1
|
Kuveljic J, Djordjevic A, Zivotic I, Dekleva M, Kolakovic A, Zivkovic M, Stankovic A, Djuric T. Expression of HMGB1, TGF-β1, BIRC3, ADAM17, CDKN1A, and FTO in Relation to Left Ventricular Remodeling in Patients Six Months after the First Myocardial Infarction: A Prospective Study. Genes (Basel) 2024; 15:1296. [PMID: 39457420 PMCID: PMC11507197 DOI: 10.3390/genes15101296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: After myocardial infarction (MI), adverse left ventricular (LV) remodeling may occur. This is followed by LV hypertrophy and eventually heart failure. The remodeling process is complex and goes through multiple phases. The aim of this study was to investigate the expression of HMGB1, TGF-β1, BIRC3, ADAM17, CDKN1A, and FTO, each involved in a specific step of LV remodeling, in association with the change in the echocardiographic parameters of LV structure and function used to assess the LV remodeling process in the peripheral blood mononuclear cells (PBMCs) of patients six months after the first MI. The expression of selected genes was also determined in PBMCs of controls. Methods: The study group consisted of 99 MI patients, who were prospectively followed-up for 6 months, and 25 controls. Cardiac parameters, measured via conventional 2D echocardiography, were evaluated at two time points: 3-5 days and 6 months after MI. The mRNA expression six-months-post-MI was detected using TaqMan® technology (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA). Results:HMGB1 mRNA was significantly higher in patients with adverse LV remodeling six-months-post-MI than in patients without adverse LV remodeling (p = 0.04). HMGB1 mRNA was significantly upregulated in patients with dilated LV end-diastolic diameter (LVEDD) (p = 0.03); dilated LV end-diastolic volume index (LVEDVi) (p = 0.03); severely dilated LV end-systolic volume index (LVESVi) (p = 0.006); impaired LV ejection fraction (LVEF) (p = 0.01); and LV enlargement (p = 0.03). It was also significantly upregulated in PBMCs from patients compared to controls (p = 0.005). TGF-β1 and BIRC3 mRNA were significantly lower in patients compared to controls (p = 0.02 and p = 0.05, respectively). Conclusions: Our results suggest that HMGB1 is involved in adverse LV remodeling six-months-post-MI, even on the mRNA level. Further research and validation are needed.
Collapse
Affiliation(s)
- Jovana Kuveljic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Ivan Zivotic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Milica Dekleva
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ana Kolakovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.K.); (I.Z.); (A.K.); (M.Z.); (A.S.); (T.D.)
| |
Collapse
|
2
|
Tona F, Civieri G, Vadori M, Masiero G, Iop L, Marra MP, Perin V, Cuciz E, Cecere A, Bernava G, Tansella D, Naumova N, Grewal S, Cozzi E, Iliceto S. Association of Angiotensin II Receptor Type 1 and Endothelin-1 Receptor Type A Agonistic Autoantibodies With Adverse Remodeling and Cardiovascular Events After Acute Myocardial Infarction. J Am Heart Assoc 2024; 13:e032672. [PMID: 38348777 PMCID: PMC11010093 DOI: 10.1161/jaha.123.032672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/27/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND The left ventricular remodeling (LVR) process has limited the effectiveness of therapies after myocardial infarction. The relationship between autoantibodies activating AT1R-AAs (angiotensin II receptor type 1-AAs) and ETAR-AAs (autoantibodies activating endothelin-1 receptor type A) with myocardial infarction has been described. Among patients with ST-segment-elevation myocardial infarction, we investigated the relationship between these autoantibodies with LVR and subsequent major adverse cardiac events. METHODS AND RESULTS In this prospective observational study, we included 131 patients with ST-segment-elevation myocardial infarction (61±11 years of age, 112 men) treated with primary percutaneous coronary intervention. Within 48 hours of admission, 2-dimensional transthoracic echocardiography was performed, and blood samples were obtained. The seropositive threshold for AT1R-AAs and ETAR-AAs was >10 U/mL. Patients were followed up at 6 months, when repeat transthoracic echocardiography was performed. The primary end points were LVR, defined as a 20% increase in left ventricular end-diastolic volume index, and major adverse cardiac event occurrence at follow-up, defined as cardiac death, nonfatal re-myocardial infarction, and hospitalization for heart failure. Forty-one (31%) patients experienced LVR. The prevalence of AT1R-AAs and ETAR-AAs seropositivity was higher in patients with versus without LVR (39% versus 11%, P<0.001 and 37% versus 12%, P=0.001, respectively). In multivariable analysis, AT1R-AAs seropositivity was significantly associated with LVR (odds ratio [OR], 4.66; P=0.002) and represented a risk factor for subsequent major adverse cardiac events (OR, 19.6; P=0.002). CONCLUSIONS AT1R-AAs and ETAR-AAs are associated with LVR in patients with ST-segment-elevation myocardial infarction. AT1R-AAs are also significantly associated with recurrent major adverse cardiac events. These initial observations may set the stage for a better pathophysiological understanding of the mechanisms contributing to LVR and ST-segment-elevation myocardial infarction prognosis.
Collapse
Affiliation(s)
- Francesco Tona
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Giovanni Civieri
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Marta Vadori
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Giulia Masiero
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Laura Iop
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Martina Perazzolo Marra
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Valentina Perin
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Elisa Cuciz
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Annagrazia Cecere
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Giacomo Bernava
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Donatella Tansella
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Nataliia Naumova
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | | | - Emanuele Cozzi
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| | - Sabino Iliceto
- Department of Cardiac, Thoracic, Vascular Sciences, and Public HealthUniversity of PaduaPaduaItaly
| |
Collapse
|
3
|
Turkieh A, Beseme O, Saura O, Charrier H, Michel JB, Amouyel P, Thum T, Bauters C, Pinet F. LIPCAR levels in plasma-derived extracellular vesicles is associated with left ventricle remodeling post-myocardial infarction. J Transl Med 2024; 22:31. [PMID: 38184604 PMCID: PMC10771704 DOI: 10.1186/s12967-023-04820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/20/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Long Intergenic noncoding RNA predicting CARdiac remodeling (LIPCAR) is a long noncoding RNA identified in plasma of patients after myocardial infarction (MI) to be associated with left ventricle remodeling (LVR). LIPCAR was also shown to be a predictor of early death in heart failure (HF) patients. However, no information regarding the expression of LIPCAR and its function in heart as well as the mechanisms involved in its transport to the circulation is known. The aims of this study are (1) to characterize the transporter of LIPCAR from heart to circulation; (2) to determine whether LIPCAR levels in plasma isolated-extracellular vesicles (EVs) reflect the alteration of its expression in total plasma and could be used as biomarkers of LVR post-MI. METHODS Since expression of LIPCAR is restricted to human species and the limitation of availability of cardiac biopsy samples, serum-free conditioned culture media from HeLa cells were first used to characterize the extracellular transporter of LIPCAR before validation in EVs isolated from human cardiac biopsies (non-failing and ischemic HF patients) and plasma samples (patients who develop or not LVR post-MI). Differential centrifugation at 20,000g and 100,000g were performed to isolate the large (lEVs) and small EVs (sEVs), respectively. Western blot and nanoparticle tracking (NTA) analysis were used to characterize the isolated EVs. qRT-PCR analysis was used to quantify LIPCAR in all samples. RESULTS We showed that LIPCAR is present in both lEVs and sEVs isolated from all samples. The levels of LIPCAR are higher in lEVs compared to sEVs isolated from HeLa conditioned culture media and cardiac biopsies. No difference of LIPCAR expression was observed in tissue or EVs isolated from cardiac biopsies obtained from ischemic HF patients compared to non-failing patients. Interestingly, LIPCAR levels were increased in lEVs and sEVs isolated from MI patients who develop LVR compared to patients who did not develop LVR. CONCLUSION Our data showed that large EVs are the main extracellular vesicle transporter of LIPCAR from heart into the circulation independently of the status, non-failing or HF, in patients. The levels of LIPCAR in EVs isolated from plasma could be used as biomarkers of LVR in post-MI patients.
Collapse
Affiliation(s)
- Annie Turkieh
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France.
| | - Olivia Beseme
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Ouriel Saura
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Henri Charrier
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | | | - Philippe Amouyel
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christophe Bauters
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Florence Pinet
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France.
| |
Collapse
|
4
|
Cao MJ, Shi FH, Yu BB, Ma XC, Zhang C, Xu L, Jiang YH, Ge H, Shen L, Pu J. Influence and mechanism of sodium-glucose cotransporter-2 inhibitors on the cardiac function: study protocol for a prospective cohort study. Front Endocrinol (Lausanne) 2023; 14:1199960. [PMID: 37538793 PMCID: PMC10395085 DOI: 10.3389/fendo.2023.1199960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023] Open
Abstract
Background Acute myocardial infarction (AMI) poses a significant threat to cardiovascular diseases (CVDs), leading to a high risk of heart failure (HF) and cardiovascular death. Growing evidence has unveiled the potential of sodium-glucose cotransporter-2 (SGLT2) inhibitors to improve cardiovascular outcomes in patients with CVD regardless of diabetes, but there is limited evidence in AMI patients. Furthermore, it is controversial whether the effects can be ascribed to the amelioration of left ventricular (LV) function, which further complicates the understanding of their underlying mechanism. Methods This study is a prospective, phase IV, open-label, parallel group, single-center trial conducted in a large tertiary teaching hospital in China. A total of 120 patients with AMI and type 2 diabetes mellitus (T2DM) will be included. Those who received SGLT2 inhibitors are considered as the experimental group, and those taking other antidiabetic agents are considered as the control group. The primary outcome is change in LV end-systolic volume index (LVESVi) measured by cardiac magnetic resonance (CMR) imaging from baseline during 1-year follow-up period. Secondary outcomes include other LV parameters such as LV mass, LV volume, and LV ejection fraction (EF); quality of life and functional capacity such as Kansas City Cardiomyopathy Questionnaire overall summary score (KCCQ-OS) and EuroQol-5 dimension (EQ-5D); biomarkers associated with diagnostic parameters of AMI and possible mechanisms on cardiovascular protection, such as creatine kinase, troponin T (TnT) level, troponin I (TnI) level, soluble suppression of tumorigenicity-2 (sST2), galectin-3 (Gal-3), fibroblast growth factor 21 (FGF21), and microRNA (miRNA) level. Discussion This study aims to investigate whether SGLT2 inhibitors could improve LV function by measuring CMR, quality of life, and functional capacity in patients with AMI in real-world settings, providing evidence on the underlying mechanism of SGLT2 inhibitors on cardioprotection. Clinical trial registration https://www.chictr.org.cn/showproj.html?proj=173672, identifier ChiCTR2200065792.
Collapse
Affiliation(s)
- Min-Jia Cao
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang-Hong Shi
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin-Bin Yu
- Department of Pharmacy, Huangyan Hospital of Wenzhou Medical University, Taizhou First People’s Hospital, Zhejiang, China
| | - Xue-Chen Ma
- Department of Pharmacy, The Third People’s Hospital of Xining, Xining, China
| | - Chen Zhang
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Hong Jiang
- Department of Endocrinology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Heng Ge
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Shen
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Identification of patient subtypes based on protein expression for prediction of heart failure after myocardial infarction. iScience 2023; 26:106171. [PMID: 36915695 PMCID: PMC10006628 DOI: 10.1016/j.isci.2023.106171] [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: 07/22/2022] [Revised: 12/19/2022] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
This study investigates the ability of high-throughput aptamer-based platform to identify circulating biomarkers able to predict occurrence of heart failure (HF), in blood samples collected during hospitalization of patients suffering from a first myocardial infarction (MI). REVE-1 (derivation) and REVE-2 (validation) cohorts included respectively 254 and 238 patients, followed up respectively 9 · 2 ± 4 · 8 and 7 · 6 ± 3 · 0 years. A blood sample collected during hospitalization was used for quantifying 4,668 proteins. Fifty proteins were significantly associated with long-term occurrence of HF with all-cause death as the competing event. k-means, an unsupervised clustering method, identified two groups of patients based on expression levels of the 50 proteins. Group 2 was significantly associated with a higher risk of HF in both cohorts. These results showed that a subset of 50 selected proteins quantified during hospitalization of MI patients is able to stratify and predict the long-term occurrence of HF.
Collapse
|
6
|
Legallois D, Hodzic A, Milliez P, Manrique A, Dolladille C, Saloux E, Beygui F. Left atrial strain quantified after myocardial infarction is associated with early left ventricular remodeling. Echocardiography 2022; 39:1581-1588. [PMID: 36376262 DOI: 10.1111/echo.15492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/23/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Left ventricular remodeling (LVR) is common and associated with adverse outcome after ST-elevation myocardial infarction (STEMI). We aimed to investigate the association between left atrial (LA) mechanical function using speckle tracking imaging and early LVR at follow-up in STEMI patients. METHODS Baseline 3D thoracic echocardiograms were performed within 48 h following admission and at a median follow-up of 7 months after STEMI. A > 20% increase in the left ventricular (LV) end-diastolic volume compared to baseline at follow-up was defined as LVR. LA global longitudinal strain was evaluated for the reservoir, conduit, and contraction (LASct) phases. RESULTS A total of 121 patients without clinical heart failure (HF) were prospectively included, between June 2015 and October 2018 (age 58.3 ± 12.5 years, male 98 (81%)). Baseline and follow-up LV ejection fraction (LVEF) were 46.8% [41.0, 52.9] and 52.1% [45.8, 57.0] respectively (p < .001). Compared to other patients, those with LVR had significantly lower values of LASct at baseline (-7.4% [-10.1, -6.5] vs. -9.9% [-12.8, -8.1], p < .01), both on univariate and baseline LV volumes-adjusted analyses. Baseline LA strain for reservoir and conduit phases were not associated with significant LVR at follow-up. Intra- and interobserver analysis showed good reproducibility of LA strain. CONCLUSIONS Baseline LASct may help identifying patients without HF after STEMI who are at higher risk of further early LVR and subsequent HF and who may benefit from more intensive management.
Collapse
Affiliation(s)
- Damien Legallois
- Department of Cardiology, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, France.,Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France
| | - Amir Hodzic
- Department of Clinical Physiology, Normandie Univ, UNICAEN, CHU de Caen Normandie, INSERM Comete, Caen, France
| | - Paul Milliez
- Department of Cardiology, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, France.,Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France
| | - Alain Manrique
- Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France.,Department of Nuclear Medicine, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, France
| | - Charles Dolladille
- Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France.,Department of Pharmacology, Normandie Univ, UNICAEN, CHU de Caen Normandie, PICARO Cardio-Oncology program, Caen, France
| | - Eric Saloux
- Department of Cardiology, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, France.,Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France
| | - Farzin Beygui
- Department of Cardiology, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, France.,Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, FHU REMOD-VHF, Caen, France.,ACTION academic research group, Pitié-Salpêtrière University Hospital, Paris, France
| |
Collapse
|
7
|
Calabretta R, Castello A, Giglioli C, Cecchi E, Cerisano G, Hacker M, Sciagrà R. Prognostic value of divergent pattern detection by 99mTc-sestamibi gated SPECT in patients with anterior acute myocardial infarction. J Nucl Cardiol 2022; 29:3115-3122. [PMID: 34914082 DOI: 10.1007/s12350-021-02874-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/27/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE In gated myocardial perfusion SPECT, apical remodeling may be identified by the presence of a divergent pattern (DP) of the left ventricle (LV). METHODS AND RESULTS We examined 150 anterior ST-elevation myocardial infarction (STEMI) patients, all successfully treated with primary percutaneous coronary interventions (PCI). Perfusion gated-SPECT to measure infarct size, LV end-diastolic (ED) and end-systolic (ES) volumes and ejection fraction (EF) was acquired before hospital discharge and repeated at 6-month follow-up. DP was observed in 26 patients, who had larger infarct size (28 ± 19% vs. 15.7 ± 17%, P < 0.02), and lower EF (33 ± 7% vs. 41 ± 10%, P < 0.001) than patients without DP. At follow-up, DP patients had significantly larger EDV (156 ± 54 vs. 107 ± 44 mL, P < 0.0001), ESV (104 ± 47 vs. 59 ± 36 mL, P < 0.0001) and lower EF (35 ± 12% vs. 48 ± 13%, P < 0.0001). 54% of DP patients developed remodeling at follow-up vs. 12% of those without DP (P < 0.001). During follow up, 7 events in the DP group (27%) and 11 events in patients without DP (9%; P < 0.02) occurred. Kaplan-Meier survival curves showed a worse prognosis for DP patients. CONCLUSION In patients with anterior AMI, early DP detection is related to subsequent LV dysfunction, larger infarct size, and worse severity. It is helpful for predicting LV remodeling at short-term follow-up and has prognostic implications.
Collapse
Affiliation(s)
- R Calabretta
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - A Castello
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Division of Nuclear Medicine, Fondazione IRCCS Ca´ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - C Giglioli
- Intensive Cardiac Coronary Unit, Heart and Vessel Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - E Cecchi
- Intensive Cardiac Coronary Unit, Heart and Vessel Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - G Cerisano
- Division of Cardiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - M Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - R Sciagrà
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
8
|
Luisi GA, Pestelli G, Lorenzoni G, Trevisan F, Smarrazzo V, Fiorencis A, Flamigni F, Ferrari R, Mele D. Severe Impairment of Left Ventricular Regional Strain in STEMI Patients Is Associated with Post-Infarct Remodeling. J Clin Med 2022; 11:jcm11185348. [PMID: 36142995 PMCID: PMC9505824 DOI: 10.3390/jcm11185348] [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: 07/22/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Measures of global left ventricular (LV) systolic function have limitations for the prediction of post-infarct LV remodeling (LVR). Therefore, we tested the association between a new measure of regional LV systolic function—the percentage of severely altered strain (%SAS)- and LVR after acute ST-elevation myocardial infarction (STEMI). As a secondary objective, we also evaluated the association between %SAS and clinical events during follow-up. Methods: Of 177 patients undergoing echocardiography within 24 h from primary percutaneous coronary angioplasty, 172 were studied for 3 months, 167 for 12 months, and 10 died. The %SAS was calculated by dividing the number of LV myocardial segments with ≥−5% peak systolic longitudinal strain by the total number of segments. LVR was defined as the increase in end-diastolic volume >20% at its first occurrence compared to baseline. Results: LVR percentage was 10.2% and 15.8% at 3 and 12 months, respectively. Based on univariable analysis, a number of clinical, laboratory, electrocardiographic and echocardiographic variables were associated with LVR. Based on multivariable analysis, %SAS and TnI peak remained associated with LVR (for %SAS 5% increase, OR 1.226, 95% CI 1.098−1.369, p < 0.0005; for TnI peak, OR 1.025, 95% CI 1.004−1.047, p = 0.022). %SAS and LVR were also associated with occurrence of clinical events at a median follow-up of 43 months (HR 1.02, 95% CI 1.0−1.04, p = 0.0165). Conclusions: In patients treated for acute STEMI, acute %SAS is associated with post-infarct LVR. Therefore, we suggest performing such evaluations on a routine basis to identify, as early as possible, STEMI patients at higher risk.
Collapse
Affiliation(s)
| | - Gabriele Pestelli
- Cardiology Unit, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Giulia Lorenzoni
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Filippo Trevisan
- Cardiology Unit, University Hospital of Ferrara, 44124 Ferrara, Italy
| | | | - Andrea Fiorencis
- Cardiology Unit, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Filippo Flamigni
- Cardiology Unit, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Roberto Ferrari
- Cardiology Unit, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Donato Mele
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-049-8218642
| |
Collapse
|
9
|
Walkowski B, Kleibert M, Majka M, Wojciechowska M. Insight into the Role of the PI3K/Akt Pathway in Ischemic Injury and Post-Infarct Left Ventricular Remodeling in Normal and Diabetic Heart. Cells 2022; 11:cells11091553. [PMID: 35563860 PMCID: PMC9105930 DOI: 10.3390/cells11091553] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
Collapse
Affiliation(s)
- Bartosz Walkowski
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
10
|
Barros-Gomes S, Roger VL, Pislaru SV, Kimura T, Pislaru C, Enriquez-Sarano M. Cardiac remodeling in acute myocardial infarction: Prospective insights from multimodality ultrasound imaging. Echocardiography 2021; 38:2032-2042. [PMID: 34845767 PMCID: PMC9059245 DOI: 10.1111/echo.15245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The severity of MI declined markedly in the last decade, and contemporary patterns of cardiac remodeling after MI are not defined. METHODS We prospectively enrolled community patients with first MI and performed comprehensive two- and three-dimensional echocardiography. Remodeling was defined as left ventricular (LV) end-systolic volume index (ESVI) above American Society of Echocardiography normal values. Remodeling patterns were characterized as an increase in LVESVI or LV end-diastolic volume index (LVEDVI), or both. RESULTS Between 2014 and 2016, 213 patients (63±13 years; 34% women) were enrolled within 3 days after MI (77% non-ST-elevation MI). Acute remodeling was present in 51% of patients. Higher troponin and wall motion score index were associated with greater remodeling (p < 0.001). Atrial annular area, leaflet tenting and papillary muscle areas increased with greater remodeling (p < 0.001). The greater the cardiac remodeling, the lower the LV ejection fraction and global longitudinal strain (p < 0.001). This decrease in LV function was accompanied by stroke volume augmentation and maintenance of cardiac index at the expense of increased LVEDVI. Different remodeling patterns were identified. Cases showing increased LVEDVI and normal LVESVI had smallest infarct size and better hemodynamics compared to cases with augmented LVESVI and normal LVEDVI. CONCLUSION Acute remodeling occurs in more than half of first MI cases and exhibits different patterns defined by cavity size and hemodynamic profile. Acute remodeling is a global phenomenon, which also involves the left atrium and the mitral apparatus.
Collapse
Affiliation(s)
| | - Véronique L. Roger
- Department of Cardiovascular Diseases Medicine, Mayo Clinic College of Medicine, Rochester, MN
- Epidemiology and Community Health Branch National Heart, Lung and Blood Institute, National Institutes of Health
| | - Sorin V. Pislaru
- Department of Cardiovascular Diseases Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | | | - Cristina Pislaru
- Department of Cardiovascular Diseases Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | - Maurice Enriquez-Sarano
- Department of Cardiovascular Diseases Medicine, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
11
|
Tanzilli G, Arrivi A, Placanica A, Viceconte N, Cammisotto V, Nocella C, Barillà F, Torromeo C, Pucci G, Acconcia MC, Granatelli A, Basili S, Dominici M, Gaudio C, Carnevale R, Mangieri E. Glutathione Infusion Before and 3 Days After Primary Angioplasty Blunts Ongoing NOX2-Mediated Inflammatory Response. J Am Heart Assoc 2021; 10:e020560. [PMID: 34533039 PMCID: PMC8649545 DOI: 10.1161/jaha.120.020560] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Glutathione is a water‐soluble tripeptide with a potent oxidant scavenging activity. We hypothesized that glutathione administration immediately before and after primary angioplasty (primary percutaneous coronary intervention) could be effective in modulating immune cell activation, thereby preventing infarct expansion. Methods and Results One hundred consecutive patients with ST‐segment–elevation myocardial infarction, scheduled to undergo primary percutaneous coronary intervention were randomly assigned before the intervention to receive an infusion of glutathione (2500 mg/25 mL over 10 minutes), followed by drug administration at the same doses at 24, 48, and 72 hours elapsing time or placebo. Total leukocytes, NOX2 (nicotinamide adenine dinucleotide phosphate oxidase 2) activation, NO bioavailability, cTpT (serum cardiac troponin T), hsCRP (high‐sensitivity C‐reactive protein), and TNF‐α (tumor necrosis factor α) levels were measured. Left ventricular size and function were assessed within 120 minutes, 5 days, and 6 months from percutaneous coronary intervention. Following reperfusion, a significant reduction of neutrophil to lymphocyte ratio (P<0.0001), hsCRP generation (P<0.0001), NOX2 activation (P<0.0001), TNF‐α levels (P<0.001), and cTpT release (P<0.0001) were found in the glutathione group compared with placebo. In treated patients, blunted inflammatory response was linked to better left ventricular size and function at follow‐up (r=0.78, P<0.005). Conclusions Early and prolonged glutathione infusion seems able to protect vital myocardial components and endothelial cell function against harmful pro‐oxidant and inflammatory environments, thus preventing maladaptive cardiac repair and left ventricular adverse remodeling. Registration URL: https://www.clinicaltrialsregister.eu; Unique identifier: 2014‐004486‐25.
Collapse
Affiliation(s)
- Gaetano Tanzilli
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Alessio Arrivi
- Department of Cardiology Interventional Cardiology Unit, "Santa Maria" Hospital Terni Italy
| | - Attilio Placanica
- Department of Cardiology "San Giovanni Evangelista" Hospital Tivoli Italy
| | - Nicola Viceconte
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Specialty Paride Stefanini Sapienza University of Rome Rome Italy
| | - Cristina Nocella
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Francesco Barillà
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Concetta Torromeo
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Giacomo Pucci
- Internal Medicine Unit, "Santa Maria" Hospital Terni Italy
| | - Maria Cristina Acconcia
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | | | - Stefania Basili
- Department of Translational and Precision Medicine SapienzaUniversity of Rome Rome Italy
| | - Marcello Dominici
- Department of Cardiology Interventional Cardiology Unit, "Santa Maria" Hospital Terni Italy
| | - Carlo Gaudio
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University Latina Italy.,Mediterranea Cardiocentro Napoli Italy
| | - Enrico Mangieri
- Department of Clinical, Internal Medicine, Anesthesiology, and Cardiovascular Sciences Sapienza University of Rome Rome Italy
| |
Collapse
|
12
|
Neves P, Pillay T, Annest L, van Bladel K, Kaiser E, Stahl F, Hanke T, Swaans M, Klein P, Ruf T, von Bardeleben RS. Patient selection for LIVE therapy: From clinical indications to multimodality imaging individual case planning. Echocardiography 2021; 38:1482-1488. [PMID: 34505315 DOI: 10.1111/echo.15182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/22/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Less Invasive Ventricular Enhancement (LIVE) with Revivent TC is an innovative therapy for symptomatic ischemic heart failure (HF). It is designed to reconstruct a negatively remodeled left ventricle (LV) after an anterior myocardial infarction (MI) by plication of the scar tissue. Its indications are specific, and as with any other structural heart intervention, the success of the procedure starts with appropriate patient selection. We aim to present the indications of the technique, crucial aspects in patient selection, and individual case planning approach. METHODS AND RESULTS After clinical evaluation, transthoracic echocardiography is the first imaging modality to be performed in a potential candidate for the therapy. However, definitive indication and detailed case planning rely on late gadolinium-enhanced cardiac magnetic resonance imaging or multiphasic contrast-enhanced cardiac computed tomography. These imaging modalities also assist with relative or absolute contra-indications for the procedure. Individual assessment is done to tailor the procedure to the specifics of the LV anatomy and location of the myocardial scar. CONCLUSION LIVE procedure is a unique intervention to treat symptomatic HF and ischemic cardiomyopathy after anterior MI. It is a highly customizable intervention that allows a patient-tailored approach, based on multimodality imaging assessment and planification.
Collapse
Affiliation(s)
- Paulo Neves
- Cardiothoracic Surgery Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | | | - Lon Annest
- BioVentrix Inc., San Ramon, California, USA
| | | | - Erhard Kaiser
- Private Practice for Internal Medicine and Cardiology, Frankfurt am Main, Germany
| | | | - Thorsten Hanke
- Department of Cardiac Surgery, Asklepios Hospital Harburg, Hamburg, Germany
| | - Martin Swaans
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Patrick Klein
- Department of Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, Netherlands
| | - Tobias Ruf
- Department of Cardiology, Heart Valve Center, University Medical Center Mainz, Mainz, Germany
| | | |
Collapse
|
13
|
Haley KE, Almas T, Shoar S, Shaikh S, Azhar M, Cheema FH, Hameed A. The role of anti-inflammatory drugs and nanoparticle-based drug delivery models in the management of ischemia-induced heart failure. Biomed Pharmacother 2021; 142:112014. [PMID: 34391184 DOI: 10.1016/j.biopha.2021.112014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022] Open
Abstract
Ongoing advancements in the treatment of acute myocardial infarction (MI) have significantly decreased MI related mortality. Consequently, the number of patients experiencing post-MI heart failure (HF) has continued to rise. Infarction size and the extent of left ventricular (LV) remodeling are largely determined by the extent of ischemia at the time of myocardial injury. In the setting of MI or acute phase of post-MI LV remodeling, anti-inflammatory drugs including intravenous immunoglobulin (IVIG) and Pentoxifylline have shown potential efficacy in preventing post-MI remodeling in-vitro and in some clinical trials. However, systemic administration of anti-inflammatory drugs are not without their off-target side effects. Herein, we explore the clinical feasibility of targeted myocardial delivery of anti-inflammatory drugs via biodegradable polymers, liposomes, hydrogels, and nano-particle based drug delivery models (NDDM) based on existing pre-clinical and clinical models. We summarize the barriers to clinical application of targeted anti-inflammatory delivery post-MI, including challenges in achieving sufficient retention and distribution, as well as the potential need for multiple dosing. Collectively, we suggest that localized delivery of anti-inflammatory agents to the myocardium using NDDM is a promising approach for successful treatment of ischemic HF. Future studies will be instrumental in determining the most effective target and delivery modalities for orchestrating NDDM-mediated treatment of HF.
Collapse
Affiliation(s)
- Kathryn E Haley
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Talal Almas
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Saeed Shoar
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Shan Shaikh
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Department of Surgery, St. Vincent's University Hospital, Dublin 4 Dublin, Ireland
| | - Faisal Habib Cheema
- HCA Healthcare Gulf Coast Division, Houston, TX, USA; University of Houston, College of Medicine, Houston, TX, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
14
|
Du H, Feng L, Xu Y, Zhan E, Xu W. Clinical Influencing Factors of Acute Myocardial Infarction Based on Improved Machine Learning. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:5569039. [PMID: 33854744 PMCID: PMC8019385 DOI: 10.1155/2021/5569039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/25/2021] [Accepted: 03/14/2021] [Indexed: 11/26/2022]
Abstract
At present, there is no method to predict or monitor patients with AMI, and there is no specific treatment method. In order to improve the analysis of clinical influencing factors of acute myocardial infarction, based on the machine learning algorithm, this paper uses the K-means algorithm to carry out multifactor analysis and constructs a hybrid model combined with the ART2 network. Moreover, this paper simulates and analyzes the model training process and builds a system structure model based on the KNN algorithm. After constructing the model system, this paper studies the clinical influencing factors of acute myocardial infarction and combines mathematical statistics and factor analysis to carry out statistical analysis of test results. The research results show that the system model constructed in this paper has a certain effect in the clinical analysis of acute myocardial infarction.
Collapse
Affiliation(s)
- Hongwei Du
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Linxing Feng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yan Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Enbo Zhan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Wei Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| |
Collapse
|
15
|
Bresson D, Roubille F, Prieur C, Biere L, Ivanes F, Bouleti C, Dubreuil O, Rioufol G, Boutitie F, Sideris G, Elbaz M, Bochaton T, De Bourguignon C, El Jonhy N, Dufay N, Dhelens C, Croisille P, Prunier F, Angoulvant D, Ovize M, Maucort-Boulch D, Mewton N. Colchicine for Left Ventricular Infarct Size Reduction in Acute Myocardial Infarction: A Phase II, Multicenter, Randomized, Double-Blinded, Placebo-Controlled Study Protocol - The COVERT-MI Study. Cardiology 2021; 146:151-160. [PMID: 33582664 DOI: 10.1159/000512772] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/30/2020] [Indexed: 11/19/2022]
Abstract
Inflammatory processes have been identified as key mediators of ischemia-reperfusion injury in ST-segment elevation myocardial infarction (STEMI). They add damage to the myocardium and are associated with clinical adverse events (heart failure and cardiovascular death) and poor myocardial recovery. Colchicine is a well-known alkaloid with potent anti-inflammatory properties. In a proof-of-concept phase II trial, colchicine has been associated with a significant 50% reduction of infarct size (assessed by creatine kinase levels) in comparison to placebo in acute STEMI patients referred for primary percutaneous coronary intervention (PPCI). The Colchicine in STEMI Patients Study (COVERT-MI) is an ongoing confirmative prospective, multicenter, randomized, double-blind trial testing whether a short course oral treatment with colchicine versus placebo decreases myocardial injury in patients presenting with STEMI referred for PPCI. Adult patients, with a first STEMI episode and an initial TIMI flow ≤1, referred for PPCI, will be randomized (n = 194) in a 1:1 ratio to receive an oral bolus of colchicine of 2 mg followed by 0.5 mg b.i.d. treatment during 5 days or matching placebo. The primary endpoint will be the reduction in infarct size as assessed by cardiac magnetic resonance at 5 ± 2 days between both groups. The main secondary endpoints will be tested between groups in hierarchical order with left ventricular ejection fraction at 5 days, microvascular obstruction presence at 5 days, and absolute adverse left ventricular remodeling between 5 days and 3 months. This academic study is being financed by a grant from the French Ministry of Health (PHRCN-16-0357). Results from this study will contribute to a better understanding of the complex pathophysiology underlying myocardial injury after STEMI. The present study describes the rationale, design, and methods of the trial.
Collapse
Affiliation(s)
- Didier Bresson
- University Hospital of Mulhouse, Hôpital Emile Muller, Mulhouse, France
| | - François Roubille
- Cardiology Department, PhyMedExp, INSERM, CNRS, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Cyril Prieur
- Coronary Care Unit, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Loic Biere
- Institut MITOVASC, CNRS 6015 INSERM U1083, CHU Angers, Service de Cardiologie, Angers Cedex, Université d'Angers, Angers, France
| | - Fabrice Ivanes
- Cardiology Department CHRU de Tours & EA4245 T2i Tours University, Tours, France
| | - Claire Bouleti
- CIC Inserm 1402n CHU de Poitiers, Université de Poitiers, Poitiers, France
| | - Olivier Dubreuil
- Invasive Cardiology Department, Centre Hospitalier Saint-Joseph Saint-Luc, Lyon, France
| | - Gilles Rioufol
- Coronary Care Unit, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France.,Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Florent Boutitie
- Biostatistical Department - Bioinformatique, Pôle de Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Georges Sideris
- Cardiology Department, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Meyer Elbaz
- Interventional Cardiology Department, CHU de Rangueil, Toulouse, France
| | - Thomas Bochaton
- Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Charles De Bourguignon
- Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Naoual El Jonhy
- Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Nathalie Dufay
- NeuroBioTec, Centre de Ressources Biologiques des HCL, Hôpital Neurologique, Bron, France
| | - Carole Dhelens
- Pharmacy Department, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Pierre Croisille
- Department Radiology, CREATIS CNRS 5220 INSERM U1206 Research Lab, Hôpital Nord University Hospital/CHU Saint Etienne, Avenue Albert Raimond, Saint-Priest en Jarez, France
| | - Fabrice Prunier
- Institut MITOVASC, CNRS 6015 INSERM U1083, CHU Angers, Service de Cardiologie, Angers Cedex, Université d'Angers, Angers, France
| | - Denis Angoulvant
- Cardiology Department CHRU de Tours & EA4245 T2i Tours University, Tours, France
| | - Michel Ovize
- Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Delphine Maucort-Boulch
- Biostatistical Department - Bioinformatique, Pôle de Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Nathan Mewton
- Centre d'Investigation Clinique, Inserm 1407, CarMeN Unit Inserm 1060, Hôpital Cardiologique Louis Pradel, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France,
| |
Collapse
|
16
|
Videira RF, da Costa Martins PA, Falcão-Pires I. Non-Coding RNAs as Blood-Based Biomarkers in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21239285. [PMID: 33291434 PMCID: PMC7730567 DOI: 10.3390/ijms21239285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
In 2020, cardiovascular diseases (CVDs) remain a leading cause of mortality and morbidity, contributing to the burden of the already overloaded health system. Late or incorrect diagnosis of patients with CVDs compromises treatment efficiency and patient's outcome. Diagnosis of CVDs could be facilitated by detection of blood-based biomarkers that reliably reflect the current condition of the heart. In the last decade, non-coding RNAs (ncRNAs) present on human biofluids including serum, plasma, and blood have been reported as potential biomarkers for CVDs. This paper reviews recent studies that focus on the use of ncRNAs as biomarkers of CVDs.
Collapse
Affiliation(s)
- Raquel Figuinha Videira
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Paula A. da Costa Martins
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Inês Falcão-Pires
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
17
|
Pezel T, Besseyre des Horts T, Schaaf M, Croisille P, Bière L, Garcia-Dorado D, Jossan C, Roubille F, Cung TT, Prunier F, Meyer E, Amaz C, Derumeaux G, de Poli F, Hovasse T, Gilard M, Bergerot C, Thibault H, Ovize M, Mewton N. Predictive value of early cardiac magnetic resonance imaging functional and geometric indexes for adverse left ventricular remodelling in patients with anterior ST-segment elevation myocardial infarction: A report from the CIRCUS study. Arch Cardiovasc Dis 2020; 113:710-720. [PMID: 33160891 DOI: 10.1016/j.acvd.2020.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/17/2020] [Accepted: 05/13/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Postinfarction adverse left ventricular (LV) remodelling is strongly associated with heart failure events. Conicity index, sphericity index and LV global functional index (LVGFI) are new LV remodelling indexes assessed by cardiac magnetic resonance (CMR). AIM To assess the predictive value of the new indexes for 1-year adverse LV remodelling in patients with anterior ST-segment elevated myocardial infarction (STEMI). METHODS CMR studies were performed in 129 patients with anterior STEMI (58±12 years; 78% men) from the randomized CIRCUS trial (CMR substudy) treated with primary percutaneous coronary intervention and followed for the occurrence of major adverse cardiovascular events (MACE) (death or hospitalization for heart failure). Conicity index, sphericity index, LVGFI, infarct size and microvascular obstruction (MVO) were assessed by CMR performed 5±4 days after coronary reperfusion. Adverse LV remodelling was defined as an increase in LV end-diastolic volume of ≥15% by transthoracic echocardiography at 1 year. RESULTS Adverse LV remodelling occurred in 27% of patients at 1 year. Infarct size and MVO were significantly predictive of adverse LV remodelling: odds ratio [OR] 1.03, 95% confidence interval [CI] 1.01-1.05 (P<0.001) and OR 1.12, 95% CI 1.05-1.22 (P<0.001), respectively. Among the newly tested indexes, only LVGFI was significantly predictive of adverse LV remodelling (OR 1.10, 95% CI 1.03-1.16; P=0.001). In multivariable analysis, infarct size remained an independent predictor of adverse LV remodelling at 1 year (OR 1.05, 95% CI 1.02-1.08; P<0.001). LVGFI and infarct size were associated with occurrence of MACE: OR 1.21, 95% CI 1.08-1.37 (P<0.001) and OR 1.02, 95% CI 1.00-1.04 (P=0.018), respectively. Conicity and sphericity indexes were not associated with MACE. CONCLUSIONS LVGFI was associated with adverse LV remodelling and MACE 1 year after anterior STEMI.
Collapse
Affiliation(s)
- Théo Pezel
- Inserm, UMRS 942, Department of Cardiology, Lariboisière Hospital, Paris University, AP-HP, 75010 Paris, France; Division of Cardiology, Johns-Hopkins University, 21287-0409 Baltimore, MD, USA
| | - Timothée Besseyre des Horts
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Mathieu Schaaf
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Pierre Croisille
- Radiology Department, University Hospital of Saint-Étienne, 42270 Saint-Priest-en-Jarez, France
| | - Loïc Bière
- Cardiology Division, University Hospital of Angers, 49100 Angers, France
| | - David Garcia-Dorado
- CIBERCV, Hospital Universitari Vall d'Hebron & Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Claire Jossan
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - François Roubille
- UMR5203, UMR661, Cardiology Division, University Hospital of Montpellier, University of Montpellier 1 and 2, 34295 Montpellier, France
| | - Thien-Tri Cung
- UMR5203, UMR661, Cardiology Division, University Hospital of Montpellier, University of Montpellier 1 and 2, 34295 Montpellier, France
| | - Fabrice Prunier
- Cardiology Division, University Hospital of Angers, 49100 Angers, France
| | - Elbaz Meyer
- Rangeuil Hospital, University Hospital of Toulouse, Paul-Sabatier University, 31400 Toulouse, France
| | - Camille Amaz
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Geneviève Derumeaux
- DHU-ATVB, Henri-Mondor Hospital, Paris-Est Créteil University, AP-HP, 94010 Créteil, France
| | - Fabien de Poli
- Cardiology Division, Haguenau Hospital, 67500 Haguenau, France
| | - Thomas Hovasse
- Cardiology Division, Jacques-Cartier Institute, 91300 Massy, France
| | - Martine Gilard
- Department of Cardiology, Brest University Hospital, 29200 Brest, France
| | - Cyrille Bergerot
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Hélène Thibault
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Michel Ovize
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France
| | - Nathan Mewton
- Inserm 1407, Clinical Investigation Centre and Heart Failure Department, Cardiovascular Hospital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard Lyon 1, 69677 Bron, France.
| |
Collapse
|
18
|
Bellis A, Mauro C, Barbato E, Di Gioia G, Sorriento D, Trimarco B, Morisco C. The Rationale of Neprilysin Inhibition in Prevention of Myocardial Ischemia-Reperfusion Injury during ST-Elevation Myocardial Infarction. Cells 2020; 9:cells9092134. [PMID: 32967374 PMCID: PMC7565478 DOI: 10.3390/cells9092134] [Citation(s) in RCA: 8] [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: 08/25/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
During the last three decades, timely myocardial reperfusion using either thrombolytic therapy or primary percutaneous intervention (pPCI) has allowed amazing improvements in outcomes with a more than halving in 1-year ST-elevation myocardial infarction (STEMI) mortality. However, mortality and left ventricle (LV) remodeling remain substantial in these patients. As such, novel therapeutic interventions are required to reduce myocardial infarction size, preserve LV systolic function, and improve survival in reperfused-STEMI patients. Myocardial ischemia-reperfusion injury (MIRI) prevention represents the main goal to reach in order to reduce STEMI mortality. There is currently no effective therapy for MIRI prevention in STEMI patients. A significant reason for the weak and inconsistent results obtained in this field may be the presence of multiple, partially redundant, mechanisms of cell death during ischemia-reperfusion, whose relative importance may depend on the conditions. Therefore, it is always more recognized that it is important to consider a "multi-targeted cardioprotective therapy", defined as an additive or synergistic cardioprotective agents or interventions directed to distinct targets with different timing of application (before, during, or after pPCI). Given that some neprilysin (NEP) substrates (natriuretic peptides, angiotensin II, bradykinin, apelins, substance P, and adrenomedullin) exert a cardioprotective effect against ischemia-reperfusion injury, it is conceivable that antagonism of proteolytic activity by this enzyme may be considered in a multi-targeted strategy for MIRI prevention. In this review, by starting from main pathophysiological mechanisms promoting MIRI, we discuss cardioprotective effects of NEP substrates and the potential benefit of NEP pharmacological inhibition in MIRI prevention.
Collapse
Affiliation(s)
- Alessandro Bellis
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Giuseppe Di Gioia
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Cardiac Catheterization Laboratory, Montevergine Clinic, 83013 Mercogliano (AV), Italy
| | - Daniela Sorriento
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Bruno Trimarco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Correspondence: ; Tel.: +39-081-746-2253; Fax: +39-081-746-2256
| |
Collapse
|
19
|
Effects of sacubitril/valsartan on ventricular remodeling in patents with left ventricular systolic dysfunction following acute anterior wall myocardial infarction. Coron Artery Dis 2020; 32:418-426. [PMID: 32732515 DOI: 10.1097/mca.0000000000000932] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the effect of sacubitril/valsartan (Sal/Val) on left ventricular (LV) remodeling in patients with LV systolic dysfunction following acute anterior wall myocardial infarction (AAMI). METHODS AAMI patients with LV systolic dysfunction were enrolled in this study. All patients underwent percutaneous coronary intervention. After hemodynamic stabilization, patients were randomly assigned either to group T (Sal/Val treatment) or group C (enalapril treatment). Changes in echocardiographic parameters and plasma biochemical markers were used to evaluate the effects of Sal/Val on LV remodeling and cardiac function. The incidence of major cardiac adverse events (MACEs) and adverse reactions during follow-ups was also recorded. RESULTS In total, 137 eligible patients were prospectively included. Compared to group C, LV ejection fraction significantly improved (P < 0.05), while the LV end-systolic volume index and wall motion score index showed a tendency to decrease in group T. There was no difference in the LV end-diastolic volume index between groups. During follow-ups, the plasma N-terminal pro-B-type natriuretic peptide and soluble suppression of tumorigenesis-2 levels in both groups decreased (all P < 0.05), and the change was more prominent in group T. Additionally, drug-related adverse effects were similar between the two groups (P > 0.05); however, the incidence of MACEs was lower in group T than in group C (39.71% vs. 53.62%, P = 0.103), although the difference was insignificant. CONCLUSION Sac/Val attenuated LV remodeling and dysfunction and was safe and effective in LV systolic dysfunction patients post AAMI.
Collapse
|
20
|
Cirillo P, Izzo R, Mancusi C, Buono F, Ziviello F, Spinelli L, Esposito G, DI Gioia G, Barbato E, Strisciuglio T, Trimarco B, Morisco C. Impact of drug-eluting stents on left ventricular wall motion after successful reperfusion of first anterior ST elevation myocardial infarction. Minerva Cardiol Angiol 2020; 69:144-153. [PMID: 32515176 DOI: 10.23736/s2724-5683.20.05176-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Timely myocardial reperfusion by primary percutaneous coronary intervention (pPCI) prevents the development of left ventricular (LV) dysfunction after myocardial infarction (MI). We aimed to investigate whether bare-metal stents (BMS) and drug eluting stents (DES) differently affect the recovery of LV function in patients with ST-elevation MI (STEMI). METHODS Overall 103anterior STEMI patients were retrospectively analyzed. All patients had single vessel disease with culprit lesion at the left anterior descending coronary artery. Patients were categorized in DES group (N.=67) and BMS group (N.=36). Changes in LV contractility were assessed by trans-thoracic echocardiogram as Left Ventricular Wall Motion Score Index (LVWMSI). Follow-up visits were performed between 6 and 12 months after hospital discharge. RESULTS Compared to baseline, LV ejection fraction (EF) remained unchanged between the two groups at the follow-up; LVWMSI significantly improved in patients treated with DES (1.95±0.25 vs. 1.78±0.38, P<0.05), whereas did not change in those treated with BMS (2.09±0.21 vs. 1.98±0.33, P: not significant). At follow-up the LVWMSI was significantly higher in patients with DES than with BMS (P=0.048). LV end-systolic and end-diastolic volumes (LVESV, LVEDV) significantly increased in patients receiving a BMS, whereas it did not change in those receiving a DES (P<0.05). Multivariate analysis adjusted for age, gender, type of stent (DES or BMS), and type of revascularization (primary PCI or rescue PCI or thrombolysis + PCI) showed that DES implantation was an independent predictor of LVWMSI improvement (OR: 3.8 [1.143-12.969] P=0.03). CONCLUSIONS DES implantation is associated with a favorable impact on LV remodeling and regional contractility.
Collapse
Affiliation(s)
- Plinio Cirillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Costantino Mancusi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Francesco Buono
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Francesca Ziviello
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Letizia Spinelli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giuseppe DI Gioia
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Emanuele Barbato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Teresa Strisciuglio
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy -
| |
Collapse
|
21
|
Duengen HD, Kim RJ, Zahger D, Orvin K, Kornowski R, Admon D, Kettner J, Shimony A, Otto C, Becka M, Kanefendt F, Romo AI, Hasin T, Ostadal P, Rojas GC, Senni M. Effects of the chymase inhibitor fulacimstat on adverse cardiac remodeling after acute myocardial infarction-Results of the Chymase Inhibitor in Adverse Remodeling after Myocardial Infarction (CHIARA MIA) 2 trial. Am Heart J 2020; 224:129-137. [PMID: 32375104 DOI: 10.1016/j.ahj.2020.01.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Adverse cardiac remodeling is a major risk factor for the development of post myocardial infarction (MI) heart failure (HF). This study investigates the effects of the chymase inhibitor fulacimstat on adverse cardiac remodeling after acute ST-segment-elevation myocardial infarction (STEMI). METHODS In this double-blind, randomized, placebo-controlled trial patients with first STEMI were eligible. To preferentially enrich patients at high risk of adverse remodeling, main inclusion criteria were a left-ventricular ejection fraction (LVEF) ≤45% and an infarct size >10% on day 5 to 9 post MI as measured by cardiac MRI. Patients were then randomized to 6 months treatment with either 25 mg fulacimstat (n = 54) or placebo (n = 53) twice daily on top of standard of care starting day 6 to 12 post MI. The changes in LVEF, LV end-diastolic volume index (LVEDVI), and LV end-systolic volume index (LVESVI) from baseline to 6 months were analyzed by a central blinded cardiac MRI core laboratory. RESULTS Fulacimstat was safe and well tolerated and achieved mean total trough concentrations that were approximately tenfold higher than those predicted to be required for minimal therapeutic activity. Comparable changes in LVEF (fulacimstat: 3.5% ± 5.4%, placebo: 4.0% ± 5.0%, P = .69), LVEDVI (fulacimstat: 7.3 ± 13.3 mL/m2, placebo: 5.1 ± 18.9 mL/m2, P = .54), and LVESVI (fulacimstat: 2.3 ± 11.2 mL/m2, placebo: 0.6 ± 14.8 mL/m2, P = .56) were observed in both treatment arms. CONCLUSION Fulacimstat was safe and well tolerated in patients with left-ventricular dysfunction (LVD) after first STEMI but had no effect on cardiac remodeling.
Collapse
Affiliation(s)
- Hans-Dirk Duengen
- Department of Internal Medicine, Cardiology, Charité-Universitaetsmedizin, Berlin, Germany
| | - Raymond J Kim
- Duke Cardiovascular Magnetic Resonance Center, Duke University Medical Center, Durham, United States
| | - Doron Zahger
- Department of Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Katia Orvin
- Rabin Medical Center - Beilinson Campus, Cardiology Division, Petah Tikva, Israel
| | - Ran Kornowski
- Rabin Medical Center - Beilinson Campus, Cardiology Division, Petah Tikva, Israel
| | - Dan Admon
- Hadassah Hebrew University Hospital Ein Kerem, Heart Institute, Jerusalem, Israel
| | - Jiri Kettner
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Avraham Shimony
- Department of Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Christiane Otto
- Experimental Medicine Cardiovascular, Bayer AG, Wuppertal, Germany.
| | - Michael Becka
- Research and Clinical Sciences Statistics, Bayer AG, Wuppertal, Germany
| | | | | | - Tal Hasin
- Shaare Zedek Medical Center, Department of Cardiology, Jerusalem, Israel
| | | | | | - Michele Senni
- Division of Cardiology, Cardiovascular Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
22
|
Cardiac remodeling after large ST-elevation myocardial infarction in the current therapeutic era. Am Heart J 2020; 223:87-97. [PMID: 32203684 DOI: 10.1016/j.ahj.2020.02.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND The evolution and clinical impact of cardiac remodeling after large ST-elevation myocardial infarction (STEMI) is not well delineated in the current therapeutic era. METHODS The PRESERVATION I trial longitudinally assessed cardiac structure and function in STEMI patients receiving primary percutaneous coronary intervention (PCI). Echocardiograms were performed immediately post-PCI and at 1, 3, 6 and 12 months after STEMI. The extent of cardiac remodeling was assessed in patients with ejection fraction (EF) ≤ 40% after PCI. Patients were stratified by the presence or absence of reverse remodeling, defined as an increase in end-diastolic volume (EDV) of ≤10 mL or decrease in EDV at 1 month, and evaluated for an association with adverse events at 1 year. RESULTS Of the 303 patients with large STEMI enrolled in PRESERVATION I, 225 (74%) had at least moderately reduced systolic function (mean EF 32 ± 5%) immediately after primary PCI. In the following year, there were significant increases in EF and LV volumes, with the greatest magnitude of change occurring in the first month. At 1 month, 104 patients (46%) demonstrated reverse remodeling, which was associated with a significantly lower rate of death, recurrent myocardial infarction and repeat cardiovascular hospitalization at 1 year (HR 0.44; 95% CI: 0.19-0.99). CONCLUSION Reduced EF after large STEMI and primary PCI is common in the current therapeutic era. The first month following primary reperfusion is a critical period during which the greatest degree of cardiac remodeling occurs. Patients demonstrating early reverse remodeling have a significantly lower rate of adverse events in the year after STEMI.
Collapse
|
23
|
Tawfik W, El-Sherif A, Bendary A, Mahros M, Salem M. Impact of global longitudinal strain on left ventricular remodeling and clinical outcome in patients with ST-segment elevation myocardial infarction (STEMI). Echocardiography 2020; 37:570-577. [PMID: 32240553 DOI: 10.1111/echo.14648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Predicting left ventricle (LV) remodeling is important for outcome prediction in patients with ST-segment elevation myocardial infarction (STEMI). Novel echocardiographic techniques may be beneficial for those patients. OBJECTIVES We hypothesized that the semiautomated calculation of baseline global longitudinal strain (GLS) can predict LV remodeling and 6-month clinical outcomes in these patients. METHODS During the period from March to December 2018, 130 patients with successful reperfusion of STEMI were prospectively included. Within 48 hours, patients underwent a baseline GLS study with follow-up study at 6 months. Patients were divided into two groups: group I: patients who showed adverse LV remodeling and group II: patients who did not. The endpoint was a composite of cardiovascular mortality, readmission due to heart failure, and urgent revascularization. RESULTS The mean baseline GLS changed from -13.1 ± 3.5% for group I and -16.8 ± 3.1% for group II, to -10.2 ± 4.7% and -12.6 ± 3.1%, respectively, at 6-month follow-up. ROC analysis demonstrated a cutoff value of baseline GLS > -12.5% predicted LV remodeling with 64.5% sensitivity and 89% specificity (AUC 0.797, 95% CI 0.690-0.904). Multivariate logistic regression analysis model using 6-month MACEs occurrence as a dependent factor showed baseline GLS value> -12.5% to be the only significant independent predictor MACEs occurrence (OR 0.704, 95% CI 0.597-0.829, P < .001). Linear regression analysis showed that for every point estimate deterioration of baseline GLS, there was a significant corresponding 2.55 mL increase in LVEDV at 6-month follow-up (CI -4.501 to -0.612, P = .01). CONCLUSION GLS measurement can predict remodeling and adverse clinical events in STEMI patients.
Collapse
Affiliation(s)
- Wael Tawfik
- Cardiology Department, Benha Faculty of Medicine, Benha University, Benha, Egypt
| | - Amr El-Sherif
- Cardiology Department, Benha Faculty of Medicine, Benha University, Benha, Egypt
| | - Ahmed Bendary
- Cardiology Department, Benha Faculty of Medicine, Benha University, Benha, Egypt
| | - Mohammed Mahros
- Cardiology Department, Benha Faculty of Medicine, Benha University, Benha, Egypt
| | - Mohamed Salem
- Cardiology Department, Benha Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
24
|
Aguiar MO, Tavares BG, Tsutsui JM, Fava AM, Borges BC, Oliveira MT, Soeiro A, Nicolau JC, Ribeiro HB, Chiang HP, Sbano JC, Goldsweig A, Rochitte CE, Lopes BB, Ramirez JA, Kalil Filho R, Porter TR, Mathias W. Sonothrombolysis Improves Myocardial Dynamics and Microvascular Obstruction Preventing Left Ventricular Remodeling in Patients With ST Elevation Myocardial Infarction. Circ Cardiovasc Imaging 2020; 13:e009536. [DOI: 10.1161/circimaging.119.009536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background:
It has recently been demonstrated that high-energy diagnostic transthoracic ultrasound and intravenous microbubbles dissolve thrombi (sonothrombolysis) and increase angiographic recanalization rates in patients with ST-segment–elevation myocardial infarction. We aimed to study the effect of sonothrombolysis on the myocardial dynamics and infarct size obtained by real-time myocardial perfusion echocardiography and their value in preventing left ventricular remodeling.
Methods:
One hundred patients with ST-segment–elevation myocardial infarction were randomized to therapy (50 patients treated with sonothrombolysis and percutaneous coronary intervention) or control (50 patients treated with percutaneous coronary intervention only). Left ventricular volumes, ejection fraction, risk area (before treatment), myocardial perfusion defect over time (infarct size), and global longitudinal strain were determined by quantitative real-time myocardial perfusion echocardiography and speckle tracking echocardiography imaging.
Results:
Risk area was similar in the control and therapy groups (19.2±10.1% versus 20.7±8.9%;
P
=0.56) before treatment. The therapy group presented a behavior significantly different than control group over time (
P
<0.001). The perfusion defect was smaller in the therapy at 48 to 72 hours even in the subgroup of patients with no recanalization at first angiography (12.9±6.5% therapy versus 18.8±9.9% control;
P
=0.015). The left ventricular global longitudinal strain was higher in the therapy than control immediately after percutaneous coronary intervention (14.1±4.1% versus 12.0±3.3%;
P
=0.012), and this difference was maintained until 6 months (17.1±3.5% versus 13.6±3.6%;
P
<0.001). The only predictor of left ventricular remodeling was treatment with sonothrombolysis: the control group was more likely to exhibit left ventricular remodeling with an odds ratio of 2.79 ([95% CI, 0.13–6.86];
P
=0.026).
Conclusions:
Sonothrombolysis reduces microvascular obstruction and improves myocardial dynamics in patients with ST-segment–elevation myocardial infarction and is an independent predictor of left ventricular remodeling over time.
Collapse
Affiliation(s)
- Miguel O.D. Aguiar
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| | - Bruno G. Tavares
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| | - Jeane M. Tsutsui
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| | - Agostina M. Fava
- University of Nebraska Medical Center, Omaha (A.M.F., A.G., T.R.P.)
| | - Bruno C. Borges
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Mucio T. Oliveira
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Alexandre Soeiro
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Jose C. Nicolau
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Henrique B. Ribeiro
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Hsu P. Chiang
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| | - João C.N. Sbano
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| | - Andrew Goldsweig
- University of Nebraska Medical Center, Omaha (A.M.F., A.G., T.R.P.)
| | - Carlos E. Rochitte
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Bernardo B.C. Lopes
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - José A.F. Ramirez
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Roberto Kalil Filho
- Heart Institute (InCor)- University of São Paulo Medical School, Brazil (B.C.B., M.T.O., A.S., J.C.N., H.B.R., C.E.R., B.B.C.L., J.A.F.R., R.K.F.)
| | - Thomas R. Porter
- University of Nebraska Medical Center, Omaha (A.M.F., A.G., T.R.P.)
| | - Wilson Mathias
- Heart Institute (InCor), University of São Paulo Medical School and Fleury Group, Brazil (M.O.D.A., B.G.T., J.M.T., H.P.C., J.C.N.S., W.M.)
| |
Collapse
|
25
|
Yang D, O’Brien CG, Ikeda G, Traverse JH, Taylor DA, Henry TD, Bolli R, Yang PC. Meta-analysis of short- and long-term efficacy of mononuclear cell transplantation in patients with myocardial infarction. Am Heart J 2020; 220:155-175. [PMID: 31821904 PMCID: PMC7173405 DOI: 10.1016/j.ahj.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Mononuclear cells (MNCs) have been tested in clinical trials across multiple cardiovascular pathologies with mixed results. Major adverse cardiac events (MACE) and markers of cardiovascular capacity have been particularly challenging to interpret because of small size. This meta-analysis is aimed to assess the efficacy of MNC therapy in randomized clinical trials to identify the markers of efficiency that could influence future trial design. METHODS PubMed, Embase, Cochrane library, and ClinicalTrials.gov were searched from inception through November 8, 2018. Changes in left ventricular ejection fraction (LVEF) and infarct size from baseline to follow-up were selected as primary outcomes. Changes in the left ventricular end-systolic volume, left ventricular end-diastolic volume, brain natriuretic peptide/N-terminal pro-B-type natriuretic peptide, 6-minute walk test, New York Heart Association class, and MACE incidences were considered secondary outcomes. RESULTS In short-term follow-up, patients treated with MNCs demonstrated a significant increase in absolute LVEF of 2.21% (95% CI 1.59-2.83; P < .001; I2 = 32%) and 6.01% (95% CI 4.45-7.57; P < .001; I2 = 0%) in acute myocardial infarction (AMI) and ischemic cardiomyopathy studies, respectively. This effect was sustained in long-term follow-up. MNC therapy significantly reduced left ventricular end-systolic volume; however, infarct size, 6-minute walk test, New York Heart Association class, and MACE rates were comparable. CONCLUSIONS MNC therapy may convey a modest but sustained increase in LVEF in ischemic cardiomyopathy patients, supporting further investigation. AMI trials, however, demonstrated minimal improvement in LVEF of unclear clinical significance, suggesting a limited role for MNC therapy in AMI.
Collapse
Affiliation(s)
- Dan Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China,Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Connor Galen O’Brien
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Gentaro Ikeda
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, MN55407, USA
| | - Doris A. Taylor
- Regenerative Medicine Research, Texas Heart Institute, PO Box 20345, Houston, TX 77225-0345 USA
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital Health Network, Cincinnati, OH45219, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, KY40202, USA
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| |
Collapse
|
26
|
Fernández-Avilés F, Sanz-Ruiz R, Bogaert J, Casado Plasencia A, Gilaberte I, Belmans A, Fernández-Santos ME, Charron D, Mulet M, Yotti R, Palacios I, Luque M, Sádaba R, San Román JA, Larman M, Sánchez PL, Sanchís J, Jiménez MF, Claus P, Al-Daccak R, Lombardo E, Abad JL, DelaRosa O, Corcóstegui L, Bermejo J, Janssens S. Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With ST-Segment Elevation Myocardial Infarction and Left Ventricular Dysfunction. Circ Res 2019; 123:579-589. [PMID: 29921651 DOI: 10.1161/circresaha.118.312823] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
RATIONALE Allogeneic cardiac stem cells (AlloCSC-01) have shown protective, immunoregulatory, and regenerative properties with a robust safety profile in large animal models of heart disease. OBJECTIVE To investigate the safety and feasibility of early administration of AlloCSC-01 in patients with ST-segment-elevation myocardial infarction. METHODS AND RESULTS CAREMI (Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With STEMI and Left Ventricular Dysfunction) was a phase I/II multicenter, randomized, double-blind, placebo-controlled trial in patients with ST-segment-elevation myocardial infarction, left ventricular ejection fraction ≤45%, and infarct size ≥25% of left ventricular mass by cardiac magnetic resonance, who were randomized (2:1) to receive AlloCSC-01 or placebo through the intracoronary route at days 5 to 7. The primary end point was safety and included all-cause death and major adverse cardiac events at 30 days (all-cause death, reinfarction, hospitalization because of heart failure, sustained ventricular tachycardia, ventricular fibrillation, and stroke). Secondary safety end points included major adverse cardiac events at 6 and 12 months, adverse events, and immunologic surveillance. Secondary exploratory efficacy end points were changes in infarct size (percentage of left ventricular mass) and indices of ventricular remodeling by magnetic resonance at 12 months. Forty-nine patients were included (92% male, 55±11 years), 33 randomized to AlloCSC-01 and 16 to placebo. No deaths or major adverse cardiac events were reported at 12 months. One severe adverse events in each group was considered possibly related to study treatment (allergic dermatitis and rash). AlloCSC-01 elicited low levels of donor-specific antibodies in 2 patients. No immune-related adverse events were found, and no differences between groups were observed in magnetic resonance-based efficacy parameters at 12 months. The estimated treatment effect of AlloCSC-01 on the absolute change from baseline in infarct size was -2.3% (95% confidence interval, -6.5% to 1.9%). CONCLUSIONS AlloCSC-01 can be safely administered in ST-segment-elevation myocardial infarction patients with left ventricular dysfunction early after revascularization. Low immunogenicity and absence of immune-mediated events will facilitate adequately powered studies to demonstrate their clinical efficacy in this setting. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT02439398.
Collapse
Affiliation(s)
- Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Ricardo Sanz-Ruiz
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | | | - Ana Casado Plasencia
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Inmaculada Gilaberte
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Ann Belmans
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Maria Eugenia Fernández-Santos
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Dominique Charron
- HLA et Medicine (HLA-MED), Hôpital Saint-Louis, Paris, France (D.C., R.A.-D.)
| | - Miguel Mulet
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Raquel Yotti
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Itziar Palacios
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Manuel Luque
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Rafael Sádaba
- Department of Cardiac Surgery, Complejo Hospitalario de Navarra, Pamplona, Spain (R.S.)
| | - J Alberto San Román
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Instituto de Ciencias del Corazón (ICICOR), Valladolid, Spain (J.A.S.R.)
| | - Mariano Larman
- Department of Cardiology, Policlínia Guipuzcoa, San Sebastián, Spain (M.L.)
| | - Pedro L Sánchez
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Hospital Clínico Universitario, Salamanca, Spain (P.L.S.)
| | - Juan Sanchís
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Hospital Clínico Universitario, Valencia, Spain (J.S.)
| | - Manuel F Jiménez
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, IBIMA, UMA, UGC Corazón Hospital Clínico Virgen de la Victoria, Málaga, Spain (M.F.J.)
| | - Piet Claus
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Reem Al-Daccak
- HLA et Medicine (HLA-MED), Hôpital Saint-Louis, Paris, France (D.C., R.A.-D.)
| | - Eleuterio Lombardo
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - José Luis Abad
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Olga DelaRosa
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Lucia Corcóstegui
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Javier Bermejo
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Stefan Janssens
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| |
Collapse
|
27
|
Rodriguez-Palomares JF, Gavara J, Ferreira-González I, Valente F, Rios C, Rodríguez-García J, Bonanad C, García Del Blanco B, Miñana G, Mutuberria M, Nuñez J, Barrabés J, Evangelista A, Bodí V, García-Dorado D. Prognostic Value of Initial Left Ventricular Remodeling in Patients With Reperfused STEMI. JACC Cardiovasc Imaging 2019; 12:2445-2456. [PMID: 31202752 DOI: 10.1016/j.jcmg.2019.02.025] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/17/2019] [Accepted: 02/22/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVES This study sought to establish the best definition of left ventricular adverse remodeling (LVAR) to predict outcomes and determine whether its assessment adds prognostic information to that obtained by early cardiac magnetic resonance (CMR). BACKGROUND LVAR, usually defined as an increase in left ventricular end-diastolic volume (LVEDV) is the main cause of heart failure after an ST-segment elevated myocardial infarction; however, the role of assessment of LVAR in predicting cardiovascular events remains controversial. METHODS Patients with ST-segment elevated myocardial infarction who received percutaneous coronary intervention within 6 h of symptom onset were included (n = 498). CMR was performed during hospitalization (6.2 ± 2.6 days) and after 6 months (6.1 ± 1.8 months). The optimal threshold values of the LVEDV increase and the LV ejection fraction decrease associated with the primary endpoint were ascertained. Primary outcome was a composite of cardiovascular mortality, hospitalization for heart failure, or ventricular arrhythmia. RESULTS The study was completed by 374 patients. Forty-nine patients presented the primary endpoint during follow-up (72.9 ± 42.8 months). Values that maximized the ability to identify patients with and without outcomes were a relative rise in LVEDV of 15% (hazard ratio [HR]: 2.1; p = 0.007) and a relative fall in LV ejection fraction of 3% (HR: 2.5; p = 0.001). However, the predictive model (using C-statistic analysis) failed to demonstrate that direct observation of LVAR at 6 months adds information to data from early CMR in predicting outcomes (C-statistic: 0.723 vs. 0.795). CONCLUSIONS The definition of LVAR that best predicts adverse cardiovascular events should consider both the increase in LVEDV and the reduction in LV ejection fraction. However, assessment of LVAR does not improve information provided by the early CMR.
Collapse
Affiliation(s)
- Jose F Rodriguez-Palomares
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Jose Gavara
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain
| | - Ignacio Ferreira-González
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain
| | - Filipa Valente
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - César Rios
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain
| | - Julián Rodríguez-García
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Clara Bonanad
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain
| | - Bruno García Del Blanco
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Gema Miñana
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain
| | - Maria Mutuberria
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Julio Nuñez
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain
| | - José Barrabés
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Artur Evangelista
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain
| | - Vicente Bodí
- Department of Cardiology, Hospital Clinico Universitario, CIBERCV, INCLIVA, University of Valencia, Valencia, Spain.
| | - David García-Dorado
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red-para enfermedades cardiovasculares, CIBERCV, Madrid, Spain.
| |
Collapse
|
28
|
Kercheva M, Ryabova T, Gusakova A, Suslova TE, Ryabov V, Karpov RS. Serum Soluble ST2 and Adverse Left Ventricular Remodeling in Patients With ST-Segment Elevation Myocardial Infarction. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2019; 13:1179546819842804. [PMID: 31065219 PMCID: PMC6487275 DOI: 10.1177/1179546819842804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/13/2019] [Indexed: 01/14/2023]
Abstract
Objective: To assess the dynamics of serum levels of soluble isoform of suppression of
tumorigenicity 2 (sST2) and N-terminal pro-brain natriuretic peptide
(NT-proBNP) and their correlations with the development of adverse left
ventricular remodeling (LVR) through 6 months in patients with primary
myocardial infarction with ST-segment elevation (STEMI). Methods: Subjects were 31 patients with STEMI (median age: 58 years), who underwent
percutaneous coronary intervention (PCI) during the first 24 hours of the
onset of myocardial infarction (MI). Blood samples and parameters of
echocardiography were assessed at days 1, 3, 7, and 14 and 6 months after
STEMI. Results: Serum levels of sST2 and NT-proBNP decreased during the 6-month period.
Levels of sST2 decreased by 48% from admission to day 7, and levels of
NT-proBNP decreased by 40% from day 7 to 6 months after STEMI. Serum levels
of sST2 at day 1 (r = 0.5, P < .05) and
day 3 (r = 0.4, P < .05) were
associated with adverse LVR by 6 months after STEMI. Logistic regression
analysis showed that a high concentration of sST2 at day 7 increased the
risk of adverse LVR (95% confidence interval [CI], 0.5-0.9; areas under
curve [AUC] = 0.8; P = .002), with 92% sensitivity and 70%
specificity. A multivariate analysis model revealed that adverse LVR was
associated with the level of sST2 (P = .003) and with
complete revascularization (P = .01) at the admission. Conclusions: The dynamics of serum levels of sST2 and NT-proBNP were different. The level
of sST2 normalized by the 7th day; NT-proBNP decreased only by the end of
the 6-month period after MI. Increased serum levels of sST2 by the 7th day
of MI were associated with the development of adverse LVR by the end of the
6-month period.
Collapse
Affiliation(s)
- Maria Kercheva
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Tamara Ryabova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Anna Gusakova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Tatiana E Suslova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Vyacheslav Ryabov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation.,National Research Tomsk State University, Tomsk, Russian Federation
| | - Rostislav S Karpov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russian Federation.,Siberian State Medical University, Tomsk, Russian Federation
| |
Collapse
|
29
|
Integrative System Biology Analyses Identify Seven MicroRNAs to Predict Heart Failure. Noncoding RNA 2019; 5:ncrna5010022. [PMID: 30866581 PMCID: PMC6468490 DOI: 10.3390/ncrna5010022] [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: 02/01/2019] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) has several etiologies including myocardial infarction (MI) and left ventricular remodeling (LVR), but its progression remains difficult to predict in clinical practice. Systems biology analyses of LVR after MI provide molecular insights into this event such as modulation of microRNA (miRNA) that could be used as a signature of HF progression. To define a miRNA signature of LVR after MI, we use 2 systems biology approaches, integrating either proteomic data generated from LV of post-MI rat induced by left coronary artery ligation or multi-omics data (proteins and non-coding RNAs) generated from plasma of post-MI patients from the REVE-2 study. The first approach predicted that 13 miRNAs and 3 of these miRNAs would be validated to be associated with LVR in vivo: miR-21-5p, miR-23a-3p and miR-222-3p. The second approach predicted that 24 miRNAs among 1310 molecules and 6 of these miRNAs would be selected to be associated with LVR in silico: miR-17-5p, miR-21-5p, miR-26b-5p, miR-222-3p, miR-335-5p and miR-375. We identified a signature of 7 microRNAs associated with LVR after MI that support the interest of integrative systems biology analyses to define a miRNA signature of HF progression.
Collapse
|
30
|
Reverse, stationary and progressive left ventricular dilatation in dilated and nondilated ventricles on admission following first anterior STEMI. Coron Artery Dis 2019; 30:11-19. [DOI: 10.1097/mca.0000000000000665] [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: 11/26/2022]
|
31
|
First Hospitalization for Heart Failure in Outpatients With Stable Coronary Artery Disease: Determinants, Role of Incident Myocardial Infarction, and Prognosis. J Card Fail 2018; 24:815-822. [DOI: 10.1016/j.cardfail.2018.09.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 09/03/2018] [Accepted: 09/27/2018] [Indexed: 11/27/2022]
|
32
|
Aboelkasem Ali Mousa M, Abdelsabour Abdallah M, Shamseddin Mohammad H, Ahmad Aly Youssef A. Early predictors of left ventricular remodeling after primary percutaneous coronary intervention. Egypt Heart J 2018; 70:403-407. [PMID: 30591763 PMCID: PMC6303352 DOI: 10.1016/j.ehj.2018.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/21/2018] [Indexed: 11/24/2022] Open
|
33
|
Ferreira JP, Bauters C, Eschalier R, Lamiral Z, Fay R, Huttin O, Girerd N, Zannad F, Pinet F, Rossignol P. Echocardiographic diastolic function evolution in patients with an anterior Q-wave myocardial infarction: insights from the REVE-2 study. ESC Heart Fail 2018; 6:70-79. [PMID: 30460754 PMCID: PMC6351891 DOI: 10.1002/ehf2.12359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/21/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022] Open
Abstract
Aims Myocardial fibrosis plays a key role in the development of adverse left ventricular remodelling after myocardial infarction (MI). This study aimed to determine whether the circulating levels of BNP, collagen peptides, and galectin‐3 are associated with diastolic function evolution (both deterioration and improvement) at 1 year after an anterior MI. Methods and results The REVE‐2 is a prospective multicentre study including 246 patients with a first anterior Q‐wave MI. Echocardiographic assessment was performed at hospital discharge and ±1 year after MI. BNP, galectin‐3, and collagen peptides were measured ±1 month after MI. Left ventricular diastolic dysfunction (DD) was defined according to the presence of at least two criteria of echocardiographic parameters: septal e′ < 8 cm/s, lateral e′ < 10 cm/s, and left atrial volume ≥ 34 mL/m2. At baseline, 87 (35.4%) patients had normal diastolic function and 159 (64.6%) patients had DD. Follow‐up of 61 patients among the 87 patients with normal diastolic function at baseline showed that 22 patients (36%) developed DD at 1 year post‐MI. The circulating levels of amino‐terminal propeptide of type III procollagen > 6 mg/L [odds ratio (OR) = 5.29; 95% confidence interval (CI) = 1.05–26.66; P = 0.044], galectin‐3 > 13 μg/L (OR = 5.99; 95% CI = 1.18–30.45; P = 0.031), and BNP > 82 ng/L (OR = 10.25; 95% CI = 2.36–44.50; P = 0.002) quantified at 1 month post‐MI were independently associated with 1 year DD. Follow‐up of the 137 patients with DD at baseline among the 159 patients showed that 36 patients (26%) had a normalized diastolic function at 1 year post‐MI. Patients with a BNP > 82 ng/L were less likely to improve diastolic function (OR = 0.06; 95% CI = 0.01–0.28; P = 0.0003). Conclusions The present study suggests that circulating levels of amino‐terminal propeptide of type III procollagen, galectin‐3, and BNP may be independently associated with new‐onset DD in post‐MI patients.
Collapse
Affiliation(s)
- João Pedro Ferreira
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France.,F-CRIN INI-CRCT, Nancy, France.,Department of Physiology, University of Porto, Porto, Portugal
| | - Christophe Bauters
- CHU Lille, Lille, France.,INSERM U1167, Institut Pasteur de Lille, Université de Lille Nord de France, FHU-REMOD-VHF, Lille, France.,Faculté de Médecine de Lille, Lille, France
| | - Romain Eschalier
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France.,F-CRIN INI-CRCT, Nancy, France.,Department of Cardiology, Université Clermont Auvergne, TGI/ISIT/CaViti, Institut Pascal and CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Zohra Lamiral
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France
| | - Renaud Fay
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France
| | - Olivier Huttin
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France
| | - Nicolas Girerd
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France.,F-CRIN INI-CRCT, Nancy, France.,Department of Cardiology, CHRU Nancy, Nancy, France.,Université de Lorraine, Nancy, France
| | - Faiez Zannad
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France.,F-CRIN INI-CRCT, Nancy, France.,Université de Lorraine, Nancy, France.,INSERM U1116, Nancy, France
| | - Florence Pinet
- F-CRIN INI-CRCT, Nancy, France.,CHU Lille, Lille, France.,INSERM U1167, Institut Pasteur de Lille, Université de Lille Nord de France, FHU-REMOD-VHF, Lille, France
| | - Patrick Rossignol
- Centre d'Investigation Clinique CIC-P 1433, INSERM, Nancy, France.,F-CRIN INI-CRCT, Nancy, France.,Université de Lorraine, Nancy, France.,INSERM U1116, Nancy, France
| |
Collapse
|
34
|
Düngen HD, Kober L, Nodari S, Schou M, Otto C, Becka M, Kanefendt F, Winkelmann BR, Gislason G, Richard F, Nielsen OW, Gheorghiade M, Senni M. Safety and Tolerability of the Chymase Inhibitor Fulacimstat in Patients With Left Ventricular Dysfunction After Myocardial Infarction-Results of the CHIARA MIA 1 Trial. Clin Pharmacol Drug Dev 2018; 8:942-951. [PMID: 30452784 DOI: 10.1002/cpdd.633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/26/2018] [Indexed: 01/29/2023]
Abstract
The chymase inhibitor fulacimstat is developed as a first-in-class treatment option for the inhibition of adverse cardiac remodeling in patients with left ventricular dysfunction (LVD) after acute myocardial infarction (MI). The aim of the study was to examine the safety and tolerability of fulacimstat in patients with LVD after remote MI. A multicenter, multinational randomized, placebo-controlled study was performed in clinically stable patients (40-79 years of age, left ventricular ejection fraction ≤ 45% because of MI in medical history) who were on stable evidence-based standard-of-care therapies for LVD post-MI including an angiotensin converting enzyme inhibitor or angiotensin receptor blocker at doses of at least half the recommended target dose. Patients were treated for 2 weeks with either placebo (n = 12) or 4 different doses of fulacimstat (5 mg twice daily, n = 9; 10 mg twice daily, n = 9; 25 mg twice daily, n = 10; 50 mg once daily, n = 9). Fulacimstat was safe and well tolerated at all examined doses. There were no clinically relevant effects on vital signs or potassium levels compared with placebo treatment. Mean plasma concentrations of fulacimstat increased with the administered dose and reached exposures predicted to be therapeutically active. The safety profile and the absence of effects on blood pressure or heart rate in a chronic patient population having similar comorbidities and receiving similar comedication as patients after acute MI support future clinical trials with fulacimstat in patients after acute MI.
Collapse
Affiliation(s)
- Hans-Dirk Düngen
- Department of Internal Medicine, Cardiology, Charité-Universitaetsmedizin, Berlin, Germany
| | - Lars Kober
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Savina Nodari
- Cardiology Section, Department of Clinical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Morten Schou
- Department of Cardiology, Cardiovascular Research Center, Gentofte and Herlev University Hospital, Herlev, Denmark
| | - Christiane Otto
- Experimental Medicine Cardiovascular and Hematology, BAYER AG, Wuppertal, Germany
| | - Michael Becka
- Research and Clinical Sciences Statistics, BAYER AG, Wuppertal, Germany
| | | | | | - Gunnar Gislason
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | | | - Olav Wendelboe Nielsen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Mihai Gheorghiade
- Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michele Senni
- Division of Cardiology 1, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
35
|
Circulating miR-1254 predicts ventricular remodeling in patients with ST-Segment-Elevation Myocardial Infarction: A cardiovascular magnetic resonance study. Sci Rep 2018; 8:15115. [PMID: 30310086 PMCID: PMC6181905 DOI: 10.1038/s41598-018-33491-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/25/2018] [Indexed: 01/26/2023] Open
Abstract
Reliable noninvasive prognostic biomarkers for left ventricular (LV) remodeling in ST-segment elevation myocardial infarction (STEMI) are needed. This study aimed to evaluate a panel of circulating microRNAs (miRNAs) as biomarkers of LV remodeling using cardiovascular magnetic resonance (CMR). We prospectively evaluated patients with a first STEMI treated with primary percutaneous coronary intervention who underwent CMR imaging at 1 week and 6 months after STEMI (n = 70). miRNAs were measured using PCR-based technologies in plasma samples collected at admission. The associations between miRNAs and LV diastolic and systolic volumes, and ejection fraction at 6-months were estimated in adjusted models. Median age was 60 years, 71.4% were male. miR-1254 was significantly associated in univariate analyses. Patients in the highest tertile of miR-1254 exhibited lower values of LVEDVI and LVESVI and higher values of LVEF at 1 week. After comprehensive multivariate adjustment including clinical, CMR variables, hs-troponin-T and NT-proBNP, miRNA-1254 was associated with decreasing LVESVI (P = 0.006), and borderline negative associated with LVEDVI (P = 0.063) at 6-months. miR-1254 also exhibited a significant positive association with increasing LVEF during follow-up (P < 0.001). Plasma miRNA-1254 predicted changes in LV volumes and LVEF at 6 months after STEMI. The value of miR-1254 to inform tailored treatment selection and monitor ongoing efficacy deserves further investigation.
Collapse
|
36
|
Wang C, Jing Q. Non-coding RNAs as biomarkers for acute myocardial infarction. Acta Pharmacol Sin 2018; 39:1110-1119. [PMID: 29698386 DOI: 10.1038/aps.2017.205] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/25/2017] [Indexed: 12/13/2022] Open
Abstract
Acute myocardial infarction (AMI) is a main threat to human lives worldwide. Early and accurate diagnoses warrant immediate medical care, which would reduce mortality and improve prognoses. Circulating non-coding RNAs have been demonstrated to serve as competent biomarkers for various diseases. Following the identification of cardiac-specific microRNA miR-208a in circulation, more non-coding RNAs (miR-1, miR-499 and miR-133) have been identified as biomarkers not only for the diagnosis of AMI but also for prognosis post infarction. Here, we summarized recent findings on non-coding RNAs as biomarkers for early diagnosis of ST-segment elevation myocardial infarction and for disease monitoring of myocardial infarction. In addition, the prognostic potential of non-coding RNAs in patients treated with percutaneous coronary intervention was also described. We also include studies based on biobanks, and build a miRNA release spectrum after AMI, which provides quantitative and time-lapse monitoring of AMI progress. With this spectrum, we are able to customize personal medical care, which prevents further damage. By constructing a network of circulating non-coding RNAs with high specificity and sensitivity, detailed diagnostic information was provided for personalized medicine. Unveiling the roles and kinetics of circulating non-coding RNAs may lead to a revolution in clinical diagnosis.
Collapse
|
37
|
Itier R, Roncalli J. New therapies for acute myocardial infarction: current state of research and future promise. Future Cardiol 2018; 14:329-342. [DOI: 10.2217/fca-2017-0047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Progress has been made into research on new therapies, mechanical and pharmacological approaches and repair/regenerative cellular therapy to treat irreversible cardiovascular pathologies, such as acute myocardial infarction. Research into cellular therapies is exploring the use of new cellular types. Although the therapeutic effects of cell therapy remain modest, results from clinical trials are encouraging. To expand this improvement, advances are being made that involve the paracrine function of stem cells, the use of growth factors, miRNA and new biomaterials. In the near future, these therapies should become part of routine clinical practice.
Collapse
Affiliation(s)
- Romain Itier
- Department of Cardiology A, Institute CARDIOMET, Clinical Center of Investigation for Biotherapies, CIC-BT 0511, INSERM 1048, University Hospital of Toulouse, Toulouse, France
| | - Jerome Roncalli
- Department of Cardiology A, Institute CARDIOMET, Clinical Center of Investigation for Biotherapies, CIC-BT 0511, INSERM 1048, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
38
|
Maciejak A, Kostarska-Srokosz E, Gierlak W, Dluzniewski M, Kuch M, Marchel M, Opolski G, Kiliszek M, Matlak K, Dobrzycki S, Lukasik A, Segiet A, Sygitowicz G, Sitkiewicz D, Gora M, Burzynska B. Circulating miR-30a-5p as a prognostic biomarker of left ventricular dysfunction after acute myocardial infarction. Sci Rep 2018; 8:9883. [PMID: 29959359 PMCID: PMC6026144 DOI: 10.1038/s41598-018-28118-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/12/2018] [Indexed: 02/06/2023] Open
Abstract
Left ventricular (LV) dysfunction after acute myocardial infarction (AMI) is associated with an increased risk of heart failure (HF) development. Diverse microRNAs (miRNAs) have been shown to appear in the bloodstream following various cardiovascular events. The aim of this study was to identify prognostic miRNAs associated with LV dysfunction following AMI. Patients were divided into subgroups comprising patients who developed or not LV dysfunction within six months of the infarction. miRNA profiles were determined in plasma and serum samples of the patients on the first day of AMI. Levels of 14 plasma miRNAs and 16 serum miRNAs were significantly different in samples from AMI patients who later developed LV dysfunction compared to those who did not. Two miRNAs were up-regulated in both types of material. Validation in an independent group of patients, using droplet digital PCR (ddPCR) confirmed that miR-30a-5p was significantly elevated on admission in those patients who developed LV dysfunction and HF symptoms six months after AMI. A bioinformatics analysis indicated that miR-30a-5p may regulate genes involved in cardiovascular pathogenesis. This study demonstrates, for the first time, a prognostic value of circulating miR-30a-5p and its association with LV dysfunction and symptoms of HF after AMI.
Collapse
Affiliation(s)
- Agata Maciejak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Edyta Kostarska-Srokosz
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Wlodzimierz Gierlak
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Miroslaw Dluzniewski
- Cardiology Department, Midtown Medical Center, Mazovia Brodno Hospital, Warsaw, Poland
| | - Marek Kuch
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Michal Marchel
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Kiliszek
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland
| | - Krzysztof Matlak
- Department of Cardiac Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Slawomir Dobrzycki
- Department of Invasive Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Lukasik
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Segiet
- 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grazyna Sygitowicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Dariusz Sitkiewicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
39
|
Kanefendt F, Thuß U, Becka M, Boxnick S, Berse M, Schultz A, Otto C. Pharmacokinetics, Safety, and Tolerability of the Novel Chymase Inhibitor BAY 1142524 in Healthy Male Volunteers. Clin Pharmacol Drug Dev 2018; 8:467-479. [PMID: 29878583 DOI: 10.1002/cpdd.579] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/19/2018] [Indexed: 12/18/2022]
Abstract
The orally available chymase inhibitor BAY 1142524 is currently being developed as a first-in-class treatment for left-ventricular dysfunction after myocardial infarction. Results from 3 randomized, single-center, phase 1 studies in healthy male volunteers examining the safety, tolerability, and pharmacokinetics of BAY 1142524 are summarized. In this first-in-human study, single oral doses of 1-200 mg were administered in fasted state as liquid service formulation or immediate release (IR) tablets. The relative bioavailability and the effect of a high-fat/high-calorie meal were investigated at the 5-mg dose. In a multiple-dose escalation study, doses of 5-50 mg twice daily and 100 mg once daily were given for 5 consecutive days. BAY 1142524 was safe and well tolerated and had no effects on heart rate or blood pressure compared with placebo. BAY 1142524 was absorbed with peak concentration 1-3 hours after administration for IR tablets; it was eliminated from plasma with a terminal half-life of 6.84-12.0 hours after administration of liquid service formulation or IR tablets. Plasma exposures appeared to be dose-linear, with a negligible food effect. There was only low accumulation of BAY 1142524 after multiple dosing. BAY 1142524 exhibited a pharmacokinetic profile allowing for once-daily dosing. The absence of blood pressure effects after administration of BAY 1142524 supports the combination of this novel anti-remodeling drug with existing standard of care in patients with left-ventricular dysfunction after acute myocardial infarction.
Collapse
Affiliation(s)
| | - Uwe Thuß
- Bioanalytics, BAYER AG, Wuppertal, Germany
| | - Michael Becka
- Research and Clinical Sciences Statistics, BAYER AG, Wuppertal, Germany
| | - Stefanie Boxnick
- CRS Clinical Research Services, Wuppertal GmbH, Wuppertal, Germany
| | - Matthias Berse
- CRS Clinical Research Services, Berlin GmbH, Berlin, Germany
| | - Armin Schultz
- CRS Clinical Research Services, Mannheim GmbH, Mannheim, Germany
| | - Christiane Otto
- Experimental Medicine Cardiovascular and Hematology, BAYER AG, Wuppertal, Germany
| |
Collapse
|
40
|
Turkieh A, Fertin M, Bouvet M, Mulder P, Drobecq H, Lemesle G, Lamblin N, de Groote P, Porouchani S, Chwastyniak M, Beseme O, Amouyel P, Mouquet F, Balligand JL, Richard V, Bauters C, Pinet F. Expression and Implication of Clusterin in Left Ventricular Remodeling After Myocardial Infarction. Circ Heart Fail 2018; 11:e004838. [DOI: 10.1161/circheartfailure.117.004838] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/16/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Annie Turkieh
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Marie Fertin
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Marion Bouvet
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Paul Mulder
- Institut Pasteur de Lille, Université de Lille, France. Inserm U1096, FHU-REMOD-VHF, Normandie University, University of Rouen, France (P.M., V.R.)
| | - Hervé Drobecq
- UMR 8161-M3T-Mechanisms of Tumorigenesis and Target Therapies, CNRS (H.D.)
| | - Gilles Lemesle
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Nicolas Lamblin
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Pascal de Groote
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
| | - Sina Porouchani
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Maggy Chwastyniak
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Olivia Beseme
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Philippe Amouyel
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- CHU Lille, Service de Santé Publique, Épidémiologie, Économie de la Santé et Prévention, France (P.A.)
| | - Frédéric Mouquet
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
| | - Jean-Luc Balligand
- Institut de Recherche Experimentale et Clinique, Pole of Pharmacology and Therapeutics and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium (J.-L.B.)
| | - Vincent Richard
- Institut Pasteur de Lille, Université de Lille, France. Inserm U1096, FHU-REMOD-VHF, Normandie University, University of Rouen, France (P.M., V.R.)
| | - Christophe Bauters
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Florence Pinet
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| |
Collapse
|
41
|
Kitamura M, Schmidt T, Kuck KH, Frerker C. Heart Failure Interventions Targeting Impaired Left Ventricles in Structural Heart Disease. Curr Cardiol Rep 2018; 20:8. [PMID: 29435772 DOI: 10.1007/s11886-018-0950-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Interventional techniques have been developed for a wide spectrum of mechanisms of heart failure (HF), especially in valvular heart disease and cardiomyopathies (ischaemic cardiomyopathy and hypertrophic cardiomyopathy). In this article, we review recent reports on catheter interventions to treat patients with HF. RECENT FINDINGS Direct modification using the Parachute device and the REVIVENT-TC device for patients with impaired left ventricle with large infarct scars improves geometry and haemodynamic efficiency, resulting in a reduction of HF symptoms. Interatrial shunt therapy improves symptoms and quality of life in HF patients. Uniquely, left ventricular outflow tract obstruction has also been targeted in patients with transcatheter mitral valve implantation. For advanced stage HF patients with prohibitively high surgical risk, emerging transcatheter interventions make it possible to modify life-limiting symptoms. Further results on HF interventions are expected from ongoing clinical trials.
Collapse
Affiliation(s)
- Mitsunobu Kitamura
- Department of Cardiology, Asklepios Klinik St. Georg, Lohmühlenstr. 5, 20099, Hamburg, Germany
| | - Tobias Schmidt
- Department of Cardiology, Asklepios Klinik St. Georg, Lohmühlenstr. 5, 20099, Hamburg, Germany
| | - Karl-Heinz Kuck
- Department of Cardiology, Asklepios Klinik St. Georg, Lohmühlenstr. 5, 20099, Hamburg, Germany
| | - Christian Frerker
- Department of Cardiology, Asklepios Klinik St. Georg, Lohmühlenstr. 5, 20099, Hamburg, Germany.
| |
Collapse
|
42
|
Jackman CP, Ganapathi AM, Asfour H, Qian Y, Allen BW, Li Y, Bursac N. Engineered cardiac tissue patch maintains structural and electrical properties after epicardial implantation. Biomaterials 2018; 159:48-58. [PMID: 29309993 DOI: 10.1016/j.biomaterials.2018.01.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/17/2017] [Accepted: 01/01/2018] [Indexed: 12/22/2022]
Abstract
Functional cardiac tissue engineering holds promise as a candidate therapy for myocardial infarction and heart failure. Generation of "strong-contracting and fast-conducting" cardiac tissue patches capable of electromechanical coupling with host myocardium could allow efficient improvement of heart function without increased arrhythmogenic risks. Towards that goal, we engineered highly functional 1 cm × 1 cm cardiac tissue patches made of neonatal rat ventricular cells which after 2 weeks of culture exhibited force of contraction of 18.0 ± 1.4 mN, conduction velocity (CV) of 32.3 ± 1.8 cm/s, and sustained chronic activation when paced at rates as high as 8.7 ± 0.8 Hz. Patches transduced with genetically-encoded calcium indicator (GCaMP6) were implanted onto adult rat ventricles and after 4-6 weeks assessed for action potential conduction and electrical integration by two-camera optical mapping of GCaMP6-reported Ca2+ transients in the patch and RH237-reported action potentials in the recipient heart. Of the 13 implanted patches, 11 (85%) engrafted, maintained structural integrity, and conducted action potentials with average CVs and Ca2+ transient durations comparable to those before implantation. Despite preserved graft electrical properties, no anterograde or retrograde conduction could be induced between the patch and host cardiomyocytes, indicating lack of electrical integration. Electrical properties of the underlying myocardium were not changed by the engrafted patch. From immunostaining analyses, implanted patches were highly vascularized and expressed abundant electromechanical junctions, but remained separated from the epicardium by a non-myocyte layer. In summary, our studies demonstrate generation of highly functional cardiac tissue patches that can robustly engraft on the epicardial surface, vascularize, and maintain electrical function, but do not couple with host tissue. The lack of graft-host electrical integration is therefore a critical obstacle to development of efficient tissue engineering therapies for heart repair.
Collapse
Affiliation(s)
| | - Asvin M Ganapathi
- Duke University Medical Center, Department of General Surgery, Durham, NC, USA
| | - Huda Asfour
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Ying Qian
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Brian W Allen
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Yanzhen Li
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Nenad Bursac
- Duke University, Department of Biomedical Engineering, Durham, NC, USA.
| |
Collapse
|
43
|
Mercier T, Bouvet M, Dubois-Deruy E, Dechaumes A, Beseme O, Richard V, Mulder P, Pinet F. Interplay Between Phosphorylation and O-GlcNAcylation of Sarcomeric Proteins in Ischemic Heart Failure. Front Endocrinol (Lausanne) 2018; 9:598. [PMID: 30344511 PMCID: PMC6182077 DOI: 10.3389/fendo.2018.00598] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/19/2018] [Indexed: 11/13/2022] Open
Abstract
Post-translational modifications (PTMs) of sarcomeric proteins could participate to left ventricular (LV) remodeling and contractile dysfunction leading in advanced heart failure (HF) with altered ejection fraction. Using an experimental rat model of HF (ligation of left coronary artery) and phosphoproteomic analysis, we identified an increase of desmin phosphorylation and a decrease of desmin O-N-acetylglucosaminylation (O-GlcNAcylation). We aim to characterize interplay between phosphorylation and O-GlcNAcylation for desmin in primary cultures of cardiomyocyte by specific O-GlcNAcase (OGA) inhibition with thiamet G and silencing O-GlcNAc transferase (OGT) and, in perfused heart perfused with thiamet G in sham- and HF-rats. In each model, we found an efficiency of O-GlcNAcylation modulation characterized by the levels of O-GlcNAcylated proteins and OGT expression (for silencing experiments in cells). In perfused heart, we found an improvement of cardiac function under OGA inhibition. But none of the treatments either in in vitro or ex vivo cardiac models, induced a modulation of desmin, phosphorylated and O-GlcNAcylated desmin expression, despite the presence of O-GlcNAc moities in cardiac desmin. Our data suggests no interplay between phosphorylation and O-GlcNAcylation of desmin in HF post-myocardial infarction. The future requires finding the targets in heart involved in cardiac improvement under thiamet G treatment.
Collapse
Affiliation(s)
- Thomas Mercier
- INSERM U1167 Unité d'Epidémiologie et de Santé Publique, Lille, France
| | - Marion Bouvet
- INSERM U1167 Unité d'Epidémiologie et de Santé Publique, Lille, France
| | | | - Arthur Dechaumes
- INSERM U1167 Unité d'Epidémiologie et de Santé Publique, Lille, France
| | - Olivia Beseme
- INSERM U1167 Unité d'Epidémiologie et de Santé Publique, Lille, France
| | - Vincent Richard
- INSERM UMR1096, Endothélium, Valvulopathies et Insuffisance Cardiaque, Rouen, France
| | - Paul Mulder
- INSERM UMR1096, Endothélium, Valvulopathies et Insuffisance Cardiaque, Rouen, France
| | - Florence Pinet
- INSERM U1167 Unité d'Epidémiologie et de Santé Publique, Lille, France
- *Correspondence: Florence Pinet
| |
Collapse
|
44
|
Ola RK, Meena CB, Ramakrishnan S, Agarwal A, Bhargava S. Detection of Left Ventricular Remodeling in Acute ST Elevation Myocardial Infarction after Primary Percutaneous Coronary Intervention by Two Dimensional and Three Dimensional Echocardiography. J Cardiovasc Echogr 2018; 28:39-44. [PMID: 29629258 PMCID: PMC5875134 DOI: 10.4103/jcecho.jcecho_32_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Left ventricular remodeling (LVR) after ST-elevation myocardial infarction (STEMI) harbingers poor prognosis. Three-dimensional echocardiography (3DE) is more accurate than 2 D echo for the assessment of left ventricle (LV) shape. We assessed LV geometry with 3D ECHO 6 months after STEMI in patients who had primary angioplasty. Materials and Methods In this prospective study, morphological and functional analysis of LV with 3D ECHO (volumes, LVEF, 3D sphericity index [SI]) was assessed up to 7 days and 6 months in 42 STEMI patients. The LVR was considered for increase >15% of the end diastolic volume of the LV (LVEDV) 6 months after the STEMI, compared to the LVEDV up to 7 days of it. Results Sixteen (38%) patients had LVR. 3D Echocardiographic measurements up to 7 days after the acute myocardial infarction (AMI) 1-LVEDV in ventricular remodeling group was 99.8 ± 19.1 ml and in no ventricular remodeling group was 87 ± 18.2 mL (P = 0.037); 2-LVEF was 0.48 ± 0.01 and 51 ± 0.02 (P <.001); 3D-SI was 0.41 ± 0.05 and 31 ± 0.05 (P < 0.001) II-after 6 months: 1-LVEDV in remodeling group was 114.2 ± 19.5 mL and no remodeling group was 94.2 ± 18.6 (P = 0.002); 2-LVEF was 0.58 ± 0.01 and 59 ± .01 (P = 0.003); 3D-sphericity was 0.35 ± 0.05 and 28 ± .05 (P < 0.001). Conclusion LVR was observed in 38% of the patients 6 months after AMI. The 3D SI has been associated with occurrence of LVR and can differentiate patients with and without subsequent development of LVR accurately and early on its basis.
Collapse
Affiliation(s)
- Rakesh Kumar Ola
- Department of Cardiology, SMS Medical College, Jaipur, Rajasthan, India
| | | | | | - Ashish Agarwal
- Department of Cardiology, SMS Medical College, Jaipur, Rajasthan, India
| | - Smriti Bhargava
- Department of Cardiology, SMS Medical College, Jaipur, Rajasthan, India
| |
Collapse
|
45
|
Liu D, Zeng X, Li X, Mehta JL, Wang X. Role of NLRP3 inflammasome in the pathogenesis of cardiovascular diseases. Basic Res Cardiol 2017; 113:5. [PMID: 29224086 DOI: 10.1007/s00395-017-0663-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
NLRP3 inflammasome is a key multiprotein signaling platform that tightly controls inflammatory responses and coordinates antimicrobial host defenses by activating caspase-1 for the subsequent maturation of pro-inflammatory cytokines, IL-1β and IL-18, and induces pyroptosis. The assembly and activation of NLRP3 inflammasome are linked to the pathogenesis of several cardiovascular disease risk factors, such as hypertension and diabetes, and their major consequences-myocardial remodeling. The study of the NLRP3 inflammasome in these cardiovascular disease states may uncover important triggers and endogenous modulators of the disease, and lead to new treatment strategies. This review outlines current insights into NLRP3 inflammasome research associated with cardiovascular diseases and discusses the questions that remain in this field.
Collapse
Affiliation(s)
- Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiang Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiao Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jawahar L Mehta
- Central Arkansas Veterans Healthcare System and the Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
46
|
Bauters C, Dubois E, Porouchani S, Saloux E, Fertin M, de Groote P, Lamblin N, Pinet F. Long-term prognostic impact of left ventricular remodeling after a first myocardial infarction in modern clinical practice. PLoS One 2017; 12:e0188884. [PMID: 29176897 PMCID: PMC5703528 DOI: 10.1371/journal.pone.0188884] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/14/2017] [Indexed: 11/19/2022] Open
Abstract
Background The association of left ventricular remodeling (LVR) after myocardial infarction (MI) with the subsequent risk of heart failure (HF) and death has not been studied in patients receiving optimal secondary prevention. Methods and results We performed a long-term clinical follow-up of patients included in 2 prospective multicentric studies on LVR after first anterior MI. At 1-year echocardiography, LVR (≥20% increase in end-diastolic volume from baseline to 1 year) occurred in 67/215 (31%) patients in cohort 1 and in 87/226 (38%) patients in cohort 2. The prescription rate of secondary prevention medications was very high (ß-blockers at 1 year: 90% and 95% for cohorts 1 and 2, respectively; angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers (ACE-I/ARB) at 1 year: 93% and 97% for cohorts 1 and 2, respectively). Median clinical follow-up after LVR assessment was 11.0 years in cohort 1 and 7.8 years in cohort 2. In both cohorts, LVR patients had a progressive increase in the risk of cardiovascular death or hospitalization for HF (p = 0.0007 in cohort 1 and 0.009 in cohort 2) with unadjusted hazard ratios of 2.52 [1.45–4.36] and 2.52 [1.23–5.17], respectively. Similar results were obtained when cardiovascular death was considered as an isolated endpoint. After adjustement on baseline characteristics including ejection fraction, the association with the composite endpoint was unchanged. Conclusion In a context of a modern therapeutic management with a large prescription of evidence-based medications, LVR remains independently associated with HF and cardiovascular death at long-term follow-up after MI.
Collapse
Affiliation(s)
- Christophe Bauters
- University of Lille, Inserm U1167, Institut Pasteur, University Hospital of Lille, Lille, France
- * E-mail:
| | - Emilie Dubois
- University of Lille, Inserm U1167, Institut Pasteur, Lille, France
| | | | - Eric Saloux
- University of Caen, EA 4650, University Hospital of Caen, Caen, France
| | | | - Pascal de Groote
- University of Lille, Inserm U1167, Institut Pasteur, University Hospital of Lille, Lille, France
| | - Nicolas Lamblin
- University of Lille, Inserm U1167, Institut Pasteur, University Hospital of Lille, Lille, France
| | - Florence Pinet
- University of Lille, Inserm U1167, Institut Pasteur, Lille, France
| |
Collapse
|
47
|
Latet SC, Van Herck PL, Claeys MJ, Van Craenenbroeck AH, Haine SE, Vandendriessche TR, Van Hoof VO, Fransen E, De Winter BY, Van Craenenbroeck EM, Heidbuchel H, Vrints CJ, Hoymans VY. Failed Downregulation of Circulating MicroRNA-155 in the Early Phase after ST Elevation Myocardial Infarction Is Associated with Adverse Left Ventricular Remodeling. Cardiology 2017; 138:91-96. [PMID: 28618412 DOI: 10.1159/000477235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/26/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND MicroRNA are noncoding RNA that have a significant role in both inflammatory and cardiovascular diseases. AIMS We aimed to assess whether the inflammation-related microRNA-155 is associated with the development of adverse left ventricular (LV) remodeling following ST elevation myocardial infarction (STEMI). METHODS Peripheral blood samples were collected in the inflammatory (day 2), proliferative (day 5), and maturation phases (6 months) after STEMI (n = 20). Granulocytes, monocytes, and lymphocytes were enumerated with flow cytometry. The changes in LV volumes were assessed with 3-D echocardiography on day 1 and after 6 months. Adverse remodeling was defined as a >20% increase in end-diastolic volume. Healthy subjects were recruited as controls. RESULTS MicroRNA-155 measured on day 5 correlated positively with the relative change in end-diastolic volume (ρ = 0.490, p = 0.028). MicroRNA-155 (day 5) was significantly higher in patients with compared to patients without adverse LV remodeling. The expression level was similar in healthy subjects (n = 8) and in patients with LV remodeling. There was a positive correlation between microRNA-155 and the amount of monocytes (day 5, ρ = 0.463, p = 0.046). CONCLUSION Impaired downregulation of microRNA-155 during the second phase of the post- STEMI inflammatory response is a determinant of the development of adverse LV remodeling.
Collapse
Affiliation(s)
- Sam C Latet
- Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hassell MECJ, Vlastra W, Robbers L, Hirsch A, Nijveldt R, Tijssen JGP, van Rossum AC, Zijlstra F, Piek JJ, Delewi R. Long-term left ventricular remodelling after revascularisation for ST-segment elevation myocardial infarction as assessed by cardiac magnetic resonance imaging. Open Heart 2017; 4:e000569. [PMID: 28861274 PMCID: PMC5577529 DOI: 10.1136/openhrt-2016-000569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/11/2017] [Accepted: 03/21/2017] [Indexed: 01/08/2023] Open
Abstract
Objective Left ventricular remodelling following a ST-segment elevated myocardial infarction (STEMI) is an adaptive response to maintain the cardiac output despite myocardial tissue loss. Limited studies have evaluated long term ventricular function using cardiac magnetic resonance imaging (CMR) after STEMI. Methods Study population consisted of 155 primary percutaneous coronary intervention treated first STEMI patients. CMR was performed at 4±2 days, 4 months and 24 months follow-up. Patients were treated with beta-blockers, ACE-inhibitors or AT-II- inhibitors, statins and dual antiplatelet according to current international guidelines. Results Mean left ventricular ejection fraction (LVEF) at baseline was 44%±8%. Twenty-one per cent of the study population had an increase of more than 5.0% after 4 months of follow-up and 21% of the cohort had a decrease of more than 5.0%. Patients with long-term LVEF deterioration have significantly larger end-systolic volumes than patients with improvement of LVEF (61±23 mL/m2 compared with 52±21 mL/m2, p=0.02) and less wall thickening in the remote zone. Patients with LVEF improvement had significantly greater improvement in wall thickening in the infarct areas and in the non-infarct or remote zone. Conclusion Contrary to previous studies, we demonstrate that myocardial remodelling after STEMI is a long-term process. Long-term LVEF deterioration is characterised by an increase in end-systolic volume and less wall thickening in the remote zones. Patients with LVEF improvement exhibit an increase in left ventricular wall thickening both in the infarct as well as in the remote zones. Trial registration The HEBE study is registered in The Netherlands Trial Register #NTR166 (www.trialregister.nl) and the International Standard Randomised Controlled Trial, #ISRCTN95796863 (https://c-d-qn9pqajji.sec.amc.nl).
Collapse
Affiliation(s)
- Mariella ECJ Hassell
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wieneke Vlastra
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lourens Robbers
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Alexander Hirsch
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Robin Nijveldt
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jan GP Tijssen
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Felix Zijlstra
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jan J Piek
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronak Delewi
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| |
Collapse
|
49
|
Lemesle G, Lamblin N, Meurice T, Tricot O, Bauters C. Angiotensin II receptor blockers versus angiotensin-converting enzyme inhibitors in patients with stable coronary artery disease: Prevalence, correlates, and prognostic impact (from the CORONOR study). J Cardiol 2017; 69:542-547. [DOI: 10.1016/j.jjcc.2016.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 11/28/2022]
|
50
|
Pinet F, Cuvelliez M, Kelder T, Amouyel P, Radonjic M, Bauters C. Integrative network analysis reveals time-dependent molecular events underlying left ventricular remodeling in post-myocardial infarction patients. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1445-1453. [PMID: 28167232 DOI: 10.1016/j.bbadis.2017.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/04/2017] [Accepted: 02/02/2017] [Indexed: 02/07/2023]
Abstract
To elucidate the time-resolved molecular events underlying the LV remodeling (LVR) process, we developed a large-scale network model that integrates the 24 molecular variables (plasma proteins and non-coding RNAs) collected in the REVE-2 study at four time points (baseline, 1month, 3months and 1year) after MI. The REVE-2 network model was built by extending the set of REVE-2 variables with their mechanistic context based on known molecular interactions (1310 nodes and 8639 edges). Changes in the molecular variables between the group of patients with high LVR (>20%) and low LVR (<20%) were used to identify active network modules within the clusters associated with progression of LVR, enabling assessment of time-resolved molecular changes. Although the majority of molecular changes occur at the baseline, two network modules specifically show an increasing number of active molecules throughout the post-MI follow up: one involved in muscle filament sliding, containing the major troponin forms and tropomyosin proteins, and the other associated with extracellular matrix disassembly, including matrix metalloproteinases, tissue inhibitors of metalloproteinases and laminin proteins. For the first time, integrative network analysis of molecular variables collected in REVE-2 patients with known molecular interactions allows insight into time-dependent mechanisms associated with LVR following MI, linking specific processes with LV structure alteration. In addition, the REVE-2 network model provides a shortlist of prioritized putative novel biomarker candidates for detection of LVR after MI event associated with a high risk of heart failure and is a valuable resource for further hypothesis generation.
Collapse
Affiliation(s)
- Florence Pinet
- Inserm, U1167, Univ. Lille, Institut Pasteur de Lille, FHU-REMOD-VHF, F-59000, Lille, France.
| | - Marie Cuvelliez
- Inserm, U1167, Univ. Lille, Institut Pasteur de Lille, FHU-REMOD-VHF, F-59000, Lille, France
| | | | - Philippe Amouyel
- Univ. Lille, Inserm, U1167, CHRU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | | | - Christophe Bauters
- Univ. Lille, Inserm, U1167, CHRU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| |
Collapse
|