1
|
Li G, He W, Wang DW. Immune cell dynamics in heart failure: implicated mechanisms and therapeutic targets. ESC Heart Fail 2025; 12:1739-1758. [PMID: 39905753 PMCID: PMC12055366 DOI: 10.1002/ehf2.15238] [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: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
The relationship between heart failure (HF) and immune activation has garnered significant interest. Studies highlight the critical role of inflammation in HF, affecting cardiac structure and function. Despite promising anti-inflammatory therapies, clinical trials have faced challenges, indicating an incomplete understanding of immune mechanisms in HF. Immune cells, which are key cytokine sources, are pivotal in HF progression. In this review, the authors provide a comprehensive overview of the complex role of different types of immune cells and their cell subtypes in HF. In addition, the authors summarize the available targets and animal experimental evidence for targeting immune cells for the treatment of HF. Future research directions will focus on the roles of immune cells and their interrelationships at different stages of HF, aiming to develop more targeted therapeutic strategies that can achieve more precise interventions in the pathological process of HF.
Collapse
Affiliation(s)
- Gen Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| |
Collapse
|
2
|
Kallikourdis M, Cochran JD, Walsh K, Condorelli G. Contributions of Noncardiac Organ-Heart Immune Crosstalk and Somatic Mosaicism to Heart Failure: Current Knowledge and Perspectives. Circ Res 2025; 136:1208-1232. [PMID: 40403105 PMCID: PMC12113531 DOI: 10.1161/circresaha.125.325489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is the final outcome of most cardiovascular diseases and shares risk factors with other cardiovascular pathologies. Among these, inflammation plays a central role in disease progression and myocardial remodeling. Over the past 2 decades, numerous studies have explored immune-related mechanisms in cardiovascular disease, highlighting the importance of immune cross-talk between the heart and extra-cardiac organs, including bone marrow, spleen, liver, gut, and adipose tissue. This review examines how immune interactions among these organs contribute to heart failure pathogenesis, with a focus on clonal hematopoiesis, an age-related alteration of hematopoietic stem cells that fosters pathological bone marrow-heart communication. Additionally, we explore recent advances in the understanding of clonal hematopoiesis and its role in heart failure, emphasizing its implications for prognosis and potential therapeutic interventions. By integrating insights from immunology, metabolism, and aging, we provide a comprehensive perspective on the immunologic determinants of heart failure, paving the way for precision medicine approaches aimed at mitigating cardiovascular risk.
Collapse
Affiliation(s)
- Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele (MI), Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano (MI), Italy
| | - Jesse D. Cochran
- Hematovascular Biology Center, Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Kenneth Walsh
- Hematovascular Biology Center, Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele (MI), Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano (MI), Italy
| |
Collapse
|
3
|
Isath A, Mehra MR. Cardio-systemic Stress and Trained Innate Immune Memory in Heart Failure. Transplantation 2025; 109:565-568. [PMID: 40013925 DOI: 10.1097/tp.0000000000005278] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Affiliation(s)
- Ameesh Isath
- Center for Advanced Heart Disease, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
4
|
Huang L, Zhao X, Wang J, Guan J, Huang B, Feng J, Li X, Zhang Y, Zhang J. Gut microbiota and risk of heart failure in European population-A comprehensive Mendelian randomization study. ESC Heart Fail 2025. [PMID: 40098322 DOI: 10.1002/ehf2.15267] [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: 07/24/2023] [Revised: 06/01/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
AIMS Gut dysbiosis is proven to be involved in the pathogenesis and progression of heart failure (HF). Hindering the detrimental effects of gut-heart axis is an emerging trend. Our goal is to investigate the causal relationship between gut microbiota and HF, with the aim of facilitating future exploration of microbiome-targeted approaches to prevent and delay the progression of HF. METHODS AND RESULTS Two-sample Mendelian randomization (MR) analysis was applied to investigate the causal association of the gut microbiome with HF among individuals of European ancestry. Genetic variants associated with the 196 bacterial taxa from MiBioGen consortium were used as exposure data, summary statistics for HF derived from Heart Failure Molecular Epidemiology for Therapeutic Targets (HERMES) consortium were used as outcome data. Five MR methods were applied, including inverse variance weighted, maximum likelihood, MR-Egger, weighted median, and weighted mode. Reverse causality of instrumental variables (IVs) was tested by MR Steiger test of directionality. Strength of IVs was evaluated by F-statistics. Cochrane's Q test, MR-Egger regression analysis, and MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) tests were used to detect heterogeneity and pleiotropy. Leave-one-out method was used for testing the stability of results. Seven microbiomes were found to be associated with HF. Five of them were associated with higher risks of developing HF, these included Order_Selenomonadales (odds ratio [OR] = 1.11, P = 0.024), Family_Peptococcaceae (OR = 1.07, P = 0.045), Genus_Eubacterium eligens group (OR = 1.14, P = 0.022), Genus_Eubacterium oxidoreducens group (OR = 1.12, P = 0.011) and Genus_Flavonifractor (OR = 1.14, P = 0.012). Genus_Anaerostipes and Order_Bacillales were associated with lower risks of HF (OR = 0.90, P = 0.014; OR = 0.95, P = 0.042, respectively). Evidence of pleiotropy or heterogeneity was not observed. CONCLUSIONS We identified seven intestinal microbiomes that were causally associated with HF at the level of gene prediction. This study will help with the discovery of potential preventive and therapeutic targets for HF.
Collapse
Affiliation(s)
- Liyan Huang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xuemei Zhao
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jing Wang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jingyuan Guan
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Boping Huang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiayu Feng
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xinqing Li
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yuhui Zhang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jian Zhang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Key Laboratory of Clinical Research for Cardiovascular Medications, National Health Committee, Beijing, China
| |
Collapse
|
5
|
Long T, Yu Y, Cheng S, Huang H, Hua W. Prognostic value of the modified Model for End-Stage Liver Disease score in patients treated with cardiac resynchronization therapy. Heart Rhythm O2 2025; 6:339-349. [PMID: 40201669 PMCID: PMC11973686 DOI: 10.1016/j.hroo.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Background Hepatorenal dysfunction is prevalent among individuals with heart failure (HF). OBJECTIVE This study investigated prognostic value of the modified Model for End-Stage Liver Disease (Model for End-Stage Liver Disease excluding international normalized ratio [MELD-XI] scores and Model for End-Stage Liver Disease with albumin replacing international normalized ratio [MELD-Albumin]) score in patients undergoing cardiac resynchronization therapy (CRT). Methods We retrospectively evaluated 365 patients (mean age 58.7 ± 11.1 years; 64.9% men) undergoing CRT implantation between 2007 and 2019. Patients were divided into 4 groups based on the modified MELD score quartiles before CRT. The primary endpoint was the combination of all-cause mortality and HF hospitalization, whereas the secondary endpoint was CRT response at 6 months. Results During mean follow-up of 3.3 years (interquartile range 1.9-5.2 years), 168 patients reached the primary endpoint. Logistic regression revealed the MELD-Albumin score was independently associated with CRT response, even after adjusting for covariates (odds ratio 1.10; 95% confidence interval [CI] 1.02-1.19; P = .013). Kaplan-Meier analysis revealed that patients with a higher MELD-XI and MELD-Albumin score had a greater risk of adverse outcomes (log-rank test: P < .001). A Cox proportional hazards analysis showed that the modified MELD score remained significantly associated with adverse outcomes after adjusting for clinical and echocardiographic factors (MELD-XI: hazard ratio 1.06, 95% CI 1.02-1.11, P = .006; MELD-Albumin: hazard ratio 1.10, 95% CI 1.05-1.16, P < .001). Furthermore, receiver-operating characteristic analysis indicated that the MELD-Albumin score provided a stronger prognostic value for long-term adverse outcomes in patients undergoing CRT than the MELD-XI score (MELD-Albumin: area under the curve 0.692, 95% CI 0.644-0.742; MELD-XI: area under the curve 0.659, 95% CI 0.608-0.715; P = .008). Conclusion The MELD-Albumin score may be useful for stratifying patients at risk for CRT response and adverse outcomes in those undergoing CRT for HF.
Collapse
Affiliation(s)
| | | | - Sijing Cheng
- Cardiac Arrhythmia Center, Department of Cardiology, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Huang
- Cardiac Arrhythmia Center, Department of Cardiology, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Hua
- Cardiac Arrhythmia Center, Department of Cardiology, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Shore S, Li H, Zhang M, Whitney R, Gross AL, Bhatt AS, Nallamothu BK, Giordani B, Briceño EM, Sussman JB, Gutierrez J, Yaffe K, Griswold M, Johansen MC, Lopez OL, Gottesman RF, Sidney S, Heckbert SR, Rundek T, Hughes TM, Longstreth WT, Levine DA. Trajectory of Cognitive Function After Incident Heart Failure. Circ Heart Fail 2025; 18:e011837. [PMID: 39963777 PMCID: PMC11992552 DOI: 10.1161/circheartfailure.124.011837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 12/13/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND The magnitude of cognitive changes after incident heart failure (HF) is unclear. We assessed whether incident HF is associated with changes in cognition after accounting for pre-HF cognitive trajectories and known determinants of cognition. METHODS This pooled cohort study included adults without HF, stroke, or dementia from 6 US population-based studies from 1971 to 2019. Linear mixed-effects models estimated cognitive change with incident HF diagnosis and the rate of cognitive change over the years after HF, controlling for pre-HF cognitive trajectories and participant factors. Outcomes included change in global cognition (primary outcome), executive function, and memory (secondary outcomes). Cognitive outcomes were standardized to a t score metric (mean [SD], 50 [10]); a 1-point difference represented a 0.1-SD difference in cognition. RESULTS We included 29 614 adults (mean [SD] age was 61 [10] years, 55% female, 70% White). During a median follow-up of 6.6 (Q1-Q3, 5.0-19.8) years, 1407 (5%) adults received an incident diagnosis of HF. Incident HF diagnosis was associated with initial decreases in global cognition (-1.1 points [95% CI, -1.4 to -0.8]) and executive function (-0.6 points [95% CI, -1.0 to -0.3]). Larger decreases in global cognition after HF were seen with older age, female sex, and White race. Participants with incident HF diagnosis demonstrated faster and long-term declines in global cognition (-0.1 points per year [95% CI, -0.2 to -0.1]) and executive function (-0.2 points per year [95% CI, -0.2 to -0.1]). The change in memory with incident HF diagnosis was not statistically significant but showed a similar trend with an initial decline of -0.5 points (95% CI, -1.4 to +0.3) and a slope of -0.1 points per year (95% CI, -0.3 to 0.0). CONCLUSIONS In this pooled cohort study, incident HF diagnosis was associated with initial decreases in global cognition and executive function and faster, persistent declines in these domains at follow-up.
Collapse
Affiliation(s)
| | - Hanyu Li
- University of Michigan, Ann Arbor, MI, USA
| | - Min Zhang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | | | - Alden L. Gross
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ankeet S. Bhatt
- Kaiser Permanente San Francisco Medical Center and Division of Research, San Francisco, CA, USA
| | | | | | | | | | | | | | - Michael Griswold
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | - Rebecca F. Gottesman
- National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA
| | - Stephen Sidney
- Kaiser Permanente San Francisco Medical Center and Division of Research, San Francisco, CA, USA
| | | | - Tatjana Rundek
- University of Miami – Miller School of Medicine, Evelyn F. McKnight Brain Institute, FL, USA
| | | | | | | |
Collapse
|
7
|
Gou D, Min C, Peng X, Wu H, Zhang L, Chen Y, Tao M. Associating factors of cognitive frailty among older people with chronic heart failure: Based on LASSO-logistic regression. J Adv Nurs 2025; 81:1399-1411. [PMID: 39078209 DOI: 10.1111/jan.16352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024]
Abstract
AIM To analyse factors associated with cognitive frailty among older chronic heart failure patients in China. DESIGN A cross-sectional design. METHODS Between August 2021 and November 2022, a total of 421 chronic heart failure patients (age ≥60 years) were randomly selected from the cardiology department of the affiliated hospital of Zunyi Medical University. The FRAIL scale, Mini-Mental State Examination, 15-item Geriatric Depression Scale, Social Support Rating Scale, Short-form Mini Nutritional Assessment and Pittsburgh Sleep Quality Index were utilized for measurement and evaluation. The demographic and clinical characteristics of patients were collected. To select initial variables, the Least Absolute Shrinkage Selection Operator was applied, and then logistic regression analysis was used to confirm associating factors. RESULTS Among 421 elderly people with chronic heart failure, 83 cases (19.7%) showed cognitive frailty. Of 31 variables, seven were selected by Least Absolute Shrinkage Selection Operator regression. Finally, multivariate logistic regression revealed that the age, monthly salary, drinking, NYHA classification, length of hospital stay, depression and malnutrition risk/malnutrition were independently associated with cognitive frailty. CONCLUSION The high proportion of cognitive frailty in older people with chronic heart failure should be concerned. Additionally, in the setting of cognitive frailty, efforts to diagnose it and develop interventions to prevent or reverse cognitive frailty status among older chronic heart failure patients are necessary. IMPACT The findings of our study highlight the necessity to evaluate cognitive frailty in older people with chronic heart failure and provide a new perspective and scientific basis for medical staff to develop individualized and specific interventions to prevent or reverse cognitive frailty status. REPORTING METHOD This study has been reported in compliance with STROBE reporting guidelines for cross-sectional studies. PATIENT OR PUBLIC CONTRIBUTION No Patient or Public Contribution.
Collapse
Affiliation(s)
- Dengqun Gou
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Changhang Min
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Xiaofeng Peng
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Hemei Wu
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Lu Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Yu Chen
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| | - Ming Tao
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, P. R. China
| |
Collapse
|
8
|
Ahmad R, Khan S, Khan A, Naveed F, Arshad F. Critical insights on "Short-term outcomes in patients with heart failure". Int J Cardiol 2025; 422:132948. [PMID: 39756622 DOI: 10.1016/j.ijcard.2024.132948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Affiliation(s)
| | | | - Ayesha Khan
- Nishtar Medical University and Hospital, Multan, Pakistan
| | | | | |
Collapse
|
9
|
Chung CC, Kao YH, Chen YC, Lin YK, Higa S, Hsu KC, Chen YJ. PCSK9 Enhances Cardiac Fibrogenesis via the Activation of Toll-like Receptor and NLRP3 Inflammasome Signaling. Int J Mol Sci 2025; 26:1921. [PMID: 40076547 PMCID: PMC11900342 DOI: 10.3390/ijms26051921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a novel target for reducing low-density lipoprotein cholesterol. PCSK9 activates the atherosclerosis process through pro-inflammation signaling. Furthermore, the serum level of PCSK9 is positively correlated with mortality in patients with heart failure (HF). Cardiac fibrosis plays a crucial role in the pathophysiology of HF. In this study, we intended to examine whether PCSK9 can increase fibroblast activities and explore what its underlying mechanisms are. Migration, proliferation analyses, and Western blotting were used on human cardiac fibroblasts with and without PCSK9. Alirocumab (a PCSK9 inhibitor, 10 mg/kg/week intra-peritoneally for 28 consecutive days) was treated in isoproterenol (100 mg/kg, subcutaneous injection)-induced HF rats. PCSK9 (50, 100 ng/mL) increased proliferation, myofibroblast differentiation capability, and collagen type I production. Compared with control cells, PCSK9 (100 ng/mL)-treated cardiac fibroblasts showed higher nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), interleukin (IL)-1, myofibroblast differentiation, and collagen production capabilities, which were attenuated by MCC950 (an NLRP3 inhibitor, 100 μmol/L). PCSK9 upregulated Myd88 and NF-κB signaling, which were reduced by TAK242 (a toll-like receptor (TLR) 4 inhibitor, 10 μmol/L). Moreover, alirocumab significantly improved left ventricular systolic function and attenuated fibrosis in HF rats. In conclusion, PCSK9 upregulates NLRP3 signaling and the profibrotic activities of cardiac fibroblasts through the activation of TLR4/Myd88/NF-κB signaling.
Collapse
Affiliation(s)
- Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-C.C.); (Y.-K.L.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-C.C.); (Y.-K.L.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa 1199, Japan;
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Jen Chen
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-C.C.); (Y.-K.L.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| |
Collapse
|
10
|
Cui XY, Zhan JK. Capsaicin and TRPV1: A Novel Therapeutic Approach to Mitigate Vascular Aging. Aging Dis 2025:AD.2024.1292. [PMID: 39965247 DOI: 10.14336/ad.2024.1292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
Vascular aging and its associated diseases represent a principal cause of mortality among the global elderly population, making the mitigation of vascular aging a significant aspiration for humanity. This article explores the intersection of nature and health, focusing on the role of the natural plant, pepper, and its principal bioactive compound, capsaicin, in combating vascular aging. By examining molecular and cellular mechanisms as well as phenotypic alterations in blood vessels, we offer a comprehensive review of the effects of capsaicin and its receptor, transient receptor potential vanilloid 1 (TRPV1), within vascular aging. We propose that capsaicin may serve as the medication with the potential to slow the progress of vascular aging and could constitute a new strategy to treat vascular aging related disease.
Collapse
Affiliation(s)
- Xing-Yu Cui
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Jun-Kun Zhan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Huang S, Wu Y, Chen M, Shen J, Zhu J, Yu H. GDF11 improves cardiac repair after myocardial infarction by reducing Macrophage infiltration and attenuating their inflammatory Properties. Int Immunopharmacol 2025; 147:113994. [PMID: 39765001 DOI: 10.1016/j.intimp.2024.113994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/29/2025]
Affiliation(s)
- Shushi Huang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Yuling Wu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Mingyao Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Jiahua Shen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China
| | - Jinyun Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China.
| | - Hong Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; State Key Laboratory of Transvascular Implantation Devices, China; Heart Regeneration and Repair Key Laboratory of Zhejiang province, China; Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
12
|
Bianchi G, Vizio B, Bosco O, Schiavello M, Cagna Vallino P, Rumbolo F, Morello F, Mengozzi G, Montrucchio G, Lupia E, Pivetta E. miR-30d Levels Predict Re-Hospitalization in Patients with Acute Cardiogenic Pulmonary Edema: A Preliminary Study. Int J Mol Sci 2025; 26:1278. [PMID: 39941043 PMCID: PMC11818144 DOI: 10.3390/ijms26031278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Acute cardiogenic pulmonary edema (ACPE) is a common and serious manifestation of heart failure (HF), representing 10-20% of all acute HF admissions. It is associated with elevated in-hospital mortality and high rates of re-hospitalization. MicroRs, like miR-30d, are of particular interest in heart failure due to their regulatory role in gene expression and potential as biomarkers for diagnosing and predicting patient outcomes, especially in high-risk cases such as ACPE. We conducted a cohort study on patients diagnosed with ACPE in the Emergency Department (ED). The circulating levels of miR-30d were analyzed at the time of hospital admission and at one-month follow-up along with other biomarkers. We enrolled 24 ACPE patients and 10 control subjects. Median age was 80.8 years (interquartile range, IQR, 8.2) in ACPE cases, and 78.5 years (IQR 9.8) in controls with a male/female ratio of 2 and 0.66, respectively. In ACPE patients, median cardiac ejection fraction was 42.5%, creatinine 1.63 mg/dL (IQR 1.24), troponin 63.5 ng/dL (58), and NT-proBNP 4243.5 pg/mL (IQR 5846) at ED evaluation. Median concentration of miR30d was 0.81 in controls, and 3.67 and 7.28 in ACPE patients at enrollment time and one month later, respectively. Re-hospitalization occurred in 7 ACPE patients in the following 3 months, and in 9 during the following year. miR-30d had a significant predictive value in assessing the risk of re-hospitalization at both 3 months and 1 year following the initial diagnosis of ACPE, while it did not in assessing the risk of death at 1 year. When compared with the other biomarkers, none of them showed a better accuracy than miR-30d. Our findings suggest that elevated levels of miR-30d are associated with an increased rate of hospital readmission at both 3 months and 1 year after discharge. Larger, multicenter studies will be needed to confirm the validity of circulating miR-30d levels as a potential biomarker useful for risk prediction in ACPE patients and its utility in improving individualized patient care.
Collapse
Affiliation(s)
- Giordano Bianchi
- Division of Emergency Medicine and High Dependency Unit, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy; (G.B.); (P.C.V.); (F.M.); (E.L.)
| | - Barbara Vizio
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Ornella Bosco
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Martina Schiavello
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Paolo Cagna Vallino
- Division of Emergency Medicine and High Dependency Unit, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy; (G.B.); (P.C.V.); (F.M.); (E.L.)
- Residency Programme in Emergency Medicine, University of Turin, 10126 Turin, Italy
| | - Francesca Rumbolo
- Clinical Biochemistry Laboratory, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy;
| | - Fulvio Morello
- Division of Emergency Medicine and High Dependency Unit, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy; (G.B.); (P.C.V.); (F.M.); (E.L.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Giulio Mengozzi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
- Clinical Biochemistry Laboratory, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy;
| | - Giuseppe Montrucchio
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Enrico Lupia
- Division of Emergency Medicine and High Dependency Unit, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy; (G.B.); (P.C.V.); (F.M.); (E.L.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| | - Emanuele Pivetta
- Division of Emergency Medicine and High Dependency Unit, Città della Salute e della Scienza di Torino, Molinette Hospital, 10126 Turin, Italy; (G.B.); (P.C.V.); (F.M.); (E.L.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (B.V.); (O.B.); (M.S.); (G.M.); (G.M.)
| |
Collapse
|
13
|
Müller ML, Brand A, Mattig I, Spethmann S, Messroghli D, Hahn K, Violano M, Mitchell JD, Hare JM, Frustaci A, Klingel K, Lüscher TF, Landmesser U, Heidecker B. Myocardial Inflammation in Cardiac Transthyretin Amyloidosis: Prevalence and Potential Prognostic Implications. Circ Heart Fail 2025; 18:e012146. [PMID: 39866106 DOI: 10.1161/circheartfailure.124.012146] [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: 06/25/2024] [Accepted: 11/14/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND Despite previous histopathologic evidence for its presence, the role of myocardial inflammation in the development and progression of cardiac transthyretin amyloidosis (ATTR-CA) remains insufficiently understood. Thus, this study sought to characterize the prevalence and potential prognostic implications of myocardial inflammation in ATTR-CA. METHODS A retrospective observational study including patients with ATTR-CA diagnosed by endomyocardial biopsy was conducted. Myocardial inflammation was diagnosed through a review of routine endomyocardial biopsy reports. Baseline characteristics were compared using the Mann-Whitney U test and the Pearson χ2 test. Clinical outcomes were monitored via follow-up visits or telephone calls. Primary outcomes were all-cause death and a composite end point of all-cause death or heart failure hospitalization. Kaplan-Meier analyses, as well as univariable and age- and sex-adjusted multivariable Cox regression analyses, were used to assess differences in overall and composite end point-free survival between patients with ATTR-CA with and without myocardial inflammation. RESULTS A total of 103 patients with ATTR-CA (100 wild type; 3 variant) were enrolled. Median follow-up was 18.2 (8.0-31.1) months. Myocardial inflammation was prevalent in 32% (n=33/103) of patients with ATTR-CA. Among evaluable patients with myocardial inflammation, 96% (n=26/27) and 31% (n=9/29) had elevated CD68 (clusters of differentiation 68)-positive macrophage and CD3 (clusters of differentiation 3)-positive T-cell counts, respectively. Overall survival (P=0.017) and composite end point-free survival (P=0.014) were significantly impaired in patients with ATTR-CA with myocardial inflammation (n=33) compared with those without (n=70). Statistical significance for both associations was sustained after adjustment for age and sex, yielding adjusted hazard ratios of 4.72 (95% CI, 1.33-16.71; P=0.016) and 2.30 (95% CI, 1.04-5.11; P=0.041) for all-cause death and the composite end point, respectively. CONCLUSIONS Our findings affirm previous evidence that myocardial inflammation is present in approximately one-third of all patients with ATTR-CA. Moreover, we provide first data indicating that myocardial inflammation may be associated with a higher risk of death and heart failure hospitalizations in ATTR-CA.
Collapse
Affiliation(s)
- Maximilian Leo Müller
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
| | - Anna Brand
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (A.B., I.M., S.S., D.M., U.L.)
| | - Isabel Mattig
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (A.B., I.M., S.S., D.M., U.L.)
- Berlin Institute of Health at Charité, Germany (I.M., K.H., B.H.)
| | - Sebastian Spethmann
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (A.B., I.M., S.S., D.M., U.L.)
| | - Daniel Messroghli
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (A.B., I.M., S.S., D.M., U.L.)
- Medical Department of Cardiology, Rhythmology, and Angiology, Medical University Lausitz-Carl Thiem, Cottbus, Germany (D.M.)
| | - Katrin Hahn
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Germany (K.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- Berlin Institute of Health at Charité, Germany (I.M., K.H., B.H.)
| | - Michele Violano
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
| | - Joshua D Mitchell
- Cardiovascular Division, John T. Milliken Department of Internal Medicine, Cardio-Oncology Center of Excellence, Washington University in St. Louis, MO (J.D.M.)
| | - Joshua M Hare
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, FL (J.M.H.)
| | - Andrea Frustaci
- Cellular and Molecular Cardiology Laboratory, IRCCS (Scientific Institute for Research, Hospitalization, and Healthcare) L. Spallanzani, Rome, Italy (A.F.)
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Germany (K.K.)
| | - Thomas F Lüscher
- Royal Brompton & Harefield Hospitals, GSTT (Guy's and St Thomas' NHS Foundation Trust), Imperial College and Kings College, London, United Kingdom (T.F.L.)
- Center for Molecular Cardiology, Schlieren Campus, University Zurich, Switzerland (T.F.L.)
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (A.B., I.M., S.S., D.M., U.L.)
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (M.L.M., U.L., B.H., D.M., A.B., I.M., S.S.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin (M.L.M., A.B., I.M., S.S., D.M., K.H., M.V., U.L., B.H.)
- Amyloidosis Center Charité Berlin, Charité-Universitätsmedizin Berlin, Germany (M.L.M., A.B., I.M., S.S., D.M., K.H., B.H.)
- Berlin Institute of Health at Charité, Germany (I.M., K.H., B.H.)
| |
Collapse
|
14
|
Yu X, Zhang X, Bilal H, Shi C, Sun L. Exploring potential biomarkers for acute myocardial infarction by combining circadian rhythm gene expression and immune cell infiltration. Sci Rep 2025; 15:4012. [PMID: 39893248 PMCID: PMC11787365 DOI: 10.1038/s41598-025-88568-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025] Open
Abstract
Current diagnostic biomarkers for acute myocardial infarction (AMI), such as troponins, often lack specificity, leading to false positives under non-cardiac conditions. Recent studies have implicated circadian rhythm and immune infiltration in the pathogenesis of AMI. This study hypothesizes that analyzing the interplay between circadian rhythm-related gene expression and immune infiltration identify highly specific diagnostic biomarkers for AMI. Our results demonstrated differential expression of 15 circadian rhythm-related genes (CRGs) between AMI patients and healthy individuals, with five key genes-JUN, NAMPT, S100A8, SERPINA1, and VCAN identified as key contributors to this process. Functional enrichment analyses suggest these genes significantly influence cytokine and chemokine production in immune responses. Immune infiltration assessments using ssGSEA indicated elevated levels of neutrophils, macrophages, and eosinophils in AMI patients. Additionally, we identified potential therapeutic implications with 13 pivotal miRNAs and 10 candidate drugs targeting these genes. The Benjamini-Hochberg method was employed to adjust for multiple testing, and the results retained statistical significance. RT-qPCR analysis further confirmed the upregulation of these five genes under hypoxic conditions, compared to controls. Collectively, our findings highlight the critical role of CRGs in AMI, providing a foundation for improved diagnostic approaches and novel therapeutic targets.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaopeng Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Hazrat Bilal
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chang Shi
- Department of Pathology, First Affiliated Hospital, Dalian, Liaoning Province, China.
| | - Lei Sun
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
15
|
Schuetze KB, Stratton MS, Bagchi RA, Hobby ARH, Felisbino MB, Rubino M, Toni LS, Reges C, Cavasin MA, McMahan RH, Alexanian M, Vagnozzi RJ, McKinsey TA. BRD4 inhibition rewires cardiac macrophages toward a protective phenotype marked by low MHC class II expression. Am J Physiol Heart Circ Physiol 2025; 328:H294-H309. [PMID: 39716819 DOI: 10.1152/ajpheart.00438.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
Bromodomain and extraterminal domain (BET) proteins, including BRD4, bind acetylated chromatin and coactivate gene transcription. A BET inhibitor, JQ1, prevents and reverses pathological cardiac remodeling in preclinical models of heart failure. However, the underlying cellular mechanisms by which JQ1 improves cardiac structure and function remain poorly defined. Here, we demonstrate that BRD4 knockdown reduced expression of genes encoding CC chemokines in cardiac fibroblasts, suggesting a role for this epigenetic reader in controlling fibroblast-immune cell cross talk. Consistent with this, JQ1 dramatically suppressed recruitment of monocytes to the heart in response to stress. Normal mouse hearts were found to have approximately equivalent numbers of major histocompatibility complex (MHC-II)high and MHC-IIlow resident macrophages, whereas MHC-IIlow macrophages predominated following JQ1 treatment. Single-cell RNA-seq data confirmed that JQ1 treatment or BRD4 knockout in CX3CR1+ cells reduced MHC-II gene expression in cardiac macrophages, and studies with cultured macrophages further illustrated a cell autonomous role for BET proteins in controlling the MHC-II axis. Bulk RNA-seq analysis demonstrated that JQ1 blocked pro-inflammatory macrophage gene expression through a mechanism that likely involves repression of NF-κB signaling. JQ1 treatment reduced cardiac infarct size in mice subjected to ischemia/reperfusion. Our findings illustrate that BET inhibition affords a powerful pharmacological approach to manipulate monocyte-derived and resident macrophages in the heart. Such an approach has the potential to enhance the reparative phenotype of macrophages to promote wound healing and limit infarct expansion following myocardial ischemia.NEW & NOTEWORTHY BRD4 inhibition blocks stress-induced recruitment of pro-inflammatory monocytes to the heart. BRD4 inhibition reprograms resident cardiac macrophages toward a reparative phenotype marked by reduced NF-κB signaling and diminished MHC-II expression. BRD4 inhibition reduces infarct size in an acute model of ischemia/reperfusion injury in mice.
Collapse
Affiliation(s)
- Katherine B Schuetze
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Matthew S Stratton
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Rushita A Bagchi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Alexander R H Hobby
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Marina B Felisbino
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Marcello Rubino
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Lee S Toni
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Caroline Reges
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Maria A Cavasin
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Rachel H McMahan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Michael Alexanian
- Gladstone Institutes, San Francisco, California, United States
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, California, United States
- Department of Pediatrics, University of California, San Francisco, California, United States
| | - Ronald J Vagnozzi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
16
|
Peng S, Cai M, Kuang H, Lin A, Ma Q, Dai X, Luo P, Liu Y, Zhang G, Bai Y. From bench to bedside: elucidating VEGF(R) inhibitor-related heart failure in cancer treatment. J Transl Med 2025; 23:109. [PMID: 39849527 PMCID: PMC11756202 DOI: 10.1186/s12967-025-06133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) inhibitors play a pivotal role in treating various tumors; however, the clinical characteristics and molecular mechanisms of their associated heart failure (HF) remain incompletely understood. METHODS We investigated the epidemiological characteristics of VEGF or VEGFR inhibitors [VEGF(R)i]-related heart failure (VirHF) using the global pharmacovigilance database Vigibase. The phenotypic features and molecular mechanisms of VirHF were characterized using VEGF(R)i-treated mouse models through a combination of echocardiography, histopathological analysis, and transcriptome sequencing. Furthermore, we performed a retrospective analysis of cardiac function parameters in patients undergoing VEGF(R)i treatment at local hospitals. RESULTS In the analysis of 1871 VirHF cases, elderly patients (≥ 65 years) and female subjects demonstrated an elevated risk of occurrence. Experimental studies in mice revealed that both acute and chronic VEGF(R)i administration resulted in reduced left ventricular EF, cardiomyocyte hypertrophy, and myocardial fibrosis. Transcriptomic analysis identified significant dysregulation of multiple key signaling pathways, including DNA repair (R = 0.46), mitochondrial ATP synthesis (R = 0.39), glycogen metabolism regulation (R = 0.45), and proteasome-mediated protein degradation (R = 0.45). Moreover, significant upregulation was observed in inflammatory pathways, specifically those involving IL-1, IL-6, TNF-α, and IRF3/IRF7-mediated immune responses. Clinical cohort analyses demonstrated significant elevations in both cardiac injury biomarkers (NT-proBNP, CK-MB, cTnT) and inflammatory mediators (CRP) following VEGF(R)i administration. CONCLUSIONS Our findings present the first comprehensive characterization of VirHF clinical features and elucidate its underlying molecular mechanisms, thereby providing a theoretical framework for optimizing the clinical safety of VEGF(R)i therapy.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - MinHong Cai
- Healthcare-Associated Infection Management Office, Sichuan Academy of Medical Sciences and Sichuan People's Hospital, Chengdu, Sichuan, China
| | - Hongyu Kuang
- Department of Cardiology, University-town Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qinghua Ma
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yijun Liu
- Department of Outpatient, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Guo Zhang
- Department of Dean's office, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
17
|
Lu Y, Gao J, Hou Y, Yang H, Wang D, Zhang G, Qin Z, Du P, Wang Z, Wang Y, Chen Q, Sun Z, Li P, Zhang J, Tang J. Targeting the NLRP3 inflammasome abrogates cardiotoxicity of immune checkpoint blockers. J Immunother Cancer 2025; 13:e010127. [PMID: 39773567 PMCID: PMC11749606 DOI: 10.1136/jitc-2024-010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many malignant tumors. However, ICI-induced hyper-immune activation causes cardiotoxicity. Traditional treatments such as glucocorticoids and immunosuppressants have limited effectiveness and may even accelerate tumor growth. This study aimed to identify approaches that effectively reduce cardiotoxicity and simultaneously preserve or enhance the antitumor immunity of ICI therapy. METHODS ICI injection in melanoma-bearing C57BL/6J female mice was used to simulate cardiotoxicity in patients with tumor undergoing immune therapy. MCC950 was used to block nod-like receptor protein 3 (NLRP3) inflammasome activity. Echocardiography, immunofluorescence, flow cytometry, and reverse transcription quantitative polymerase chain reaction were used to assess cardiac function, immune cell populations, and inflammatory factor levels. Bulk and single-cell RNA sequencing was used to detect the changes in cardiac transcriptome and immunological network. RESULTS NLRP3 inhibition reduced inflammatory response and improved cardiac function. Notably, NLRP3 inhibition also resulted in a pronounced suppression of tumor growth. Single-cell RNA sequencing elucidated that MCC950 treatment reduced the cardiac infiltration of pathogenic macrophages, cytotoxic T cells, activated T cells, and their production of inflammatory cytokines, while enhancing the presence of reparative macrophages and naive T cells. In addition, MCC950 attenuated cardiotoxicity induced by dual programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunotherapy and promoted tumor regression, and showed efficacy in treating established cardiotoxicity. CONCLUSIONS Our findings provide a promising clinical approach for preventing and treating cardiotoxicity induced by ICIs, dissociating the antitumor efficacy of ICI-based therapies from their cardiotoxic side effects.
Collapse
Affiliation(s)
- Yang Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiamin Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yachen Hou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Han Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Dashuai Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Pengchong Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yunzhe Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Quanzhou Chen
- Department of Nutrition, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaowei Sun
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Ping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| |
Collapse
|
18
|
Yazaki K, Dewar M, Dauz J, Akazawa Y, Hui L, Sun M, Hui W, Kabir G, Dejardin JF, Connelly KA, Heximer SP, Friedberg MK. Serial and regional assessment of the right ventricular molecular and functional response to pressure loading. Am J Physiol Heart Circ Physiol 2025; 328:H58-H74. [PMID: 39422363 DOI: 10.1152/ajpheart.00322.2024] [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: 05/21/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Right ventricular (RV) function determines outcomes in RV pressure loading. A better understanding of the time-course and regional distribution of RV remodeling may help optimize targets and timing for therapeutic intervention. We sought to characterize RV remodeling between zero and 6 wk after the initiation of RV pressure loading. Thirty-six rats were randomized to either sham surgery or to pulmonary artery banding (PAB). After echocardiography and conductance catheter studies, groups of rats were euthanized at 1 wk, 3 wk, and 6 wk after sham surgery, or induction of RV pressure loading, for RV histological, RNA, and molecular analysis. A vigorous inflammatory response characterized by increased RV inflammatory cytokines, chemokines, and macrophage markers was observed at 1 wk following PAB. Metabolic changes, transforming growth factor-β (TGF-β)1 canonical signaling, collagenous fibrosis deposition, and apoptosis were already significantly increased by 1 wk after PAB. Genes marking fibroblast activation were upregulated at 1 wk but not at 6 wk post-PAB surgery. Mitochondrial dysfunction was evidenced by increased pyruvate dehydrogenase kinase (PDK) activity and decreased pyruvate dehydrogenase (PDH) phosphorylation significantly at 6-wk post-PAB. These processes preceded the development of overt myocardial hypertrophy and impaired echo parameters of systolic and diastolic function that occurred significantly from 3 wk after PAB. RV myocardial inflammation, metabolic shift, metabolic gene transcription, and profibrotic signaling occur early after initiation of pressure loading when RV pressures are only moderately elevated, before the development of overt myocardial hypertrophy and dysfunction, suggesting that adaptive hypertrophy and maladaptive remodeling occur simultaneously. These results suggest that therapeutic intervention to reduce adverse RV remodeling may be needed earlier and at lower thresholds than currently used.NEW & NOTEWORTHY Exploring the dynamics of right ventricular remodeling: unveiling the intricate interplay between inflammation, metabolic shifts, and fibrotic signaling in response to pressure loading. Through a comprehensive study spanning from initiation to 6 wk post-pressure loading, our research sheds light on the early onset of crucial molecular processes preceding overt hypertrophy and dysfunction. These findings challenge conventional intervention timing, advocating for early, targeted therapeutic strategies to mitigate adverse remodeling in right ventricular pressure loading.
Collapse
MESH Headings
- Animals
- Ventricular Remodeling
- Male
- Ventricular Function, Right
- Rats, Sprague-Dawley
- Fibrosis
- Rats
- Ventricular Pressure
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Disease Models, Animal
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/etiology
- Time Factors
- Apoptosis
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Signal Transduction
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Cytokines/metabolism
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
Collapse
Affiliation(s)
- Kana Yazaki
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Michael Dewar
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John Dauz
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Yohei Akazawa
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Hui
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mei Sun
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Wei Hui
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Golam Kabir
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jean-Francois Dejardin
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kim A Connelly
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Scott P Heximer
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mark K Friedberg
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Yin Z, Fu L, Wang Y, Tai S. Impact of gut microbiota on cardiac aging. Arch Gerontol Geriatr 2025; 128:105639. [PMID: 39312851 DOI: 10.1016/j.archger.2024.105639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Recent research has suggested imbalances in gut microbiota composition as contributors to cardiac aging. An individual's physical condition, along with lifestyle-associated factors, including diet and medication, are significant determinants of gut microbiota composition. This review discusses evidence of bidirectional associations between aging and gut microbiota, identifying gut microbiota-derived metabolites as potential regulators of cardiac aging. It summarizes the effects of gut microbiota on cardiac aging diseases, including cardiac hypertrophy and fibrosis, heart failure, and atrial fibrillation. Furthermore, this review discusses the potential anti-aging effects of modifying gut microbiota composition through dietary and pharmacological interventions. Lastly, it underscores critical knowledge gaps and outlines future research directions. Given the current limited understanding of the direct relationship between gut microbiota and cardiac aging, there is an urgent need for preclinical and clinical investigations into the mechanistic interactions between gut microbiota and cardiac aging. Such endeavors hold promise for shedding light on the pathophysiology of cardiac aging and uncovering new therapeutic targets for cardiac aging diseases.
Collapse
Affiliation(s)
- Zhiyi Yin
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Liyao Fu
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yongjun Wang
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| | - Shi Tai
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
20
|
Thorp EB, Filipp M. Contributions of Inflammation to Cardiometabolic Heart Failure with Preserved Ejection Fraction. ANNUAL REVIEW OF PATHOLOGY 2025; 20:143-167. [PMID: 39357068 DOI: 10.1146/annurev-pathmechdis-111523-023405] [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: 10/04/2024]
Abstract
The most common form of heart failure is heart failure with preserved ejection fraction (HFpEF). While heterogeneous in origin, the most common form of HFpEF is the cardiometabolic manifestation. Obesity and aging promote systemic inflammation that appears integral to cardiometabolic HFpEF pathophysiology. Accumulation of immune cells within the heart, fueled by an altered metabolome, contribute to cardiac inflammation and fibrosis. In spite of this, broad anti-inflammatory therapy has not shown significant benefit in patient outcomes. Thus, understanding of the nuances to metabolic and age-related inflammation during HFpEF is paramount for more targeted interventions. Here, we review clinical evidence of inflammation in the context of HFpEF and summarize our mechanistic understanding of immunometabolic inflammation, highlighting pathways of therapeutic potential along the way.
Collapse
Affiliation(s)
- Edward B Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; ,
| | - Mallory Filipp
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; ,
| |
Collapse
|
21
|
Madureira S, Gouveia R, Elias C, Neves A, Ribeirinho-Soares P, Amorim M, Soares M, Pereira J, Almeida J, Lourenço P. Hyperuricemia Is Associated With Higher Mortality in Non-diabetic Heart Failure Patients. Cureus 2024; 16:e75394. [PMID: 39781167 PMCID: PMC11709702 DOI: 10.7759/cureus.75394] [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] [Accepted: 11/09/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Hyperuricemia (HU) is associated with an increased risk of incident heart failure (HF) and adverse HF outcomes. Patients with diabetes mellitus (DM) have a greater prevalence of HU. AIMS We evaluated the prognostic impact of HU in patients with HF according to the coexistence of DM. METHODS A retrospective cohort study of ambulatory patients with HF with left ventricular systolic dysfunction (LVSD) was conducted from January 2012 to May 2018. The end point was all-cause mortality; follow-up was until January 2021. A Cox regression analysis was used to assess the prognostic impact of elevated uric acid (UA) levels. The cut-off for HU was 8.2 mg/dL. A multivariate model was built accounting for confounders. The analysis was stratified according to DM, and interaction between DM and UA levels was tested. RESULTS We studied 538 patients, of whom 66% were males. Of the patients, 45% had ischemic HF, 41% had DM, and 11% were receiving urate-lowering therapies. The median (interquartile range (IQR)) admission UA was 5.5 (5.8-9.2) mg/dL, and 40% had UA > 8.2 mg/dL. During a median 46-month follow-up, 48.5% of patients died. Patients with UA > 8.2 mg/dL had a multivariate-adjusted hazard ratio (HR) (95% confidence interval (CI)) of all-cause mortality of 1.75 (1.20-2.55; p=0.003). The interaction between DM and UA levels was significant (p=0.04). The independent association of hyperuricemia with mortality persisted only in non-DM patients (HR: 1.70, 95% CI: 1.16-2.51; p=0.007). In those with DM, hyperuricemia portended no survival disadvantage. CONCLUSION DM appears to influence the prognostic impact of HU in chronic HF. The risk of all-cause mortality in hyperuricemic HF patients without DM increases by 70% when compared with those with normal UA levels.
Collapse
Affiliation(s)
- Sergio Madureira
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Rita Gouveia
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Catarina Elias
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Ana Neves
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | | | - Marta Amorim
- Department of Internal Medicine, Centro Hospitalar Póvoa de Varzim - Vila do Conde, Vila do Conde, PRT
| | - Marta Soares
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Joana Pereira
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Jorge Almeida
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
| | - Patrícia Lourenço
- Department of Internal Medicine, Unidade Local de Saúde de São João, Porto, PRT
- Department of Medicine, Faculty of Medicine - University of Porto, Porto, PRT
| |
Collapse
|
22
|
Zheng Y, Nie Z, Zhang Y, Guo Z. The association between heart failure and systemic inflammatory response index: A cross-sectional study. J Natl Med Assoc 2024; 116:662-672. [PMID: 39537471 DOI: 10.1016/j.jnma.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The systemic inflammatory response index (SIRI) is a recently developed composite index that assesses the entire extent of inflammation in the body, closely linked to heart failure (HF). This study aimed to evaluate the potential association between SIRI and HF. METHODS The cross-sectional study utilized data from the National Health and Nutrition Examination Survey (NHANES) database from 2001 to 2018. SIRI is calculated based on the counts of monocytes, neutrophils, and lymphocytes. A weighted multiple-variable linear regression model examined the correlation between SIRI and HF. Using restrained cubic splines explored the nonlinear relationship between the two, and the robustness of the results was verified by subgroup analysis and interaction tests. RESULTS Our study included 30,294 participants, 814 of whom were diagnosed with HF and 29,480 with non-HF. The multiple linear regression analysis showed that SIRI was positively correlated with HF (OR = 1.66; 95 % CI, 1.21, 2.29) and that there was no nonlinear relationship between the two. This relationship persisted in subgroup analyses. CONCLUSIONS The results indicate a linear positive correlation between SIRI and HF. Further extensive prospective studies are needed to validate these findings.
Collapse
Affiliation(s)
- Yu Zheng
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Hunan University of Chinese Medicine, Yuelu District, Changsha, Hunan 410000, China
| | - Zixing Nie
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Hunan University of Chinese Medicine, Yuelu District, Changsha, Hunan 410000, China
| | - Yifan Zhang
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Hunan University of Chinese Medicine, Yuelu District, Changsha, Hunan 410000, China
| | - Zhihua Guo
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China; Hunan University of Chinese Medicine, Yuelu District, Changsha, Hunan 410000, China; Key Laboratory of Chinese Medicine Intelligent Diagnosis and Treatment of Chronic Diseases in Hunan Province, Hunan University of Traditional Chinese Medicine, Changsha 410208, China; Joint Postgraduate Training Base for Intelligent Application of Internet + Chronic Disease Chinese Medicine Diagnosis and Treatment and Wellness, Changsha 410208, China.
| |
Collapse
|
23
|
Liu SQ, Xie SY, Zhang T, Zhang H, Chen MY, Xing Y, Zhao N, Li L, Chen S, Wang SS, Zeng XF, Deng W, Tang QZ. Impeding Nucleotide-Binding Oligomerization Domain-Like Receptor 3 Inflammasome Ameliorates Cardiac Remodeling and Dysfunction in Obesity-Associated Cardiomyopathy. J Am Heart Assoc 2024:e035234. [PMID: 39604027 DOI: 10.1161/jaha.124.035234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Inflammation and metabolic disturbances are key culprits in the pathogenesis of obesity-associated cardiomyopathy. The NLRP3 (nucleotide-binding oligomerization domain-like receptor 3) inflammasome mediates the release of the proinflammatory cytokines IL-1β (interleukin-1β) and IL-18 by activating caspase-1, which is strongly implicated in metabolic disturbances. We here sought to determine whether NLRP3 inflammasome inhibition could ameliorate obesity cardiomyopathy and if so, to further explore its underlying mechanisms. METHODS AND RESULTS Male mice were fed a high-fat diet for 24 weeks to induce obesity cardiomyopathy. MCC950 was used to inhibit NLRP3 inflammasome activation. Recombinant adeno-associated virus serotype 9 encoding TXNIP (thioredoxin-interacting protein) under cTnT (cardiac troponin T) promoter and the mitochondrial-targeted antioxidant MitoTEMPO were injected into obese mice to investigate the specific mechanism. To mimic obesity cardiomyopathy in vitro, neonatal rat ventricular myocytes transfected with the small interfering RNA against TXNIP were incubated with 400 μmol palmitic acid for 24 hours. NLRP3 inflammasome was significantly increased in obese hearts. NLRP3 inflammasome inhibition by NLRP3 deletion or MCC950 prevented obesity-induced cardiac systolic and diastolic dysfunction, myocardial hypertrophy and fibrosis, and excessive lipid accumulation in male mice. Conversely, TXNIP overexpression worsened obesity-associated cardiomyopathy. Similarly, MCC950 treatment or TXNIP knockdown reduced palmitic acid-induced NLRP3 inflammasome activation and lipid storage. Mechanistically, abnormal NF-κB (nuclear factor kappa B) pathway activation, increased mitochondrial reactive oxygen species, and elevated TXNIP levels led to excessive NLRP3 inflammasome activation. CONCLUSIONS Our study confirms that aberrant NLRP3 inflammasome activation in cardiomyocytes worsens obesity-associated cardiomyopathy and implicates inhibition of NLRP3 inflammasome as a potent therapeutic approach for obesity cardiomyopathy.
Collapse
Affiliation(s)
- Shi-Qiang Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Sai-Yang Xie
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Tong Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Heng Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Meng-Ya Chen
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Yun Xing
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Nan Zhao
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Lanlan Li
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Sha-Sha Wang
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Xiao-Feng Zeng
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Wei Deng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Qi-Zhu Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| |
Collapse
|
24
|
Wu Z, Liu W, Si X, Liang J. Screening of key genes related to M6A methylation in patients with heart failure. BMC Cardiovasc Disord 2024; 24:565. [PMID: 39415091 PMCID: PMC11481427 DOI: 10.1186/s12872-024-04228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVE This study aims to identify m6A methylation-related and immune cell-related key genes with diagnostic potential for heart failure (HF) by leveraging various bioinformatics techniques. METHODS The GSE116250 and GSE141910 datasets were sourced from the Gene Expression Omnibus (GEO) database. Correlation analysis was conducted between differentially expressed genes (DEGs) in HF and control groups, alongside differential m6A regulatory factors, to identify m6A-related DEGs (m6A-DEGs). Subsequently, candidate genes were narrowed down by intersecting key module genes derived from weighted gene co-expression network analysis (WGCNA) with m6A-DEGs. Key genes were then identified through the Least Absolute Shrinkage and Selection Operator (LASSO) analysis. Correlation analyses between key genes and differentially expressed immune cells were performed, followed by the validation of key gene expression levels in public datasets. To ensure clinical applicability, five pairs of blood samples were collected for quantitative real-time fluorescence PCR (qRT-PCR) validation. RESULTS A total of 93 m6A-DEGs were identified (|COR| > 0.6, P < 0.05), and five key genes (LACTB2, NAMPT, SCAMP5, HBA1, and PRKAR2A) were selected for further analysis. Correlation analysis revealed that differential immune cells were negatively associated with the expression of LACTB2, NAMPT, and PRKAR2A (P < 0.05), while positively correlated with SCAMP5 and HBA1 (P < 0.05). Subsequent expression validation confirmed significant differences in key gene expression between the HF and control groups, with consistent expression trends observed across both training and validation sets. The expression trends of LACTB2, PRKAR2A, and HBA1 in blood samples from the qRT-PCR assay aligned with the results derived from public databases. CONCLUSION This study successfully identified five m6A methylation-related key genes with diagnostic significance, providing a theoretical foundation for further exploration of m6A methylation's molecular mechanisms in HF.
Collapse
Affiliation(s)
- Zelan Wu
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wupeng Liu
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Xiaoyun Si
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jinfeng Liang
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
25
|
Cotter G, Davison B, Freund Y, Mebazaa A, Voors A, Edwards C, Novosadova M, Takagi K, Hayrapetyan H, Mshetsyan A, Mayranush D, Cohen‐Solal A, ter Maaten JM, Biegus J, Ponikowski P, Filippatos G, Chioncel O, Pagnesi M, Simon T, Metra M, Mann DL. Corticosteroid burst therapy in patients with acute heart failure: Design of the CORTAHF pilot study. ESC Heart Fail 2024; 11:2672-2680. [PMID: 38943232 PMCID: PMC11424380 DOI: 10.1002/ehf2.14930] [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: 04/01/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024] Open
Abstract
AIMS Inflammation has emerged as a potential key pathophysiological mechanism in heart failure (HF) in general and acute HF (AHF) specifically, with inflammatory biomarkers shown to be highly predictive of adverse outcomes in these patients. The CORTAHF study builds on both these data and the fact that steroid burst therapy has been shown to be effective in the treatment of respiratory diseases and COVID-19. Our hypothesis is that in patients with AHF and elevated C-reactive protein (CRP) levels without symptoms or signs of infection, a 7-day course of steroid therapy will lead to reduced inflammation and short-term improvement in quality of life and a reduced risk of worsening HF (WHF) events. METHODS AND RESULTS The study, which is currently ongoing, will include 100 patients with AHF ages 18-85, regardless of ejection fraction, screened within 12 h of presentation. Patients will be included who have NT-proBNP > 1500 pg/mL and CRP > 20 mg/L at screening. Exclusion criteria include haemodynamic instability and symptoms and signs of infection. After signed consent, eligible patients will be randomized according to a central randomization scheme stratified by centre 1:1 to either treatment once daily for 7 days with 40 mg prednisone orally or to standard care. Patients will be assessed at study day 2, day 4 or at discharge if earlier, and at days 7 and 31 at the hospital; and at day 91 through a telephone follow-up. The primary endpoint is the change in CRP level from baseline to day 7, estimated from a mixed model for repeated measures (MMRM) including all measured timepoints, in patients without a major protocol violation. Secondary endpoints include the time to the first event of WHF adverse event, readmission for HF, or death through day 91; and changes to day 7 in EQ-5D visual analogue scale score and utility index. Additional clinical and laboratory measures will be assessed. CONCLUSIONS The results of the study will add to the knowledge of the role of inflammation in AHF and potentially inform the design of larger studies with possibly longer duration of anti-inflammatory therapies in AHF.
Collapse
Affiliation(s)
- Gad Cotter
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Heart InitiativeDurhamNCUSA
- Momentum Research, Inc.DurhamNCUSA
| | - Beth Davison
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Heart InitiativeDurhamNCUSA
- Momentum Research, Inc.DurhamNCUSA
| | - Yonathan Freund
- IMProving Emergency Care FHUSorbonne UniversitéParisFrance
- Emergency Department and Service Mobile d'Urgence et de Réanimation (SMUR)Hôpital Pitié‐Salpêtrière, Assistance Publique‐Hôpitaux de Paris (AP‐HP)ParisFrance
| | - Alexandre Mebazaa
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Department of Anesthesiology and Critical Care and Burn Unit, Saint‐Louis and Lariboisière HospitalsFHU PROMICE, DMU Parabol, APHP NordParisFrance
| | - Adriaan Voors
- Department of CardiologyUniversity Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | | | | | | | | | | | | | - Alain Cohen‐Solal
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Department of Cardiology, APHP NordLariboisière University HospitalParisFrance
| | - Jozine M. ter Maaten
- Department of CardiologyUniversity Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Jan Biegus
- Institute of Heart DiseasesWroclaw Medical UniversityWroclawaPoland
| | - Piotr Ponikowski
- Institute of Heart DiseasesWroclaw Medical UniversityWroclawaPoland
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens, School of MedicineAttikon University HospitalHaidariGreece
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’University of Medicine ‘Carol Davila’BucharestRomania
| | - Matteo Pagnesi
- Department of Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Tabassome Simon
- IMProving Emergency Care FHUSorbonne UniversitéParisFrance
- Department of Clinical Pharmacology and Clinical Research Platform Paris‐East (URCEST‐CRC‐CRB)St Antoine Hospital, APHPParisFrance
| | - Marco Metra
- Department of Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Douglas L. Mann
- Department of Medicine, Division of Cardiovascular, Center for Cardiovascular ResearchWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
26
|
van Dijck P, Hannemann C, Dreger H, Stangl V, Stangl K, Ludwig A, Hewing B. Increased Expression of Inactive Rhomboid Protein 2 in Circulating Monocytes after Acute Myocardial Infarction. J Cardiovasc Transl Res 2024; 17:1059-1066. [PMID: 38743187 PMCID: PMC11519168 DOI: 10.1007/s12265-024-10519-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/11/2024] [Indexed: 05/16/2024]
Abstract
Increased TNF-α levels following acute myocardial infarction (AMI) contribute to impaired recovery of myocardial function. Interaction of inactive rhomboid protein 2 (iRhom2) with TNF-α converting enzyme (TACE) is required for TNF-α shedding from immune cells. We hypothesized that iRhom2 expression increases in circulating monocytes following AMI. Transcript levels of iRhom2, TACE and TNF-α were evaluated by quantitative real-time PCR in isolated monocytes of 50 AMI patients at admission (d1) and 3 days (d3) after. We observed a significant increase in levels of iRhom2 mRNA expression in monocytes between d1-3, while TNF-α and TACE mRNA expression remained unchanged. At d3, iRhom2 mRNA expression positively correlated with levels of intermediate monocytes or serum TNF-α, and negatively with LV systolic function. iRhom2 may contribute to regulation of post-infarction inflammation and is associated with LV dysfunction following AMI. iRhom2 modulation should be evaluated as a potential therapeutic strategy to attenuate cardiac remodeling following AMI.
Collapse
Affiliation(s)
- Phillip van Dijck
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
| | - Carmen Hannemann
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Henryk Dreger
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow Klinikum, Deutsches Herzzentrum der Charité, Berlin, Germany
- Structural Heart Interventions Program (SHIP), Deutsches Herzzentrum der Charité, Berlin, Germany
| | - Verena Stangl
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Karl Stangl
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Antje Ludwig
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Bernd Hewing
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Mitte, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany.
- Berlin Institute of Health (BIH), 10178, Berlin, Germany.
- Zentrum Für Kardiologie, Kardiologische Gemeinschaftspraxis, Muenster, Germany.
- Department of Cardiology III - Adult Congenital and Valvular Heart Disease, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
27
|
Delcea C, Adrian Buzea C, Dobrev D, Andrei Dan G. Prognostic roles of neutrophil-lymphocyte, monocyte-lymphocyte and platelet-lymphocyte ratios for long-term all-cause mortality in heart failure. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VASCULATURE 2024; 54:101502. [PMID: 39280696 PMCID: PMC11402304 DOI: 10.1016/j.ijcha.2024.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024]
Abstract
Background Heart failure (HF) and inflammation have a bidirectional relation leading to activation and adaptation of multiple cellular lines, including leucocyte subtypes and platelets. We aimed to assess and compare the predictive value of the neutrophil-lymphocyte (NLR), monocyte-lymphocyte (MLR) and platelet-lymphocyte (PLR) ratios for all-cause long-term mortality in HF. Methods This is an observational retrospective cohort study that included patients from the HI-HF cohort that survived the initial hospitalization. Vital status and survival time were assessed in June 2020. Results We analyzed 1018 HF patients with a mean age of 72.32 ± 10.29 years and 53.54 % women. All-cause long-term mortality was 38.21 % after a median follow-up time of 68 [38 - 82] months. NLR (AUC 0.667, 95 %CI 0.637 - 0.697), MLR (AUC 0.670, 95 %CI 0.640 - 0.700) and PLR (AUC 0.606, 95 %CI 0.574 - 0.636) were predictors of all-cause mortality. In multivariable Cox proportional hazards analysis, NLR≥3.56 was the only hematological index independent predictor of fatality (HR 1.36, 95 %CI 1.05 - 1.76). Conclusions Of the three hematological indices, NLR was the only independent predictor of all-cause long-term mortality of HF patients. We suggest NLR≥3.56 as an auxiliary prognostic biomarker for the evaluation of HF patients.
Collapse
Affiliation(s)
- Caterina Delcea
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Catalin Adrian Buzea
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Gheorghe Andrei Dan
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
28
|
Cristescu L, Tilea I, Iancu DG, Stoica F, Moldovan DA, Capriglione V, Varga A. Insights into the Neutrophil-to-Lymphocyte Ratio and the Platelet-to-Lymphocyte Ratio as Predictors for the Length of Stay and Readmission in Chronic Heart Failure Patients. Diagnostics (Basel) 2024; 14:2102. [PMID: 39335781 PMCID: PMC11431337 DOI: 10.3390/diagnostics14182102] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Chronic heart failure (CHF) is characterized by complex pathophysiology, leading to increased hospitalizations and mortality. Inflammatory biomarkers such as the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) provide valuable diagnostic insights. METHODS This study evaluates the prognostic relationship between NLR, PLR, and, in a specific subcohort, N-terminal pro B-type natriuretic peptide (NT-proBNP), alongside length of stay (LOS) and 90-day readmission rates in CHF patients, irrespective of heart failure phenotype. A retrospective analysis of 427 CHF admissions (males = 57.84%) was conducted. RESULTS The mean age of the entire population was 68.48 ± 11.53 years. The average LOS was 8.33 ± 5.26 days, with a readmission rate of 73 visits (17.09%) for 56 patients. The NLR (3.79 ± 3.32) showed a low but positive correlation with the LOS (r = 0.222, p < 0.001). Conversely, the PLR (144.84 ± 83.08) did not demonstrate a significant association with the LOS. The NLR presented a low negative correlation for days until the next admission (r = -0.023, p = 0.048). In a prespecified subanalysis of 323 admissions, the NT-proBNP exhibited a low positive Pearson correlation with the NLR (r = 0.241, p < 0.001) and PLR (r = 0.151, p = 0.006). CONCLUSIONS The impact of the NLR across heart failure phenotypes may suggest the role of systemic inflammation in understanding and managing CHF.
Collapse
Affiliation(s)
- Liviu Cristescu
- Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (D.-A.M.); (V.C.); (A.V.)
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.-G.I.); (F.S.)
- Department of Internal Medicine II-Cardiology, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
| | - Ioan Tilea
- Department of Internal Medicine II-Cardiology, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Dragos-Gabriel Iancu
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.-G.I.); (F.S.)
- Department of Internal Medicine II-Cardiology, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Florin Stoica
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.-G.I.); (F.S.)
- Department of Internal Medicine II-Cardiology, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
| | - Diana-Andreea Moldovan
- Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (D.-A.M.); (V.C.); (A.V.)
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.-G.I.); (F.S.)
- Department of Cardiology I, The Emergency Institute for Cardiovascular Diseases and Transplantation, 540136 Targu Mures, Romania
| | - Vincenzo Capriglione
- Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (D.-A.M.); (V.C.); (A.V.)
| | - Andreea Varga
- Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (D.-A.M.); (V.C.); (A.V.)
- Department of Internal Medicine II-Cardiology, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
| |
Collapse
|
29
|
Chen G, Yao Y, Liu Y, Zhang R, Wen C, Zhou Q, Xu Y, Wang W, Jiang H, Tao Z, Chen W, Qiu Z, Chen X. IKKα-STAT3-S727 axis: a novel mechanism in DOX-induced cardiomyopathy. Cell Mol Life Sci 2024; 81:406. [PMID: 39287798 PMCID: PMC11408453 DOI: 10.1007/s00018-024-05439-1] [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: 01/29/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic drug, but its use can lead to cardiomyopathy, which is the leading cause of mortality among cancer patients. Macrophages play a role in DOX-induced cardiomyopathy (DCM), but the mechanisms undlerlying this relationship remain unclear. This study aimed to investigate how IKKα regulates macrophage activation and contributes to DCM in a mouse model. Specifically, the role of macrophage IKKα was evaluated in macrophage-specific IKKα knockout mice that received DOX injections. The findings revealed increased expression of IKKα in heart tissues after DOX administration. In mice lacking macrophage IKKα, myocardial injury, ventricular remodeling, inflammation, and proinflammatory macrophage activation worsened in response to DOX administration. Bone marrow transplant studies confirmed that IKKα deficiency exacerbated cardiac dysfunction. Macrophage IKKα knockout also led to mitochondrial damage and metabolic dysfunction in macrophages, thereby resulting in increased cardiomyocyte injury and oxidative stress. Single-cell sequencing analysis revealed that IKKα directly binds to STAT3, leading to the activation of STAT3 phosphorylation at S727. Interestingly, the inhibition of STAT3-S727 phosphorylation suppressed both DCM and cardiomyocyte injury. In conclusion, the IKKα-STAT3-S727 signaling pathway was found to play a crucial role in DOX-induced cardiomyopathy. Targeting this pathway could be a promising therapeutic strategy for treating DOX-related heart failure.
Collapse
Affiliation(s)
- Ganyi Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yiwei Yao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yafeng Liu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Ruoyu Zhang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Chenghao Wen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Qiang Zhou
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yueyue Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Wuwei Wang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Hongwei Jiang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Zhonghao Tao
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| | - Zhibing Qiu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| |
Collapse
|
30
|
Paronetto MP, Crescioli C. Rethinking of phosphodiesterase 5 inhibition: the old, the new and the perspective in human health. Front Endocrinol (Lausanne) 2024; 15:1461642. [PMID: 39355618 PMCID: PMC11442314 DOI: 10.3389/fendo.2024.1461642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
The phosphodiesterases type 5 (PDE5) are catalytic enzymes converting the second messenger cyclic guanosine monophosphate (cGMP) to 5' GMP. While intracellular cGMP reduction is associated with several detrimental effects, cGMP stabilization associates with numerous benefits. The PDE5 specific inhibitors, PDE5i, found their explosive fortune as first-line treatment for erectile dysfunction (ED), due to their powerful vasoactive properties. The favorable effect for ED emerged as side-effect when PDE5i were originally proposed for coronary artery disease (CAD). From that point on, the use of PDE5i captured the attention of researchers, clinicians, and companies. Indeed, PDE5-induced intracellular cGMP stabilization offers a range of therapeutic opportunities associated not only with vasoactive effects, but also with immune regulatory/anti-inflammatory actions. Chronic inflammation is acknowledged as the common link underlying most non-communicable diseases, including metabolic and cardiac diseases, autoimmune and neurodegenerative disorders, cancer. In this scenario, the clinical exploitation of PDE5i is undeniably beyond ED, representing a potential therapeutic tool in several human diseases. This review aims to overview the biological actions exerted by PDE5i, focusing on their ability as modulators of inflammation-related human diseases, with particular attention to inflammatory-related disorders, like cardiac diseases and cancer.
Collapse
Affiliation(s)
- Maria Paola Paronetto
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
| |
Collapse
|
31
|
Srabanti MG, Garcia J. Quadratic stratification of left ventricular hypertrophy and association with mitral insufficiency grading: a retrospective study using cardiac magnetic resonance. Cardiovasc Diagn Ther 2024; 14:589-608. [PMID: 39263481 PMCID: PMC11384468 DOI: 10.21037/cdt-23-466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/14/2024] [Indexed: 09/13/2024]
Abstract
Background Chronic primary mitral regurgitation (MR) is caused by the defect in >1 component of the mitral valve, potentially leading to left ventricular hypertrophy (LVH). The relationship between LVH subtypes and the insufficiency grading of chronic MR remains unclear. Thus, we aimed to investigate this association and explore the impact of unhealthy habits on LVH development in patients with chronic primary MR through a cross-sectional study. Methods Cardiac magnetic resonance (CMR) data was retrospectively collected from 3T magnetic resonance imaging (MRI) scanners in 71 patients with chronic primary MR (range, 20-84 years, 52% men). Considered patients (with mild-to-severe MR) were enrolled between March 2015 and September 2022 from the Cardiovascular Imaging Registry of Calgary (CIROC) database. Left ventricle (LV) function was assessed using cvi42 v5.11.5. Patients were categorized into 'mild-to-severe' MR using regurgitation fraction (RF), according to the current imaging guidelines. LVH subtypes were determined using mass-to-volume (M/V) calculations. IBM SPSS was used to run all the statistical analyses. This study employed normality checks by using the Shapiro-Wilk test; one-way analysis of variance (ANOVA) and Kruskal-Wallis tests with post-hoc pairwise comparisons; Chi-squared tests, Fisher's Exact test, crosstabulation analysis, and multinomial logistic regression to examine relationships between MR severity, LVH types, and impact of lifestyle factors, significance at P<0.05. Results Eccentric LVH was significantly associated with increased severity of MR, while concentric remodeling (CR) was linked to decreased MR severity (χ2=13.276, P=0.03, stratified by sex χ2=7.729, P=0.005). Sex differences emerged in the overall study population. Eccentric LVH was dominantly higher than CR in both males and females (females: 57.7% vs. 42.3%, P=0.05, males: 82.8% vs. 17.2%, P=0.26). No differences were observed between age groups ('Young-Middle' = under 60 years, and 'Middle-Old' = over 60 years). Still, there were notable differences in LVH prevalence within the 'Young-Middle' age group for mild-moderate (P=0.01) and moderate-severe MR (P=0.02). Eccentric LVH was associated with higher body mass index (BMI), smoking, and frequent alcohol consumption [odds ratio (OR) 1.02, 95% confidence interval (CI): 0.56-1.26; OR 1.65, 95% CI: 1.31-6.52; OR 1.15, 95% CI: 0.26-1.34], while CR was solely associated with increased BMI (smokers OR =1.84, 95% CI: 1.25-3.91 and alcohol consumers OR =1.32, 95% CI: 0.86-2.48). Nicotine and caffeine consumption did not appear to be a risk factor for LVH (nicotine: eccentric, OR =0.99, 95% CI: 0.65-1.86; CR, OR =0.97, 95% CI: 0.69-2.39 and caffeine: eccentric, OR =0.69, 95% CI: 0.48-1.61; CR, OR =0.97, 95% CI: 0.78-4.01). Conclusions This study reveals sex-based associations between LVH subtypes and severity of chronic primary MR. Lifestyle factors such as cigarette smoking, alcohol consumption, and elevated BMI influence LVH risk, while nicotine and caffeine consumption exhibit minimal effects.
Collapse
Affiliation(s)
- Monisha Ghosh Srabanti
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- Stephenson Cardiac Imaging Centre, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Julio Garcia
- Stephenson Cardiac Imaging Centre, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
- Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| |
Collapse
|
32
|
Duni A, Kitsos A, Bechlioulis A, Lakkas L, Markopoulos G, Tatsis V, Koutlas V, Tzalavra E, Baxevanos G, Vartholomatos G, Mitsis M, Naka KK, Dounousi E. Identification of Novel Independent Correlations between Cellular Components of the Immune System and Strain-Related Indices of Myocardial Dysfunction in CKD Patients and Kidney Transplant Recipients without Established Cardiovascular Disease. Int J Mol Sci 2024; 25:9162. [PMID: 39273110 PMCID: PMC11395156 DOI: 10.3390/ijms25179162] [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: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The role of immune system components in the development of myocardial remodeling in chronic kidney disease (CKD) and kidney transplantation remains an open question. Our aim was to investigate the associations between immune cell subpopulations in the circulation of CKD patients and kidney transplant recipients (KTRs) with subclinical indices of myocardial performance. We enrolled 44 CKD patients and 38 KTRs without established cardiovascular disease. A selected panel of immune cells was measured by flow cytometry. Classical and novel strain-related indices of ventricular function were measured by speckle-tracking echocardiography at baseline and following dipyridamole infusion. In CKD patients, the left ventricular (LV) relative wall thickness correlated with the CD14++CD16- monocytes (β = 0.447, p = 0.004), while the CD14++CD16+ monocytes were independent correlates of the global radial strain (β = 0.351, p = 0.04). In KTRs, dipyridamole induced changes in global longitudinal strain correlated with CD14++CD16+ monocytes (β = 0.423, p = 0.009) and CD4+ T-cells (β = 0.403, p = 0.01). LV twist and untwist were independently correlated with the CD8+ T-cells (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively) in CKD patients, whereas the CD14++CD16+ monocytes were independent correlates of LV twist and untwist in KTRs (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively). Immune cell subsets independently correlate with left ventricular strain and torsion-related indices in CKD patients and KTRs without established CVD.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Athanasios Kitsos
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Aris Bechlioulis
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Lampros Lakkas
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Georgios Markopoulos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Tatsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Koutlas
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Eirini Tzalavra
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Gerasimos Baxevanos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
- Department of Internal Medicine, General Hospital of Ioannina, G. Chatzikosta, 454 45 Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Michail Mitsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| |
Collapse
|
33
|
Llerena-Velastegui J, Santamaria-Lasso M, Mejia-Mora M, Santander-Aldean M, Granda-Munoz A, Hurtado-Alzate C, de Jesus ACFS, Baldelomar-Ortiz J. Efficacy of Beta-Blockers and Angiotensin-Converting Enzyme Inhibitors in Non-Ischemic Dilated Cardiomyopathy: A Systematic Review and Meta-Analysis. Cardiol Res 2024; 15:281-297. [PMID: 39205958 PMCID: PMC11349132 DOI: 10.14740/cr1653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 09/04/2024] Open
Abstract
Background Non-ischemic dilated cardiomyopathy (NIDCM) is a form of heart failure with a poor prognosis and unclear optimal management. The aim of the study was to systematically review the literature and assess the efficacy and safety of beta-blockers and angiotensin-converting enzyme (ACE) inhibitors in the management of chronic heart failure secondary to NIDCM and explore their putative mechanisms of action. Methods Studies from 1990 to 2023 were reviewed using PubMed and EMBASE, focusing on their effects on left ventricular ejection fraction (LVEF) in NIDCM patients, according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results Beta-blockers showed a significant beneficial effect on LVEF improvement in NIDCM, with an overall effect size of Cohen's d = 1.30, 95% confidence interval (CI) (0.76, 1.84), high heterogeneity (Tau2 = 0.90; Chi2 = 162.05, df = 13, P < 0.00001; I2 = 92%), and a significant overall effect (Z = 4.72, P < 0.00001). ACE inhibitors also showed a beneficial role, but with less heterogeneity (Tau2 = 0.02; Chi2 = 1.09, df = 1, P = 0.30; I2 = 8%) and a nonsignificant overall effect (Z = 1.36, P = 0.17), 95% CI (-0.24, 1.31). Conclusions The study highlights the efficacy of carvedilol in improving LVEF in NIDCM patients over ACE inhibitors, recommends beta-blockers as first-line therapy, and advocates further research on ACE inhibitors.
Collapse
Affiliation(s)
- Jordan Llerena-Velastegui
- Medical School, Pontifical Catholic University of Ecuador, Quito, Ecuador
- Research Center, Center for Health Research in Latin America (CISeAL), Quito, Ecuador
| | | | - Melany Mejia-Mora
- Medical School, Pontifical Catholic University of Ecuador, Quito, Ecuador
| | | | | | | | | | | |
Collapse
|
34
|
Obeid RA, Mohammed RA, Kaskoos RA, Zair A. Exploring the antioxidant potential of Moringa oleifera leaf extracts mitigating doxorubicin-induced cardiotoxicity in male rats. J Adv Pharm Technol Res 2024; 15:166-170. [PMID: 39290548 PMCID: PMC11404441 DOI: 10.4103/japtr.japtr_531_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/11/2024] [Accepted: 05/18/2024] [Indexed: 09/19/2024] Open
Abstract
Doxorubicin (DOX) is a commonly used drug in chemotherapy for cancer treatment. However, it can cause the threatening side effect of cardiotoxicity. This study investigates whether the hydro-alcoholic leaves of Moringa oleifera have any protective potential against DOX-induced cardiotoxicity. The phytochemical analysis showed that the plant extracts contained bioactive compounds with antioxidant activities. The DOX-treated group confirmed a significant increment in cardiac troponin I (cTnI) and proinflammatory cytokine interleukin-6 (IL-6) levels, which indicates damage to the cardiomyocytes and also inflammation. However, treatment with the M. oleifera extracts significantly inhibited DOX-induced cardiomyocyte damage, as indicated by the significantly low cTnI release. Furthermore, treatment with M. oleifera extracts further increased antioxidant activities, thereby decreasing oxidative stress and lipid peroxidation. Moreover, DOX was found to increase the IL-6 level, and treatment with M. oleifera extracts had a significant impact on the inhibition of IL-6 levels. These results indicate that the M. oleifera extracts have a cardioprotective effect and can play a role as an adjunct drug in mitigating DOX-induced cardiotoxicity, thus providing new prospects for the improvement of safety and efficacy in the treatment of cancer.
Collapse
Affiliation(s)
- Ruaa Ali Obeid
- Department of Chemistry and Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia, Iraq
| | - Rusul Ahmed Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Al Mustaqbal University, Hillah, Iraq
| | - Raad A Kaskoos
- Department of Pharmacognosy and Phytochemistry, Pharmacy, Al-Manara College for Medical Sciences, Amarah, Iraq
| | - Amira Zair
- Department of Biochemistry, Faculty of Medicine, University of Sousse, Sousse, Tunisia, Iraq
| |
Collapse
|
35
|
Miao H, Tang X, Cui Y, Shi J, Xiong X, Wang C, Zhang Y. Obeticholic Acid Inhibit Mitochondria Dysfunction Via Regulating ERK1/2-DRP Pathway to Exert Protective Effect on Lipopolysaccharide-Induced Myocardial Injury. Adv Biol (Weinh) 2024; 8:e2300576. [PMID: 38728002 DOI: 10.1002/adbi.202300576] [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: 10/25/2023] [Revised: 04/19/2024] [Indexed: 07/13/2024]
Abstract
Farnesoid X receptor (FXR) plays critical regulatory roles in cardiovascular physiology/pathology. However, the role of FXR agonist obeticholic acid (OCA) in sepsis-associated myocardial injury and underlying mechanisms remain unclear. C57BL/6J mice are treated with OCA before lipopolysaccharide (LPS) administration. The histopathology of the heart and assessment of FXR expression and mitochondria function are performed. To explore the underlying mechanisms, H9c2 cells, and primary cardiomyocytes are pre-treated with OCA before LPS treatment, and extracellular signal-regulated protein kinase (ERK) inhibitor PD98059 is used. LPS-induced myocardial injury in mice is significantly improved by OCA pretreatment. Mechanistically, OCA pretreatment decreased reactive oxygen species (ROS) levels and blocked the loss of mitochondrial membrane potential (ΔΨm) in cardiomyocytes. The expression of glutathione peroxidase 1 (GPX1), superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2), and nuclear factor erythroid 2-related factor 2 (NRF-2) increased in the case of OCA pretreatment. In addition, OCA improved mitochondria respiratory chain with increasing Complex I expression and decreasing cytochrome C (Cyt-C) diffusion. Moreover, OCA pretreatment inhibited LPS-induced mitochondria dysfunction via suppressing ERK1/2-DRP signaling pathway. FXR agonist OCA inhibits LPS-induced mitochondria dysfunction via suppressing ERK1/2-DRP signaling pathway to protect mice against LPS-induced myocardial injury.
Collapse
Affiliation(s)
- Huijie Miao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Xiaomeng Tang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Yun Cui
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Jingyi Shi
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Xi Xiong
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| |
Collapse
|
36
|
Shi J, Shao MJ, Yu M, Tang BP. The Inflammation-Fibrosis Combined Index: A Novel Marker for Predicting Left Ventricular Reverse Remodeling and Prognosis in Patients with HFrEF. J Inflamm Res 2024; 17:3967-3982. [PMID: 38915807 PMCID: PMC11194169 DOI: 10.2147/jir.s460641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024] Open
Abstract
Background Inflammation and cardiac fibrosis are important pathogenic drivers of heart failure. The fibrosis-4 index (FIB-4) is associated with a higher degree of fibrosis. The systemic immune inflammation index (SII) is associated with a higher degree of systemic inflammation status. Previous studies have shown that they are associated with a poor prognosis for cardiovascular disease. We sought to investigate the value of FIB-4 combined with the SII as a novel inflammation-fibrosis combined index (IFCI) in predicting left ventricular reverse remodeling (LVRR) and prognosis among reduced ejection fraction heart failure (HFrEF) patients. Methods A total of 895 patients with HFrEF were continuously recruited. Receiver operating characteristic curves were drawn to assess the abilities of inflammation-fibrosis indicators to predict LVRR. Multivariable Cox regression analysis was used to examine independent predictors of composite cardiac events and all-cause death. Results After six months of follow-up, 344 (38.4%) patients experienced LVRR. The IFCI had the largest area under the curve (0.835, P < 0.001). In multivariate-adjusted logistic regression analyses, FIB-4, SII, and IFCI were predictive of LVRR (P value < 0.05). The IFCI was associated with a 3.686-fold higher risk of non-LVRR (odds ratio [OR] = 3.686, P < 0.001). Moreover, an increased IFCI predicted a poor prognosis in HFrEF patients. The highest risk of composite cardiac events (hazard ratio [HR] = 2.716, P < 0.001) was observed in the top IFCI-tertile group, and similar results were found regarding independent risk indicators of all-cause death. Conclusion In summary, this study indicated that increased IFCI at admission offers good predictability regarding non-LVRR and predicts the risk of all-cause mortality or composite cardiovascular events due to HFrEF patients and could be used as a novel marker.
Collapse
Affiliation(s)
- Jia Shi
- Cardiac Pacing and Physiological Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Meng-Jiao Shao
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Miao Yu
- Cardiac Pacing and Physiological Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Bao-Peng Tang
- Cardiac Pacing and Physiological Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
37
|
Shao HH, Yin RX. Pathogenic mechanisms of cardiovascular damage in COVID-19. Mol Med 2024; 30:92. [PMID: 38898389 PMCID: PMC11186295 DOI: 10.1186/s10020-024-00855-2] [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: 03/09/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND COVID-19 is a new infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Since the outbreak in December 2019, it has caused an unprecedented world pandemic, leading to a global human health crisis. Although SARS CoV-2 mainly affects the lungs, causing interstitial pneumonia and severe acute respiratory distress syndrome, a number of patients often have extensive clinical manifestations, such as gastrointestinal symptoms, cardiovascular damage and renal dysfunction. PURPOSE This review article discusses the pathogenic mechanisms of cardiovascular damage in COVID-19 patients and provides some useful suggestions for future clinical diagnosis, treatment and prevention. METHODS An English-language literature search was conducted in PubMed and Web of Science databases up to 12th April, 2024 for the terms "COVID-19", "SARS CoV-2", "cardiovascular damage", "myocardial injury", "myocarditis", "hypertension", "arrhythmia", "heart failure" and "coronary heart disease", especially update articles in 2023 and 2024. Salient medical literatures regarding the cardiovascular damage of COVID-19 were selected, extracted and synthesized. RESULTS The most common cardiovascular damage was myocarditis and pericarditis, hypertension, arrhythmia, myocardial injury and heart failure, coronary heart disease, stress cardiomyopathy, ischemic stroke, blood coagulation abnormalities, and dyslipidemia. Two important pathogenic mechanisms of the cardiovascular damage may be direct viral cytotoxicity as well as indirect hyperimmune responses of the body to SARS CoV-2 infection. CONCLUSIONS Cardiovascular damage in COVID-19 patients is common and portends a worse prognosis. Although the underlying pathophysiological mechanisms of cardiovascular damage related to COVID-19 are not completely clear, two important pathogenic mechanisms of cardiovascular damage may be the direct damage of the SARSCoV-2 infection and the indirect hyperimmune responses.
Collapse
Affiliation(s)
- Hong-Hua Shao
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Rui-Xing Yin
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China.
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
38
|
Li J, Liu L, Luo Q, Zhou W, Zhu Y, Jiang W. Exploring the causal relationship between immune cell and all-cause heart failure: a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1363200. [PMID: 38938655 PMCID: PMC11210391 DOI: 10.3389/fcvm.2024.1363200] [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: 01/21/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
Background and objectives Heart failure (HF) is a disease with numerous genetic and environmental factors that affect it. The results of previous studies indicated that immune phenotypes are associated with HF, but there have been inconclusive studies regarding a causal relationship. Therefore, Mendelian randomization (MR) analyses were undertaken to confirm the causal connections between immune phenotypes and HF, providing genetic evidence supporting the association of immune cell factors with HF risk. Methods We selected instrumental variables that met the criteria based on data from the results of genome-wide association studies (GWAS) of immune phenotype and all-cause HF. An evaluation of the causal association between 731 immune cell factors and HF risk was carried out using the inverse variance weighted (IVW), MR-Egger regression (MR-Egger), and weighted median (WM) analysis methods. To determine the horizontal pleiotropy, heterogeneity, and stability of the genetic variants, the MR-Egger intercept test, Cochran's Q test, MR-PRESSO, and leave-one-out sensitivity analysis were performed. Results MR principal method (IVW) analysis showed that a total of 38 immune cell-related factors were significantly causally associated with HF. Further analyses combining three methods (IVW, MR-Egger and WME) showed that six exposure factors significantly associated with heart failure, as shown below. The effect of Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell% Dendritic cell, CD39+ CD8+ T cell% CD8+ T cell, CD3 on Central Memory CD4+ T cell on heart failure was positive. Whereas, a reverse effect was observed for CD14+ CD16+ monocyte% monocyte. Conclusion We investigated the causal relationship between immune phenotypes and all-cause HF. According to the results, Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell% Dendritic cell, CD39+ CD8+ T cell% CD8+ T cell, CD3 on Central Memory CD4+ T cell aggravate HF, and the risk of HF is decreased by CD14+ CD16+ monocyte% monocyte. These phenotypes may serve as new biomarkers, providing new therapeutic insights for the prevention and treatment of all-cause HF.
Collapse
Affiliation(s)
| | | | | | | | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Mann DL. The Emerging Field of Cardioimmunology: Past, Present and Foreseeable Future. Circ Res 2024; 134:1663-1680. [PMID: 38843286 PMCID: PMC11160976 DOI: 10.1161/circresaha.123.323656] [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: 12/11/2023] [Accepted: 02/08/2024] [Indexed: 06/09/2024]
Abstract
Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.
Collapse
Affiliation(s)
- Douglas L Mann
- Cardiovascular Division, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
40
|
Fernandez-Patron C, Lopaschuk GD, Hardy E. A self-reinforcing cycle hypothesis in heart failure pathogenesis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:627-636. [PMID: 39196226 DOI: 10.1038/s44161-024-00480-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/25/2024] [Indexed: 08/29/2024]
Abstract
Heart failure is a progressive syndrome with high morbidity and mortality rates. Here, we suggest that chronic exposure of the heart to risk factors for heart failure damages heart mitochondria, thereby impairing energy production to levels that can suppress the heart's ability to pump blood and repair mitochondria (both energy-consuming processes). As damaged mitochondria accumulate, the heart becomes deprived of energy in a 'self-reinforcing cycle', which can persist after the heart is no longer chronically exposed to (or after antagonism of) the risk factors that initiated the cycle. Together with other previously described pathological mechanisms, this proposed cycle can help explain (1) why heart failure progresses, (2) why it can recur after cessation of treatment, and (3) why heart failure is often accompanied by dysfunction of multiple organs. Ideally, therapy of heart failure syndrome would be best attempted before the self-reinforcing cycle is triggered or designed to break the self-reinforcing cycle.
Collapse
Affiliation(s)
- Carlos Fernandez-Patron
- Cardiovascular Research Centre, Department of Biochemistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
41
|
Hattori Y, Hattori K, Ishii K, Kobayashi M. Challenging and target-based shifting strategies for heart failure treatment: An update from the last decades. Biochem Pharmacol 2024; 224:116232. [PMID: 38648905 DOI: 10.1016/j.bcp.2024.116232] [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: 01/22/2024] [Revised: 03/31/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Heart failure (HF) is a major global health problem afflicting millions worldwide. Despite the significant advances in therapies and prevention, HF still carries very high morbidity and mortality, requiring enormous healthcare-related expenditure, and the search for new weapons goes on. Following initial treatment strategies targeting inotropism and congestion, attention has focused on offsetting the neurohormonal overactivation and three main therapies, including angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor antagonists, β-adrenoceptor antagonists, and mineralocorticoid receptor antagonists, have been the foundation of standard treatment for patients with HF. Recently, a paradigm shift, including angiotensin receptor-neprilysin inhibitor, sodium glucose co-transporter 2 inhibitor, and ivabradine, has been added. Moreover, soluble guanylate cyclase stimulator, elamipretide, and omecamtiv mecarbil have come out as a next-generation therapeutic agent for patients with HF. Although these pharmacologic therapies have been significantly successful in relieving symptoms, there is still no complete cure for HF. We may be currently entering a new era of treatment for HF with animal experiments and human clinical trials assessing the value of antibody-based immunotherapy and gene therapy as a novel therapeutic strategy. Such tempting therapies still have some challenges to be addressed but may become a weighty option for treatment of HF. This review article will compile the paradigm shifts in HF treatment over the past dozen years or so and illustrate current landscape of antibody-based immunotherapy and gene therapy as a new therapeutic algorithm for patients with HF.
Collapse
Affiliation(s)
- Yuichi Hattori
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Tobetsu, Japan; Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.
| | - Kohshi Hattori
- Department of Anesthesiology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masanobu Kobayashi
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Tobetsu, Japan
| |
Collapse
|
42
|
Perticone M, Gigliotti S, Shehaj E, Maio R, Suraci E, Miceli S, Andreozzi F, Matera G, Perticone F. Gut Permeability and Immune-Mediated Inflammation in Heart Failure. Biomedicines 2024; 12:1217. [PMID: 38927424 PMCID: PMC11200601 DOI: 10.3390/biomedicines12061217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Heart failure (HF) is characterized by low-grade immune-mediated inflammation due to increased Toll-like receptor (TLR) expression as response to endotoxin increase and dysregulated gut barrier permeability. We investigated TLR expression and possible gut dysbiosis in HF patients compared to a control group. We enrolled 80 Caucasian HF patients and 20 controls. Low-grade immune-mediated inflammation was evaluated by TLR expression, while gut dysbiosis by the detection of zonulin and bacterial endotoxin activity in a semi-quantitative (endotoxin activity assay [EAA]) and quantitative (limulus amebocyte lysate [LAL] test) way. Compared to controls, patients with HF showed significantly higher age and blood pressure values, worse metabolic profile and kidney function, higher inflammatory biomarkers levels, and lower levels of zonulin and endotoxin activity. When dividing failing patients in those with reduced ejection fraction (HF-rEF) and those with preserved ejection fraction (HF-pEF), HF-rEF patients showed significantly higher values of inflammatory biomarkers and TLR expression than HF-pEF patients. Gut permeability biomarkers inversely correlated with the severity of HF and positively with renal function. eGFR was retained as an independent predictor of zonulin variation in all the three groups of failing patients. Present data work to extend current knowledge about the role of gut microbiota in immune-mediated inflammation in HF.
Collapse
Affiliation(s)
- Maria Perticone
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| | - Simona Gigliotti
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (S.G.); (G.M.)
| | - Ermal Shehaj
- Cardiology and Cardiovascular Intensive Care Unit, Presidio Ospedaliero “Giovanni Paolo II” di Lamezia Terme, Azienda Sanitaria Provinciale di Catanzaro, 88046 Lamezia Terme, Italy;
| | - Raffaele Maio
- Geriatrics Unit, P.O. Germaneto, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy; (R.M.); (S.M.)
| | - Edoardo Suraci
- Internal Medicine Unit, P.O. Pugliese-Ciaccio, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy;
| | - Sofia Miceli
- Geriatrics Unit, P.O. Germaneto, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy; (R.M.); (S.M.)
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| | - Giovanni Matera
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (S.G.); (G.M.)
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| |
Collapse
|
43
|
Biegus J, Szenborn L, Zymliński R, Zakliczyński M, Reczuch K, Guzik M, Urban S, Rosiek-Biegus M, Jankowiak B, Iwanek G, Fudim M, Ponikowski P. The early safety profile of simultaneous vaccination against influenza and Respiratory Syncytial Virus (RSV) in patients with high-risk heart failure. Vaccine 2024; 42:2937-2940. [PMID: 38531725 DOI: 10.1016/j.vaccine.2024.03.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
The safety of simultaneous vaccination for Respiratory Syncytial Virus (RSV) and influenza in vulnerable high-risk heart failure (HF) patients remains unclear. In an open-label, prospective study, 105 patients received concurrent influenza (Vaxigrip Tetra, season 2023/2024, Sanofi) and RSV (Arexvy, GSK) vaccinations from September 15th to November 17th, 2023. Adverse events were collected on the fourth-day post-vaccination. Overall, the vaccination was well tolerated, with the most common reaction being injection site pain (63 %). General symptoms occurred in 33 % of patients, predominantly fatigue (23 %), myalgia (12 %), and headache (9 %). Grade 3 reactions were observed in 6 % of patients, and a few experienced temperature elevation or flu-like symptoms, managing them with antipyretics. Notably, there were no exacerbations of HF, hospitalizations, or deaths within a week post-vaccination. This study indicates the safety of simultaneous influenza and RSV vaccination in high-risk HF patients, with a low incidence of mild adverse events.
Collapse
Affiliation(s)
- Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland.
| | - Leszek Szenborn
- Department of Paediatrics and Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Robert Zymliński
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Krzysztof Reczuch
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Mateusz Guzik
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Szymon Urban
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Rosiek-Biegus
- Department and Clinic of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Berenika Jankowiak
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Gracjan Iwanek
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Marat Fudim
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
44
|
Luo X, Shi Y, Ma Y, Liu Y, Jing P, Cao X, Wang J, Hu Z, Cai H. Exploring the mechanism of ShenGui capsule in treating heart failure based on network pharmacology and molecular docking: A review. Medicine (Baltimore) 2024; 103:e37512. [PMID: 38579077 PMCID: PMC10994518 DOI: 10.1097/md.0000000000037512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/13/2023] [Indexed: 04/07/2024] Open
Abstract
ShenGui capsule (SGC), as a herbal compound, has significant effects on the treatment of heart failure (HF), but its mechanism of action is unclear. In this study, we aimed to explore the potential pharmacological targets and mechanisms of SGC in the treatment of HF using network pharmacology and molecular docking approaches. Potential active ingredients of SGC were obtained from the traditional Chinese medicine systems pharmacology database and analysis platform database and screened by pharmacokinetic parameters. Target genes of HF were identified by comparing the toxicogenomics database, GeneCards, and DisGeNET databases. Protein interaction networks and gene-disorder-target networks were constructed using Cytoscape for visual analysis. Gene ontology and Kyoto Encyclopedia of Genes and Genomes were also performed to identify protein functional annotations and potential target signaling pathways through the DAVID database. CB-DOCK was used for molecular docking to explore the role of IL-1β with SGC compounds. Sixteen active ingredients in SGC were screened from the traditional Chinese medicine systems pharmacology database and analysis platform, of which 36 target genes intersected with HF target genes. Protein-protein interactions suggested that each target gene was closely related, and interleukin-1β (IL-1β) was identified as Hub gene. The network pharmacology analysis suggested that these active ingredients were well correlated with HF. Kyoto Encyclopedia of Genes and Genomes enrichment analysis suggested that target genes were highly enriched in pathways such as inflammation. Molecular docking results showed that IL-1β binds tightly to SGC active components. This experiment provides an important research basis for the mechanism of action of SGC in the treatment of HF. In this study, the active compounds of SGC were found to bind IL-1β for the treatment of heart failure.
Collapse
Affiliation(s)
- Xiang Luo
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yixi Liu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pan Jing
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingyu Cao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jincheng Wang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Hu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hongyan Cai
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
45
|
Chen X, Qian J, Liang S, Qian J, Luo W, Shi Y, Zhu H, Hu X, Wu G, Li X, Liang G. Hyperglycemia activates FGFR1 via TLR4/c-Src pathway to induce inflammatory cardiomyopathy in diabetes. Acta Pharm Sin B 2024; 14:1693-1710. [PMID: 38572108 PMCID: PMC10985127 DOI: 10.1016/j.apsb.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 04/05/2024] Open
Abstract
Protein tyrosine kinases (RTKs) modulate a wide range of pathophysiological events in several non-malignant disorders, including diabetic complications. To find new targets driving the development of diabetic cardiomyopathy (DCM), we profiled an RTKs phosphorylation array in diabetic mouse hearts and identified increased phosphorylated fibroblast growth factor receptor 1 (p-FGFR1) levels in cardiomyocytes, indicating that FGFR1 may contribute to the pathogenesis of DCM. Using primary cardiomyocytes and H9C2 cell lines, we discovered that high-concentration glucose (HG) transactivates FGFR1 kinase domain through toll-like receptor 4 (TLR4) and c-Src, independent of FGF ligands. Knocking down the levels of either TLR4 or c-Src prevents HG-activated FGFR1 in cardiomyocytes. RNA-sequencing analysis indicates that the elevated FGFR1 activity induces pro-inflammatory responses via MAPKs-NFκB signaling pathway in HG-challenged cardiomyocytes, which further results in fibrosis and hypertrophy. We then generated cardiomyocyte-specific FGFR1 knockout mice and showed that a lack of FGFR1 in cardiomyocytes prevents diabetes-induced cardiac inflammation and preserves cardiac function in mice. Pharmacological inhibition of FGFR1 by a selective inhibitor, AZD4547, also prevents cardiac inflammation, fibrosis, and dysfunction in both type 1 and type 2 diabetic mice. These studies have identified FGFR1 as a new player in driving DCM and support further testing of FGFR1 inhibitors for possible cardioprotective benefits.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Shiqi Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yujuan Shi
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hong Zhu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiang Hu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Guang Liang
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| |
Collapse
|
46
|
Liu J, Chen X, Zeng L, Zhang L, Wang F, Peng C, Huang X, Li S, Liu Y, Shou W, Li X, Cao D. Targeting S100A9 Prevents β-Adrenergic Activation-Induced Cardiac Injury. Inflammation 2024; 47:789-806. [PMID: 38446361 DOI: 10.1007/s10753-023-01944-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] [Accepted: 12/05/2023] [Indexed: 03/07/2024]
Abstract
Altered cardiac innate immunity is highly associated with the progression of cardiac disease states and heart failure. S100A8/A9 is an important component of damage-associated molecular patterns (DAMPs) that is critically involved in the pathogenesis of heart failure, thus considered a promising target for pharmacological intervention. In the current study, initially, we validated the role of S100A8/A9 in contributing to cardiac injury and heart failure via the overactivation of the β-adrenergic pathway and tested the potential use of paquinimod as a pharmacological intervention of S100A8/A9 activation in preventing cardiac dysfunction, collagen deposition, inflammation, and immune cell infiltration in β-adrenergic overactivation-mediated heart failure. This finding was further confirmed by the cardiomyocyte-specific silencing of S100A9 via the use of the adeno-associated virus (AAV) 9-mediated short hairpin RNA (shRNA) gene silencing system. Most importantly, in the assessment of the underlying cellular mechanism by which activated S100A8/A9 cause aggravated progression of cardiac fibrosis and heart failure, we discovered that the activated S100A8/A9 can promote fibroblast-macrophage interaction, independent of inflammation, which is likely a key mechanism leading to the enhanced collagen production. Our results revealed that targeting S100A9 provides dual beneficial effects, which is not only a strategy to counteract cardiac inflammation but also preclude cardiac fibroblast-macrophage interactions. The findings of this study also indicate that targeting S100A9 could be a promising strategy for addressing cardiac fibrosis, potentially leading to future drug development.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Xin Chen
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Lijun Zeng
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Laiping Zhang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Cuiping Peng
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Ying Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA.
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China.
| | - Dayan Cao
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
47
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
48
|
Grushko OG, Cho S, Tate AM, Rosenson RS, Pinsky DJ, Haus JM, Hummel SL, Goonewardena SN. Glycocalyx Disruption Triggers Human Monocyte Activation in Acute Heart Failure Syndromes. Cardiovasc Drugs Ther 2024; 38:305-313. [PMID: 36260206 DOI: 10.1007/s10557-022-07390-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE Acute heart failure (AHF) syndromes manifest increased inflammation and vascular dysfunction; however, mechanisms that integrate the two in AHF remain largely unknown. The glycocalyx (GAC) is a sugar-based shell that envelops all mammalian cells. Much GAC research has focused on its role in vascular responses, with comparatively little known about how the GAC regulates immune cell function. METHODS In this study, we sought to determine if GAC degradation products are elevated in AHF patients, how these degradation products relate to circulating inflammatory mediators, and whether the monocyte GAC (mGAC) itself modulates monocyte activation. Inflammatory markers and GAC degradation products were profiled using ELISAs. Flow cytometry was used to assess the mGAC and RNA-seq was employed to understand the role of the mGAC in regulating inflammatory activation programs. RESULTS In a cohort of hospitalized AHF patients (n = 17), we found that (1) the GAC degradation product heparan sulfate (HS) was elevated compared with age-matched controls (4396 and 2903 ng/mL; p = 0.01) and that (2) HS and soluble CD14 (a marker of monocyte activation) levels were closely related (Pearson's r = 0.65; p = 0.002). Mechanistically, Toll-like receptor (TLR) activation of human monocytes results in GAC remodeling and a decrease in the mGAC (71% compared with no treatment; p = 0.0007). Additionally, we found that ex vivo enzymatic removal of HS and disruption of the mGAC triggers human monocyte activation and amplifies monocyte inflammatory responses. Specifically, using RNA-seq, we found that enzymatic degradation of the mGAC increases transcription of inflammatory (IL6, CCL3) and vascular (tissue factor/F3) mediators. CONCLUSION These studies indicate that the mGAC is dynamically remodeled during monocyte activation and that mGAC remodeling itself may contribute to the heightened inflammation associated with AHF.
Collapse
Affiliation(s)
- Olga G Grushko
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA
| | - Steven Cho
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA
| | - Ashley M Tate
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA
| | - Robert S Rosenson
- Metabolism and Lipids Unit, Icahn School of Medicine at Mount Sinai, Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Mount Sinai, NY, USA
| | - David J Pinsky
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA
| | - Jacob M Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Scott L Hummel
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA
- VA Ann Arbor Health System, Ann Arbor, MI, USA
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- University of Michigan Frankel Cardiovascular Center, 1500 East Medical Center Drive, SPC 5853, Ann Arbor, MI, 48109-5853, USA.
- VA Ann Arbor Health System, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Zhang QL, Chen XH, Zhou SJ, Lei YQ, Chen Q, Cao H. Relationship between heart failure and intestinal inflammation in infants with congenital heart disease. BMC Microbiol 2024; 24:98. [PMID: 38528458 DOI: 10.1186/s12866-024-03229-0] [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: 07/28/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
OBJECTIVE The association between heart failure (HF) and intestinal inflammation caused by a disturbed intestinal microbiota in infants with congenital heart disease (CHD) was investigated. METHODS Twenty infants with HF and CHD who were admitted to our hospital between October 2021 and March 2022 were included in this study. Twenty age- and sex-matched infants without HF at our hospital were selected as the control group. Faecal samples were obtained from each participant and analysed by enzyme-linked immunoassay and 16 S rDNA sequencing to assess intestinal inflammatory factors and the microbiota. RESULTS The levels of intestinal inflammatory factors, including IL-1β, IL-4, IL-6, IL-17 A and TNF-α, were greatly increased, while the levels of IL-10 were significantly decreased in the HF group compared to the control group (p < 0.05). The intestinal microbial diversity of patients in the HF group was markedly lower than that in the control group (p < 0.05). The abundance of Enterococcus was significantly increased in the HF group compared to the control group (p < 0.05), but the abundance of Bifidobacterium was significantly decreased in the HF group compared to the control group (p < 0.05). The diversity of the intestinal microbiota was negatively correlated with the levels of IL-1β, IL-4, IL-6 and TNF-α in the intestinal tract but was positively correlated with that of IL-10. The abundance of Enterococcus was positively associated with the levels of IL-1β, IL-4, IL-6 and TNF-α in the intestinal tract but was negatively correlated with that of IL-10. NT-proBNP was positively associated with the levels of IL-1β, IL-4, IL-6 and TNF-α in the HF group but was negatively correlated with that of IL-10. The heart function score was positively associated with the levels of IL-1β, IL-4, IL-6 and TNF-α in the HF group but was negatively correlated with that of IL-10. CONCLUSIONS Infants with CHD-related HF had a disordered intestinal microbiota, decreased diversity of intestinal microbes, increased levels of pathogenic bacteria and decreased levels of beneficial bacteria. The increased abundance of Enterococcus and the significant decrease in the diversity of the intestinal microbiota may exacerbate the intestinal inflammatory response, which may be associated with the progression of HF.
Collapse
Affiliation(s)
- Qi-Liang Zhang
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| | - Xiu-Hua Chen
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Si-Jia Zhou
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yu-Qing Lei
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Qiang Chen
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
50
|
Azhar G, Nagano K, Patyal P, Zhang X, Verma A, Wei JY. Deletion of Interleukin-1β Converting Enzyme Alters Mouse Cardiac Structure and Function. BIOLOGY 2024; 13:172. [PMID: 38534442 DOI: 10.3390/biology13030172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024]
Abstract
Interleukin-1β converting enzyme (ICE, caspase-1) is a thiol protease that cleaves the pro-inflammatory cytokine precursors of IL-1β and IL-18 into active forms. Given the association between caspase-1 and cardiovascular pathology, we analyzed the hearts of ICE knockout (ICE KO) mice to test the hypothesis that caspase-1 plays a significant role in cardiac morphology and function. We characterized the histological and functional changes in the hearts of ICE KO mice compared to the Wild type. The cardiomyocytes from the neonatal ICE KO mice showed an impaired response to oxidative stress. Subsequently, the hearts from the ICE KO mice were hypertrophied, with a significant increase in the left ventricular and septal wall thickness and a greater LV mass/body weight ratio. The ICE KO mice hearts exhibited irregular myofibril arrangements and disruption of the cristae in the mitochondrial structure. Proapoptotic proteins that were significantly increased in the hearts of ICE KO versus the Wild type included pErk, pJNK, p53, Fas, Bax, and caspase 3. Further, the antiapoptotic proteins Bag-1 and Bcl-2 are activated in ICE KO hearts. Functionally, there was an increase in the left ventricular epicardial diameter and volume in ICE KO. In conclusion, our findings support the important role of caspase-1 in maintaining cardiac health; specifically, a significant decrease in caspase-1 is detrimental to the cardiovascular system.
Collapse
Affiliation(s)
- Gohar Azhar
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Koichiro Nagano
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Pankaj Patyal
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xiaomin Zhang
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ambika Verma
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|