated patients, 74.0% used DOACs and 62.5% had extended OAC treatment. Factors associated with being untreated with OACs in the first month from VTE (odds ratio; 95% confidence interval) included Hispanic ethnicity (vs. White;1.35; 1.29-1.42), having part D low-income subsidy (1.14; 1.07, 1.20), and comorbidities such as cardiovascular diseases. Among the OAC-treated cohort, patients with index VTE diagnosis in the emergency room (vs. outpatient) setting had higher odds of receiving DOAC vs. warfarin; patients with pulmonary embolism diagnosis (vs. deep vein thrombosis) had higher odds of extended OAC treatment. CONCLUSIONS In this study of Medicare patients newly diagnosed with VTE, half of the patients were not treated with OAC in the first month from initial diagnosis. Factors such as Hispanic ethnicity, having low-income subsidy, and comorbidity burden were found to be associated with being untreated with OAC. Among OAC-treated patients, the majority were treated with DOAC vs. warfarin. Interestingly, more than a third of OAC-treated patients were not treated beyond 3 months, which warrants further investigation.
Collapse
Affiliation(s)
- James C. Coons
- Department of Pharmacy, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Aolin Wang
- Analysis Group, Inc., New York, New York, United States of America
| | | | - Cristina Russ
- Pfizer Inc., New York, New York, United States of America
| | - Dong Cheng
- Bristol Myers Squibb, Lawrenceville, New Jersey, United States of America
| | - Robert Stellhorn
- Bristol Myers Squibb, Lawrenceville, New Jersey, United States of America
| | - Feng Dai
- Pfizer Inc., New York, New York, United States of America
| | - David R. Steffen
- Analysis Group, Inc., New York, New York, United States of America
| | - Abigail Zion
- Analysis Group, Inc., Boston, Massachusetts, United States of America
| | - Serina Deeba
- Pfizer Inc., New York, New York, United States of America
| | | |
Collapse
|
56
|
Kuku KO, Shearer JJ, Joo J, Remaley AT, Connelly MA, Bielinski SJ, Roger VL. Cross-sectional evaluation of the metabolic vulnerability index in heart failure populations. BMC Cardiovasc Disord 2025; 25:292. [PMID: 40247156 PMCID: PMC12004792 DOI: 10.1186/s12872-025-04758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The Metabolic Vulnerability Index (MVX) is a novel multi-marker risk score derived from nuclear magnetic resonance (NMR) measures and has shown predictive value for mortality in heart failure. Hence, we aimed to evaluate the distribution of MVX and its clinical correlates within a clinical trial population and a comparable subpopulation of patients with heart failure with reduced ejection fraction and ischemic heart disease within a community cohort. METHODS We studied a clinical trial (2016-2018) and a community cohort (2003-2012), matched based on ejection fraction category and presence of ischemic heart failure. NMR LipoProfile analyses of plasma from both populations provided measures of valine, leucine, isoleucine, citrate, GlycA, and small high-density lipoprotein particles used to compute sex-specific MVX scores. Univariable and multivariable regression models assessed the relationship between MVX (modeled continuously), and selected demographic and clinical covariates. RESULTS Clinical trial patients (N = 101, median age: 63, 93% male, median EF: 28%) were younger and predominantly male compared to the cohort (N = 288, median age: 75, 70% male, median EF: 30%). The median MVX score was lower in the clinical trial (50, 42-61) compared to the cohort (66, 58-73). Male sex and hyperlipidemia were linked to higher MVX scores in the clinical trial, while obesity and NT-proBNP were linked to lower and higher MVX scores, respectively, in the cohort (p <.05). After adjusting for significant covariates from univariable analyses and age in multivariable analyses, only the associations between male sex and MVX scores in the clinical trial, and NT-proBNP levels with MVX in the cohort remained significant. CONCLUSION This study highlights significant differences in MVX distribution and its clinical correlates between a clinical trial and a community cohort despite matched heart failure subtypes. These findings have important implications for interpreting and applying the score in diverse study settings.
Collapse
Affiliation(s)
- Kayode O Kuku
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joseph J Shearer
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jungnam Joo
- Office of Biostatistics Research National Heart, Lung, and Blood Institute, National Institutes, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Suzette J Bielinski
- Division of Epidemiology, Department of Quantitative Health Sciences Mayo Clinic, Rochester, MN, USA
| | - Véronique L Roger
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
57
|
Jakob P, Lansky AJ, Basir MB, Schonning MJ, Falah B, Zhou Z, Batchelor WB, Abu-Much A, Grines CL, O'Neill WW, Stähli BE. Characteristics and Outcomes of Older Patients Undergoing Protected Percutaneous Coronary Intervention With Impella. J Am Heart Assoc 2025:e038509. [PMID: 40240978 DOI: 10.1161/jaha.124.038509] [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: 08/23/2024] [Accepted: 02/07/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND In patients undergoing high-risk percutaneous coronary intervention, Impella has become an important adjunctive tool to support revascularization. The impact of age on the outcomes of patients undergoing high-risk percutaneous coronary intervention is limited. The aim of this study is to describe the characteristics and outcomes of patients ≥75 years of age undergoing Impella-supported high-risk percutaneous coronary intervention. METHODS AND RESULTS Baseline characteristics and outcomes of patients ≥75 years of age versus those of patients <75 years of age in patients enrolled in the cVAD PROTECT III (Catheter-Based Ventricular Assist Device Prospective, Multi-Center, Randomized Controlled Trial of the IMPELLA RECOVER LP 2.5 System Versus Intra Aortic Balloon Pump in Patients Undergoing Non Emergent High Risk Percutaneous Coronary Intervention) study (NCT04136392). Major adverse cardiovascular and cerebral events (composite of all-cause death, nonfatal myocardial infarction, stroke/transient ischemic attack, and repeat revascularization) were assessed at 30 and 90 days and all-cause death at 1 year. Out of 1237 patients, 493 (39.9%) patients were ≥75 years of age. Patients ≥75 years of age had less diabetes and prior myocardial infarction, more hypertension and dyslipidemia, worse renal function, more severe valvular heart disease, but higher left ventricular ejection fraction (P<0.05 for all comparisons). Baseline Synergy Between Percutaneous Coronary Intervention With Taxus and Cardiac Surgery scores were similar between groups. Older patients underwent more left main percutaneous coronary intervention (58% versus 39%; P<0.0001), atherectomy (32% versus 22%; P<0.0001), and femoral access (87% versus 79%, P=0.0003) as compared with younger patients. In-hospital vascular complications did not differ, but rates of respiratory failure, pericardial tamponade, and cardiogenic shock were higher in older patients. Rates of all-cause death and major adverse cardiovascular and cerebral events did not differ between groups at 30 and 90 days. Rates of all-cause death at 1 year were higher in patients ≥75 years (adjusted hazard ratio, 1.99 [95% CI, 1.24-3.18], P=0.004). CONCLUSIONS Impella-supported high-risk percutaneous coronary intervention in older patients is feasible with an acceptable safety profile. However, age ≥75 years remained a statistically significant predictor for all-cause death at 1 year. REGISTRATION URL: https://clinicaltrials.gov; Unique Identifier: NCT04136392.
Collapse
Affiliation(s)
- Philipp Jakob
- Department of Cardiology, University Heart Center, University Hospital Zurich and the Center for Translational and Experimental Cardiology (CTEC) University of Zurich Zurich Switzerland
| | - Alexandra J Lansky
- Department of Cardiology Yale University School of Medicine New Haven CT USA
| | - Mir B Basir
- Center for Structural Heart Disease, Division of Cardiology Henry Ford Health System Detroit MI USA
| | | | - Batla Falah
- Clinical Trials Center Cardiovascular Research Foundation New York NY USA
| | - Zhipeng Zhou
- Clinical Trials Center Cardiovascular Research Foundation New York NY USA
| | - Wayne B Batchelor
- Inova Center of Outcomes Research Inova Heart and Vascular Institute Falls Church VA USA
| | - Arsalan Abu-Much
- Clinical Trials Center Cardiovascular Research Foundation New York NY USA
| | - Cindy L Grines
- Department of Cardiology Northside Hospital Cardiovascular Institute Atlanta GA USA
| | - William W O'Neill
- Center for Structural Heart Disease, Division of Cardiology Henry Ford Health System Detroit MI USA
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital Zurich and the Center for Translational and Experimental Cardiology (CTEC) University of Zurich Zurich Switzerland
| |
Collapse
|
58
|
Daida YG, Rosales AG, Frankland TB, Bacong AM, Waitzfelder B, Li J, Keawe'aimoku Kaholokula J, Palaniappan L, Fortmann SP. Differences in Coronary Heart Disease and Stroke Incidence Among Single-Race and Multiracial Asian and Pacific Islander Subgroups in Hawaii and California: A Retrospective Cohort Study. J Am Heart Assoc 2025:e039076. [PMID: 40240959 DOI: 10.1161/jaha.124.039076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Little is known about clinical and sociodemographic factors affecting coronary heart disease (CHD) and stroke incidence in single-race and multiracial American Asian, Native Hawaiian, and Pacific Islander subgroups. As the US population becomes more diverse, it is important to characterize differences in risks for CHD and stroke, and their contributing factors, in these populations. METHODS AND RESULTS The study population included 303 958 patients from Kaiser Permanente Hawaii and Palo Alto Medical Foundation in California. Self-reported race and ethnicity were derived from electronic health records and 12 mutually exclusive single-race and multiracial groups were created for analyses. Cox proportional hazard models were used to compare CHD and stroke incidence. Unadjusted models were compared with models adjusted for age, income, education, body mass index, smoking, and comorbidities. We found up to a 4-fold variation in CHD and stroke rates among American Asian, Native Hawaiian, and Pacific Islander subgroups. Multiracial subgroups had higher rates than single-race groups. While most single-race American Asian, Native Hawaiian, and Pacific Islander groups had lower CHD and stroke risks, middle-aged Asian Indian men and Native Hawaiian women had higher stroke risks than non-Hispanic White controls. Income, education, body mass index, smoking, and comorbidities contributed significantly to risks in all groups, especially in Native Hawaiian, Pacific Islander, and multiracial groups. CONCLUSIONS Risks for CHD and stroke vary by racial and ethnic subgroups, demonstrating the need to unmask risks by disaggregating racial and ethnic subgroups. Multiracial American Asian, Native Hawaiian, and Pacific Islander groups had higher risks that were only partially explained by modifiable risk factors. Future studies should further explore lifestyle, psychosocial, and sociocultural factors.
Collapse
Affiliation(s)
- Yihe G Daida
- Center for Integrated Health Care Research Kaiser Permanente Honolulu HI USA
| | | | - Timothy B Frankland
- Center for Integrated Health Care Research Kaiser Permanente Honolulu HI USA
| | | | - Beth Waitzfelder
- Center for Integrated Health Care Research Kaiser Permanente Honolulu HI USA
| | - Jiang Li
- Palo Alto Medical Foundation Research Institute, Sutter Health Palo Alto CA USA
| | | | | | - Stephen P Fortmann
- Kaiser Permanente Center for Health Research Portland OR USA
- Stanford University School of Medicine Stanford CA USA
- Kaiser Permanente Bernard J. Tyson School of Medicine Pasadena CA USA
| |
Collapse
|
59
|
Dabravolski SA, Popov MA, Utkina AS, Babayeva GA, Maksaeva AO, Sukhorukov VN, Orekhov AN. Preclinical and mechanistic perspectives on adipose-derived stem cells for atherosclerotic cardiovascular disease treatment. Mol Cell Biochem 2025:10.1007/s11010-025-05285-0. [PMID: 40234340 DOI: 10.1007/s11010-025-05285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Adipose-derived mesenchymal stem cells (AD-MSCs) are a promising therapeutic modality for cardiovascular diseases due to their immunomodulatory, anti-inflammatory, and pro-angiogenic properties. This manuscript explores the current status, challenges, and future directions of AD-MSC therapies, focusing on their application in atherosclerosis (AS), myocardial infarction (MI), and heart failure (HF). Preclinical studies highlight AD-MSC's ability to stabilise atherosclerotic plaques, reduce inflammation, and enhance myocardial repair through mechanisms such as macrophage polarisation, endothelial protection, and angiogenesis. Genetically and pharmacologically modified AD-MSCs, including those overexpressing SIRT1, IGF-1, and PD-L1 or primed with bioactive compounds, exhibit superior efficacy compared to unmodified cells. These modifications enhance cell survival, immunopotency, and reparative capacity, showcasing the potential for tailored therapies. However, clinical translation faces significant hurdles. While recent clinical trials have confirmed the safety of AD-MSC therapy, their efficacy remains inconsistent, necessitating further optimisation of patient selection, dosing strategies, and delivery methods. Donor variability, particularly in patients with co-morbidities like type 2 diabetes (T2D) or obesity, impairs AD-MSC efficacy. Emerging research on extracellular vesicles (EVs) derived from AD-MSC offers a promising cell-free alternative, retaining the therapeutic benefits while mitigating risks. Future perspectives emphasise the need for multidisciplinary approaches to overcome these limitations. Strategies include refining genetic modifications, exploring EV-based therapies, and integrating personalised medicine and advanced diagnostic tools. By addressing these challenges, AD-MSC therapies hold the potential to revolutionise the treatment of cardiovascular diseases, providing innovative solutions to improve patient outcomes.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51P.O. Box 78, 2161002, Karmiel, Israel.
| | - Mikhail A Popov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
| | - Aleksandra S Utkina
- Department of Commodity Expertise and Customs Business, Plekhanov Russian University of Economics, 36, Stremyanny Lane, 115054, Moscow, Russia
| | - Gulalek A Babayeva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552, Moscow, Russia
| | - Anastasia O Maksaeva
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
- Sechenov First Moscow State Medical University, 8, Trubetskaya Street Building 2, 119991, Moscow, Russia
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
- Institute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, 119991, Moscow, Russia
| | - Alexander N Orekhov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street Building 4, 117418, Moscow, Russia
| |
Collapse
|
60
|
Wang S, Pei G, Shen J, Fang Z, Chen T, Wang L, Cheng H, Li H, Pei H, Feng Q, Fan Q, He C, Fu C, Guo Y, Wei Q. Pulsed electromagnetic fields treatment ameliorates cardiac function after myocardial infarction in mice and pigs. J Adv Res 2025:S2090-1232(25)00263-2. [PMID: 40250556 DOI: 10.1016/j.jare.2025.04.019] [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: 05/23/2024] [Revised: 09/21/2024] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
INTRODUCTION Ischemic heart disease (IHD) is a prominent contributor to mortality worldwide, with myocardial infarction (MI) representing its most severe manifestation. Pulsed electromagnetic fields (PEMF) treatment shows promise for treating IHD. Nevertheless, the therapeutic impact and underlying mechanism of PEMF in MI are not fully understood. OBJECTIVES To investigate the efficacy, safety, and mechanisms of PEMF for MI. METHODS We established MI models in both mice and pigs and performed serial echocardiography and cardiac magnetic resonance follow-up to demonstrate the benefit of PEMF treatment after MI. The pathological environment after myocardial infarction was simulated in vitro to observe changes in various cells exposed to PEMF. Gene knockout (TLR4-/-) mice and inhibitors were used to compare the differences in the efficacy of PEMF treatment relative to that of gene knockout/inhibitor treatments. Agonists were used to further explore the mechanism of PEMF treatment. RESULTS In post-MI mice, PEMF treatment enhanced cardiac function and reduced scar formation. PEMF reduced the macrophage inflammatory response, improved cardiomyocyte survival in an inflammatory environment, and decreased collagen secretion by fibroblasts in vitro. Importantly, in the clinically relevant porcine model, PEMF treatment inhibited the inflammatory response and alleviated adverse left ventricular remodeling. Moreover, PEMF could exert therapeutic effects similar to those of gene knockout or inhibitor treatments. In the presence of TLR4 knockout or pyrrolidine dithiocarbamate (an NF-κB inhibitor) administration, PEMF could still improve cardiac function in post-MI mice. Mechanistically, the anti-inflammatory effect of PEMF was reversed when RS09 (a TLR4 agonist) was administered, and the antifibrotic effect of PEMF was attenuated after treatment with SRI-011381 (a TGF-β signaling pathway agonist). CONCLUSIONS PEMF treatment exhibits considerable promise as a noninvasive physical therapy modality, warranting further investigation into its potential implications for managing patients with IHD.
Collapse
Affiliation(s)
- Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gaiqin Pei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayu Shen
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhi Fang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingyu Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanbin Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongliang Pei
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Qipu Feng
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingwen Fan
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenying Fu
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
61
|
Zhang Y, Wang J, Yang S, Kou H, Liu P. Tanshinone IIA alleviate atherosclerosis and hepatic steatosis via down-regulation of MAPKs/NF-κB signaling pathway. Int Immunopharmacol 2025; 152:114465. [PMID: 40090083 DOI: 10.1016/j.intimp.2025.114465] [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: 12/24/2024] [Revised: 03/09/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVES Tanshinone IIA (Tan IIA) exhibits therapeutic potential for atherosclerosis (AS) and hepatic steatosis (HS). The study aims to explore the mechanisms underlying the anti-atherosclerosis and anti-hepatic steatosis effects of Tan IIA. METHODS The LDLR-/-mice were divided into control, model, low/high Tan IIA and atorvastatin group, which fed with High-fat diet to build NAFLD-associated AS model, then administrated with 0.9 % saline, Tan IIA or atorvastatin. RAW264.7 cells divided into control, LPS, LPS plus low/high Tan IIA and LPS plus Tan IIA plus JNK activator group. The different goups' pathological changes visualized with H&E, Oil Red O and Immunofluorescence staining. The therapeutic effect of Tan IIA was reflected by lipids metabolism changes, hepatic indexes, inflammation levels. ELISA, RT-qPCR and Western blot assay were used to determine the inflammatory factors and upstream proteins. Molecular docking was used to reconfirm the importance of genes studied and locate the specific gene will study. RESULTS Tan IIA alleviated LDLR-/-mice AS and HS by reducing AS plaque area, lowering serum &liver lipid levels (TC, TG), improving liver function (AST, ALT). Tan IIA decreased serum inflammation levels (IL-1β, IL-6, TNF-α) and aorta & liver inflammatory-related cytokines levels (iNOS, VCAM-1, IL-6) and inhibited the phosphorylation of aorta & liver protein ERK1/2, JNK, p38 and NF-κB p65, which were validated in the LPS-stimulated macrophages supernatant and cells. CONCLUSIONS The study indicated that Tan IIA can alleviate atherosclerosis and hepatic steatosis via down-regulating MAPKs/NF-κB signaling pathway. This provides a potential therapeutic strategy for the co-existing situation of atherosclerosis and hepatic steatosis.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiarou Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuo Yang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haixin Kou
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
62
|
Jering KS, Claggett BL, Braunwald E, Granger CB, Køber L, Landmesser U, Lewis EF, Maggioni AP, Mann DL, McMurray JJ, Mehran R, Petrie MC, Prescott MF, Rouleau JL, Schou M, Solomon SD, Steg PG, von Lewinski D, Pfeffer MA. NT-proBNP in the Early Convalescent Phase after High-Risk Myocardial Infarction Is Associated with Adverse Cardiovascular Outcomes: the PARADISE-MI Trial. J Card Fail 2025:S1071-9164(25)00164-2. [PMID: 40250826 DOI: 10.1016/j.cardfail.2025.03.018] [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: 12/12/2024] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND N-terminal pro-B-type natriuretic peptide (NT-proBNP) is associated with heart failure (HF) hospitalizations and death when measured during a myocardial infarction (MI). However, NT-proBNP concentrations change following the initial ischemic insult and less is known about the prognostic importance of NT-proBNP in the early convalescent phase. METHODS PARADISE-MI randomized 5661 patients with MI complicated by LVEF ≤40% and/or pulmonary congestion to sacubitril/valsartan or ramipril. Patients with available week 2 NT-proBNP concentrations and without incident HF between randomization and week 2 (n=1062) were analyzed. Associations of week 2 NT-proBNP with subsequent clinical outcomes were evaluated in landmark analyses using Cox models adjusted for clinical characteristics including LVEF, baseline NT-proBNP and atrial fibrillation. RESULTS Median 2-week NT-proBNP was 1391 [676 - 2507] ng/L. Patients in the highest NT-proBNP quartile (≥2507 ng/L) were older, had lower LVEF and eGFR, higher Killip class and more atrial fibrillation. Higher NT-proBNP concentrations were independently associated with greater risk of CV death or incident HF (adjusted HR [aHR] 1.65 per doubling of NT-proBNP; 95% CI, 1.31-2.09), HF hospitalization (aHR 1.87; 95% CI, 1.38-2.54), recurrent MI (aHR 1.46; 95% CI, 1.09-1.95) and all-cause death (aHR 1.85; 95% CI, 1.35-2.53). CONCLUSIONS Patients with elevated NT-proBNP concentrations approximately two weeks after a high-risk MI are at heightened risk of incident HF, recurrent coronary events and death, independent of baseline NT-proBNP concentrations and clinical characteristics. Elevations in NT-proBNP concentrations in the early convalescent phase may assist in risk stratification and identification of patients in need for more advanced preventive treatment approaches. LAY SUMMARY N-terminal pro-B-type natriuretic peptide (NT-proBNP) is a marker of ventricular wall stress and in the acute phase of a myocardial infarction (MI) is strongly associated with adverse cardiovascular outcomes. NT-proBNP concentrations change dynamically after MI but they are not routinely remeasured during follow-up. In a contemporary post-MI population enrolled in the PARADISE-MI trial, NT-proBNP measured approximately two weeks following MI was independently associated with heightened risk of death, incident heart failure and recurrent MI. NT-proBNP concentrations in the early convalescent phase may help risk stratify patients following MI and identify those in need for closer follow-up and more aggressive therapies.
Collapse
Affiliation(s)
- Karola S Jering
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| | - Brian L Claggett
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Eugene Braunwald
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA; TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | | | - Lars Køber
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiology, Angiology and Intensive Care Medicine; Charité Universitätsmedizin Berlin, Campus Benjamin-Franklin (CBF), 12203 Berlin, Germany
| | - Eldrin F Lewis
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aldo P Maggioni
- ANMCO Research Center, Heart Care Foundation, Florence, Italy
| | - Douglas L Mann
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - John Jv McMurray
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Roxana Mehran
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, and Cardiovascular Research Foundation, New York, NY, USA
| | - Mark C Petrie
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | | | - Jean L Rouleau
- Institut de Cardiologie de Montréal, Université de Montréal, Montreal, Canada
| | - Morten Schou
- Department of Cardiology, Herlev-Gentofte University Hospital, Hellerup, Denmark
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Phillippe Gabriel Steg
- Université Paris-Cité, Institut Universitaire de France, AP-HP (Assistance Publique-Hôpitaux de Paris), FACT (French Alliance for Cardiovascular Trials) and INSERM U-1148, Paris, France
| | | | - Marc A Pfeffer
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
63
|
Ji W, Li L, Cheng Y, Yuan Y, Zhao Y, Wang K, Chen B, Wang Y, Yang Y, Zhou Y. Air pollution, lifestyle, and cardiovascular disease risk in northwestern China: A cohort study of over 5.8 million participants. ENVIRONMENT INTERNATIONAL 2025; 199:109459. [PMID: 40253932 DOI: 10.1016/j.envint.2025.109459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Evidence on the combined impact of air pollution and lifestyle on cardiovascular disease (CVD) risk is limited. We employed the Space-Time Extra-Trees model, an ensemble learning method for spatiotemporal data, to estimate the annual average concentrations of five air pollutants from 2017 to 2019. Cox proportional hazards models were used to assess the associations between air pollutant exposure and CVD incidence. A lifestyle score, based on body mass index, waist circumference, diet, physical activity, alcohol consumption, and smoking, was developed to examine the moderating effect of lifestyle on the air pollution-CVD relationship. Among 5,838,833 baseline participants without CVD, 414,218 developed CVD during follow-up. Long-term exposure to particulate matter (PM1, PM2.5, PM10), ozone (O3), and carbon monoxide (CO) was significantly associated with increased CVD risk. Stratified analyses revealed that exercise had the most significant impact on this association, with exercisers showing a notable reduction in risk compared to non-exercisers. An interaction between air pollution and lifestyle was observed (P-interaction < 0.001). Compared to individuals with a relatively healthy lifestyle and low air pollution exposure, those with an unhealthy lifestyle and high exposure had the highest risk of developing CVD (PM1: HR = 1.660, PM2.5: HR = 1.891, PM10: HR = 1.755, O3: HR = 1.970, CO: HR = 1.426). Further analysis revealed a synergistic additive interaction between lifestyle and air pollution, leading to relative excess risks of 0.151, 0.154, 0.137, 0.171, and 0.095 in groups with relatively unhealthy lifestyles and high exposure to PM1, PM2.5, PM10, O3, and CO, respectively. Thus, in addition to controlling major air pollutant emissions, promoting healthy lifestyle adoption is crucial.
Collapse
Affiliation(s)
- Weidong Ji
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Lin Li
- School of Nursing, Xinjiang Medical University, Urumqi 830054 Xinjiang, China
| | - Yinlin Cheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Yujuan Yuan
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830000, China; Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yu Zhao
- School of Public Health, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Kai Wang
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830011, China
| | - Baoyu Chen
- School of Geography and Planning, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Yushan Wang
- Center of Health Management, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830000, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830000, China.
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830000, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830000, China.
| | - Yi Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080 Guangdong, China.
| |
Collapse
|
64
|
Kim Y, Jang S, Ullahansari S, Vo J, Hyun K, Fadel PJ. Neighborhood Safety and Hypertension Risk: A Systematic Review. J Am Heart Assoc 2025; 14:e035381. [PMID: 40178095 DOI: 10.1161/jaha.124.035381] [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: 03/16/2024] [Accepted: 01/17/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Responding to the increasing focus on residential environments, our systematic review aimed to consolidate existing empirical evidence regarding the impact of neighborhood safety on blood pressure. We also summarized the mediating and moderating mechanisms through which neighborhood safety influences blood pressure, alongside their direct effects, to offer insights for future research. METHODS We searched 5 electronic databases (PubMed, Ovid MEDLINE, CINAHL Complete, ProQuest Dissertations and Theses Global, and Web of Science) for the period up to and including December 27, 2022. The initial search yielded 4944 studies reviewed, of which 19 met our criteria and were reviewed. RESULTS Our findings consistently show that living in a safe neighborhood is associated with lower blood pressure outcomes. While most cross-sectional studies found that the association was not statistically significant (7/10 studies showed insignificant results), longitudinal studies that tracked changes in neighborhood safety over time (4/5 studies) showed significant negative associations between neighborhood safety and blood pressure. Additionally, some studies identified sex (n=3), age (n=2), and neighborhood characteristics (n=4) as significant moderators, with the strength of the association between neighborhood safety and blood pressure varying across different demographic groups and neighborhood contexts. CONCLUSIONS Our findings suggest that unsafe neighborhoods may increase blood pressure and hypertension risk, warranting further research and interventions. This review also highlights the importance of adopting longitudinal designs, especially those using time-varying measures of neighborhood environments.
Collapse
Affiliation(s)
- Yeonwoo Kim
- Department of Kinesiology University of Texas at Arlington Arlington TX USA
| | - Soeun Jang
- School of Social Work University of Texas at Arlington Arlington TX USA
| | - Shaikh Ullahansari
- Department of Kinesiology University of Texas at Arlington Arlington TX USA
| | - Jimmy Vo
- Department of Kinesiology University of Texas at Arlington Arlington TX USA
| | - Kate Hyun
- Department of Civil Engineering University of Texas at Arlington Arlington TX USA
| | - Paul J Fadel
- Department of Kinesiology University of Texas at Arlington Arlington TX USA
| |
Collapse
|
65
|
Dai J, Zhao J, Xu X, Chen Y, Sun S, Li S, Cui L, Wang Y, Li L, Guo R, Huang D, Ma X, Zhao R, Yu H, Chen T, Tan J, Liu X, Jiang S, Hou J, Fang C, Mintz GS, Yu B. Long-Term Prognostic Implications of Non-Culprit Lesions in Patients Presenting With an Acute Myocardial Infarction: Is It the Angiographic Stenosis Severity or the Underlying High-Risk Morphology? Circulation 2025; 151:1098-1110. [PMID: 39886764 DOI: 10.1161/circulationaha.124.071855] [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: 08/13/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Patients with acute myocardial infarction and angiographically obstructive non-culprit lesions are at high risk for recurrent major adverse cardiac events (MACEs). However, it remains largely unknown whether events are due to stenosis severity or due to the underlying high-risk lesion morphology. METHODS Between January 2017 and December 2021, 1312 patients with acute myocardial infarction underwent optical coherence tomography of all the 3 main epicardial arteries after successful percutaneous coronary intervention. Patients and lesions were categorized according to the presence or absence of (1) 1 or more non-culprit angiographic obstructive stenoses with a visual diameter stenosis of ≥50% and (2) 1 or more lesions with an underlying high-risk morphology defined as an optical coherence tomography thin-cap fibroatheroma (TCFA). Patients were followed for up to 5 years (median 4.1 [interquartile range: 3.0-5.0] years). MACEs comprised cardiac death, non-fatal myocardial infarction, and unplanned coronary revascularization. RESULTS Overall, 492 patients had at least 1 obstructive non-culprit lesion, 352 had a single lesion, and 140 had multiple obstructive non-culprit lesions. The presence and number of angiographic obstructive non-culprit lesions correlated with the proportion and number of optical coherence tomography-derived TCFAs. At the lesion level, the prevalence of TCFA was twice as high in obstructive lesions compared with nonobstructive lesions. Patients with obstructive non-culprit lesions had an increased risk of overall MACEs (17.7% versus 12.8%; hazard ratio, 1.39 [95% CI, 1.02-1.91]) and non-culprit lesion-related MACEs (8.7% versus 3.9%; HR, 2.13 [95% CI, 1.26-3.59). Results were similar when patients were categorized on the basis of the underlying TCFA. A proportionally higher rate of overall and non-culprit lesion-related MACEs was observed as the number of obstructive stenoses or TCFAs in non-culprit segments increased. The lesion-specific HRs for obstructive lesion and TCFA were 2.03 (95% CI, 1.06-3.89) and 2.39 (95% CI, 1.29-4.43), respectively. Optical coherence tomography-derived TCFA, but not angiographic obstructive stenosis, was independently predictive of recurrent MACEs in both patient-level and lesion-level multivariable models in which these 2 characteristics were introduced simultaneously. CONCLUSIONS The long-term prognostic implications of the presence and extent of angiographic obstructive non-culprit lesions in patients with acute myocardial infarction are primarily due to their correlation with the underlying high-risk morphology, which confers an increased risk of recurrent MACEs.
Collapse
Affiliation(s)
- Jiannan Dai
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- International Medical Center, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., Y.W.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China (J.D., J.H., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Jiawei Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Xueming Xu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Yuzhu Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Sibo Sun
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Shuang Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Lina Cui
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Yini Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- International Medical Center, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., Y.W.)
| | - Lulu Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
| | - Ruirong Guo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Dongxu Huang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Xianqin Ma
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Rui Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Huai Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
| | - Tao Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
| | - Jinfeng Tan
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
| | - Xiaohui Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
| | - Senqing Jiang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Jingbo Hou
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China (J.D., J.H., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Chao Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| | - Gary S Mintz
- Cardiovascular Research Foundation, New York, NY, USA (G.S.M.)
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., Y.W., L.L., R.G., D.H., X.M., R.Z., H.Y., T.C., J.T., X.L., S.J., J.H., C.F., B.Y.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China (J.D., J.H., B.Y.)
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China (J.D., J.Z., X.X., Y.C., S.S., S.L., L.C., R.G., D.H., X.M., R.Z., S.J., J.H., C.F., B.Y.)
| |
Collapse
|
66
|
Zhevlakova I, Liu H, Dudiki T, Gao D, Yakubenko V, Tkachenko S, Cherepanova O, Podrez EA, Byzova TV. Mechanisms and consequences of myeloid adhesome dysfunction in atherogenesis. Cardiovasc Res 2025; 121:62-76. [PMID: 39393814 PMCID: PMC11999018 DOI: 10.1093/cvr/cvae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/13/2024] Open
Abstract
AIMS In the context of atherosclerosis, macrophages exposed to oxidized low-density lipoproteins (oxLDLs) exhibit cellular abnormalities, specifically in adhesome functions, yet the mechanisms and implications of these adhesive dysfunctions remain largely unexplored. METHODS AND RESULTS This study reveals a significant depletion of Kindlin3 (K3) or Fermt3, an essential component of the adhesome regulating integrin functions, in macrophages located within atherosclerotic plaques in vivo and following oxLDL exposure in vitro. To examine the effects of K3 deficiency, the study utilized hyperlipidaemic bone marrow chimeras devoid of myeloid Kindlin3 expression. The absence of myeloid K3 increased atherosclerotic plaque burden in the aortas in vivo and enhanced lipid accumulation and lipoprotein uptake in macrophages from Kindlin3-null chimeric mice in vitro. Importantly, re-expression of K3 in macrophages ameliorated these abnormalities. RNA sequencing of bone marrow-derived macrophages (BMDM) from K3-deficient mice revealed extensive deregulation in adhesion-related pathways, echoing changes observed in wild-type cells treated with oxLDL. Notably, there was an increase in Olr1 expression [encoding the lectin-like oxidized LDL receptor-1 (LOX1)], a gene implicated in atherogenesis. The disrupted K3-integrin axis in macrophages led to a significant elevation in the LOX1 receptor, contributing to increased oxLDL uptake and foam cell formation. Inhibition of LOX1 normalized lipid uptake in Kindlin3-null macrophages. A similar proatherogenic phenotype, marked by increased macrophage LOX1 expression and foam cell formation, was observed in myeloid-specific Itgβ1-deficient mice but not in Itgβ2-deficient mice, underscoring the critical role of K3/Itgβ1 interaction. CONCLUSION This study shows that the loss of Kindlin3 in macrophages upon exposure to oxLDL leads to adhesome dysfunction in atherosclerosis and reveals the pivotal role of Kindlin3 in macrophage function and its contribution to the progression of atherosclerosis, providing valuable insights into the molecular mechanisms that could be targeted for therapeutic interventions.
Collapse
Affiliation(s)
- Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Huan Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37684, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, 2109 Adelbert Rd Building, Cleveland, OH 44106, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
67
|
Jung C, Han JW, Lee SJ, Kim KH, Oh JE, Bae S, Lee S, Nam YJ, Kim S, Dang C, Kim J, Chu N, Lee EJ, Yoon YS. Novel Directly Reprogrammed Smooth Muscle Cells Promote Vascular Regeneration as Microvascular Mural Cells. Circulation 2025; 151:1076-1094. [PMID: 39945059 PMCID: PMC11996609 DOI: 10.1161/circulationaha.124.070217] [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: 09/06/2024] [Accepted: 01/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Although cell therapy has emerged as a promising approach to promote neovascularization, its effects are mostly limited to capillaries. To generate larger or more stable vessels, layering of mural cells such as smooth muscle cells (SMCs) or pericytes is required. Recently, direct reprogramming approaches have been developed for generating SMCs. However, such reprogrammed SMCs lack genuine features of contractile SMCs, a native SMC phenotype; thus, their therapeutic and vessel-forming potential in vivo was not explored. Therefore, we aimed to directly reprogram human dermal fibroblasts toward contractile SMCs (rSMCs) and investigated their role for generating vascular mural cells in vivo and their therapeutic effects on ischemic disease. METHODS We applied myocardin and all-trans retinoic acid with specific culture conditions to directly reprogram human dermal fibroblasts into rSMCs. We characterized their phenotype as contractile SMCs through quantitative reverse-transcriptase polymerase chain reaction, flow cytometry, and immunostaining. We then explored their contractility using a vasoconstrictor, carbachol, and through transmission electron microscope and bulk RNA sequencing. Next, we evaluated whether transplantation of rSMCs improves blood flow and induces vessel formation as mural cells in a mouse model of hindlimb ischemia with laser Doppler perfusion imaging and histological analysis. We also determined their paracrine effects. RESULTS Our novel culture conditions using myocardin and all-trans retinoic acid efficiently reprogrammed human dermal fibroblasts into SMCs. These rSMCs displayed characteristics of contractile SMCs at the mRNA, protein, and cellular levels. Transplantation of rSMCs into ischemic mouse hind limbs enhanced blood flow recovery and vascular repair and improved limb salvage. Histological examination showed that vascular density was increased and the engrafted rSMCs were incorporated into the vascular wall as pericytes and vascular SMCs, thereby contributing to formation of more stable and larger microvessels. Quantitative reverse-transcriptase polymerase chain reaction analysis revealed that these transplanted rSMCs exerted pleiotropic effects, including angiogenic, arteriogenic, vessel-stabilizing, and tissue regenerative effects, on ischemic limbs. CONCLUSIONS A combination of myocardin and all-trans retinoic acid in defined culture conditions efficiently reprogrammed human fibroblasts into contractile and functional SMCs. The rSMCs were shown to be effective for vascular repair and contributed to neovascularization through mural cells and various paracrine effects. These human rSMCs could represent a novel source for cell-based therapy and research.
Collapse
Affiliation(s)
- Cholomi Jung
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji Woong Han
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shin-Jeong Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyung Hee Kim
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jee Eun Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seongho Bae
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sangho Lee
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Young-Jae Nam
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chaewon Dang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jaehyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Nakhyung Chu
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eun Jig Lee
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Endocrinology, Division of Endocrinology and Metabolism, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young-sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
68
|
Chen JH, Li KX, Fan CF, Yang H, Zhang ZR, Chen YH, Qi C, Li AH, Lin AQ, Chen X, Luo P. Integration of machine learning and experimental validation reveals new lipid-lowering drug candidates. Acta Pharmacol Sin 2025:10.1038/s41401-025-01539-1. [PMID: 40234619 DOI: 10.1038/s41401-025-01539-1] [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: 11/16/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025]
Abstract
Hyperlipidemia, a major risk factor for cardiovascular diseases, is associated with limitations in clinical lipid-lowering medications. Drug repurposing strategies expedite the research process and mitigate development costs, offering an innovative approach to drug discovery. This study employed systematic literature and guidelines review to compile a training set comprising 176 lipid-lowering drugs and 3254 non-lipid-lowering drugs. Multiple machine learning models were developed to predict the lipid-lowering potential of drugs. A multi-tiered validation strategy was implemented, encompassing large-scale retrospective clinical data analysis, standardized animal studies, molecular docking simulations and dynamics analyses. Through a comprehensive screening analysis utilizing machine learning, 29 FDA-approved drugs with lipid-lowering potential were identified. Clinical data analysis confirmed that four candidate drugs, with Argatroban as the representative, demonstrated lipid-lowering effects. In animal experiments, the candidate drugs significantly improved multiple blood lipid parameters. Molecular docking and dynamics simulations elucidated the binding patterns and stability of candidate drugs in interaction with related targets. We successfully identified multiple non-lipid-lowering drugs with lipid-lowering potential by integrating state-of-the-art machine learning techniques with multi-level validation methods, thereby providing new insights into lipid-lowering drugs, establishing a paradigm for AI-based drug repositioning research, and expanding the repertoire of lipid-lowering medications available to clinicians.
Collapse
Affiliation(s)
- Jing-Hong Chen
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China
| | - Ke-Xin Li
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Chao-Fan Fan
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hong Yang
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Zhi-Rou Zhang
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Yi-Han Chen
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Chang Qi
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China
- The University of Hong Kong, Hong Kong, China
| | - Ang-Hua Li
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - An-Qi Lin
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China.
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China.
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China.
| |
Collapse
|
69
|
Stopa V, Dafou D, Karagianni K, Nossent AY, Farrugia R, Devaux Y, Sopic M. Epitranscriptomics in atherosclerosis: Unraveling RNA modifications, editing and splicing and their implications in vascular disease. Vascul Pharmacol 2025; 159:107496. [PMID: 40239855 DOI: 10.1016/j.vph.2025.107496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Atherosclerosis remains a leading cause of morbidity and mortality worldwide, driven by complex molecular mechanisms involving gene regulation and post-transcriptional processes. Emerging evidence highlights the critical role of epitranscriptomics, the study of chemical modifications occurring on RNA molecules, in atherosclerosis development. Epitranscriptomics provides a new layer of regulation in vascular health, influencing cellular functions in endothelial cells, smooth muscle cells, and macrophages, thereby shedding light on the pathogenesis of atherosclerosis and presenting new opportunities for novel therapeutic targets. This review provides a comprehensive overview of the epitranscriptomic landscape, focusing on key RNA modifications such as N6-methyladenosine (m6A), 5-methylcytosine (m5C), pseudouridine (Ψ), RNA editing mechanisms including A-to-I and C-to-U editing and RNA isoforms. The functional implications of these modifications in RNA stability, alternative splicing, and microRNA biology are discussed, with a focus on their roles in inflammatory signaling, lipid metabolism, and vascular cell adaptation within atherosclerotic plaques. We also highlight how these modifications influence the generation of RNA isoforms, potentially altering cellular phenotypes and contributing to disease progression. Despite the promise of epitranscriptomics, significant challenges remain, including the technical limitations in detecting RNA modifications in complex tissues and the need for deeper mechanistic insights into their causal roles in atherosclerotic pathogenesis. Integrating epitranscriptomics with other omics approaches, such as genomics, proteomics, and metabolomics, holds the potential to provide a more holistic understanding of the disease.
Collapse
Affiliation(s)
- Victoria Stopa
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Dimitra Dafou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Korina Karagianni
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - A Yaël Nossent
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg.
| | - Miron Sopic
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg; Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
70
|
Volpi JJ, Kasner SE, Neervoort J, Wolters LF, Louwsma T, Marti AK, Landaas EJ. The Annual Economic Burden of Patent Foramen Ovale-Associated Stroke in the United States. J Stroke Cerebrovasc Dis 2025:108319. [PMID: 40239827 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Stroke is the second leading cause of death worldwide and the third leading cause of death and disability combined. Among ischemic strokes, 25% to 40% are classified as cryptogenic, with patent foramen ovale (PFO) identified as a potential underlying cause. PFO is found in 25% of the general population. Despite the significant occurrence of PFO-associated strokes, the associated costs remain largely unexplored. AIM This study aimed to evaluate the annual economic burden of PFO-associated stroke in the US. METHODS A cost-of-illness study was performed, encompassing the direct and indirect costs of PFO-associated stroke on both society and the healthcare system. The model adopted a top-down approach and a one-year, US societal perspective. A payer perspective and bottom-up costing approach were included as scenario analyses. Data was obtained by a targeted literature review. RESULTS The societal model, assuming 32.5% cryptogenic strokes, incurs an annual cost of over $1.3 billion in the US, with over $1.0 billion coming from new strokes and over $300 million from recurrent strokes. The majority of the costs are indirect-50% from productivity losses due to premature death and 27% from other productivity losses. Direct costs constitute 23% of the total. From the payer perspective, the annual costs for PFO-associated strokes in the US were estimated at $302 million, with hospitalization costs comprising 44%, followed by prescriptions and outpatient care at 19% and 16%, respectively. CONCLUSION The economic burden of PFO-associated strokes in the US is substantial, exceeding $1.3 billion per year. Different types of medical management or surgical treatments for PFO-associated stroke could lead to gains in both costs and health outcomes.
Collapse
Affiliation(s)
- John J Volpi
- The Houston Methodist Institute for Academic Medicine, Houston, TX, USA
| | - Scott E Kasner
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Timon Louwsma
- Asc Academics B.V, Groningen, Netherlands; Department of Health Sciences, University Medical Center Groningen, Groningen, Netherlands
| | | | | |
Collapse
|
71
|
Assenza MR, Gaggi G, di Credico A, Ghinassi B, Barbagallo F. The Effect of Endocrine Disruptors on the Cardiovascular System: does sex matter? ENVIRONMENTAL RESEARCH 2025; 277:121612. [PMID: 40239736 DOI: 10.1016/j.envres.2025.121612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Endocrine disruptors (EDs) are environmental chemicals that interfere with hormone function, posing significant risks to human health, including the cardiovascular system. This review comprehensively examines the impact of EDs on cardiovascular health, with a specific focus on sex differences observed in various models. Utilizing in-vitro studies, in vivo animal models, and human clinical data, we delineate how sex-specific hormonal environments influence the cardiovascular effects of ED exposure. In vitro studies highlight cellular and molecular mechanisms that differ between male and female-derived cells. In vivo models reveal distinct physiological responses and susceptibilities to EDs, influenced by sex hormones. Human studies provide epidemiological evidence and clinical observations that underscore the variability in cardiovascular outcomes between men and women. This review underscores the necessity of considering sex as a critical factor in understanding the cardiovascular implications of ED exposure, advocating for gender-specific risk assessment and therapeutic strategies. The findings aim to enhance awareness and inform future research and policy-making to mitigate the adverse cardiovascular effects of EDs across different sexes.
Collapse
Affiliation(s)
- Maria Rita Assenza
- Department of Medicine and Surgery, "Kore" University of Enna "Kore", 94100, Enna, Italy
| | - Giulia Gaggi
- Cell Reprogramming and Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | - Andrea di Credico
- Cell Reprogramming and Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | - Barbara Ghinassi
- Cell Reprogramming and Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Federica Barbagallo
- Department of Medicine and Surgery, "Kore" University of Enna "Kore", 94100, Enna, Italy.
| |
Collapse
|
72
|
Canonico ME, Avvedimento M, Piccolo R, Hess CN, Bardi L, Ilardi F, Giugliano G, Franzone A, Gargiulo G, Berkowitz SD, Cannon CP, Esposito G, Bonaca MP. Long-term Antithrombotic Therapy in Patients With Chronic Coronary Syndrome: An Updated Review of Current Evidence. Clin Ther 2025:S0149-2918(25)00086-4. [PMID: 40229176 DOI: 10.1016/j.clinthera.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/21/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE Despite improvements in the secondary prevention of atherothrombosis in patients with coronary artery disease during the past decade, it is estimated that approximately 19 million people annually die from cardiovascular diseases worldwide. Atherothrombosis remains the core pathobiology of acute complications including myocardial infarction (MI), and therefore, antithrombotic therapy plays a pivotal role in the strategies for major adverse cardiovascular event (MACE) prevention. Unlike early antithrombotic management after acute coronary syndrome, less evidence is available on long-term antithrombotic therapy in patients with chronic coronary syndrome (CCS). In addition, greater recognition of the impact of bleeding complications of such therapies has led to a more complex and personalized approach to their application. The purpose of this article is to review the available evidence on long-term antithrombotic therapy in patients with CCS including those with high-risk characteristics such as prior MI or polyvascular disease. METHODS A comprehensive literature review was performed in major databases including PubMed, Embase, and the Cochrane Library. The main focus of this narrative review was on available data from guidelines, meta-analysis, randomized controlled trials, and observational studies that assessed the efficacy and safety profile of long-term antithrombotic therapy in patients with CCS. FINDINGS Several studies suggest that long-term antithrombotic therapy is effective in reducing the risk of recurrent MACEs in patients with CCS. Current clinical guidelines recommend single antiplatelet therapy with aspirin as a first-line long-term strategy for patients without indication for oral anticoagulation. However, novel approaches focused on P2Y12 inhibitor monotherapy are emerging. More intensive antithrombotic strategies including long-term dual antiplatelet therapy and dual pathway inhibition further reduce ischemic risk but at the cost of increased bleeding. IMPLICATIONS This review highlights the importance of close monitoring and regular reassessment of the risk-benefit balance of antithrombotic therapy in patients with CCS. Overall, long-term antithrombotic therapy with either single antiplatelet therapy or dual antiplatelet therapy/dual pathway inhibition is effective in reducing the risk of MACEs in patients with CCS. The choice of antithrombotic therapy should be individualized based on the patient's clinical profile, particularly for thrombohemorrhagic risk. Future research should focus on identifying the optimal antithrombotic regimen for specific subgroups of patients with prior MI particularly for those with high bleeding risk.
Collapse
Affiliation(s)
- Mario Enrico Canonico
- CPC Clinical Research, Aurora, Colorado; Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.
| | - Marisa Avvedimento
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy; Quebec Heart & Lung Institute, Laval University, Quebec City, Quebec, Canada
| | - Raffaele Piccolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Connie N Hess
- CPC Clinical Research, Aurora, Colorado; Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Luca Bardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Federica Ilardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Giugliano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Anna Franzone
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Gargiulo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Scott D Berkowitz
- CPC Clinical Research, Aurora, Colorado; Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Christopher P Cannon
- CPC Clinical Research, Aurora, Colorado; Cardiovascular Division, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Marc P Bonaca
- CPC Clinical Research, Aurora, Colorado; Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
73
|
Barreto G, Sánchez P, Dias R, Baltar A, Shirahige L, Fragoso de Andrade R, Cavalcanti de Oliveira KS, Santana CA, Piscitelli D, Monte-Silva K. The impact of the number of sessions and stimulation parameters on repetitive transcranial magnetic stimulation efficacy for post-stroke upper extremity recovery: A systematic review and meta-analysis. Clin Rehabil 2025:2692155251328945. [PMID: 40223300 DOI: 10.1177/02692155251328945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
ObjectiveTo identify the optimal number of sessions and parameters of repetitive transcranial magnetic stimulation frequency, stimulation intensity, number of pulses per session/treatment that promotes a greater effect on post-stroke upper extremity function.Data sourcesMEDLINE (PubMed), Cumulative Index to Nursing and Allied Health Literature, Physiotherapy Evidence Database, Latin American and Caribbean Health Science Literature, Scientific Electronic Library Online, and Scopus up to November 2024.Review methodsWe conducted a meta-analysis on randomized controlled trials of repetitive transcranial magnetic stimulation combined with other therapies for post-stroke upper extremity function, assessed risk-of-bias using the Physiotherapy Evidence Database scale, calculated standard mean differences (SMD) with 95% confidence intervals (CI), and determined parameter cutoff points using a receiver operating characteristic curve.ResultsThirty-five randomized clinical trials involving 897 patients were included. Compared to control groups, repetitive transcranial magnetic stimulation combined with other therapies improved upper extremity motor function with a higher effect size for: ≥ 15 sessions (SMD 0.72 95% CI, 0.37 to 1.08; p < 0.0001]), > 1 Hz frequencies (0.98 [95% CI, 0.48 to 1.48; p = 0001]), intensities of <100% of resting motor threshold (SMD 0.52 [95% CI, 0.29 to 0.75; p < 0.00001]), ≥ 1000 pulses per session (SMD 0.61 [95% CI, 0.40 to 0.82; p < 0.00001]), and ≥12,000 pulses per treatment (SMD 0.64 [95% CI, 0.36 to 0.93; p < 0.0001]).ConclusionTo enhance post-stroke upper extremity function, the optimal repetitive transcranial magnetic stimulation protocol involves at least 15 sessions, frequencies above 1 Hz, intensities below 100% of resting motor threshold, at least 1000 pulses per session, and 12,000 pulses per treatment.
Collapse
Affiliation(s)
- Gabriel Barreto
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- NAPeN Network (Núcleo de Assistência e Pesquisa em Neuromodulação), Recife, Brazil
| | - Paz Sánchez
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Rebeca Dias
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Adriana Baltar
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- NAPeN Network (Núcleo de Assistência e Pesquisa em Neuromodulação), Recife, Brazil
| | - Lívia Shirahige
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- NAPeN Network (Núcleo de Assistência e Pesquisa em Neuromodulação), Recife, Brazil
| | | | | | | | - Daniele Piscitelli
- Doctor of Physical Therapy Program, Department of Kinesiology, University of Connecticut, Storrs, USA
| | - Kátia Monte-Silva
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- NAPeN Network (Núcleo de Assistência e Pesquisa em Neuromodulação), Recife, Brazil
| |
Collapse
|
74
|
Zhang Z, Li H, Wan Z, Su M, Zhang Y, Yang T, Ji X, Men J, Xing G, Han J, Ji Y, Zhang W, Chen H, Xu X, Fei J, Dong C, Yang Y, Wu Y, Yi Q, Pang W, Zhang M, Shi C, Zhen K, Wang D, Lei J, Wu S, Shu S, Zhang Y, Zhang S, Gao Q, Wan J, Xie W, Yang P, Zhang P, Zuo X, Jiang T, Zhai Z, Wang C. Whole genome sequencing identifies pathogenic genetic variants in Han Chinese patients with familial venous thromboembolism. Commun Biol 2025; 8:604. [PMID: 40221599 PMCID: PMC11993696 DOI: 10.1038/s42003-025-07935-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 03/13/2025] [Indexed: 04/14/2025] Open
Abstract
Genetic factors play a pivotal role in determining venous thromboembolism (VTE) risk, particularly in cases of unprovoked early-onset VTE and those with a family history. While genome-wide association studies (GWAS) has advanced our understanding, high-quality whole-genome sequencing (WGS) from family-based studies is essential to elucidate the role of rare variants. In this study, we performed WGS on 216 individuals from 35 Han Chinese VTE pedigrees and validated findings in 99 high-heritability VTE cases using whole-exome sequencing. Functional impact was assessed via qPCR and Western Blot in HEK293T cells. Classical genes explained partial familial inheritance (20/35), while non-classical genes showed comparable effects on VTE recurrence and CTEPH. From 36 rare variants, 34 pedigrees (97%) were interpreted, with 29 variants reported for the first time. Notably, three novel variants, GP6 (c.G1094A:p.R365H), TET2 (c.G3451T:p.E1151X), and JAK2 (c.G380A:p.G127D), shared in two unrelated pedigrees each and are classified as low frequency in East Asians. Functional analyses revealed significant changes in GP6 and TET2 expression compared to the wild type. These findings provide novel insights into the genetic architecture of VTE and highlight GP6, TET2, and JAK2 as potential risk factors in East Asian populations, underscoring the clinical relevance of rare variants in VTE pathogenesis.
Collapse
Affiliation(s)
- Zhu Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Haobo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhaoman Wan
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China
| | - Mingming Su
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yu Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Tao Yang
- Department of Vascular Surgery, Shanxi Bethune Hospital, Shanxi, China
| | - Xiaofan Ji
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianlong Men
- Precision Medicine Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Guoqiang Xing
- Department of Pulmonary and Critical Care Medicine, Zibo First Hospital, Zibo, Shandong, China
| | - Jing Han
- Department of Pulmonary and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yingqun Ji
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Hong Chen
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaomao Xu
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, Beijing, China
| | - Jianwen Fei
- Department of Pulmonary and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong, China
| | - Chunling Dong
- Department of Pulmonary and Critical Care Medicine, Second Hospital, Jilin University, Jilin, China
| | - Yuanhua Yang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital Affiliated to Nanjing, Wuxi, Jiangsu, China
| | - Qun Yi
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wenyi Pang
- Department of Pulmonary and Critical Care Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chong Shi
- Beijing University of Chinese Medicine, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Kaiyuan Zhen
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dingyi Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Clinical research and Data management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jieping Lei
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Clinical research and Data management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Sinan Wu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Clinical research and Data management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shi Shu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yunxia Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shuai Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qian Gao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jun Wan
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wanmu Xie
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Peng Zhang
- Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China.
| | - Taijiao Jiang
- State Key Laboratory of Respiratory Disease, The Key laboratory of Advanced Interdisciplinary Studies Center, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, China.
| | - Zhenguo Zhai
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Chen Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Respiratory Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
75
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
76
|
Zhang H, Li M, Liu W, Yuan H. Dementia-related adverse events associated with direct oral anticoagulants use: a real-world, pharmacovigilance study based on the FAERS database. Expert Opin Drug Saf 2025:1-10. [PMID: 40207729 DOI: 10.1080/14740338.2025.2490847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Direct oral anticoagulants (DOACs) are commonly used to prevent and treat thromboembolic diseases. This study aimed to assess and compare dementia related adverse events (AEs) associated with DOACs. RESEARCH DESIGN AND METHODS AEs related to DOACs from January 2014 to June 2023 were extracted from the FDA Adverse Event Reporting System (FAERS) database. Disproportionality analysis methods, including reporting odds ratio (ROR), proportional reporting ratio, Bayesian Confidence Propagation Neural Network, and Multi-Item Gamma Poisson Shrinker, were used to evaluate the association between DOACs and dementia-related AEs. RESULTS There were 12,692,968 AEs reported in FAERS after deduplication. Among these, 165, 206, 1574, and 12 dementia-related AEs that were attributed to dabigatran, rivaroxaban, apixaban, and edoxaban, respectively. Apixaban showed the strongest association with dementia-related AEs (ROR 7.66, 95% confidence interval (CI) 7.27-8.06), while rivaroxaban had the lowest ROR (0.95, 95%CI 0.83-1.09). Women exhibited higher RORs for all DOACs, with apixaban showing the most significant correlation. Subgroup analysis indicated a significant link between apixaban and dementia, dementia Alzheimer's type and senile dementia. CONCLUSIONS Apixaban appears most associated with dementia-related AEs among DOACs, whereas rivaroxaban poses a lower risk. Further research is needed to validate these findings through large-scale prospective studies.
Collapse
Affiliation(s)
- Hanxu Zhang
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengya Li
- Department of Pharmacy, Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Wei Liu
- Department of Pharmacy, Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
77
|
Zeng Z, Liu T, Zeng P, Xie Y, Li L, Tan J, Wang H, Liu S, Bian Q, Xiao H, Liang S, Chen J, Chen Y, Lu L. Enhancing vascular implants with heparin-polylysine-copper nanozyme coating for synergistic anticoagulation and antirestenotic activity. Int J Biol Macromol 2025; 309:143048. [PMID: 40216105 DOI: 10.1016/j.ijbiomac.2025.143048] [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: 12/30/2024] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Re-establishing blood flow through vascular implants, such as stents, faces significant challenges, including late stent thrombosis (LST) and in-stent restenosis (ISR). Strategies to overcome these issues focus on enhancing stent surfaces with anticoagulant, pro-endothelialization, and anti-neointimal hyperplasia (NIH) properties. However, achieving all of these functionalities typically requires complex surface modifications. In this study, we developed nanozyme particles by assembling heparin, polylysine (PLL) and copper ions, which catalytically release nitric oxide (NO) in situ. A functional coating was then formed on pre-deposited polydopamine (PDA) transition layer. Our nanozyme coating not only exhibits robust anticoagulant activity but also enables sustained, in situ release of NO, a critical gas molecule for maintaining vascular health and patency. In vitro results showed that the coating significantly inhibited platelet aggregation, remarkably prolonged activated partial thromboplastin time (APTT), and selectively promoted endothelial cell growth over smooth muscle cells. Ex vivo blood circulation models confirmed its superior anti-thrombotic efficacy, while in vivo experiments further validated its ability to facilitate endothelial regeneration and suppress NIH. This technology offers significant potential for improving the safety and outcomes of cardiovascular implants.
Collapse
Affiliation(s)
- Zheng Zeng
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China; Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Tao Liu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Peiying Zeng
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Yinhong Xie
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Huanran Wang
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Qihao Bian
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Hongkai Xiao
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Siyu Liang
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yin Chen
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lei Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
78
|
Mao T, Jiang K, Pang Y, Pan Y, Jia W, Gao Q, Lin Q. Hydroxysafflor yellow A for ischemic heart diseases: a systematic review and meta-analysis of animal experiments. Front Pharmacol 2025; 16:1510657. [PMID: 40271057 PMCID: PMC12014549 DOI: 10.3389/fphar.2025.1510657] [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: 10/13/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hydroxysafflor yellow A (HSYA) possesses a variety of pharmacological activities which has been demonstrated to be effective against ischemic heart disease (IHD). This study aimed to comprehensively examine the efficacy and summarize the potential mechanisms of HSYA against IHD in animal models. Methods We conducted electronic searches for preclinical studies on PubMed, Embase, Web of Science, Cochrane Library, CNKI, SinoMed, Wanfang, and Chinese VIP databases from inception to 31 January 2024. The CAMARADES checklist was chosen to assess the quality of evidence. STATA 14.0 software was utilized to analyze the data. The underlying mechanisms were categorized and summarized. Results Twenty-eight studies involving 686 rodents were included and the mean score of methodology quality was 5.04 (range from 4 to 7). Meta-analysis observed that HSYA could decrease myocardial infarction size (SMD: -2.82, 95%CI: -3.56 to -2.08, p < 0.001) and reduce the levels of biomarkers of myocardial injury including cTnI (SMD: -3.82, 95%CI: -5.20 to -2.44, p < 0.001) and CK-MB (SMD: -2.74, 95%CI: -3.58 to -1.91, p < 0.001). HSYA displayed an improvement in cardiac function indicators including LVEF, LVSP, +dp/dt max and -dp/dt max. Furthermore, HSYA was able to reduce the levels of MDA, TNF-α and IL-6, while increasing SOD and NO levels. Mechanistically, the protective effect of HSYA in alleviating myocardial injury after ischemia may be associated with NLRP3 inflammasome, Bcl-2, Bax, caspase-3, eNOS proteins, and TLR/NF-κB, Nrf2/HO-1, JAK/STAT, PI3K/Akt, AMPK/mTOR, VEGFA pathways. Conclusion This study demonstrates that HSYA exerts cardioprotective effects in decreasing infarct size, reducing myocardial enzymes and improving cardiac function, which may be mediated by anti-inflammatory, antioxidant, anti-apoptotic, regulation of autophagy, improvement of microcirculation and promotion of angiogenesis. However, the absence of safety assessment, lack of animal models of co-morbidities, and inconsistency between timing of administration and clinical practice are limitations of preclinical studies. Systematic Review Registration clinicaltrials.gov, Identifier, CRD42023460790.
Collapse
Affiliation(s)
- Tianshi Mao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaixin Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanting Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenhao Jia
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Qun Gao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
79
|
Bozkurt B, Mullens W, Leclercq C, Russo AM, Savarese G, Böhm M, Hill L, Kinugawa K, Sato N, Abraham WT, Bayes-Genis A, Mebazaa A, Rosano GMC, Zieroth S, Linde C, Butler J. Cardiac rhythm devices in heart failure with reduced ejection fraction - role, timing, and optimal use in contemporary practice. European Journal of Heart Failure expert consensus document. Eur J Heart Fail 2025. [PMID: 40204670 DOI: 10.1002/ejhf.3641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/10/2025] [Accepted: 02/23/2025] [Indexed: 04/11/2025] Open
Abstract
Guidelines for management of heart failure with reduced ejection fraction (HFrEF) emphasize personalized care, patient engagement, and shared decision-making. Medications and cardiac rhythm management (CRM) devices are recommended with a high level of evidence. However, there are significant disparities: patients who could benefit from devices are frequently referred too late or not at all. Misconceptions about device therapy and the notion that the needs of patients (especially the prevention of sudden cardiac death) can now be met by expanding drug therapies may play a role in these disparities. This state-of-the-art review is produced by members of the DIRECT HF initiative, a patient-centred, expert-led educational programme that aims to advance guideline-directed use of CRM devices in patients with HFrEF. This review discusses the latest evidence on the role of CRM devices in reducing HFrEF mortality and morbidity, and provides practical guidance on patient referral, device selection, implant timing and patient-centred follow-up.
Collapse
Affiliation(s)
| | - Wilfried Mullens
- Ziekenhuis Oost Limburg, Genk, Belgium
- Hasselt University, Hasselt, Belgium
| | | | - Andrea M Russo
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | | | | | | | | | - Naoki Sato
- Kawaguchi Cardiovascular and Respiratory Hospital, Saitama, Japan
| | | | | | | | - Giuseppe M C Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, Italy
- IRCCS San Raffaele Roma, Rome, Italy
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Institute, City St George's, University of London, London, UK
| | - Shelley Zieroth
- Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| |
Collapse
|
80
|
Majee S, Sahni A, Pal JD, McIntyre EE, Mukherjee D. Understanding embolus transport and source to destination mapping of thromboemboli in hemodynamics driven by left ventricular assist device. Sci Rep 2025; 15:12150. [PMID: 40204915 PMCID: PMC11982188 DOI: 10.1038/s41598-025-88653-6] [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: 10/02/2024] [Accepted: 01/29/2025] [Indexed: 04/11/2025] Open
Abstract
Left Ventricular Assist Devices (LVADs) are a key treatment option for patients with advanced heart failure, but they carry a significant risk of thromboembolic complications. While improved LVAD design, and systemic anticoagulation regimen, have helped mitigate thromboembolic risks, ischemic stroke due to adverse thromboembolic events remains a major concern with current LVAD therapies. Improved understanding of embolic events, and embolus movement to the brain, is critical to develop techniques to minimize risks of occlusive embolic events such as a stroke after LVAD implantation. Here, we address this need, and devise a quantitative in silico framework to characterize thromboembolus transport and distrbution in hemodynamics driven by an operating LVAD. We conduct systematic numerical experiments to establish that our framework can quantify the source-to-destination transport patterns of thromboemboli as a function of: LVAD outflow graft anastomosis, LVAD operating pulse modulation, thromboembolus sizes, and origin locations of emboli. Additionally, we demonstrate how the resulting embolus distribution patterns compare and correlate with descriptors based solely on hemodynamic patterns such as helicity, vorticity, and wall shear stress. Using the concepts of size-dependent embolus-hemodynamics interactions, and jet impingement driven flow for hemodynamics under LVAD operation as established in our prior works, we gain valuable insights on departure of thromboembolus distribution from flow distribution, and establish that our in silico model can generate deep insights into embolus dynamics which is not otherwise available from standard of care imaging and clinical data.
Collapse
Affiliation(s)
- Sreeparna Majee
- Paul M Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, USA
| | - Akshita Sahni
- Paul M Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, USA
| | - Jay D Pal
- Department of Surgery, University of Washington Seattle, Seattle, USA
| | - Erin E McIntyre
- Division of Cardiothoracic Surgery, University of Colorado, Anschutz Medical Campus, Colorado, USA
| | - Debanjan Mukherjee
- Paul M Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, USA.
| |
Collapse
|
81
|
Hou D, Hu Y, Yun T, Yu D, Yang G. USP7 promotes PINK1/Parkin-dependent mitophagy to ameliorate cerebral ischemia-reperfusion injury by deubiquitinating and stabilizing SIRT1. Brain Res 2025; 1858:149638. [PMID: 40216374 DOI: 10.1016/j.brainres.2025.149638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Cerebral ischemia-reperfusion (CI/R) injury, a major complication of ischemic stroke, is characterized by mitochondrial dysfunction and neuronal apoptosis, and understanding its underlying molecular mechanisms is essential for the development of effective therapeutic strategies. This study aimed to investigate the role of ubiquitin-specific protease 7 (USP7) in CI/R injury and elucidate its regulatory mechanisms. METHODS A rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) and an in vitro neuronal model subjected to oxygen-glucose deprivation/reperfusion (OGD/R) were used to mimic CI/R injury. USP7 was overexpressed or knocked down, with or without co-treatment, using the autophagy inhibitor 3-methyladenine (3-MA). Neurological function was evaluated using standardized scoring systems, and cerebral infarct volume was quantified by TTC staining. Histopathological changes in the cortex and hippocampus were assessed using hematoxylin-eosin (HE) and Nissl staining. Neuronal viability and apoptosis were measured by CCK-8 assay, TUNEL staining, and flow cytometry. To assess cellular metabolism and oxidative stress, ATP and LDH levels, along with antioxidant markers (including SOD, GSH, and GSH-Px), were analyzed using commercial biochemical kits. Mitochondrial morphology and autophagosome formation were visualized using transmission electron microscopy. Gene and protein expression levels were quantified by qRT-PCR and Western blotting, respectively. Immunofluorescence microscopy was performed to evaluate the subcellular localization of target proteins and co-localization with mitochondrial membrane markers. Lastly, protein-protein interactions and ubiquitination modification were analyzed by co-immunoprecipitation assays. RESULTS USP7 overexpression significantly alleviated neurological deficits, reduced infarct volume, attenuated histological damage, and decreased neuronal apoptosis in the MCAO/R model. Similarly, in the OGD/R model, USP7 overexpression markedly enhanced neuronal viability, suppressed apoptosis, restored ATP production, improved antioxidant capacity (as evidenced by increased levels of SOD, GSH, and GSH-Px), and reduced LDH release. Mechanistically, USP7 stabilized SIRT1 protein expression through deubiquitination, which in turn activated the PINK1/Parkin pathway and enhanced mitophagy. This activation was demonstrated by an increased LC3II/LC3I ratio, elevated ATG5 expression, enhanced co-localization of Tomm20 and Parkin, and increased autophagosome formation. Moreover, these protective effects were abolished when either 3-MA treatment was applied or SIRT1/PINK1 expression was knocked down. CONCLUSION USP7 mitigates CI/R injury by promoting PINK1/Parkin-dependent mitophagy through SIRT1 deubiquitination and stabilization, suggesting USP7 as a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Dan Hou
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Yujie Hu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Tian Yun
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Dan Yu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China.
| | - Guoshuai Yang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China.
| |
Collapse
|
82
|
Marinho-Buzelli AR, Vijayakumar A, Wasilewski MB, Linkewich E, Hitzig SL. Implementation considerations of aquatic therapy post-stroke: A qualitative study from Ontario, Canada. Complement Ther Clin Pract 2025; 60:101984. [PMID: 40267747 DOI: 10.1016/j.ctcp.2025.101984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND AND PURPOSE Aquatic therapy is an effective alternative approach to therapy on land in facilitating mobility and independence after a stroke. Despite the evidence reporting on the effectiveness of many aquatic interventions, there is a lack of studies elucidating essential implementation factors for aquatic therapy post-stroke. MATERIALS AND METHODS A purposive sample of people with lived experience of stroke and healthcare providers participated in semi-structured interviews about their experiences with aquatic therapy. A deductive content analysis approach using the Consolidated Framework for Implementation Research (CFIR) was used to identify the essential aspects of aquatic therapy implementation post-stroke. RESULTS Twenty-three participants (9 people with stroke experience and 14 healthcare providers) participated in the study, from across the province of Ontario in Canada. The interview data were all relevant to the five main CFIR constructs and to some of the sub-constructs. Although participants were highly favorable to aquatic therapy, several implementation considerations in terms of costs, program structure, provider qualifications, and pool access were identified at the patient, provider, and system level. CONCLUSION The CFIR proved valuable in identifying contextual factors relevant to implementing aquatic therapy with the stroke population. These factors included the importance of education, barriers to accessing aquatic therapy in rehabilitation and community settings, and internal and external factors influencing the application of aquatic therapy post-stroke.
Collapse
Affiliation(s)
- Andresa R Marinho-Buzelli
- St. John's Rehab Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.
| | - Abirami Vijayakumar
- St. John's Rehab Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Marina B Wasilewski
- St. John's Rehab Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Occupational Science and Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Elizabeth Linkewich
- Department of Occupational Science and Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; University Hospital, London Health Sciences Centre, Canada; School of Occupational Therapy, Faculty of Health Sciences, University of Western Ontario, Canada
| | - Sander L Hitzig
- St. John's Rehab Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Occupational Science and Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
83
|
Zhang B, Lai J, Li D, Li Y, Wang P, Cai S, Ren Q, Li D. Inverse association between triglyceride-glucose index and maximal oxygen uptake in US young and middle-aged population: a cross-sectional study. Front Cardiovasc Med 2025; 12:1583614. [PMID: 40271127 PMCID: PMC12014587 DOI: 10.3389/fcvm.2025.1583614] [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: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
Background The triglyceride-glucose (TyG) index has been linked to impaired cardiovascular fitness (CVF). However, the available evidence regarding the direct relationship between the TyG index and maximal oxygen uptake (VO2max) is limited. This study aims to investigate the association between the TyG index and VO2max. Methods We conducted a retrospective cross-sectional study involving 3,571 participants who completed a CVF examination as part of the National Health and Nutrition Examination Survey (NHANES) 1999-2004. Data on triglycerides, glucose, and VO2max were collected from all participants. The TyG index was calculated using the formula: Ln[triglyceride (TG)(mg/dl) × fasting plasma glucose (FPG)(mg/dl)/2]. Linear regression analysis was utilized to substantiate the research objectives. Results The complex sampling design and mobile examination center sample weights were considered. In multivariable linear regression analyses, each 1 unit increase in the TyG index was associated with a decrease in VO2max [β = -1.24, 95% CI (-1.97, -0.51), p = 0.002] when expressed as a continuous variable, independent of confounders. The TyG index was converted into a categorical variable based on four quartiles. Compared with the lowest TyG quintile (Q1: 6.750-7.887), the fully adjusted β for Q4 (8.672-12.481) was -1.91 (95% CI: -3.24, -0.57, p < 0.007). A significant interaction (p = 0.007) between sex and the TyG index for VO2max was found in the population using subgroup analysis. The results of the sensitivity analysis remained stable. Mediation analysis showed the direct effect of the TyG index was -1.467 (-2.019, -0.948), with a total effect of -1.813 (-2.377, -1.286). The mediation effect of diastolic blood pressure (DBP), white blood cell count (WBC), and C-reactive protein (CRP) was -0.389 (-0.526, -0.268), -0.308 (-0.432, -0.177), and -0.252 (-0.453, -0.135), respectively. HGB was found to exert a suppressing effect on the relationship between the TyG index and VO2max, with a value of 1.469 (1.252, 1.702). The p-values for all the above effects were <0.05. Conclusions In the US young and middle-aged population, the TyG index was significantly adversely associated with VO2max levels. Females may exert an interaction on TyG. Evidence supported DBP, WBC, and CRP as intervening variables through which the TyG index exerts its influence on VO2max. HGB may overrule the potential inverse association between the TyG index and VO2max.NCHS IRB/ERB Protocol Number: Protocol #98-12.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junxing Lai
- Department of Cardiovascular Disease and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
| | - Dan Li
- Department of Electrocardiogram, Jiangmen Central Hospital, Jiangmen, China
| | - Yongfeng Li
- Department of Medical Records, Jiangmen Central Hospital, Jiangmen, China
| | - Peng Wang
- Department of Information, Jiangmen Central Hospital, Jiangmen, China
| | - Shangan Cai
- Department of Urology, Jiangmen Central Hospital, Jiangmen, China
| | - Qiang Ren
- Department of Cardiovascular Disease and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
| | - Dong Li
- Department of Intensive Care Unit, Jiangmen Central Hospital, Jiangmen, China
| |
Collapse
|
84
|
Zhang J, Li Y, Xiang Z, Pu H, Ji C, Ren X, Fu D, Wang Y. In Situ H 2S-Releasing Stents Optimize Vascular Healing. ACS NANO 2025; 19:12864-12882. [PMID: 40159867 DOI: 10.1021/acsnano.4c16345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Stent implantation remains a cornerstone of interventional cardiology, providing a minimally invasive solution to restore blood flow in occluded vessels. However, current stents face persistent challenges in simultaneously preventing neointimal hyperplasia and promoting reendothelialization, compromising their long-term efficacy. To address these limitations, we developed an in situ H2S-releasing polymer brush-coated stent that actively modulates material-blood interactions, creating a favorable microenvironment for vascular healing. H2S enhances the stent's antithrombotic properties by inhibiting fibrinogen binding and platelet activation, while also mitigating oxidative stress and promoting macrophage polarization toward the anti-inflammatory M2 phenotype. In vivo, the H2S-releasing stents significantly improved vascular healing by accelerating endothelialization and inhibiting smooth muscle cell overproliferation, resulting in a thinner neointima with functional endothelial coverage. Transcriptomic analysis further elucidated the underlying mechanisms, revealing H2S-mediated modulation of key biological pathways that support vascular healing. These findings underscore the potential of in situ H2S release as an effective strategy for optimizing vascular implants and improving long-term outcomes.
Collapse
Affiliation(s)
- Jiayi Zhang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Yang Li
- Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhen Xiang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Hongxia Pu
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Cheng Ji
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Xingrong Ren
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Daihua Fu
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
85
|
Chen Q, Zheng A, Xu X, Shi Z, Yang M, Sun S, Wang L, Wang Y, Zhao H, Xiao Q, Zhang L. Nrf3-Mediated Mitochondrial Superoxide Promotes Cardiomyocyte Apoptosis and Impairs Cardiac Functions by Suppressing Pitx2. Circulation 2025; 151:1024-1046. [PMID: 40099370 DOI: 10.1161/circulationaha.124.070286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/31/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Myocardial infarction (MI) elicits mitochondria reactive oxygen species (ROS) production and cardiomyocyte (CM) apoptosis. Nrf3 (nuclear factor erythroid 2-related factor 3) has an established role in regulating redox signaling and tissue homeostasis. Here, we aimed to evaluate the role and mechanism of Nrf3 in injury-induced pathological cardiac remodeling. METHODS Global (Nrf3-KO) and CM-specific (Nrf3△CM) Nrf3 knockout mice were subjected to MI or ischemia/reperfusion injury, followed by functional and histopathological analysis. Primary neonatal mouse and rat ventricular myocytes and CMs derived from human induced pluripotent stem cells were used to evaluate the impact of Nrf3 on CM apoptosis and mitochondrial ROS production. Chromatin immunoprecipitation sequencing and immunoprecipitation-mass spectrometry analysis were used to uncover potential targets of Nrf3. MitoParaquat administration and CM-specific adeno-associated virus vectors were used to further confirm the in vivo relevance of the identified signal pathways. RESULTS Nrf3 was expressed mainly in CMs in healthy human hearts, and an increased level of Nrf3 was observed in CMs within the border zone of infarcted human hearts and murine cardiac tissues after MI. Both global and CM-specific Nrf3 knockout significantly decreased injury-induced mitochondrial ROS production, CM apoptosis, and pathological cardiac remodeling, consequently improving cardiac functions. In addition, cardiac-specific Nrf3 overexpression reversed the ameliorative cardiac phenotypes observed in Nrf3-KO mice. Functional studies showed that Nrf3 promoted neonatal mouse ventricular myocyte, neonatal rat ventricular myocyte, and CMs derived from human induced pluripotent stem cell apoptosis by increasing mitochondrial ROS production. Critically, augmenting mitochondrial ROS with MitoParaquat blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling. Mechanistically, a redox regulator Pitx2 (paired-like homeodomain transcription factor 2) was identified as one of the main target genes of Nrf3. Specifically, Nrf3 binds to Pitx2 promoter, where it increases DNA methylation through recruiting heterogeneous nuclear ribonucleoprotein K and DNA-methyltransferase 1 complex, thereby inhibiting Pitx2 expression. CM-specific knockdown of Pitx2 blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling, and cardiac-specific Pitx2 overexpression attenuated MI-induced mitochondrial ROS production and CM apoptosis, as well as preserved cardiac functions after MI. CONCLUSIONS Nrf3 promotes injury-induced CM apoptosis and deteriorates cardiac functions by increasing mitochondrial ROS production through suppressing Pitx2 expression. Targeting the Nrf3-Pitx2-mitochondrial ROS signal axis may therefore represent a novel therapeutic approach for MI treatment.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Ancheng Zheng
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Xiaolei Xu
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Zhenning Shi
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Mei Yang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Shasha Sun
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Leyu Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Yumeng Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Haige Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (H.Z.)
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Li Zhang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| |
Collapse
|
86
|
Ning Z, Jiang X, Huang H, Ma H, Luo J, Yang X, Zhang B, Liu Y. Machine learning integration of multimodal data identifies key features of circulating NT-proBNP in people without cardiovascular diseases. Sci Rep 2025; 15:12015. [PMID: 40199949 PMCID: PMC11978906 DOI: 10.1038/s41598-025-96689-x] [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: 10/04/2024] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
N-Terminal Pro-Brain Natriuretic Peptide (NT-proBNP) is important for diagnosing and predicting heart failure or many other diseases. However, few studies have comprehensively assessed the factors correlated with NT-proBNP levels in people with cardiovascular health. We used data from the 1999-2004 National Health and Nutrition Examination Survey (NHANES). Machine learning was employed to assess 66 factors that associated with NT-proBNP levels, including demographic, anthropometric, lifestyle, biochemical, blood, metabolic, and disease characteristics. The predictive power of the model was assessed using five-fold cross-validation. The optimal features predicting NT-proBNP levels were identified using univariate and step-forward multivariate models. Weighted least squares regression (WLS) was applied for supplementary analysis. Finally, the relationship between the corresponding features and NT-proBNP was validated using weighted and adjusted generalized additive models (GAM). We included 12, 526 participants without cardiovascular diseases. In the univariate model, age exhibited the highest association with NT-proBNP levels (the coefficient of determination (R2) = 36.91%). The multivariate models revealed that age, gender, red blood cell count, race/ethnicity, systolic blood pressure, and total protein level were the top six predictors of NT-proBNP. GAM demonstrated a noteworthy non-linear association between NT-proBNP and age, red blood cell count, systolic blood pressure, and total protein. Our study contributes to explaining the biological mechanisms of NT-proBNP and will facilitate the design of relevant cohort studies. We underscore the significance of assessing various population subgroups when employing NT-proBNP as a biomarker, and the need for developing innovative clinical algorithms to establish personalized levels.
Collapse
Affiliation(s)
- Zhiyuan Ning
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Xuanfei Jiang
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Huan Huang
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Honggang Ma
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Ji Luo
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Xiangyan Yang
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China
| | - Bing Zhang
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China.
| | - Ying Liu
- Department of Neurology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, 1558 Third Ring North Road, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
87
|
Yohe EO, Alonso A, Drane DL, Patel SS, Schwinne M, Epenge E, Gikelekele G, Herve E, Kavugho I, Tshengele N, Mampunza S, Mananga L, Zhao L, Qiu D, Stringer A, Saindane AM, Ikanga JN. Predictors of white matter hyperintensities in the elderly Congolese population. Front Aging Neurosci 2025; 17:1491477. [PMID: 40264461 PMCID: PMC12011861 DOI: 10.3389/fnagi.2025.1491477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction White matter hyperintensities (WMHs) are strongly linked to cardiovascular risk factors and other health conditions such as Alzheimer's disease. However, there is a dearth of research on this topic in low-income countries and underserved populations, especially in the Democratic Republic of Congo (DRC) where the population is aging rapidly with increasing cardiovascular risk factors and dementia-related diseases. This study evaluates health factors associated with WMH in the elderly Sub-Saharan Africa (SSA), specifically Congolese adults. Methods In a cross-sectional study of 77 people from the DRC, participants underwent neuroimaging to analyze WMHs volume and completed clinical evaluation, laboratory-based blood exams, self-reported questionnaires, and interviews. A simple linear regression model was conducted to test the association between WMHs and potential predictors (dementia, age, sex, hypertension, diabetes, tobacco abuse, stroke, high cholesterol, cardiovascular medication, and alcohol abuse). Stepwise selection and backward elimination analyses were performed to obtain the final model. Finally, a multiple linear regression model was conducted to assess the association between WMHs and variables retained in the final model (dementia, sex, and age). Results Of the 77 individuals, 47 (61%) had dementia, 40 (52.6%) were males, and the mean age was 73 years (± 8.0 years standard deviation). In simple linear regression models, WMHs was significantly associated with dementia (expβ1 = 1.75, 95% CI = 1.14-2.71, p-value = 0.01) though it had a weak association with age (expβ1 = 1.03, 95% CI = 1.00-1.05, p-value = 0.05) and sex (male) (expβ1 = 0.66, 95% CI = 0.43-1.01, p-value = 0.05). In multiple linear regression models, WMHs was statistically significantly associated with dementia (expβ1 = 1.97, 95% CI = 1.31-2.95, p-value =0.001), male sex (expβ2 = 0.54, 95% CI = 0.36-0.80, p-value = 0.003), and age (expβ3 = 1.03, 95% CI = 1.00-1.06, p-value = 0.03). However, WMHs was not significantly associated with common cardiovascular risk factors, such as high blood pressure, diabetes, tobacco use, obesity, and high cholesterol levels. Discussion WMHs is significantly associated with dementia, sex, and age in the Congolese population. Understanding these predictors may improve our ability to diagnose, assess, and develop preventative treatments for white matter disease in SSA/DRC populations, where neuroimaging is difficult to obtain.
Collapse
Affiliation(s)
- Emile Omba Yohe
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Daniel L. Drane
- Departments of Neurology and Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Saranya Sundaram Patel
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
- OneRehab, Dallas, TX, United States
| | - Megan Schwinne
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, United States
| | - Emmanuel Epenge
- Department of Neurology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Guy Gikelekele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Esambo Herve
- Department of Neurology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | | | - Nathan Tshengele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Samuel Mampunza
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Lelo Mananga
- Department of Neurology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Deqiang Qiu
- Department of Radiology and Imaging Sciences & Department of Biomedical Engineering, School of Medicine, Emory University, Atlanta, GA, United States
| | - Anthony Stringer
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Amit M. Saindane
- Departments of Radiology and Imaging Sciences and Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Jean N. Ikanga
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| |
Collapse
|
88
|
Xu C, Yu XH, Wang G, Luo W, Chen L, Xia XD. The m 7G methylation modification: An emerging player of cardiovascular diseases. Int J Biol Macromol 2025; 309:142940. [PMID: 40210060 DOI: 10.1016/j.ijbiomac.2025.142940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Cardiovascular diseases severely endanger human health and are closely associated with epigenetic dysregulation. N7-methylguanosine (m7G), one of the common epigenetic modifications, is present in many different types of RNA molecules and has attracted significant attention due to its impact on various physiological and pathological processes. Recent studies have demonstrated that m7G methylation plays an important role in the occurrence and development of multiple cardiovascular diseases. Application of small molecule inhibitors to target m7G modification mediated by methyltransferase-like protein 1 (METTL1) has shown potentiality in the treatment of cardiovascular diseases. In this review, we summarize the basic knowledge about m7G modification and discuss its role and therapeutic potential in diverse cardiovascular diseases, aiming to provide a theoretical foundation for future research and therapeutic intervention.
Collapse
Affiliation(s)
- Can Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Wei Luo
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Lei Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China.
| | - Xiao-Dan Xia
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| |
Collapse
|
89
|
Yang H, Huang C, Sawano M, Herrin J, Chen G, Li Z, Spatz ES, Krumholz HM, Lu Y. Systolic Blood Pressure and Cardiovascular Mortality in U.S. Adults Aged 80+ Taking Antihypertensive Medications. J Am Coll Cardiol 2025; 85:1455-1458. [PMID: 40100170 DOI: 10.1016/j.jacc.2025.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 03/20/2025]
Affiliation(s)
- Huanhuan Yang
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Chenxi Huang
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mitsuaki Sawano
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jeph Herrin
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Guochong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhihui Li
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Erica S Spatz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Harlan M Krumholz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA; Department of Health Policy and Management, Yale School of Public Health, New Haven, Connecticut, USA
| | - Yuan Lu
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
90
|
Peng X, Sun B, Tang C, Shi C, Xie X, Wang X, Jiang D, Li S, Jia Y, Wang Y, Tang H, Zhong S, Piao M, Cui X, Zhang S, Wang F, Wang Y, Na R, Huang R, Jiang Y, Zhang W, Xu J, Lin K, Guo J, Pan Z, Wang K, Zhao Q, Liu H, Yu B, Ji Y, Zhang J, Li S, Tian J. HMOX1-LDHB interaction promotes ferroptosis by inducing mitochondrial dysfunction in foamy macrophages during advanced atherosclerosis. Dev Cell 2025; 60:1070-1086.e8. [PMID: 39731912 DOI: 10.1016/j.devcel.2024.12.011] [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: 01/20/2024] [Revised: 08/27/2024] [Accepted: 12/04/2024] [Indexed: 12/30/2024]
Abstract
Advanced atherosclerosis is the pathological basis for acute cardiovascular events, with significant residual risk of recurrent clinical events despite contemporary treatment. The death of foamy macrophages is a main contributor to plaque progression, but the underlying mechanisms remain unclear. Bulk and single-cell RNA sequencing demonstrated that massive iron accumulation in advanced atherosclerosis promoted foamy macrophage ferroptosis, particularly in low expression of triggering receptor expressed on myeloid cells 2 (TREM2low) foamy macrophages. This cluster exhibits metabolic characteristics with low oxidative phosphorylation (OXPHOS), increasing ferroptosis sensitivity. Mechanically, upregulated heme oxygenase 1 (HMOX1)-lactate dehydrogenase B (LDHB) interaction enables Lon peptidase 1 (LONP1) to degrade mitochondrial transcription factor A (TFAM), leading to mitochondrial dysfunction and ferroptosis. Administration of the mitochondria-targeted reactive oxygen species (ROS) scavenger MitoTEMPO (mitochondrial-targeted TEMPO) or LONP1 inhibitor bortezomib restored mitochondrial homeostasis in foamy macrophages and alleviated atherosclerosis. Collectively, our study elucidates the cellular and molecular mechanism of foamy macrophage ferroptosis, offering potential therapeutic strategies for advanced atherosclerosis.
Collapse
Affiliation(s)
- Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Bin Sun
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chaohui Tang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Chengyu Shi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Xianwei Xie
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Dingsheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuo Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Ying Jia
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yani Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China
| | - Huifang Tang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang 421001, China
| | - Shan Zhong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Xiuru Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Shenghao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Fan Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Yan Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Ruisi Na
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China; Heilongjiang Province Key Laboratory of Research on Molecular Targeted Anti-Tumor Drugs, Harbin 150081, China
| | - Renping Huang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yanan Jiang
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin 150081, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Kaiyang Lin
- Department of Cardiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Junli Guo
- Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences, Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhenwei Pan
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin 150081, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Huibin Liu
- Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, 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, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| | - Yong Ji
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| | - Jian Zhang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Shuijie Li
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China; Heilongjiang Province Key Laboratory of Research on Molecular Targeted Anti-Tumor Drugs, Harbin 150081, China.
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| |
Collapse
|
91
|
Garcia MI, Dame K, Charwat V, Siemons BA, Finsberg H, Bhardwaj B, Yokosawa R, Goswami I, Bruckner D, Wall ST, Ford KA, Healy KE, Ribeiro AJS. Human induced pluripotent stem cell-derived cardiomyocytes and their use in a cardiac organ-on-a-chip to assay electrophysiology, calcium and contractility. Nat Protoc 2025:10.1038/s41596-025-01166-4. [PMID: 40195549 DOI: 10.1038/s41596-025-01166-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025]
Abstract
Cardiac organs-on-a-chip (OoCs) or microphysiological systems have the potential to predict cardiac effects of new drug candidates, including unanticipated cardiac outcomes, which are among the main causes for drug attrition. This protocol describes how to prepare and use a cardiac OoC containing cardiomyocytes differentiated from human induced pluripotent stem cells (hiPS cells). The use of cells derived from hiPS cells as reliable sources of human cells from diverse genetic backgrounds also holds great potential, especially when cultured in OoCs that are physiologically relevant culture platforms. To promote the broad adoption of hiPS cell-derived cardiac OoCs in the drug development field, there is a need to first ensure reproducibility in their preparation and use. This protocol aims to provide key information on how to reduce sources of variability during hiPS cell maintenance, differentiation, loading and maturation in OoCs. Variability in these procedures can lead to inconsistent purity after differentiation and variable function between batches of microtissues formed in OoCs. This protocol also focuses on describing the handling and functional assessment of cardiac microtissues using live-cell microscopy approaches to quantify parameters of cellular electrophysiology, calcium transients and contractility. The protocol consists of five stages: (1) thaw and maintain hiPS cells, (2) differentiate hiPS cell cardiomyocytes, (3) load differentiated cells into OoCs, (4) maintain and characterize loaded cells, and (5) evaluate and utilize cardiac OoCs. Execution of the entire protocol takes ~40 days. The required skills to carry out the protocol are experience with sterile techniques, mammalian cell culture and maintaining hiPS cells in a pluripotent state.
Collapse
Affiliation(s)
- M Iveth Garcia
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| | - Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Brian A Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Henrik Finsberg
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Bhavya Bhardwaj
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ryosuke Yokosawa
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Dylan Bruckner
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Samuel T Wall
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Kevin A Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, CA, USA
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
92
|
Wu L, Pang C, Zeng G, Liu D, Li C, Liu K, Huang P, Wang W, Zhu J, Dai Z, Qiu Z, Jiang Y. 10-year trajectories of triglyceride-glucose index and progression of vertebrobasilar artery stenosis: A multicenter hospital-based prospective longitudinal cohort study. Neuroscience 2025; 571:44-51. [PMID: 40021078 DOI: 10.1016/j.neuroscience.2025.02.054] [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: 11/05/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND The association between the triglyceride-glucose (TyG) index and the atherosclerosis has been validated by numerous evidences. However, the prospective relationship between long-term dynamic changes in the TyG index and the progression of vertebrobasilar artery (VBA) atherosclerotic plaques remained unclear. METHODS This multicenter, hospital-based, prospective longitudinal cohort study included 1,336 patients with suspected stroke from January 1, 2004 to December 31, 2022. Baseline characteristics and vascular examinations were collected and performed at baseline. Patients were followed up for 10 years. The latent class trajectory modeling method was used to analyze the TyG index trajectories over the follow-up period. Cox regression was used to analyze the association of the baseline and trajectory of the TyG index with the progression of VBA plaques. RESULTS During the follow-up (10.80 ± 2.30 years), VBA plaque progression was found in 175 participants. Cox regression analysis indicated there was a significant positive association between the baseline TyG index and the VBA plaque progression (HR 1.886, 95 % CI 1.166-3.050, P = 0.010). Two trajectories of the TyG index were identified in a total of 1,336 participants, the low-stable group and high-increasing group. Compared with the low-stable group, the odds ratio for the high-increasing group had a 3.156-fold (95 % CI 1.629-6.112, P = 0.001) risk of VBA plaque progression. CONCLUSIONS Our findings suggested that higher baseline and the high-increasing trajectory of the TyG index were associated with VBA plaque progression. People with high-increasing trajectories of the TyG index predispose to develop VBA plaque progression and deserve more attention and more aggressive preventive therapies.
Collapse
Affiliation(s)
- Li Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Chunmei Pang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China.
| | - Guanfeng Zeng
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Dezhi Liu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chenghao Li
- Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Kang Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Pengyue Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Wenbo Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215300, China.
| | - Zheng Dai
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Zhihua Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China.
| | - Yongjun Jiang
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi 214023, Jiangsu, China.
| |
Collapse
|
93
|
Halaseh R, Sun GK, Bhatt AS, Chang AJ, Svetlichnaya J, Adatya S, Fudim M, Greene SJ, Bensimhon DR, Adler ED, Alexy T, Biegus J, Sauer AJ, Pang PS, Collins SP, Pandey A, Butler J, Ambrosy AP. Outpatient worsening heart failure: innovative decongestion strategies and health equity implications. Heart Fail Rev 2025:10.1007/s10741-025-10509-y. [PMID: 40188318 DOI: 10.1007/s10741-025-10509-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Worsening heart failure (WHF) is a major clinical and economic challenge, contributing to high rates of hospitalization and significant healthcare costs. While WHF has traditionally been managed through hospitalization, recent approaches are shifting toward outpatient care to maximize patient time spent at home and optimize allocation of hospital resources. Emerging treatments like subcutaneous furosemide and intranasal bumetanide offer promising alternatives for safe, well-tolerated, and effective diuresis outside the hospital. However, these novel strategies face several challenges, including the need for clinician/staff training, patient education, logistical difficulties, and a lack of evidence in diverse populations. To ensure equitable management, it is also essential to address healthcare disparities, particularly in socioeconomically disadvantaged and rural populations. While these new treatments have the potential to improve care delivery, additional research is necessary to assess their comparative effectiveness and overcome current limitations fully.
Collapse
Affiliation(s)
- Rami Halaseh
- Department of Medicine, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Grace K Sun
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
| | - Ankeet S Bhatt
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Alex J Chang
- Department of Medicine, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Jana Svetlichnaya
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
| | - Sirtaz Adatya
- Department of Cardiology, Kaiser Permanente Santa Clara Medical Center, Santa Clara, CA, USA
| | - Marat Fudim
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Stephen J Greene
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | | | - Eric D Adler
- Division of Cardiology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Tamas Alexy
- Division of Cardiology, University of Minnesota, Minneapolis, MN, USA
| | - Jan Biegus
- Institute of Heart Diseases, WrocłAw Medical University, Wrocław, Poland
| | - Andrew J Sauer
- Saint Luke'S mid America Heart Institute, Kansas City, MO, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Peter S Pang
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sean P Collins
- Emergency Medicine, Education and Clinical Center (GRECC), Vanderbilt University Medical Center and Veterans Affairs Tennessee Valley Healthcare System, Geriatric Research, Nashville, TN, USA
| | - Ambarish Pandey
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Baylor, Scott, & White Research Institute, Dallas, TX, USA
| | - Javed Butler
- Baylor, Scott, & White Research Institute, Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| | - Andrew P Ambrosy
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA.
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA.
| |
Collapse
|
94
|
Li D, Cui TR, Liu JH, Shao WC, Liu X, Chen ZK, Xu ZG, Li X, Xu SY, Xie ZY, Jian JM, Wang X, Tao LQ, Wu XM, Cheng ZW, Dong ZR, Liu HF, Yang Y, Zhou J, Ren TL. Motion-unrestricted dynamic electrocardiogram system utilizing imperceptible electronics. Nat Commun 2025; 16:3259. [PMID: 40188239 PMCID: PMC11972297 DOI: 10.1038/s41467-025-58390-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/13/2025] [Indexed: 04/07/2025] Open
Abstract
Electrocardiogram (ECG) plays a vital role in the prevention, diagnosis, and prognosis of cardiovascular diseases (CVDs). However, the lack of a user-friendly and accurate long-term dynamic electrocardiogram (DCG) device in motion has made it challenging to perform many daily cardiovascular risk screenings and assessments, such as sudden cardiac arrest, resulting in additional economic burdens on society. Here, we present a motion-unrestricted dynamic electrocardiogram (MU-DCG) system, which employs skin-conformal, imperceptible electronics for long-term, comfortable, and accurate 12-lead DCG monitoring. To facilitate assembly for use on the skin, the MU-DCG system features a pressure-activated flexible skin socket for stably soft-connecting the on-skin soft module and the off-skin stiff module during dynamic movements. Crucially, blinded cardiologist evaluations confirm minimal motion artifacts in MU-DCG-acquired ECG signals. Our results demonstrate that the MU-DCG system, with large-area, ultra-thin on-skin electrodes/leads, and an off-skin module, accomplishes anti-motion interference acquisition and in-situ analysis while retaining wearing imperceptibility.
Collapse
Affiliation(s)
- Ding Li
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Tian-Rui Cui
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Jia-Hao Liu
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Wan-Cheng Shao
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Xiao Liu
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhi-Kang Chen
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Zi-Gan Xu
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Xin Li
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Shuo-Yan Xu
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Zi-Yi Xie
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin-Ming Jian
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Xu Wang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu-Qi Tao
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, China
| | - Xiao-Ming Wu
- School of Integrated Circuit, Tsinghua University, Beijing, China
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, China
| | - Zhong-Wei Cheng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zi-Rui Dong
- School of Integrated Circuit, Tsinghua University, Beijing, China
| | - Hou-Fang Liu
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, China.
| | - Yi Yang
- School of Integrated Circuit, Tsinghua University, Beijing, China.
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, China.
| | - Jun Zhou
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China.
| | - Tian-Ling Ren
- School of Integrated Circuit, Tsinghua University, Beijing, China.
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, China.
| |
Collapse
|
95
|
Gao J, Wang X, Wan Q. One nanoparticle delivers two different neuroprotective amino acids into ischemic brain and protects against neuronal death in rat cerebral ischemia injury. Mol Cell Neurosci 2025; 133:104009. [PMID: 40194569 DOI: 10.1016/j.mcn.2025.104009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Previous studies have proven that glycine and proline are neuroprotective but have very low permeability through the blood-brain barrier (BBB), which is a major barrier to the application of these neuroprotective amino acids in the therapy of brain injury. In this study, we aimed to develop a therapeutic strategy by which one chitosan nanoparticle could deliver two different neuroprotective amino acids, glycine and proline, into the rat ischemic brain to confer neuroprotection in a rat model of cerebral ischemia-reperfusion (I/R) injury. Using the ion cross-linking method, we developed a preparation in which one chitosan nanoparticle was simultaneously loaded with glycine and proline (AA-NPs). We evaluated the therapeutic potential of AA-NPs in both cell and animal models of cerebral ischemic stroke. We found that the levels of glycine and proline were decreased in the brain tissues of I/R rats. AA-NPs delivered both glycine and proline into the ischemic brain and reduced ischemic neuronal death in both in vitro and in vivo. These results indicated that the dual delivery of glycine and proline via AA-NPs mediated neuroprotective effects, as evidenced by the reduction of neuronal death in both cellular and animal models of ischemic stroke. AA-NPs provide an efficient and potential delivery strategy by which multiple neuroprotective amino acids can be transported into the ischemic brain simultaneously for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jingchen Gao
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Xiyuran Wang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Qi Wan
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
96
|
Enogela EM, Bowling CB, Levitan EB, Safford MM, Sterling MR. Reserve and Resilience: A Framework to Inform Cardiovascular Disease Outcomes Research Among Older Adults. Circ Cardiovasc Qual Outcomes 2025:e011396. [PMID: 40184149 DOI: 10.1161/circoutcomes.124.011396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Affiliation(s)
- Ene M Enogela
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham (E.M.E., E.B.L.)
| | - C Barrett Bowling
- Durham Veterans Affairs Geriatric Research Education and Clinical Center, Durham Veterans Affairs Medical Center, NC (C.B.B.)
- Department of Medicine, Duke University School of Medicine, Durham, NC (C.B.B.)
| | - Emily B Levitan
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham (E.M.E., E.B.L.)
| | - Monika M Safford
- Division of General Internal Medicine, Weill Medical College of Cornell University, New York, NY (M.M.S., M.R.S.)
| | - Madeline R Sterling
- Division of General Internal Medicine, Weill Medical College of Cornell University, New York, NY (M.M.S., M.R.S.)
| |
Collapse
|
97
|
Hochrainer K, Hansen A, Garcia-Bonilla L. Keep your guard up: blood-brain barrier protection by empagliflozin after acute ischaemic stroke. Cardiovasc Res 2025:cvaf049. [PMID: 40177812 DOI: 10.1093/cvr/cvaf049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Affiliation(s)
- Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Ashley Hansen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| |
Collapse
|
98
|
Kang CM, Zhao JJ, Xie XX, Yu KW, Lai BC, Wang YX, Li TT, Ke PF, Huang XZ. Unveiling the role of GATA4 in endothelial cell senescence and atherosclerosis development. Atherosclerosis 2025; 404:119183. [PMID: 40209341 DOI: 10.1016/j.atherosclerosis.2025.119183] [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: 08/05/2024] [Revised: 03/17/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND AND AIMS Cellular senescence is intimately linked to atherosclerosis development and progression. However, the mechanism is not well known. GATA4 is a classical regulator in human fibroblast senescence. This study aimed to determine the role of GATA4 in endothelial cell (EC) senescence and atherosclerosis development and the mechanisms by which it acts. METHODS Senescence ECs were induced using H2O2 by isolating human primary umbilical vein ECs from umbilical veins. The level of GATA4 was examined in endothelial progenitor cells (EPCs), ECs of arterial tissue from older individuals (>65 years), and aged mice (>24 months). Adeno-associated virus with EC-selective Tie1 promoter, an EC-specific gene transduction system, was used to explore the role of GATA4 in EC senescence and atherosclerosis development in ApoE-/- mice. RT-qPCR, Western blot, ChIP-PCR, and ELISA were conducted to further explore the mechanism of GATA4 in EC senescence and atherosclerosis development. RESULTS GATA4 protein levels are elevated in EC senescence induced by H2O2 and EPCs in older individuals. Additionally, GATA4 protein levels are increased in the ECs of arterial tissue from older individuals and aged mice and are strongly correlated with the progression of atherosclerosis plaques. Knockdown of GATA4 decreased EC senescence, dysfunction, and monocyte adhesion. Mechanistically, we found that GATA4 activates NFκB2 transcription and induces senescence-associated secretory phenotype (SASP) expression (IL-6, IL-8, CXCL1, CXCL3, ICAM-1). In vivo experiments on ApoE-/- mice demonstrated that GATA4 overexpression in ECs contributes to higher SASP expression, vascular senescence, atherosclerotic plaque formation, and impaired cardiac function. CONCLUSIONS Taken together, our findings indicate that elevated EC GATA4 levels contribute to the progression of atherosclerosis through the GATA4-NFκB2-SASP pathway, suggesting potential therapeutic targets for atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Chun-Min Kang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Jing-Jing Zhao
- Department of Laboratory Medicine, Nanfang Hospital Affiliated to Southern Medical University, Guangdong, 510515, China
| | - Xi-Xi Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Ke-Wei Yu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Bai-Cong Lai
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Yun-Xiu Wang
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Ting Ting Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Pei-Feng Ke
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xian-Zhang Huang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China; Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
99
|
Li J, Tang X, Wang X. LT-YOLO: long-term temporal enhanced YOLO for stenosis detection on invasive coronary angiography. Front Mol Biosci 2025; 12:1558495. [PMID: 40242408 PMCID: PMC12001240 DOI: 10.3389/fmolb.2025.1558495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 04/18/2025] Open
Abstract
Coronary artery stenosis detection by invasive coronary angiography plays a pivotal role in computer-aided diagnosis and treatment. However, it faces the challenge of stenotic morphology confusion stemming from coronary-background similarity, varied morphology, and small-area stenoses. Furthermore, existing automated methods ignore long-temporal information mining. To address these limitations, this paper proposes a long-term temporal enhanced You Only Look Once (YOLO) method for automatic stenosis detection and assessment in invasive coronary angiography. Our approach integrates long-term temporal information and spatial information for stenosis detection with state-space models and YOLOv8. First, a spatial-aware backbone based on a dynamic Transformer and C2f Convolution of YOLOv8 combines the local and global feature extraction to distinguish the coronary regions from the background. Second, a spatial-temporal multi-level fusion neck integrates the long-term temporal and spatial features to handle varied stenotic morphology. Third, a detail-aware detection head leverages low-level information for accurate identification of small stenoses. Extensive experiments on 350 invasive coronary angiography (ICA) video sequences demonstrate the model's superior performance over seven state-of-the-art methods, particularly in detecting small stenoses (<50%), which were previously underexplored.
Collapse
Affiliation(s)
- Jiaxin Li
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, China
| | - Xiang Tang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, China
| | - Xuesong Wang
- College of Mining Engineering, University of Science and Technology Liaoning, Anshan, China
| |
Collapse
|
100
|
Wang CH, Wu CY, Tay J, Wu MC, Ho LT, Lin WH, Lin JJ, Yeh HF, Tsai CL, Huang CH, Chen WJ. Association between post-arrest 12-lead electrocardiographic features and neurologically intact survival for patients of in-hospital cardiac arrest. Intern Emerg Med 2025:10.1007/s11739-025-03936-0. [PMID: 40172790 DOI: 10.1007/s11739-025-03936-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
Twelve-lead electrocardiogram (ECG) may provide prognostic information for in-hospital cardiac arrest (IHCA). This study aimed to identify post-arrest ECG features and their temporal changes associated with IHCA outcomes. This single-center retrospective study included patients experiencing IHCA between 2005 and 2022. Post-arrest ECGs were obtained within 48 h after an IHCA, admission ECGs upon hospital admission, and pre-arrest ECGs within 72 h before an IHCA. Multivariable logistic regression analyses were conducted to identify ECG features associated with neurologically intact survival. A total of 708 patients were included, with 131 (18.5%) achieving neurologically intact survival. The median age was 70.4 years (interquartile range: 59.2-82.6), and 362 (62.7%) patients were male. Four post-arrest ECG features were associated with survival: sinus rhythm (odds ratio [OR]: 1.81, 95% confidence interval [CI]: 1.11-2.93), QRS duration between 80 and 120 ms (OR: 1.91, 95% CI 1.19-3.08), low QRS voltage (OR: 0.50, 95% CI 0.25-0.99), and prolonged QTc (OR: 1.89, 95% CI 1.08-3.28). Comparing with admission ECGs, new-onset right bundle branch block (OR: 0.39, 95% CI 0.16-0.95) and increases in the number of leads with ST depression (OR: 0.85, 95% CI 0.77-0.94) on post-arrest ECGs were inversely associated with survival. Compared with pre-arrest ECGs, increases in the number of leads with ST depression (OR: 0.91, 95% CI 0.88-0.96) on post-arrest ECGs were also inversely associated with survival. Post-arrest ECGs may serve as a valuable prognostic tool for IHCA. Further exploration is warranted to determine whether incorporating these ECG features can enhance the performance of prediction models for IHCA outcomes.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
- Department of Emergency Medicine, College of Medicine, National Taiwan University, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
- Department of Emergency Medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Cheng-Yi Wu
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Joyce Tay
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Meng-Che Wu
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Li-Ting Ho
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Wei-Han Lin
- Department of Emergency Medicine, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Jr-Jiun Lin
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
- Department of Emergency Medicine, College of Medicine, National Taiwan University, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Huang-Fu Yeh
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Chu-Lin Tsai
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.).
- Department of Emergency Medicine, College of Medicine, National Taiwan University, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.).
| | - Chien-Hua Huang
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
- Department of Emergency Medicine, College of Medicine, National Taiwan University, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.)
| | - Wen-Jone Chen
- Department of Emergency Medicine, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.).
- Department of Emergency Medicine, College of Medicine, National Taiwan University, No.7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100, Taiwan (R.O.C.).
- Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan.
| |
Collapse
|