1
|
Bi X, Wang Z, He J. Recent advances in biomimetic nanodelivery systems for the treatment of myocardial ischemia reperfusion injury. Colloids Surf B Biointerfaces 2025; 247:114414. [PMID: 39626610 DOI: 10.1016/j.colsurfb.2024.114414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025]
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is a significant challenge in the treatment of myocardial infarction, a leading cause of global mortality due to irreversible cardiac damage. Biomimetic nanodelivery systems offer promising therapeutic strategies to address MIRI. In this review, we comprehensively investigate the underlying pathophysiological mechanisms of MIRI and discuss recent advances in biomimetic nanodelivery systems including cell membrane-coated nanoparticles, exosomes, and nanoenzymes as innovative approaches for MIRI treatment. We emphasize the advantages and potential of biomimetic strategies in enhancing therapeutic efficacy, assess the preclinical effectiveness of these nanodelivery systems, and discuss the challenges associated with translating these approaches into clinical practice. This paper aims to provide new perspectives on biomimetic strategies for MIRI treatment, contributing to the development of effective drug delivery systems.
Collapse
Affiliation(s)
- Xiaojun Bi
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Ze Wang
- Dalian Medical University, Liaoning 116044, China
| | - Jingteng He
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
2
|
Björkenheim A, Sunnefeldt E, Finke K, Smith DR, Fröbert O, Brasier N. Biomarkers of inflammation in sweat after myocardial infarction. Sci Rep 2025; 15:5564. [PMID: 39955425 PMCID: PMC11829942 DOI: 10.1038/s41598-025-90240-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
ST-elevation myocardial infarction (STEMI) triggers a significant inflammatory response. Sweat may offer a novel, non-invasive medium for monitoring inflammation. In this prospective study, we characterized the inflammatory signatures in plasma and sweat collected from the skin surface of two patient groups: (1) 18 STEMI patients immediately following percutaneous coronary intervention (exposure) and (2) six patients who underwent outpatient angiography without subsequent intervention (control). Levels of 92 biomarkers were measured using a high-throughput proteomic assay and reassessed after 4-6 weeks in STEMI patients. Adjusting for patient group, sweat biomarkers did not show significant changes over time. In plasma, hepatocyte growth factor and interleukin-6 showed a significant decrease from the acute phase to follow-up, adjusted for patient group. STAM binding protein was significantly higher in the sweat of STEMI patients compared to controls, adjusted for time effects. While sweat was less sensitive than plasma for detecting biomarker levels in the setting of STEMI, its longitudinal analysis via wearable sensors holds promise for detecting specific markers.Trial registration: The trial is registered on www.clinicaltrials.gov with the trial registration number NCT05843006.
Collapse
Affiliation(s)
- Anna Björkenheim
- Department of Cardiology, School of Medical Sciences, Örebro University, 701 82, Örebro, Sweden.
| | - Erik Sunnefeldt
- Department of Cardiology, School of Medical Sciences, Örebro University, 701 82, Örebro, Sweden
| | - Karl Finke
- Department III of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Daniel Robert Smith
- Clinical Epidemiology and Biostatistics, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, 701 82, Örebro, Sweden
| | - Ole Fröbert
- Department of Cardiology, School of Medical Sciences, Örebro University, 701 82, Örebro, Sweden
- Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, 8000, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8000, Aarhus, Denmark
| | - Noé Brasier
- Department of Health Science and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Diaz-Arocutipa C, Bueno H, Moreno G, Olmos VJ, Vicent L. DIFFERENCES ON IN-HOSPITAL OUTCOMES IN PATIENTS WITH CARDIOGENIC SHOCK DUE TO STEMI VERSUS NSTEMI USING A NATIONWIDE DATABASE. Shock 2025; 63:36-42. [PMID: 39671550 DOI: 10.1097/shk.0000000000002480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
ABSTRACT Background: Our study aims to compare in-hospital management and outcomes in patients with cardiogenic shock due to ST-segment elevation myocardial infarction (STEMI) versus non-ST-segment elevation myocardial infarction (NSTEMI). Methods: We conducted a retrospective cohort study using the National Inpatient Sample database between 2016-2019, including patients with STEMI/NSTEMI complicated by cardiogenic shock. An inverse probability treatment weighting analysis was performed to compare in-hospital management and outcomes between patients with STEMI and NSTEMI. Adjusted relative risks (aRR) with their 95% confidence intervals (CIs) were estimated. Results: A total of 150,395 patients with cardiogenic shock due to acute myocardial infarction were included, of whom 52.8% had STEMI. The median age was 68 years (60-77) and 35% were female. Percutaneous coronary intervention, intra-aortic balloon counterpulsation, percutaneous ventricular assist device, extracorporeal membrane oxygenation, and mechanical ventilation use were significantly higher in the STEMI group compared to NSTEMI. Coronary artery bypass grafting, renal replacement therapy, length of hospital stay, and total costs were lower in the STEMI group. Pulmonary arterial catheterization and cardiac transplantation were similar between both groups. Inverse probability treatment weighting analysis showed that in-hospital mortality was significantly higher in the STEMI group compared to NSTEMI (34.2% vs. 28.8%, aRR 1.19, 95% CI 1.14-1.23) and also major bleeding. Conclusion: In conclusion, patients with cardiogenic shock due to STEMI had worse prognosis, higher use of percutaneous coronary intervention/mechanical circulatory support, and major bleeding than the NSTEMI group. In contrast, patients with NSTEMI had greater use of coronary artery bypass grafting and hospital resources.
Collapse
Affiliation(s)
- Carlos Diaz-Arocutipa
- Unidad de Revisiones Sistemáticas y Meta-análisis (URSIGET), Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima, Peru
| | | | | | | | | |
Collapse
|
4
|
Liu L, Hu J, Lei H, Qin H, Wang C, Gui Y, Xu D. Regulatory T Cells in Pathological Cardiac Hypertrophy: Mechanisms and Therapeutic Potential. Cardiovasc Drugs Ther 2024; 38:999-1015. [PMID: 37184744 DOI: 10.1007/s10557-023-07463-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/29/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Pathological cardiac hypertrophy is linked to immune-inflammatory injury, and regulatory T cells (Tregs) play a crucial role in suppressing immune-inflammatory responses. However, the precise role of Tregs in pathological cardiac hypertrophy remains unclear. OBJECTIVE To summarize the current knowledge on the role and mechanisms of Tregs in pathological cardiac hypertrophy and explore their perspectives and challenges as a new therapeutic approach. RESULTS Treg cells may play an important protective role in pressure overload (hypertension, aortic stenosis), myocardial infarction, metabolic disorders (diabetes, obesity), acute myocarditis, cardiomyopathy (hypertrophic cardiomyopathy, storage diseases), and chronic obstructive pulmonary disease-related pathological cardiac hypertrophy. Although some challenges remain, the safety and efficacy of Treg-based therapies have been confirmed in some clinical trials, and engineered antigen-specific Treg cells may have better clinical application prospects due to stronger immunosuppressive function and stability. CONCLUSION Targeting the immune-inflammatory response via Treg-based therapies might provide a promising and novel future approach to the prevention and treatment of pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Huali Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chunfang Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yajun Gui
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
5
|
Matetic A, Kyriacou T, Mamas MA. Machine-learning clustering analysis identifies novel phenogroups in patients with ST-elevation acute myocardial infarction. Int J Cardiol 2024; 411:132272. [PMID: 38880421 DOI: 10.1016/j.ijcard.2024.132272] [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: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Machine learning clustering of patients with ST-elevation acute myocardial infarction (STEMI) may provide important insights into their risk profile, management and prognosis. METHODS All adult discharges for STEMI in the National Inpatient Sample (October 2015 to December 2019) were included, excluding patients with prior myocardial infarction. Machine-learning clustering analysis was used to define clusters based on 21 clinical attributes of interest. Main outcomes of the study were cluster-based comparison of risk profile, in-hospital clinical outcomes and utilization of invasive management. Binomial hierarchical multivariable logistic regression with adjusted odds ratios (aOR) and 95% confidence intervals (95% CI) was used to detect the between-cluster differences. RESULTS Out of overall 470,960 STEMI cases, the machine-learning analysis revealed 4 different clusters with 205,640 (cluster 0: 'behavioural risk cluster'), 146,400 (cluster 1: 'least comorbidity cluster'), 45,100 (cluster 2: 'diabetes with end-organ damage cluster') and 73,820 (cluster 3: 'cardiometabolic cluster') cases. Attributes with the highest importance for clustering were hypertension and diabetes. After multivariable adjustment, patients from 'diabetes with end-organ damage cluster' exhibited the worst mortality, MACCE and ischemic stroke (p < 0.001 for all), as well as the lowest utilization of invasive management (p < 0.001 for all), in comparison to other clusters. Patients from 'behavioural risk cluster' exhibited the best in-hospital prognosis and the highest utilization of invasive management, compared to other clusters (p < 0.001 for all). CONCLUSIONS Machine learning driven clustering of inpatients with STEMI reveals important population subgroups with distinct prevalence, risk profile, prognosis and management. Data driven approaches may identify high risk phenogroups and warrants further study.
Collapse
Affiliation(s)
- Andrija Matetic
- Department of Cardiology, University Hospital of Split, Split, Croatia; Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, United Kingdom
| | - Theocharis Kyriacou
- School of Computer Science and Mathematics, Keele University, Keele, United Kingdom
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, United Kingdom; National Institute for Health and Care Research (NIHR), Birmingham Biomedical Research Centre, United Kingdom.
| |
Collapse
|
6
|
Zarei B, Bozorgi A, Khoshfetrat M, Arefizadeh R, Mohsenizadeh SA, Mousavi SH, Jalali A, Shafiee A. Incidence and predictors of new-onset atrial fibrillation in ST-elevation myocardial infarction: A single-center study. Health Sci Rep 2024; 7:e2226. [PMID: 38957860 PMCID: PMC11217016 DOI: 10.1002/hsr2.2226] [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: 10/14/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
Background and Aims Atrial fibrillation (AF) is a common arrhythmia that occurs following ST-elevation myocardial infarction (STEMI) and can significantly impact clinical outcomes. We investigated the incidence and predictors of AF following STEMI in patients, as well as its association with major adverse cardiac and cerebrovascular events (MACCE). Methods We conducted a retrospective cohort study, including all STEMI patients who presented under code 247 to Tehran Heart Center between 2016 and 2020 and completed a 1-year follow-up. Patients were divided into two groups based on the development of AF during follow-up, and their baseline and clinical characteristics were compared. We used multivariable regression models to identify predictors of MACCE. Results Out of 3647 STEMI patients, 84 (2.3%) developed new-onset AF (NOAF). Patients with AF were significantly older and had lower levels of total and low-density lipoprotein cholesterol, triglyceride, and hemoglobin, but higher levels of fasting blood sugar and creatinine. AF patients were also more likely to have a history of hypertension, chronic kidney disease (CKD), congestive heart failure, and cerebrovascular accidents. The multivariable logistic regression model identified the CHA2DS2-VASc score and CKD as independent predictors of NOAF following primary percutaneous coronary intervention. Furthermore, the incidence of MACCE was higher in the AF group, and AF independently predicted MACCE with a hazard ratio of 2.766. Conclusion The CHA2DS2-VASc score and the presence of CKD can serve as useful predictors of NOAF among patients with STEMI. Early detection and appropriate management are crucial to improve outcomes.
Collapse
Affiliation(s)
- Behrouz Zarei
- Department of Cardiology, School of MedicineAJA University of Medical SciencesTehranIran
| | - Ali Bozorgi
- Department of Cardiology, Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Mehran Khoshfetrat
- Department of Cardiology, School of MedicineAJA University of Medical SciencesTehranIran
| | - Reza Arefizadeh
- Department of Cardiology, School of MedicineAJA University of Medical SciencesTehranIran
| | | | - Seyyed Hossein Mousavi
- Department of Cardiology, School of MedicineAJA University of Medical SciencesTehranIran
| | - Arash Jalali
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Akbar Shafiee
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
7
|
Chen P, Gao Z, Guo M, Pan D, Zhang H, Du J, Shi D. Efficacy and safety of Panax notoginseng saponin injection in the treatment of acute myocardial infarction: a systematic review and meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1353662. [PMID: 38576488 PMCID: PMC10991745 DOI: 10.3389/fphar.2024.1353662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/16/2024] [Indexed: 04/06/2024] Open
Abstract
Purpose: This study aimed to assess the efficacy and safety of Panax notoginseng saponin (PNS) injection, when combined with conventional treatment (CT), for acute myocardial infarction (AMI). Methods: Comprehensive searches were conducted in seven databases from inception until 28 September 2023. The search aimed to identify relevant randomized controlled trials (RCTs) focusing on PNS injection in the context of AMI. This meta-analysis adhered to the PRISMA 2020 guidelines, and its protocol was registered with PROSPERO (number: CRD42023480131). Result: Twenty RCTs involving 1,881 patients were included. The meta-analysis revealed that PNS injection, used adjunctively with CT, significantly improved treatment outcomes compared to CT alone, as evidenced by the following points: (1) enhanced total effective rate [OR = 3.09, p < 0.05]; (2) decreased incidence of major adverse cardiac events [OR = 0.32, p < 0.05]; (3) reduction in myocardial infarct size [MD = -6.53, p < 0.05]; (4) lower ST segment elevation amplitude [MD = -0.48, p < 0.05]; (5) mitigated myocardial injury as indicated by decreased levels of creatine kinase isoenzymes [MD = -11.19, p < 0.05], cardiac troponin T [MD = -3.01, p < 0.05], and cardiac troponin I [MD = -10.72, p < 0.05]; (6) enhanced cardiac function, reflected in improved brain natriuretic peptide [MD = -91.57, p < 0.05], left ventricular ejection fraction [MD = 5.91, p < 0.05], left ventricular end-diastolic dimension [MD = -3.08, p < 0.05], and cardiac output [MD = 0.53, p < 0.05]; (7) reduced inflammatory response, as shown by lower levels of C-reactive protein [MD = -2.99, p < 0.05], tumor necrosis factor-α [MD = -6.47, p < 0.05], interleukin-6 [MD = -24.46, p < 0.05], and pentraxin-3 [MD = -2.26, p < 0.05]; (8) improved vascular endothelial function, demonstrated by decreased endothelin-1 [MD = -20.56, p < 0.05] and increased nitric oxide [MD = 1.33, p < 0.05]; (9) alleviated oxidative stress, evidenced by increased superoxide dismutase levels [MD = 25.84, p < 0.05]; (10) no significant difference in adverse events [OR = 1.00, p = 1.00]. Conclusion: This study highlighted the efficacy and safety of adjunctive PNS injections in enhancing AMI patient outcomes beyond CT alone. Future RCTs need to solidify these findings through rigorous methods. Systematic Review Registration: (https://www.crd.york.ac.uk/PROSPERO/), identifier (CRD42023480131).
Collapse
Affiliation(s)
- Pengfei Chen
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuye Gao
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ming Guo
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Deng Pan
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianpeng Du
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital, Cohina Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Nogic J, Cailes B, Yeoh J, Yudi M, Tong D, Farouque O, Brennan A, Dinh D, Brown AJ, Clark D. Natural History and Clinical Outcomes After ST-Segment Elevation Myocardial Infarction Without Stent Insertion. Am J Cardiol 2023; 209:60-65. [PMID: 37863114 DOI: 10.1016/j.amjcard.2023.09.096] [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: 06/13/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
After restoration of coronary perfusion in patients presenting with ST-segment elevation myocardial infarction (STEMI), discrete severe stenotic coronary lesions are not always apparent. There remains ambiguity whether drug-eluting stent (DES) insertion or initial medical management is best practice. We sought to assess short-term clinical outcomes in patients presenting with STEMI without initial stent insertion. Patients who underwent percutaneous coronary intervention for STEMI between 2014 and 2020 were prospectively enrolled and assessed for inclusion. Patients presenting with in-stent restenosis or stent thrombosis, or who did not survive to hospital discharge were excluded. Of 13,871 patients presenting, 456 (3.3%) were treated without initial stenting. These patients were older than those treated with DES (66.1 ± 13.6 vs 62.3 ± 12.4 years, p <0.001), had higher rates of diabetes (23.5% vs 16.0%, p <0.001) and previous revascularization with either percutaneous coronary intervention (14.0% vs 7.3%, p <0.001) or coronary artery bypass graft (3.5% vs 1.8%, p = 0.008). Thirty-day mortality was elevated in patients treated without stenting compared to those receiving DES (4.2% vs 0.9%, p <0.001), as were rates of myocardial infarction (1.3% vs 0.5%, p = 0.026) and major adverse cardiac events (10.5% vs 2.4%, p <0.001). After propensity matching, a trend toward increased mortality remained (4.2% vs 2.0%, p = 0.055). In conclusion, a no-stenting initial strategy, compared with DES insertion, is associated with increased 30-day mortality in those presenting with STEMI without severe stenosis. These data suggest when appropriate, current-generation DES insertion should be undertaken.
Collapse
Affiliation(s)
- Jason Nogic
- Department of Cardiology, Eastern Health, Melbourne, Victoria, Australia; Monash Cardiovascular Research Centre, Monash University and Victorian Heart Hospital, Monash Health, Melbourne, Victoria, Australia; Monash Cardiovascular Research Centre, Monash University and Victorian Heart Hospital, Monash Health, Melbourne, Victoria, Australia.
| | - Benjamin Cailes
- Department of Cardiology, Austin Hospital Clinical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Julian Yeoh
- Department of Cardiology, Austin Hospital Clinical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Matias Yudi
- Department of Cardiology, Austin Hospital Clinical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - David Tong
- Department of Cardiology, Eastern Health, Melbourne, Victoria, Australia
| | - Omar Farouque
- Department of Cardiology, Austin Hospital Clinical School, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Diem Dinh
- Monash University, Melbourne, Victoria, Australia
| | - Adam J Brown
- Monash Cardiovascular Research Centre, Monash University and Victorian Heart Hospital, Monash Health, Melbourne, Victoria, Australia
| | - David Clark
- Department of Cardiology, Austin Hospital Clinical School, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Aleem MA, Macintyre CR, Rahman B, Islam AKMM, Akhtar Z, Chowdhury F, Qadri F, Chughtai AA. Association of recent respiratory illness and influenza with acute myocardial infarction among the Bangladeshi population: A case-control study. Epidemiol Infect 2023; 151:e204. [PMID: 38031480 PMCID: PMC10753452 DOI: 10.1017/s0950268823001863] [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: 07/15/2023] [Revised: 10/25/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023] Open
Abstract
Current evidence suggests that recent acute respiratory infections and seasonal influenza may precipitate acute myocardial infarction (AMI). This study examined the potential link between recent clinical respiratory illness (CRI) and influenza, and AMI in Bangladesh. Conducted during the 2018 influenza season at a Dhaka tertiary-level cardiovascular (CV) hospital, it included 150 AMI cases and two control groups: 44 hospitalized cardiac patients without AMI and 90 healthy individuals. Participants were matched by gender and age groups. The study focused on self-reported CRI and laboratory-confirmed influenza ascertained via quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) within the preceding week, analyzed using multivariable logistic regression. Results showed that cases reported CRI, significantly more frequently than healthy controls (27.3% vs. 13.3%, adjusted odds ratio (aOR): 2.21; 95% confidence interval (CI): 1.05-4.06), although this was not significantly different from all controls (27.3% vs. 22.4%; aOR: 1.19; 95% CI: 0.65-2.18). Influenza rates were insignificantly higher among cases than controls. The study suggests that recent respiratory illnesses may precede AMI onset among Bangladeshi patients. Infection prevention and control practices, as well as the uptake of the influenza vaccine, may be advocated for patients at high risk of acute CV events.
Collapse
Affiliation(s)
- Mohammad Abdul Aleem
- School of Population Health, UNSW Medicine, The University of New South Wales, Sydney, NSW, Australia
- Program for Emerging Infections, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, Bangladesh
| | - C. Raina Macintyre
- Biosecurity Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Bayzidur Rahman
- Biosecurity Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - A. K. M. Monwarul Islam
- Department of Cardiology, National Institute of Cardiovascular Diseases (NICVD), Dhaka, Bangladesh
| | - Zubair Akhtar
- Program for Emerging Infections, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, Bangladesh
- Biosecurity Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Fahmida Chowdhury
- Program for Emerging Infections, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Firdausi Qadri
- Respiratory and Enteric Infections, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (ICDDR, B), Dhaka, Bangladesh
| | - Abrar Ahmad Chughtai
- School of Population Health, UNSW Medicine, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
10
|
Shiraz Rizvi SM, Sunny S, Wani IA, Mahdi F, Zaidi ZH, Rajasekaran NS. Influence of electrolyte imbalance on regional wall motion abnormalities in STEMI patients of North Indian origin. Front Cardiovasc Med 2023; 10:1223954. [PMID: 38099220 PMCID: PMC10720728 DOI: 10.3389/fcvm.2023.1223954] [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: 05/17/2023] [Accepted: 10/10/2023] [Indexed: 12/17/2023] Open
Abstract
Assessing regional wall motion abnormalities (RWMA) in the myocardium may provide early diagnosis and treat chronic remodeling in STEMI patients. We assessed RWMA in 217 subjects with anterior STEMI admitted to Era University Hospital in Lucknow, UP, India. Besides abnormalities in the LAD territory, sub-sets of patients exhibited diffuse regional myocardial dysfunction. Interestingly, variations in serum electrolytes, specifically sodium and potassium, significantly affected the distribution and frequency of RWMA. Notably, RWMA occurred in the basal septum, apical septum, apex, and lateral wall in the anterior STEMI group. Additionally, the rate of regional dysfunction varied with serum urea and creatinine levels. This suggests that anterior STEMI can manifest myocardial abnormalities beyond the LAD territory. These findings indicate that ST-segment elevation might not be specific, possibly influenced by electrolyte changes affecting cardiac rhythm. Therefore, diagnosing and correcting region-specific wall motion abnormalities and electrolyte imbalances may improve outcomes in STEMI patients.
Collapse
Affiliation(s)
- S. Mohd. Shiraz Rizvi
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Sini Sunny
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, United States
| | - Irshad A. Wani
- Department of Cardiology, Era’s Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Farzana Mahdi
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Zeeshan H. Zaidi
- Department of Community Medicine, Era’s Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Namakkal S. Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
11
|
Zhang J, Liu L, Dong Z, Lu X, Hong W, Liu J, Zou X, Gao J, Jiang H, Sun X, Hu K, Yang Y, Ge J, Luo X, Sun A. An ischemic area-targeting, peroxynitrite-responsive, biomimetic carbon monoxide nanogenerator for preventing myocardial ischemia-reperfusion injury. Bioact Mater 2023; 28:480-494. [PMID: 37408796 PMCID: PMC10318466 DOI: 10.1016/j.bioactmat.2023.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
Myocardial ischemia-reperfusion (MI/R) injury is common in patients who undergo revascularization therapy for myocardial infarction, often leading to cardiac dysfunction. Carbon monoxide (CO) has emerged as a therapeutic molecule due to its beneficial properties such as anti-inflammatory, anti-apoptotic, and mitochondrial biogenesis-promoting properties. However, its clinical application is limited due to uncontrolled release, potential toxicity, and poor targeting efficiency. To address these limitations, a peroxynitrite (ONOO-)-triggered CO donor (PCOD585) is utilized to generate a poly (lactic-co-glycolic acid) (PLGA)-based, biomimetic CO nanogenerator (M/PCOD@PLGA) that is coated with the macrophage membrane, which could target to the ischemic area and neutralize proinflammatory cytokines. In the ischemic area, local produced ONOO- triggers the continuous release of CO from M/PCOD@PLGA, which efficiently ameliorates MI/R injury by clearing harmful ONOO-, attenuating the inflammatory response, inhibiting cardiomyocyte apoptosis, and promoting mitochondrial biogenesis. This study provides a novel insight into the safe therapeutic use of CO for MI/R injury by utilizing a novel CO donor combined with biomimetic technology. The M/PCOD@PLGA nanogenerator offers targeted delivery of CO to the ischemic area, minimizing potential toxicity and enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Jinyan Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Liwei Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xicun Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenxuan Hong
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiaoyi Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Jinfeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Hao Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Youjun Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiao Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| |
Collapse
|
12
|
Harm T, Rath D, Kreisselmeier KP, Baas L, Prang C, Gekeler S, Schröder S, Gawaz MP, Geisler T, Müller II, Müller KAL. Thrombus architecture is influenced by the antiplatelet loading treatment in patients with acute myocardial infarction. Thromb Res 2023; 230:45-54. [PMID: 37634310 DOI: 10.1016/j.thromres.2023.08.004] [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: 06/05/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Intracoronary thrombus formation is a main cause of acute myocardial infarction triggered by platelet activation. However, there are no data on the impact of different treatment strategies with antiplatelet agents before percutaneous coronary intervention (PCI) on histological characteristics of thrombus formation. OBJECTIVE In this study, we investigate the impact of preinterventional administration of the P2Y12-inhibitors clopidogrel and prasugrel on thrombus composition, highlighting significant changes associated with the antiplatelet pre-treatment. METHODS We prospectively enrolled 104 consecutive patients with ST-segment elevation myocardial infarction (STEMI) undergoing immediate PCI and thrombus aspiration by immunohistochemical staining along with RNA-sequencing employing Nanostring analysis. Fifty-two patients were treated with either prasugrel loading (60 mg) or clopidogrel loading (600 mg) prior to PCI, respectively. RESULTS In Patients with STEMI, intracoronary thrombus architecture was significantly altered between patients pre-treated with prasugrel when compared to clopidogrel. Fibrin content of thrombi was significantly decreased (41.8 % versus 66.7 %, p = 0.009) after pre-treatment with prasugrel compared to clopidogrel. Furthermore, levels of MPO positive cells in intracoronary thrombi were significantly decreased in patients with prasugrel pre-treatment (90.5 versus 201.1, p = 0.014) indicating an association of antiplatelet pre-treatment and the inflammatory responses during thrombus formation. Most strikingly, we observed significant differences among both pre-treatment groups regarding altered RNA expression and signaling pathways of thrombo-inflammatory processes within the thrombotic material, which were independently associated with antiplatelet strategies. CONCLUSIONS Our study elucidates the impact of antiplatelet pre-treatment on thrombus remodeling and architecture, thereby lowering the risk of recurrent adverse cardiovascular events in prasugrel-treated patients.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Klaus-Peter Kreisselmeier
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Livia Baas
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Carolin Prang
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Sarah Gekeler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Stephen Schröder
- Department of Cardiology and Angiology, Klinik am Eichert, Eichertstraße 3, 73035 Göppingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany.
| | - Iris Irmgard Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| |
Collapse
|
13
|
Kelly C, Lan NSR, Phan J, Hng C, Matthews A, Rankin JM, Schultz CJ, Hillis GS, Reid CM, Dwivedi G, Figtree GA, Ihdayhid AR. Characteristics and Outcomes of Young Patients With ST-Elevation Myocardial Infarction Without Standard Modifiable Risk Factors. Am J Cardiol 2023; 202:81-89. [PMID: 37423175 DOI: 10.1016/j.amjcard.2023.06.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 06/11/2023] [Indexed: 07/11/2023]
Abstract
Patients with ST-elevation myocardial infarction (STEMI) with no standard modifiable risk factors (SMuRFs: hypertension, diabetes mellitus, hypercholesterolemia, and smoking) have worse short-term mortality than those with SMuRFs. Whether this association extends to younger patients is unclear. A retrospective cohort study was performed of patients aged 18 to 45 years with STEMI at 3 Australian hospitals between 2010 and 2020. Nonatherosclerotic causes of STEMI were excluded. The primary outcome was 30-day all-cause mortality. Secondary outcomes included 1 and 2-year mortality. Cox proportional hazards analysis was used. Of 597 patients, the median age was 42 (interquartile range 38 to 44) years, 85.1% were men and 8.4% were SMuRF-less. Patients who are SMuRF-less were >2 times more likely to have cardiac arrest (28.0% vs 12.6%, p = 0.003); require vasopressors (16.0% vs 6.8%, p = 0.018), mechanical support (10.0% vs 2.3%, p = 0.046), or intensive care admission (20.0% vs 5.7%, p <0.001); and have higher rate of left anterior descending artery infarcts than those with SMuRFs (62.0% vs 47.2%, p = 0.045). No significant differences in thrombolysis or percutaneous intervention were observed. Guideline-directed medical therapy at discharge was high (>90%), and not different in the SMuRF-less. 30-day mortality was almost fivefold higher in the SMuRF-less (hazard ratio 4.70, 95% confidence interval 1.66 to 13.35, p = 0.004), remaining significant at 1 and 2 years. In conclusion, young patients who are SMuRF-less have a higher 30-day mortality after STEMI than their counterparts with SMuRFs. This may be partially mediated by higher rates of cardiac arrest and left anterior descending artery territory events. These findings further highlight the need for improved prevention and management of SMuRF-less STEMI.
Collapse
Affiliation(s)
- Ciaran Kelly
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
| | - Nick S R Lan
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia; Department of Cardiology, Royal Perth Hospital, Perth, Australia; Medical School, The University of Western Australia, Perth, Australia; Harry Perkins Institute of Medical Research, Perth, Australia
| | - Jane Phan
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
| | - Cherng Hng
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
| | - Amy Matthews
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
| | - James M Rankin
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
| | - Carl J Schultz
- Department of Cardiology, Royal Perth Hospital, Perth, Australia; Medical School, The University of Western Australia, Perth, Australia
| | - Graham S Hillis
- Department of Cardiology, Royal Perth Hospital, Perth, Australia; Medical School, The University of Western Australia, Perth, Australia
| | | | - Girish Dwivedi
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia; Medical School, The University of Western Australia, Perth, Australia; Harry Perkins Institute of Medical Research, Perth, Australia
| | - Gemma A Figtree
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | - Abdul Rahman Ihdayhid
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia; Harry Perkins Institute of Medical Research, Perth, Australia; Curtin Medical School, Curtin University, Perth, Australia.
| |
Collapse
|
14
|
Lindsey ML, Becirovic‐Agic M. Skin wound healing as a mirror to cardiac wound healing. Exp Physiol 2023; 108:1003-1010. [PMID: 37093202 PMCID: PMC10948174 DOI: 10.1113/ep090888] [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: 03/13/2023] [Accepted: 04/06/2023] [Indexed: 04/25/2023]
Abstract
NEW FINDINGS What is the topic of this review? Wound healing is a general response of the body to injury and can be divided into three phases: inflammation, inflammation resolution and repair. In this review, we compare the wound-healing response of the skin after an injury and the wound-healing response of the heart after a myocardial infarction. What advances does it highlight? We highlight differences and similarities between skin and cardiac wound healing and summarize how skin can be used to provide information about the heart. ABSTRACT Wound healing is a general response of the body to injury. All organs share in common three response elements to wound healing: inflammation to prevent infection and stimulate the removal of dead cells, active anti-inflammatory signalling to turn off the inflammatory response, and a repair phase characterized by extracellular matrix scar formation. The extent of scar formed depends on the ability of endogenous cells that populate each organ to regenerate. The skin has keratinocytes that have regenerative capacity, and in general, wounds are fully re-epithelialized. Heart, in contrast, has cardiac myocytes that have little to no regenerative capacity, and necrotic myocytes are entirely replaced by scars. Despite differences in tissue regeneration, the skin and heart share many wound-healing properties that can be exploited to predict the cardiac response to pathology. We summarize in this review article our current understanding of how the response of the skin to a wounding event can inform us about the ability of the myocardium to respond to a myocardial infarction.
Collapse
Affiliation(s)
- Merry L. Lindsey
- School of Graduate StudiesMeharry Medical CollegeNashvilleTennesseeUSA
- Research ServiceNashville VA Medical CenterNashvilleTennesseeUSA
| | - Mediha Becirovic‐Agic
- Integrative Physiology, Department of Medical Cell BiologyUppsala UniversityUppsalaSweden
| |
Collapse
|
15
|
Savovic Z, Pindovic B, Nikolic M, Simic I, Davidovic G, Ignjatovic V, Vuckovic J, Zornic N, Nikolic Turnic T, Zivkovic V, Srejovic I, Bolevich S, Jakovljevic V, Iric Cupic V. Prognostic Value of Redox Status Biomarkers in Patients Presenting with STEMI or Non-STEMI: A Prospective Case-Control Clinical Study. J Pers Med 2023; 13:1050. [PMID: 37511663 PMCID: PMC10381258 DOI: 10.3390/jpm13071050] [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: 04/03/2023] [Revised: 04/26/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: The aim of our study was to determine the role of oxidative stress (OS) during early evaluation of acute ST-elevated myocardial infarction (STEMI) and non-ST-elevated myocardial infarction (NSTEMI) patients in order to define the role of redox balance in profiling the development of myocardial infarction (MI). (2) Methods: This prospective observational case-control study included 40 consecutive STEMI and 39 NSTEMI patients hospitalized in the coronary care unit of the cardiology clinic at the Kragujevac Clinical Center, Serbia, between 1 January 2016 and 1 January 2017. Blood samples were collected from all patients for measuring cardio-specific enzymes at admission and 12 h after admission to evaluate systemic oxidative stress biomarkers and the activity of antioxidant enzymes. (3) Results: In this study, participants were predominately female (52%), with a mean age of 56.17 ± 1.22 years old in the STEMI group and 69.17 ± 3.65 in the non-STEMI group. According to the Killip classification, the majority of patients (>50%) were at the second and third level. We confirmed the elevation of superoxide anion radicals in the non-STEMI group 6 h after admission in comparison with the STEMI and CTRL groups, but levels had decreased 12 h after admission. Levels of hydrogen peroxide were statistically significantly increased in the NSTEMI group. A positive correlation of superoxide anion radicals and levels of troponin I at admission was observed (r = 0.955; p = 0.045), as well as an inverse correlation between reduced glutathione and levels of NT-pBNP measured 6 h after admission (r = -0.973; p = 0.027). (4) Conclusions: We confirmed that superoxide anion radicals and reduced glutathione observed together with hs-troponin I at admission and NT-pBNP during hospital treatment could be predictors of ST evolution.
Collapse
Affiliation(s)
- Zorica Savovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
| | - Bozidar Pindovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Maja Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Simic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Goran Davidovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Vladimir Ignjatovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Jelena Vuckovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Nenad Zornic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Tamara Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- N.A. Semashko Public Health and Healthcare Department, F. F. Erismann Institute of Public Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Clinical Pharmacology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Clinical Pharmacology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Sergej Bolevich
- Department of Human Pathology, 1st Moscow State Medical, University I. M. Sechenov, 119991 Moscow, Russia
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical, University I. M. Sechenov, 119991 Moscow, Russia
| | - Violeta Iric Cupic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
16
|
Carbone F, Liberale L, Libby P, Montecucco F. Vitamin D in atherosclerosis and cardiovascular events. Eur Heart J 2023; 44:2078-2094. [PMID: 36943351 PMCID: PMC10281557 DOI: 10.1093/eurheartj/ehad165] [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: 07/22/2022] [Revised: 01/30/2023] [Accepted: 03/04/2023] [Indexed: 03/23/2023] Open
Abstract
Both experimental and clinical findings linking vitamin D to cardiovascular (CV) risk have prompted consideration of its supplementation to improve overall health. Yet several meta-analyses do not provide support for the clinical effectiveness of this strategy. Meanwhile, the understanding of the roles of vitamin D in the pathophysiology of CV diseases has evolved. Specifically, recent work has revealed some non-classical pleiotropic effects of vitamin D, increasing the complexity of vitamin D signalling. Within particular microenvironments (e.g. dysfunctional adipose tissue and atherosclerotic plaque), vitamin D can act locally at cellular level through intracrine/autocrine/paracrine feedforward and feedback circuits. Within atherosclerotic tissues, 'local' vitamin D levels may influence relevant systemic consequences independently of its circulating pool. Moreover, vitamin D links closely to other signalling pathways of CV relevance including those driving cellular senescence, ageing, and age-related diseases-among them CV conditions. This review updates knowledge on vitamin D biology aiming to clarify the widening gap between experimental and clinical evidence. It highlights the potential reverse causation confounding correlation between vitamin D status and CV health, and the need to consider novel pathophysiological concepts in the design of future clinical trials that explore the effects of vitamin D on atherosclerosis and risk of CV events.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Peter Libby
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa—Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
17
|
Yang T, Zhang D. Research progress on the effects of novel hypoglycemic drugs in diabetes combined with myocardial ischemia/reperfusion injury. Ageing Res Rev 2023; 86:101884. [PMID: 36801379 DOI: 10.1016/j.arr.2023.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Acute myocardial infarction (AMI) reperfusion is associated with ischemia/reperfusion (I/R) injury, which leads to enlarged myocardial infarction size, poor healing of the infarcted myocardium, and poor left ventricular remodeling, thus increasing the risk of major adverse cardiovascular events (MACEs). Diabetes increases myocardial susceptibility to I/R injury, decreases myocardial responsiveness to cardioprotective strategies, exacerbates myocardial I/R injury, and expands the infarct size of AMI, thereby increasing the incidence of malignant arrhythmias and heart failure. Currently, evidence regarding pharmacological interventions for diabetes combined with AMI and I/R injury is lacking. Traditional hypoglycemic drugs have a limited role in the prevention and treatment of diabetes combined with I/R injury. Current evidence suggests that novel hypoglycemic drugs may exert a preventive effect on diabetes combined with myocardial I/R injury, especially glucagon-like peptide-1 receptor agonists (GLP-1 RA) and sodium-dependent glucose transporter protein 2 inhibitors (SGLT2i), which may increase coronary blood flow, reduce acute thrombosis, attenuate I/R injury, decrease myocardial infarction size, inhibit structural and functional remodeling of the ischemic heart, improve cardiac function, and reduce the occurrence of MACEs of diabetes patients combined with AMI via mechanisms such as reduction of inflammatory response, inhibition of oxidative stress, and improvement of vascular endothelial function. This paper will systematically elaborate the protective role and molecular mechanisms of GLP-1 RA and SGLT2i in diabetes combined with myocardial I/R injury, aiming to provide clinical assistance.
Collapse
Affiliation(s)
- Tiangui Yang
- Department of Cardiology, Shengjing Hospital of China Medical University, China.
| | - Daqing Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
18
|
Schindler TH, Bhandiwad AR, Jain S. Role of cardiac magnetic resonance imaging in the prediction of left ventricular remodeling after infarction: Emerging new concepts? Int J Cardiol 2023; 374:127-128. [PMID: 36640961 DOI: 10.1016/j.ijcard.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023]
Affiliation(s)
- Thomas H Schindler
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, MO, USA; Cardiovascular Division, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Anita R Bhandiwad
- Cardiovascular Division, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sudhir Jain
- Cardiovascular Division, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
Interaction between Acute Hepatic Injury and Early Coagulation Dysfunction on Mortality in Patients with Acute Myocardial Infarction. J Clin Med 2023; 12:jcm12041534. [PMID: 36836066 PMCID: PMC9966038 DOI: 10.3390/jcm12041534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/04/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND In acute myocardial infarction (AMI), acute hepatic injury is an independent risk factor for prognosis and is associated with complex coagulation dynamics. This study aims to determine the interaction between acute hepatic injury and coagulation dysfunction on outcomes in AMI patients. METHODS The Medical Information Mart for Intensive Care (MIMIC-III) database was used to identify AMI patients who underwent liver function testing within 24 h of admission. After ruling out previous hepatic injury, patients were divided into the hepatic injury group and the nonhepatic injury group based on whether the alanine transaminase (ALT) level at admission was >3 times the upper limit of normal (ULN). The primary outcome was intensive care unit (ICU) mortality. RESULTS Among 703 AMI patients (67.994% male, median age 65.139 years (55.757-76.859)), acute hepatic injury occurred in 15.220% (n = 107). Compared with the nonhepatic injury group, patients with hepatic injury had a higher Elixhauser comorbidity index (ECI) score (12 (6-18) vs. 7 (1-12), p < 0.001) and more severe coagulation dysfunction (85.047% vs. 68.960%, p < 0.001). In addition, acute hepatic injury was associated with increased in-hospital mortality (odds ratio (OR) = 3.906; 95% CI: 2.053-7.433; p < 0.001), ICU mortality (OR = 4.866; 95% CI: 2.489-9.514; p < 0.001), 28-day mortality (OR = 4.129; 95% CI: 2.215-7.695; p < 0.001) and 90-day mortality (OR = 3.407; 95% CI: 1.883-6.165; p < 0.001) only in patients with coagulation disorder but not with normal coagulation. Unlike patients with coagulation disorder and normal liver, patients with both coagulation disorder and acute hepatic injury had greater odds of ICU mortality (OR = 8.565; 95% CI: 3.467-21.160; p < 0.001) than those with normal coagulation. CONCLUSIONS The effects of acute hepatic injury on prognosis are likely to be modulated by early coagulation disorder in AMI patients.
Collapse
|
20
|
Deng T, Zhang T, Lu H, Chen J, Liu X, He W, Yao X. Evaluation and subgroup analysis of the efficacy and safety of intensive rosuvastatin therapy combined with dual antiplatelet therapy in patients with acute ischemic stroke. Eur J Clin Pharmacol 2023; 79:389-397. [PMID: 36580143 PMCID: PMC9941271 DOI: 10.1007/s00228-022-03442-8] [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: 11/01/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVES We investigated the efficacy of intensive rosuvastatin therapy plus 7-day dual antiplatelet therapy (DAPT) in reducing stroke recurrence for patients with acute ischemic stroke (AIS) and compared subgroups of patients. METHODS We enrolled patients with AIS whose time of onset to medication was ≤ 72 h, and the baseline scores of NIHSS (bNIHSS) were 0-10. The patients received intensive rosuvastatin therapy plus 7-day DAPT with aspirin and clopidogrel (study group) or rosuvastatin plus single antiplatelet therapy (SAPT, control group). The primary outcomes were recurrence of ischemic stroke, bleeding, statin-induced liver injury, and statin-associated myopathy (SAM) within 90 days. We also performed a subgroup analysis to assess the heterogeneity of the two therapy regimens in reducing recurrent stroke. RESULTS Recurrent stroke occurred in 10 patients in the study group and 42 patients in the control group (hazard ratio [HR], 0.373, 95% confidence interval [CI], 0.178-0.780; P = 0.009). Bleeding events occurred in 9 patients in the study group and 14 patients in the control group (HR, 1.019; 95%CI, 0.441-2.353; P = 0.966). Statin-induced liver injury and SAM were not recorded. Intensive rosuvastatin plus 7-day DAPT was generally effective in reducing the risk of recurrent stroke, except in the subgroup with bNIHSS ≤ 2. The therapy was particularly efficient in the elderly, male, high-bNIHSS, and hypertension, diabetes, and hyperlipidemia subgroups, with P < 0.02. CONCLUSIONS Without increasing bleeding and statin-associated adverse events, intensive rosuvastatin therapy plus 7-day DAPT significantly reduced the risk of recurrent stroke, especially for subgroups with high-risk factors. CLINICAL TRIAL REGISTRATION China Clinical Trial Registration Center (ChiCTR1800017809).
Collapse
Affiliation(s)
- Ting Deng
- Emergency Department, China Rehabilitation Research Center Beijing Bo’ai Hospital, Beijing, 100068 China
| | - Tong Zhang
- Neurology Department, China Rehabilitation Research Center Beijing Bo'ai Hospital, Beijing, 100068, China.
| | - Haitao Lu
- Neurology Department, China Rehabilitation Research Center Beijing Bo'ai Hospital, Beijing, 100068, China.
| | - Jingmian Chen
- Emergency Department, China Rehabilitation Research Center Beijing Bo’ai Hospital, Beijing, 100068 China
| | - Xiaomeng Liu
- Emergency Department, China Rehabilitation Research Center Beijing Bo’ai Hospital, Beijing, 100068 China
| | - Wei He
- Emergency Department, China Rehabilitation Research Center Beijing Bo’ai Hospital, Beijing, 100068 China
| | - Xiaohua Yao
- Emergency Department, China Rehabilitation Research Center Beijing Bo’ai Hospital, Beijing, 100068 China
| |
Collapse
|
21
|
Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD. Heart Disease and Ageing: The Roles of Senescence, Mitochondria, and Telomerase in Cardiovascular Disease. Subcell Biochem 2023; 103:45-78. [PMID: 37120464 DOI: 10.1007/978-3-031-26576-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Laura K Booth
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael E Redgrave
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Helen M Phillips
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
22
|
Bianco HT, Povoa R, Izar MC, Alves CMR, Barbosa AHP, Bombig MTN, Gonçalves I, Luna B, Aguirre AC, Moraes PIDM, Almeida D, Moreira FT, Povoa FF, Stefanini E, Caixeta AM, Bacchin AS, Moisés VA, Fonseca FA. Pharmaco-invasive Strategy in Myocardial Infarction: Descriptive Analysis, Presentation of Ischemic Symptoms and Mortality Predictors. Arq Bras Cardiol 2022; 119:691-702. [PMID: 36453760 PMCID: PMC9750212 DOI: 10.36660/abc.20211055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/15/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND ST-segment elevation myocardial infarction (STEMI) is defined by symptoms accompanied by typical electrocardiogram changes. However, the characterization of ischemic symptoms is unclear, especially in subgroups such as women and the elderly. OBJECTIVES To analyze the typification of ischemic symptoms, temporal metrics and observe the occurrence of in-hospital outcomes, in the analysis of predictive scores, in patients with STEMI, in a drug-invasive strategy. METHODS Study involving 2,290 patients. Types of predefined clinical presentations: typical pain, atypical pain, dyspnea, syncope. We measured the time between the onset of symptoms and demand for care and the interval between arrival at the medical unit and thrombolysis. Odds-ratios (OR; CI-95%) were estimated in a regression model. ROC curves were constructed for mortality predictors. The adopted significance level (alpha) was 5%. RESULTS Women had a high prevalence of atypical symptoms; longer time between the onset of symptoms and seeking care; delay between arrival at the emergency room and fibrinolysis. Hospital mortality was 5.6%. Risk prediction by Killip-Kimball classification: AUC: [0.77 (0.73-0.81)] in class ≥II. Subgroups studied [OR (CI-95%)]: women [2.06 (1.42-2.99); p=0.01]; chronic renal failure [3.39 (2.13-5.42); p<0.001]; elderly [2.09 (1.37-3.19) p<0.001]; diabetics [1.55 (1.04-2.29); p=0.02]; obese 1.56 [(1.01-2.40); p=0.04]: previous stroke [2.01 (1.02-3.96); p=0.04] correlated with higher mortality rates. CONCLUSION Despite higher mortality rates in some subgroups, significant disparity persists in women, with delays in symptom recognition and prompt thrombolysis. We highlight the applicability of the Killip-Kimball score in prediction, regardless of the clinical presentation.
Collapse
Affiliation(s)
- Henrique Tria Bianco
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Rui Povoa
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Maria Cristina Izar
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Claudia Maria Rodrigues Alves
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Adriano Henrique Pereira Barbosa
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Maria Teresa Nogueira Bombig
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Iran Gonçalves
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Bráulio Luna
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Ana Caroline Aguirre
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Pedro Ivo de Marqui Moraes
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Dirceu Almeida
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Flávio Tocci Moreira
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Fernando Focaccia Povoa
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Edson Stefanini
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Adriano Mendes Caixeta
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Amanda S. Bacchin
- Universidade Federal de São PauloCardiologiaSão PauloSPBrasilUniversidade Federal de São Paulo – Cardiologia, São Paulo, SP – Brasil
| | - Valdir Ambrósio Moisés
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| | - Francisco A.H. Fonseca
- Universidade Federal de São Paulo Escola Paulista de MedicinaMedicinaSão PauloSPBrasilUniversidade Federal de São Paulo Escola Paulista de Medicina – Medicina, São Paulo, SP – Brasil
| |
Collapse
|
23
|
Sacubitril/valsartan attenuates myocardial ischemia/reperfusion injury via inhibition of the GSK3β/NF-κB pathway in cardiomyocytes. Arch Biochem Biophys 2022; 730:109415. [PMID: 36179911 DOI: 10.1016/j.abb.2022.109415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022]
Abstract
In ischemia/reperfusion (I/R) injury, both inflammation and apoptosis play a vital role, and the inhibition of excessive inflammation and apoptosis show substantial clinical potential in the treatment of I/R disease. The role of sacubitril/valsartan (SAC/VAL)-a first-in-class angiotensin receptor-neprilysin inhibitor (ARNI)-in inflammation regulation and apoptosis in the context of I/R injury needs to be further explored. In this study, we investigate the short- and long-term effects of SAC/VAL administration in treating adult murine I/R injury both in vivo and in vitro. Our results verified that the application of SAC/VAL could reduce infarct size and suppress apoptosis and the inflammatory response in the acute phase post I/R. Long-term application of SAC/VAL for four weeks significantly improved ventricular function and reversed pathological ventricular remodeling. Mechanistically, SAC/VAL treatment induces the inhibition of the GSK3β-mediated NF-κB pathway through synergistically blocking angiotensin 1 receptor (AT1R) and activating natriuretic peptide receptor (NPR). In summary, we reported the therapeutic role of SAC/VAL in regulating the GSK3β/NF-κB signaling pathway to suppress the inflammatory response and apoptosis, thereby reducing cardiac dysfunction and remodeling post I/R.
Collapse
|
24
|
Kotronias RA, Fielding K, Greenhalgh C, Lee R, Alkhalil M, Marin F, Emfietzoglou M, Banning AP, Vallance C, Channon KM, De Maria GL. Machine learning assisted reflectance spectral characterisation of coronary thrombi correlates with microvascular injury in patients with ST-segment elevation acute coronary syndrome. Front Cardiovasc Med 2022; 9:930015. [PMID: 36204570 PMCID: PMC9530633 DOI: 10.3389/fcvm.2022.930015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/25/2022] [Indexed: 12/04/2022] Open
Abstract
Aims We set out to further develop reflectance spectroscopy for the characterisation and quantification of coronary thrombi. Additionally, we explore the potential of our approach for use as a risk stratification tool by exploring the relation of reflectance spectra to indices of coronary microvascular injury. Methods and results We performed hyperspectral imaging of coronary thrombi aspirated from 306 patients presenting with ST-segment elevation acute coronary syndrome (STEACS). Spatially resolved reflected light spectra were analysed using unsupervised machine learning approaches. Invasive [index of coronary microvascular resistance (IMR)] and non-invasive [microvascular obstruction (MVO) at cardiac magnetic resonance imaging] indices of coronary microvascular injury were measured in a sub-cohort of 36 patients. The derived spectral signatures of coronary thrombi were correlated with both invasive and non-invasive indices of coronary microvascular injury. Successful machine-learning-based classification of the various thrombus image components, including differentiation between blood and thrombus, was achieved when classifying the pixel spectra into 11 groups. Fitting of the spectra to basis spectra recorded for separated blood components confirmed excellent correlation with visually inspected thrombi. In the 36 patients who underwent successful thrombectomy, spectral signatures were found to correlate well with the index of microcirculatory resistance and microvascular obstruction; R 2: 0.80, p < 0.0001, n = 21 and R 2: 0.64, p = 0.02, n = 17, respectively. Conclusion Machine learning assisted reflectance spectral analysis can provide a measure of thrombus composition and evaluate coronary microvascular injury in patients with STEACS. Future work will further validate its deployment as a point-of-care diagnostic and risk stratification tool for STEACS care.
Collapse
Affiliation(s)
- Rafail A. Kotronias
- Oxford Heart Centre, National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Kirsty Fielding
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Mohammad Alkhalil
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- Cardiothoracic Centre, Freeman Hospital, Newcastle, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Federico Marin
- Oxford Heart Centre, National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Maria Emfietzoglou
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Adrian P. Banning
- Oxford Heart Centre, National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Claire Vallance
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Keith M. Channon
- Oxford Heart Centre, National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Giovanni Luigi De Maria
- Oxford Heart Centre, National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Short- and long-term survival after ST-elevation myocardial infarction treated with pharmacoinvasive versus primary percutaneous coronary intervention strategy: a prospective cohort study. BMJ Open 2022. [PMCID: PMC9301816 DOI: 10.1136/bmjopen-2022-061590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective Compare survival in patients with ST-elevation myocardial infarction (STEMI) treated with a pharmacoinvasive (PI) or primary percutaneous coronary intervention (pPCI) strategy based on estimated time to PCI. Design Prospective observational cohort study. Consecutive STEMI patients were registered on admission to our PCI centre and classified in a PI or pPCI group, based on the reperfusion strategy chosen in the prehospital or local hospital location. Time and cause of death was provided by the Norwegian Cause of Death registry. Mortality at 30 days, Kaplan-Meier survival and incidence of cardiovascular (CV) death was estimated. Adjusted effect of PI versus pPCI strategy on survival was estimated using logistic and Cox regression and propensity score weighting. Setting Single-centre registry in Norway during 2005–2011, within a regional STEMI network allocating patients to a PI strategy if estimated time to PCI >120 min. Primary outcomes 30-day mortality and survival during follow-up. Secondary outcome Incidence of CV death during follow-up. Results 4061 STEMI patients <80 years were included, 527 (13%) treated with a PI strategy and 3534 (87%) with a pPCI strategy. Median symptom-to-needle time was 110 min (25–75th percentile 75–163) in the PI group vs symptom-to-balloon 230 min (149–435) in the pPCI group. 30-day mortality was 3.2% and 5.0% in the PI and pPCI groups (ORadjusted0.58 (95% CI 0.30 to 1.13)) and 8-year survival was 85.9% (95% CI 80.9% to 89.6%) and 79.3% (95% CI 76.9% to 81.6%), respectively (HRadjusted 0.72 (95% CI 0.53 to 0.99)). Unadjusted incidence of 8-year CV death was 7.0% (95% CI 4.4% to 10.4%) in the PI group vs 12.4% (95% CI 9.9% to 15.2%) in the pPCI group. Adjusted long-term CV death was also lower in the PI group. Conclusion STEMI patients treated with a PI strategy experienced better survival compared with a pPCI strategy, also when adjusting for baseline characteristics. This supports using a PI strategy for eligible STEMI patients when pPCI cannot be performed within 120 min.
Collapse
|
26
|
Volpe M, Gallo G. Myocardial Infarction in Patients Without Cardiovascular Risk Factors: Advanced Prediction Models to Unlock the Clinical Dilemma? JACC Cardiovasc Interv 2022; 15:1176-1178. [PMID: 35680198 DOI: 10.1016/j.jcin.2022.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Massimo Volpe
- Cardiology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | - Giovanna Gallo
- Cardiology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
27
|
Pan Q, Liu Y, Ma W, Kan R, Zhu H, Li D. Cardioprotective Effects and Possible Mechanisms of Luteolin for Myocardial Ischemia-Reperfusion Injury: A Systematic Review and Meta-Analysis of Preclinical Evidence. Front Cardiovasc Med 2022; 9:685998. [PMID: 35548432 PMCID: PMC9081501 DOI: 10.3389/fcvm.2022.685998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAt present, effective clinical therapies for myocardial ischemia-reperfusion injury (MIRI) are lacking. We investigated if luteolin conferred cardioprotective effects against MIRI and elucidated the potential underlying mechanisms.MethodFour databases were searched for preclinical studies of luteolin for the treatment of MIRI. The primary outcomes were myocardial infarct size (IS) and intracardiac hemodynamics. The second outcomes were representative indicators of apoptosis, oxidative stress, and inflammatory. The Stata and RevMan software packages were utilized for data analysis.ResultsLuteolin administration was confirmed to reduce IS and ameliorate hemodynamics as compared to the control groups (p < 0.01). IS had decreased by 2.50%, 2.14%, 2.54% in three subgroups. Amelioration of hemodynamics was apparent in two different myocardial infarct models (model of left anterior descending branch ligation and model of global heart ischemia), as left ventricular systolic pressure improved by 21.62 and 35.40 mmHg respectively, left ventricular end-diastolic pressure decreased by 7.79 and 4.73 mmHg respectively, maximum rate of left ventricular pressure rise increased by 737.48 and 750.47 mmHg/s respectively, and maximum rate of left ventricular pressure decrease increased by 605.66 and 790.64 mmHg/s respectively. Apoptosis of cardiomyocytes also significantly decreased, as indicated by thelevels of MDA, an oxidative stress product, and expression of the inflammatory factor TNF-α (p < 0.001).ConclusionPooling of the data demonstrated that luteolin exerts cardioprotective effects against MIRI through different signaling pathways. As possible mechanisms, luteolin exerts anti-apoptosis, anti-oxidation, and anti-inflammation effects against MIRI.
Collapse
Affiliation(s)
- Qinyuan Pan
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Wenrui Ma
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Rongsheng Kan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Hong Zhu
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dongye Li
| |
Collapse
|
28
|
CPE Regulates Proliferation and Apoptosis of Primary Myocardial Cells Mediated by Ischemia and Hypoxia Injury. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3155171. [PMID: 35340224 PMCID: PMC8942647 DOI: 10.1155/2022/3155171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/23/2022] [Indexed: 11/28/2022]
Abstract
Objective To observe the effect of carboxypeptidase E (CPE) on the ischemia and hypoxia (I/H) injury of primary cardiomyocytes. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) technology was used to detect the expression of CPE in sham and myocardial infarction (MI) rat heart tissue, and the plasmid was transferred into primary cardiomyocytes by transfection technology. The apoptosis rate of cardiomyocytes was detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining, Annexin V-PI staining, and Cell Counting Kit-8 (CCK-8) assay. In addition, Caspase kit and qRT-PCR technology were used to detect the expression of apoptosis-related factors. The cell proliferation was detected by 5-ethynyl-2'-deoxyuridine (EdU) staining, flow cytometry, and qRT-PCR technology. In addition, Western blotting (WB) and qRT-PCR techniques were used to detect the Wnt/β-catenin pathway. Results First, we found that the expression of CPE in the marginal zone of MI was obviously reduced. Overexpression of CPE in primary cardiomyocytes can effectively inhibit ischemia/hypoxia (I/H)-induced apoptosis and decreased cell activity. In addition, CPE can promote cell proliferation and relieve the inhibitory effect of I/H on cardiomyocytes. At the same time, CPE can promote the expression of β-catenin and c-myc. Conclusion Overexpression of CPE in primary cardiomyocytes can effectively alleviate the decreased cell activity, increased apoptosis, and decreased proliferation caused by I/H and regulated by Wnt/β-catenin pathway.
Collapse
|
29
|
Yin X, Wang X, Wang S, Xia Y, Chen H, Yin L, Hu K. Screening for Regulatory Network of miRNA–Inflammation, Oxidative Stress and Prognosis-Related mRNA in Acute Myocardial Infarction: An in silico and Validation Study. Int J Gen Med 2022; 15:1715-1731. [PMID: 35210840 PMCID: PMC8863347 DOI: 10.2147/ijgm.s354359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Acute myocardial infarction (AMI), which commonly leads to heart failure, is among the leading causes of mortality worldwide. The aim of this study was to find potential regulatory network for miRNA-inflammation, oxidative stress and prognosis-related mRNA to uncover molecular mechanisms of AMI. Methods The expression profiles of miRNA and mRNA in the blood samples from AMI patients were downloaded from the Gene Expression Omnibus (GEO) dataset for differential expression analysis. Weighted gene co-expression network analysis (WGCNA) was used to further identify important mRNAs. The negatively regulatory network construction of miRNA–inflammation, oxidative stress and prognosis-related mRNAs was performed, followed by protein–protein interaction (PPI) and functional analysis of mRNAs. Results A total of three pairs of negatively regulatory network of miRNA–inflammation and prognosis-related mRNAs (hsa-miR-636/hsa-miR-491-3p/hsa-miR-188-5p/hsa-miR-188-3p-AQP9, hsa-miR-518a-3p-C5AR1 and hsa-miR-509-3-5p/hsa-miR-127-5p-PLAUR), two pairs of negatively regulatory network of miRNA–oxidative stress and prognosis-related mRNAs (hsa-miR-604-TLR4 and hsa-miR-139-5p-CXCL1) and three pairs of negatively regulatory network of miRNA-inflammation, oxidative stress and prognosis-related mRNA (hsa-miR-634/hsa-miR-591-TLR2, hsa-miR-938-NFKBIA and hsa-miR-520h/hsa-miR-450b-3p-ADM) were identified. In the KEGG analysis, some signaling pathways were identified, such as complement and coagulation cascades, pathogenic Escherichia coli infection, chemokine signaling pathway and cytokine–cytokine receptor interaction and Toll-like receptor signaling pathway. Conclusion Identified negatively regulatory network of miRNA-inflammation/oxidative stress and prognosis-related mRNA may be involved in the process of AMI. Those inflammation/oxidative stress and prognosis-related mRNAs may be diagnostic and prognostic biomarkers for AMI.
Collapse
Affiliation(s)
- Xunli Yin
- Department of Cardiovascular Medicine, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Xuebing Wang
- Department of Cardiovascular Medicine, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Shiai Wang
- Department of Cardiovascular Medicine, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Youwei Xia
- Department of Critical Care Medicine, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Huihui Chen
- Department of Cardiovascular Medicine, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Ling Yin
- Department of Conduit Room, The Seventh People’s Hospital of Jinan, Jinan, 250100, People’s Republic of China
| | - Keqing Hu
- Cardiovascular Department, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
- Correspondence: Keqing Hu, Central Hospital Affiliated to Shandong First Medical University, Cardiovascular Department,105#, Jiefang Road, Jinan 250013, Shandong, China, Tel +86 0531-85695114, Fax +86 0531-86942457 Email
| |
Collapse
|
30
|
Kucuk U, Volina E. The relationship between H2FPEF score and thrombus burden in patients with ST elevation myocardial infarction. INTERNATIONAL JOURNAL OF THE CARDIOVASCULAR ACADEMY 2022. [DOI: 10.4103/ijca.ijca_15_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
|
31
|
The Spiked Helmet Sign Predicting a Poor Outcome in a Patient with Non-Myocardial Infarction ST-Segment Elevation. Medicina (B Aires) 2021; 57:medicina57111184. [PMID: 34833402 PMCID: PMC8622399 DOI: 10.3390/medicina57111184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022] Open
Abstract
Spiked helmet sign is a novel electrocardiogram marker that reflects a poor prognosis, and may mimic myocardial infarction, especially in patients with an acute alteration of mental status or out-of-hospital cardiac arrest. In cases where a spiked helmet sign is missed, there may be a delay in surgical intervention for the underlying conditions because of unnecessary cardiac catheterization. In addition, antiplatelet agents for acute coronary syndrome in such cases can lead to catastrophic complications. Therefore, early recognition of spiked helmet sign is useful for timely correction of the underlying disease and prevention of poor outcomes. Herein, we describe a rare case of a patient with internal bleeding and subarachnoid hemorrhage presenting with spiked helmet sign on an electrocardiogram.
Collapse
|
32
|
Xu LN, Wang SH, Su XL, Komal S, Fan HK, Xia L, Zhang LR, Han SN. Targeting Glycogen Synthase Kinase 3 Beta Regulates CD47 Expression After Myocardial Infarction in Rats via the NF-κB Signaling Pathway. Front Pharmacol 2021; 12:662726. [PMID: 34349643 PMCID: PMC8327268 DOI: 10.3389/fphar.2021.662726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the effects of the GSK-3β/NF-κB pathway on integrin-associated protein (CD47) expression after myocardial infarction (MI) in rats. An MI Sprague Dawley rat model was established by ligating the left anterior descending coronary artery. The rats were divided into three groups: Sham, MI, and SB + MI (SB216763) groups. Immunohistochemistry was used to observe the changes in cardiac morphology. A significant reduction in the sizes of fibrotic scars was observed in the SB + MI group compared to that in the MI group. SB216763 decreased the mRNA and protein expression of CD47 and NF-κB during MI. Primary rat cardiomyocytes (RCMs) and the H9c2 cell line were used to establish in vitro hypoxia models. Quantitative real-time PCR and western blotting analyses were conducted to detect mRNA and protein expression levels of CD47 and NF-κB and apoptosis-related proteins, respectively. Apoptosis of hypoxic cells was assessed using flow cytometry. SB216763 reduced the protein expression of CD47 and NF-κB in RCMs and H9c2 cells under hypoxic conditions for 12 h, and alleviated hypoxia-induced apoptosis. SN50 (an NF-κB inhibitor) also decreased CD47 protein expression in RCMs and H9c2 cells under hypoxic conditions for 12 h and protected cells from apoptosis. GSK-3β upregulates CD47 expression in cardiac tissues after MI by activating NF-κB, which in turn leads to myocardial cell damage and apoptosis.
Collapse
Affiliation(s)
- Li-Na Xu
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shu-Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xue-Ling Su
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Sumra Komal
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hong-Kun Fan
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Li Xia
- Department of Anesthesiology in Surgery Branch, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Sheng-Na Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev 2021; 198:111540. [PMID: 34237321 PMCID: PMC8387860 DOI: 10.1016/j.mad.2021.111540] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.
Collapse
|
34
|
Scărlătescu AI, Micheu MM, Popa-Fotea NM, Dorobanțu M. MicroRNAs in Acute ST Elevation Myocardial Infarction-A New Tool for Diagnosis and Prognosis: Therapeutic Implications. Int J Mol Sci 2021; 22:4799. [PMID: 33946541 PMCID: PMC8124280 DOI: 10.3390/ijms22094799] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Despite diagnostic and therapeutic advances, coronary artery disease and especially its extreme manifestation, ST elevation myocardial infarction (STEMI), remain the leading causes of morbidity and mortality worldwide. Early and prompt diagnosis is of great importance regarding the prognosis of STEMI patients. In recent years, microRNAs (miRNAs) have emerged as promising tools involved in many pathophysiological processes in various fields, including cardiovascular diseases. In acute coronary syndromes (ACS), circulating levels of miRNAs are significantly elevated, as an indicator of cardiac damage, making them a promising marker for early diagnosis of myocardial infarction. They also have prognostic value and great potential as therapeutic targets considering their key function in gene regulation. This review aims to summarize current information about miRNAs and their role as diagnostic, prognostic and therapeutic targets in STEMI patients.
Collapse
Affiliation(s)
- Alina Ioana Scărlătescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.-M.P.-F.); (M.D.)
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| | - Nicoleta-Monica Popa-Fotea
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.-M.P.-F.); (M.D.)
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| | - Maria Dorobanțu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.-M.P.-F.); (M.D.)
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| |
Collapse
|
35
|
Mean platelet volume/platelet count ratio as a predictor of stent thrombosis in patients with ST-segment-elevation myocardial infarction. Ir J Med Sci 2021; 190:1095-1102. [PMID: 33893611 DOI: 10.1007/s11845-021-02626-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/08/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Despite the important role of some haematological parameters in tendency to thrombosis is known, their relationship with long-term stent thrombosis (ST) remains unclear. AIMS This study aimed to investigate the association between the mean platelet volume (MPV) to platelet count (PC) ratio and long-term ST and mortality in patients with ST-segment-elevation myocardial infarction (STEMI) treated successfully by primary percutaneous coronary intervention (pPCI). METHODS In a retrospective cohort study, according to their baseline MPV/PC ratios, 3667 consecutive STEMI patients undergoing pPCI were divided into three groups: tertile 1 (T1) (n = 1222, 0.357 ≥ MPV/PC ≥ 0.043), tertile 2 (T2) (n = 1222, 0.033 < MPV/PC < 0.043) and tertile 3 (T3) (n = 1223, 0.009 ≤ MPV/PC ≤ 0.032). Patients were followed up with for 5 years, focusing on ST and all-cause mortality outcomes. RESULTS Patients with T1 displayed a greater 5-year ST rate, including a 2.76-fold greater (95% confidence interval 1.68-10.33) rate than that of patients with T3, who had the lowest rates and were used as the reference group. Meanwhile, the 5-year mortality rate was similarly higher among patients with T1 by 1.72 times (95% confidence interval 1.33-2.22) relative to that among patients with T3. These significant relationships persisted even after adjustment for all confounders. CONCLUSION We found that higher MPV/PC ratios were associated with long-term ST and mortality. The MPV/PC ratio may constitute both a rapid and an easily obtainable parameter for identifying reliably high-risk patients who have undergone pPCI.
Collapse
|
36
|
Coronary Microvascular Dysfunction: PET, CMR and CT Assessment. J Clin Med 2021; 10:jcm10091848. [PMID: 33922841 PMCID: PMC8123021 DOI: 10.3390/jcm10091848] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 01/05/2023] Open
Abstract
Microvascular dysfunction is responsible for chest pain in various kinds of patients, including those with obstructive coronary artery disease and persistent symptoms despite revascularization, or those with myocardial disease without coronary stenosis. Its diagnosis can be performed with an advanced imaging technique such as positron emission tomography, which represents the gold standard for diagnosing microvascular abnormalities. In recent years, cardiovascular magnetic resonance and cardiac computed tomography have demonstrated to be emerging modalities for microcirculation assessment. The identification of microvascular disease represents a fundamental step in the characterization of patients with chest pain and no epicardial coronary disease: its identification is important to manage medical strategies and improve prognosis. The present overview summarizes the main techniques and current evidence of these advanced imaging strategies in assessing microvascular dysfunction and, if present, their relationship with invasive evaluation.
Collapse
|
37
|
Alsagaff MY, Kusumawardhani N. Wrapped left anterior descending artery STEMI: time to revisit. BMJ Case Rep 2021; 14:14/1/e237839. [PMID: 33431456 PMCID: PMC7802692 DOI: 10.1136/bcr-2020-237839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We report three cases of acute myocardial infarction caused by left anterior descending (LAD) artery occlusion presenting as ST elevation in the inferior. Therefore, coronary angiography showed an occlusion of the LAD coronary artery. Our cases show the rare occurrence of left coronary circulation dominance affecting inferior leads. These cases show an unusual and very rare form of left dominance coronary circulation where LAD is wrapped around the apex and continuing as a posterior descending artery. This would make inferior myocardial infarction because of occluded LAD or determine as wrapped LAD.
Collapse
Affiliation(s)
- Mochamad Yusuf Alsagaff
- Department of Cardiology and Vascular Medicine, Airlangga University, Surabaya, East Java, Indonesia,Department of Cardiology and Vascular Medicine, Universitas Airlangga Hospital, Surabaya, East Java, Indonesia
| | - Novia Kusumawardhani
- Department of Cardiology and Vascular Medicine, Airlangga University, Surabaya, East Java, Indonesia
| |
Collapse
|
38
|
He L, Wang Z, Zhou R, Xiong W, Yang Y, Song N, Qian J. Dexmedetomidine exerts cardioprotective effect through miR-146a-3p targeting IRAK1 and TRAF6 via inhibition of the NF-κB pathway. Biomed Pharmacother 2021; 133:110993. [PMID: 33220608 DOI: 10.1016/j.biopha.2020.110993] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion (I/R) injury is a common cause of mortality. Cardiac miR-146a is emerging as a potent regulator of myocardial function. Dexmedetomidine preconditioning provides cardioprotective effects, of which mechanisms related to miR-146a-3p are unclear. METHODS A myocardial I/R model in rats and a cellular anoxia/reoxygenation (A/R) model in H9C2 cells were established and preconditioned with dexmedetomidine or not. H9C2 cells were transfected with mimics, inhibitor, or negative controls of miR-146a-3p, and siRNAs of IRAK1 or TRAF6. Relative expressions of miR-146a-3p were determined by quantitative real-time polymerase chain reaction. The apoptosis rates and reactive oxygen species (ROS) levels in H9C2 cells were examined by flow cytometry. Protein expressions of IRAK1, TRAF6, cleaved Caspase-3, BAX, BCL-2, NF-κB p65, phosphorylated NF-κB p65 (p-NF-κB p65), IκBα, and phosphorylated IκBα (p-IκBα) in H9C2 cells were detected by Western blot. RESULTS Dexmedetomidine decreased myocardial infarction size and apoptosis rates of H9C2 cells. Dexmedetomidine upregulated expression of miR-146a-3p. Dexmedetomidine significantly decreased protein expressions of IRAK1, TRAF6, cleaved Caspase-3, BAX, and NF-κB p65, but increased expressions of BCL-2 in H9C2 cells. miR-146a-3p overexpression strengthened the anti-apoptotic effect induced by dexmedetomidine in H9C2 cells via decreasing protein levels of IRAK1, TRAF6, cleaved Caspase-3, BAX, NF-κB p65, p-NF-κB p65, and p-IκBα and increasing protein level of BCL-2. Downregulation of miR-146a-3p reversed the changes in these proteins in H9C2 cells. Expressions of NF-κB p65 and p-NF-κB p65 were further decreased following knockdown of IRAK1 or TRAF6. ROS emission was significantly increased after A/R, while significantly decreased following dexmedetomidine preconditioning in H9C2 cells transfected with siIRAK1 or siTRAF6. CONCLUSION miR-146a-3p targeting IRAK1 and TRAF6 through inhibition of NF-κB signaling pathway and ROS emission is involved in cardioprotection induced by dexmedetomidine pretreatment.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China; Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming Medical University, Kunming, Yunnan Province, 650051, China
| | - Zhuoran Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Rui Zhou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Ning Song
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
39
|
Argirò A, Olivotto I. The coronary microcirculation in sepsis: not of micro-importance. Glob Cardiol Sci Pract 2020; 2020:e202030. [PMID: 33598490 PMCID: PMC7868102 DOI: 10.21542/gcsp.2020.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Alessia Argirò
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| |
Collapse
|
40
|
Matin E, Ghaffari S, Garjani A, Roshanravan N, Matin S, Mesri Alamdari N, Safaie N. Oxidative stress and its association with ST resolution and clinical outcome measures in patients with ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention. BMC Res Notes 2020; 13:525. [PMID: 33176844 PMCID: PMC7656688 DOI: 10.1186/s13104-020-05350-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/21/2020] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Reperfusion of ischemic myocardium generates oxidative stress, which itself can mediate myocardial injury. So, in this study, we investigated the level of oxidative stress markers and its association with clinical outcomes in patients with ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention. RESULTS As indicated in the results, Post MI (Myocardial Infarction) heart failure was significantly higher in the group A (11% vs 4%, p = 0.047). Complete STR (ST-segment resolution) was observed to be significantly higher in the group B (36% vs 17%, p = 0.006). The SOD (Superoxide dismutase) and GPX (Glutathione peroxidase) levels were significantly higher in the group B compared to the other group (1547.51 ± 328.29 vs. 1449.97 ± 246.06, p = 0.019 and 60.62 ± 11.95 vs 57.41 ± 10.14, p = 0.042). The levels of GPX and SOD were shown to be directly related with complete STR and post PCI (Percutaneous coronary intervention)TIMI(Thrombolysis in Myocardial Infarction) flow 3 in the group A (p = 0.002 and p < 0.01, p = 0.005 and p < 0.02, respectively).
Collapse
Affiliation(s)
- Elmira Matin
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Department of Pharmacology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somaieh Matin
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Naimeh Mesri Alamdari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Naser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Li M, Tang X, Liu X, Cui X, Lian M, Zhao M, Peng H, Han X. Targeted miR-21 loaded liposomes for acute myocardial infarction. J Mater Chem B 2020; 8:10384-10391. [PMID: 33112352 DOI: 10.1039/d0tb01821j] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acute and persistent myocardial ischemia is the main cause of acute myocardial infarction (AMI) and heart failure. MicroRNA-21(miR-21) contributes to the pathophysiological consequences of acute myocardial infarction by targeting downstream crucial regulators. Thus, miR-21 mimics are a promising strategy for the treatment of AMI. However, their poor stability and insufficient cellular uptake are the major challenges. Herein, we encapsulated miR-21 mimics into liposomes modified with the cardiac troponin T (cTnT) antibody for targeted delivery of miR-21(cT-21-LIPs) to the ischemic myocardium. The cT-21-LIPs exhibited enhanced targeting efficiency to hypoxia primary cardiomyocytes in vitro and improved accumulation in the ischemic heart of AMI rats after injection via the tail vein due to the specifical target to overexpressed troponin. The cT-21-LIPs could significantly improve the cardiac function and decrease the infarct size after AMI, while maintaining the viability of cardiomyocytes. This design provides a novel strategy for delivering small nucleotide drugs specifically to the infarcted heart, which may find great potential in clinics.
Collapse
Affiliation(s)
- Minghui Li
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China and School of Chemistry and Chemical Engineering, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150001, China.
| | - Xuefeng Tang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
| | - Xiaoying Liu
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
| | - Xinyu Cui
- School of Chemistry and Chemical Engineering, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150001, China.
| | - Mingming Lian
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
| | - Man Zhao
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
| | - Xiaojun Han
- School of Chemistry and Chemical Engineering, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
42
|
Crea F. The conundrum of acute coronary syndromes: why does a stable plaque become unstable? Eur Heart J 2020; 41:3489-3493. [DOI: 10.1093/eurheartj/ehaa853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
43
|
Rusiecka OM, Montgomery J, Morel S, Batista-Almeida D, Van Campenhout R, Vinken M, Girao H, Kwak BR. Canonical and Non-Canonical Roles of Connexin43 in Cardioprotection. Biomolecules 2020; 10:biom10091225. [PMID: 32842488 PMCID: PMC7563275 DOI: 10.3390/biom10091225] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022] Open
Abstract
Since the mid-20th century, ischemic heart disease has been the world’s leading cause of death. Developing effective clinical cardioprotection strategies would make a significant impact in improving both quality of life and longevity in the worldwide population. Both ex vivo and in vivo animal models of cardiac ischemia/reperfusion (I/R) injury are robustly used in research. Connexin43 (Cx43), the predominant gap junction channel-forming protein in cardiomyocytes, has emerged as a cardioprotective target. Cx43 posttranslational modifications as well as cellular distribution are altered during cardiac reperfusion injury, inducing phosphorylation states and localization detrimental to maintaining intercellular communication and cardiac conduction. Pre- (before ischemia) and post- (after ischemia but before reperfusion) conditioning can abrogate this injury process, preserving Cx43 and reducing cell death. Pre-/post-conditioning has been shown to largely rely on the presence of Cx43, including mitochondrial Cx43, which is implicated to play a major role in pre-conditioning. Posttranslational modifications of Cx43 after injury alter the protein interactome, inducing negative protein cascades and altering protein trafficking, which then causes further damage post-I/R injury. Recently, several peptides based on the Cx43 sequence have been found to successfully diminish cardiac injury in pre-clinical studies.
Collapse
Affiliation(s)
- Olga M. Rusiecka
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; (O.M.R.); (J.M.); (S.M.)
| | - Jade Montgomery
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; (O.M.R.); (J.M.); (S.M.)
| | - Sandrine Morel
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; (O.M.R.); (J.M.); (S.M.)
| | - Daniela Batista-Almeida
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal; (D.B.-A.); (H.G.)
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (R.V.C.); (M.V.)
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (R.V.C.); (M.V.)
| | - Henrique Girao
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal; (D.B.-A.); (H.G.)
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Brenda R. Kwak
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; (O.M.R.); (J.M.); (S.M.)
- Correspondence:
| |
Collapse
|
44
|
Xiang H, Yang J, Li J, Yuan L, Lu F, Liu C, Tang Y. Citrate pretreatment attenuates hypoxia/reoxygenation-induced cardiomyocyte injury via regulating microRNA-142-3p/Rac1 aix. J Recept Signal Transduct Res 2020; 40:560-569. [PMID: 32456513 DOI: 10.1080/10799893.2020.1768548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose: Citrate has a positive effect on improving the pathophysiological changes of cardiomyocytes such as cardiac failure and auricular fibrillation. However, the underlying mechanism remains still unclear.Methods: Rat cardiomyocytes were used to establish hypoxia/reoxygenation (H/R) cell model. Citrate was conduct to pretreat with cardiomyocytes, and microRNA-142-3p (miR-142-3p) knockdown and overexpression were used to determine the underlying mechanism of their functions in cardiomyocytes. Cell viability and apoptosis were respectively detected by CCK-8 and flow cytometry. Protein and mRNA levels were determined by Western blot and qRT-PCR. Luciferase reporter assay and Targetscan were performed to study the regulation of miR-142-3p and Rac1.Results: The level of miR-142-3p was down-regulated in H/R model, but up-regulated in cardiomyocytes following citrate treatment. Citrates attenuated H/R injury induced miR-142-3p level and cell viability, and also inhibited H/R injury induced apoptosis, LDH, MDA and autophagy. Cell viability was improved, and autophagy was suppressed by miR-142-3p mimic, while inhibitor had opposite results. Compared with H/R + miR-142-3p inhibitor group, cell viability was higher, and apoptosis and autophagy were lower in Cit + H/R + miR-142-3p inhibitor group. Furthermore, Rac1 was target gene of miR-142-3p, and decreased by citrate, in comparison with H/R + miR-142-3p inhibitor group.Conclusion: Taken together, our findings indicated that citrate ameliorates H/R injury-induced cardiomyocytes autophagy by regulating miR-142-3p/Rac1 aix.
Collapse
Affiliation(s)
- Haiyan Xiang
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juesheng Yang
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Linhui Yuan
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Lu
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chen Liu
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanhua Tang
- Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
45
|
Duncan SE, Gao S, Sarhene M, Coffie JW, Linhua D, Bao X, Jing Z, Li S, Guo R, Su J, Fan G. Macrophage Activities in Myocardial Infarction and Heart Failure. Cardiol Res Pract 2020; 2020:4375127. [PMID: 32377427 PMCID: PMC7193281 DOI: 10.1155/2020/4375127] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Heart diseases remain the major cause of death worldwide. Advances in pharmacological and biomedical management have resulted in an increasing proportion of patients surviving acute heart failure (HF). However, many survivors of HF in the early stages end up increasing the disease to chronic HF (CHF). HF is an established frequent complication of myocardial infarction (MI), and numerous influences including persistent myocardial ischemia, shocked myocardium, ventricular remodeling, infarct size, and mechanical impairments, as well as hibernating myocardium trigger the development of left ventricular systolic dysfunction following MI. Macrophage population is active in inflammatory process, yet the clear understanding of the causative roles for these macrophage cells in HF development and progression is actually incomplete. Long ago, it was thought that macrophages are of importance in the heart after MI. Also, though inflammation is as a result of adverse HF in patients, but despite the fact that broad immunosuppression therapeutic target has been used in various clinical trials, no positive results have showed up, but rather, the focus on proinflammatory cytokines has proved more benefits in patients with HF. Therefore, in this review, we discuss the recent findings and new development about macrophage activations in HF, its role in the healthy heart, and some therapeutic targets for myocardial repair. We have a strong believe that there is a need to give maximum attention to cardiac resident macrophages due to the fact that they perform various tasks in wound healing, self-renewal of the heart, and tissue remodeling. Currently, it has been discovered that the study of macrophages goes far beyond its phagocytotic roles. If researchers in future confirm that macrophages play a vital role in the heart, they can be therapeutically targeted for cardiac healing.
Collapse
Affiliation(s)
- Sophia Esi Duncan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Michael Sarhene
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Joel Wake Coffie
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Deng Linhua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Xingru Bao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Zhang Jing
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Sheng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Rui Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Jing Su
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| |
Collapse
|
46
|
Liu T, Wang Q, Yao K. Huoxue Wentong Formula ameliorates myocardial infarction in rats through inhibiting CaMKII oxidation and phosphorylation. Chin Med 2020; 15:3. [PMID: 31938036 PMCID: PMC6954496 DOI: 10.1186/s13020-020-0285-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background The Chinese medicine Huoxue Wentong Formula (HXWTF) was used to treat thoracic obstruction and angina pectoris in clinic, which has not been investigated in myocardial ischemia-induced apoptosis and angiogenic function. Here we aimed to investigate the roles of HXWTF in rats with myocardial ischemia-induced apoptosis and angiogenesis disorders, as well as to reveal the potential mechanisms. Methods Male SD rats were subjected to coronary artery ligation followed by HXWTF (420, 840 and 1680 mg/kg/day, p.o.) or isosorbide mononitrate (6.3 mg/kg/day, p.o.) treatment for 4 weeks. Electrocardiogram (ECG) and Echocardiography (ECHO) were used to measure cardiac function. Hematoxylin and eosin (H&E) staining and CD34/α-SMA immunohistochemical staining were performed to observe the ischemic heart sections pathological changes and angiogenesis. Then, the effects on cardiomyocyte apoptosis of H9c2 and tube formation of HCMECs were observed, as well as the changes in the levels of total calmodulin dependent protein kinase II (t-CaMKII), phosphorylated CaMKII (p-CaMKII), oxidized CaMKII (ox-CaMKII), CD34, and Bcl-2/Bax ratio were detected. Results Rats with coronary artery ligation exhibited abnormal cardiac function, enlarged myocardial space, disorderly arranged myocardial fibers, inflammatory cells infiltrated, and aggravated myocardial cell apoptosis, along with angiogenesis dysfunction. The expressions of CD34, p-CaMKII, and ox-CaMKII were elevated and Bcl-2/Bax ratio was diminished in ischemic hearts and H/SD-treated H9c2 or HCMECs, while HXWTF treatment completely rescued angiogenic dysfunction, inhibited cardiomyocyte apoptosis, and down-regulated cardiac CaMKII oxidation and phosphorylation activities. Conclusion Our study demonstrates that HXWTF improves myocardial infarction possibly through inhibiting CaMKII oxidation and phosphorylation levels, facilitating angiogenic function and alleviating cardiomyocyte apoptosis. Thus, therapeutics targeting CaMKII activities may be a promising strategy for rescuing ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Tiantian Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixian'ge, District of Xi Cheng, Beijing, 100053 China
| | - Qingqing Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixian'ge, District of Xi Cheng, Beijing, 100053 China
| | - Kuiwu Yao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixian'ge, District of Xi Cheng, Beijing, 100053 China
| |
Collapse
|
47
|
Qiao B, Nie JJ, Shao Y, Li Y, Zhang C, Hao W, Li S, Chen D, Yu B, Li HH, Xu FJ, Du J. Functional Nanocomplexes with Vascular Endothelial Growth Factor A/C Isoforms Improve Collateral Circulation and Cardiac Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905925. [PMID: 31880079 DOI: 10.1002/smll.201905925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/02/2019] [Indexed: 06/10/2023]
Abstract
Protein-based therapies are potential treatments for cancer, immunological, and cardiovascular diseases. However, effective delivery systems are needed because of their instability, immunogenicity, and so on. Crosslinked negatively charged heparin polysaccharide nanoparticle (HepNP) is proposed for protein delivery. HepNP can efficiently condense vascular endothelial growth factor (VEGF) because of the unique electronegative sulfonic acid and carboxyl domain of heparin. HepNP is then assembled with VEGF-C (Hep@VEGF-C) or VEGF-A (Hep@VEGF-A) protein for the therapy of myocardial infarction (MI) via intravenous (iv) injection. Hep@VEGF-A-mediated improvement of cardiac function by promoting angiogenesis is limited because of elevated vascular permeability, while Hep@VEGF-C effectively promotes lymphangiogenesis and reduces edema. On this basis, a graded delivery of VEGF-C (0.5-1 h post-MI) and VEGF-A (5 d post-MI) using HepNP is developed. At the dose ratio of 3:1 (Hep@VEGF-C vs Hep@VEGF-A), Hep@VEGF functional complexes substantially reduce the scar formation (≈-39%; p < 0.05) and improve cardiac function (≈+74%; p < 0.05). Such a HepNP delivery system provides a simple and effective therapeutic strategy for cardiovascular diseases by delivering functional proteins. Because of the unique binding ability of heparin with cytokines and growth factors, HepNP also has considerable application prospects in protein therapy for other serious diseases.
Collapse
Affiliation(s)
- Bokang Qiao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Jing-Jun Nie
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yihui Shao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Yulin Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Congcong Zhang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Wenjing Hao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Bingran Yu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| |
Collapse
|
48
|
Liberale L, Bonaventura A, Montecucco F, Dallegri F, Carbone F. Impact of Red Wine Consumption on Cardiovascular Health. Curr Med Chem 2019; 26:3542-3566. [PMID: 28521683 DOI: 10.2174/0929867324666170518100606] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/05/2017] [Accepted: 03/05/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The devastating effects of heavy alcohol drinking have been long time recognized. In the last decades, potential benefits of modest red wine drinking were suggested. In European countries in which red wide intake is not negligible (such as France), the association between cholesterol and cardiovascular (CV) risk was less evident, suggesting the action of some protective molecules in red wine or other foods and drinks. METHODS This narrative review is based on the material searched for and obtained via PubMed up to May 2016. The search terms we used were: "red wine, cardiovascular, alcohol" in combination with "polyphenols, heart failure, infarction". RESULTS Epidemiological and mechanistic evidence of a J-shaped relationship between red wine intake and CV risk further supported the "French paradox". Specific components of red wine both in vitro and in animal models were discovered. Polyphenols and especially resveratrol largely contribute to CV prevention mainly through antioxidant properties. They exert beneficial effects on endothelial dysfunction and hypertension, dyslipidemia, metabolic diseases, thus reducing the risk of adverse CV events such as myocardial infarction ischemic stroke and heart failure. Of interest, recent studies pointed out the role of ethanol itself as a potential cardioprotective agent, but a clear epidemiological evidence is still missing. The aim of this narrative review is to update current knowledge on the intracellular mechanism underlying the cardioprotective effects of polyphenols and ethanol. Furthermore, we summarized the results of epidemiological studies, emphasizing their methodological criticisms and the need for randomized clinical trials able to clarify the potential role of red wine consumption in reducing CV risk. CONCLUSION Caution in avowing underestimation of the global burden of alcohol-related diseases was particularly used.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.,IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.,IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
49
|
Luo X, Song N, He S, Wei X, Feng Y, He Y, Chen X. Acute ST-segment elevation myocardial infarction due to extrinsic compression of left coronary artery from pulmonary epithelioid hemangioendothelioma: A case report. Medicine (Baltimore) 2019; 98:e18158. [PMID: 31770259 PMCID: PMC6890309 DOI: 10.1097/md.0000000000018158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/11/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
RATIONALE Acute myocardial infarction is usually caused by coronary atherosclerotic plaque disruption (rupture or erosion), also including other uncommon etiologies. Pulmonary epithelioid hemangioendothelioma (PEH) is a rare low to intermediate malignant vascular tumor originating from vascular endothelial cells. Here, we report a rare case of acute ST-segment elevation myocardial infarction (STEMI) due to extrinsic compression of left coronary artery from PEH. PATIENT CONCERNS A 63-year-old woman with pulmonary nodules received left pulmonary nodulectomy, and the pathological examination indicated PEH. Five months after the pulmonary nodulectomy, the patient was admitted due to progressive dyspnea. DIAGNOSIS Electrocardiography showed the obvious ST-segment elevation in the leads I, aVL, and V1-3, and laboratory tests revealed the elevated level of cardiac troponin T. Emergent coronary angiography and the contrast-enhanced computed tomography scan conformed STEMI due to extrinsic compression of left coronary artery from PEH. INTERVENTIONS The patient did not undergo further therapy after the pulmonary nodulectomy. During the present hospitalization, she received basic life support and nutritional support treatment. OUTCOMES The patient deteriorated rapidly into multi-organ failure and eventually died. LESSONS Acute STEMI could be caused by extrinsic compression of the coronary artery from the mass effects of PEH, and active therapy and close follow-up should be considered for patients with PEH.
Collapse
Affiliation(s)
- Xiaojia Luo
- The Second People's Hospital of Chengdu, Chengdu 610017
- Department of Cardiovascular Medicine
| | - Ningying Song
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sen He
- Department of Cardiovascular Medicine
| | - Xin Wei
- Department of Cardiovascular Medicine
| | - Yuan Feng
- Department of Cardiovascular Medicine
| | - Yong He
- Department of Cardiovascular Medicine
| | | |
Collapse
|
50
|
Du Y, Zhao Y, Zhu Y, Hu C, Zhang J, Ji Q, Liu W, Han H, Yang L, Zhang D, Tong S, Wang Z, Guo Y, Liu X, Zhou Y. High Serum Secreted Frizzled-Related Protein 5 Levels Associates with Early Improvement of Cardiac Function Following ST-Segment Elevation Myocardial Infarction Treated by Primary Percutaneous Coronary Intervention. J Atheroscler Thromb 2019; 26:868-878. [PMID: 30773518 PMCID: PMC6800391 DOI: 10.5551/jat.47019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
AIM Several members of secreted frizzled-related protein (SFRP) are involved in the process of myocardial ischemia-reperfusion injury. However, little is known about the role of SFRP5 in patients with acute ST-segment elevation myocardial infarction (STEMI). METHODS In this cross-sectional study, 85 patients with first-time anterior STEMI who underwent timely primary percutaneous coronary intervention (PCI) and 35 patients without coronary artery disease (CAD) were enrolled. Serum SFRP5 levels were measured using an enzyme-linked immunosorbent assay kit. Patients with STEMI were divided into low-SFRP5 and high-SFRP5 groups according to their median baseline serum SFRP5 levels. To evaluate cardiac function and structure after infarction, the left ventricular ejection fraction (LVEF) and left ventricular end-diastolic volume (LVEDV) were measured using echocardiography. The associations between changes in LVEF and reduced LVEF (≤ 50%) and clinical variables were determined by univariate and multivariate analyses. RESULTS Baseline serum SFRP5 levels were significantly higher in patients with STEMI than in those without CAD (23.3 ng/mL vs 19.8 ng/mL, P=0.008), although they decreased over time. Also, baseline serum SFRP5 levels were inversely correlated with peak hypersensitive cardiac troponin I (hs-cTnI) levels (r=-0.234, P=0.025) and peak hypersensitive C-reactive protein (hs-CRP) levels (r=-0.262, P=0.015). A multivariate linear regression model showed that changes in LVEF were positively correlated with serum SFRP5 levels at baseline (β= 0.249, 95% confidence interval (CI) 0.018-0.245, P=0.024) and 24 h after admission (β=0.220, 95% CI 0.003-0.264, P=0.045). At 3 months, LVEF in patients with high SFRP5 levels was significantly improved over baseline [(60.8±7.1) % vs (56.1±7.5) %, P=0.001]. LVEF was also significantly higher in patients with high SFRP5 levels than in those with low at the 3-month follow-up [(60.8±7.1) % vs (56.8±8.9) %, P=0.028]. Consequently, high serum SFRP5 levels at baseline were associated with a decreased risk of reduced LVEF at 3 months, independent of peak hs-cTnI and baseline cardiac function (hazard ratio 0.190, 95% CI 0.036-0.996; P=0.049). CONCLUSIONS High serum SFRP5 levels measured during the acute phase of STEMI were significantly associated with promoting myocardial recovery at an early phase following primary PCI, suggesting that SFRP5 is a potential therapeutic target in acute STEMI.
Collapse
Affiliation(s)
- Yu Du
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yingxin Zhao
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yong Zhu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Chenping Hu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Jianwei Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Qingwei Ji
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Wei Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Hongya Han
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Lixia Yang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Dai Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Shan Tong
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Zhijian Wang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yonghe Guo
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Xiaoli Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yujie Zhou
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| |
Collapse
|