1
|
Sen P, Hamers J, Sittig T, Shashikadze B, d'Ambrosio L, Stöckl JB, Bierschenk S, Zhang H, d'Alessio C, Zandbergen LM, Pauly V, Clauss S, Wolf E, Dendorfer A, Fröhlich T, Merkus D. Oxidative stress initiates hemodynamic change in CKD-induced heart disease. Basic Res Cardiol 2024; 119:957-971. [PMID: 39404904 PMCID: PMC11628585 DOI: 10.1007/s00395-024-01085-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
Chronic kidney disease (CKD) predisposes to cardiac remodeling and coronary microvascular dysfunction. Studies in swine identified changes in microvascular structure and function, as well as changes in mitochondrial structure and oxidative stress. However, CKD was combined with metabolic derangement, thereby obscuring the contribution of CKD alone. Therefore, we studied the impact of CKD on the heart and combined proteome studies with measurement of cardiac function and perfusion to identify processes involved in cardiac remodeling in CKD. CKD was induced in swine at 10-12 weeks of age while sham-operated swine served as controls. 5-6 months later, left ventricular (LV) function and coronary flow reserve were measured. LC-MS-MS-based proteomic analysis of LV tissue was performed. LV myocardium and kidneys were histologically examined for interstitial fibrosis and oxidative stress. Renal embolization resulted in mild chronic kidney injury (increased fibrosis and urinary NGAL). PV loops showed LV dilation and increased wall stress, while preload recruitable stroke work was impaired in CKD. Quantitative proteomic analysis of LV myocardium and STRING pre-ranked functional analysis showed enrichments in pathways related to contractile function, reactive oxygen species, and extracellular matrix (ECM) remodeling, which were confirmed histologically and associated with impaired total anti-oxidant capacity. H2O2 exposure of myocardial slices from CKD, but not normal swine, impaired contractile function. Furthermore, in CKD, mitochondrial proteins were downregulated suggesting mitochondrial dysfunction which was associated with higher basal coronary blood flow. Thus, mild CKD induces alterations in mitochondrial proteins along with contractile proteins, oxidative stress and ECM remodeling, that were associated with changes in cardiac function and perfusion.
Collapse
Affiliation(s)
- Payel Sen
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Jules Hamers
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Theresa Sittig
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Laura d'Ambrosio
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Susanne Bierschenk
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Hengliang Zhang
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
| | - Chiara d'Alessio
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
| | - Lotte M Zandbergen
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Valerie Pauly
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Department of Medicine I, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Department of Medicine I, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | | | - Andreas Dendorfer
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany
| | | | - Daphne Merkus
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, Marchioninistrasse 68, 81377, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany.
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Murai T, Hikita H, Yamaguchi M, Ito A, Warisawa T, Ikeda H, Takahashi K, Yano H, Chang J, Watanabe T, Yoshikawa H, Kanno Y, Hishikari K, Takahashi A, Fujii H, Yonetsu T, Sasano T, Kakuta T. Basal Coronary Microvascular Resistance Predicting Death and Heart Failure in Patients Without Functional Coronary Stenosis. Circ J 2024; 88:1788-1797. [PMID: 38897975 DOI: 10.1253/circj.cj-24-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND Abnormal coronary microcirculation is linked to poor patient prognosis, so the aim of the present study was to assess the prognostic relevance of basal microvascular resistance (b-IMR) in patients without functional coronary stenosis. METHODS AND RESULTS Analyses of 226 patients who underwent intracoronary physiological assessment of the left anterior descending artery included primary endpoints of all-cause death and heart failure, as well as secondary endpoints of cardiovascular death and atherosclerotic vascular events. During a median follow-up of 2 years, there were 12 (5.3%) primary and 21 (9.3 %) secondary endpoints. The optimal b-IMR cutoff for the primary endpoints was 47.1 U. Kaplan-Meier curve analysis demonstrated worse event-free survival of the primary endpoints in patients with a b-IMR below the cutoff (χ2=21.178, P<0.001). b-IMR was not significantly associated with the secondary endpoints (P=0.35). A low coronary flow reserve (CFR; <2.5) had prognostic value for both endpoints (primary endpoints: χ2=11.401, P=0.001; secondary endpoints: (χ2=6.015; P=0.014), and high hyperemic microvascular resistance (≥25) was associated only with the secondary endpoints (χ2=4.420; P=0.036). Incorporating b-IMR into a clinical model that included CFR improved the Net Reclassification Index and Integrated Discrimination Improvement for predicting the primary endpoints (P<0.001 and P=0.034, respectively). CONCLUSIONS b-IMR may be a specific marker of the risk of death and heart failure in patients without functional coronary stenosis.
Collapse
Affiliation(s)
| | | | | | - Aki Ito
- Cardiovascular Center, Yokosuka Kyosai Hospital
| | | | | | | | | | | | | | - Hiroshi Yoshikawa
- Department of Cardiovascular Medicine, Tokyo Medical Dental University Hospital
| | - Yoshinori Kanno
- Department of Cardiovascular Medicine, Tokyo Medical Dental University Hospital
| | | | | | - Hiroyuki Fujii
- Department of Cardiology, Yokohama Minami Kyosai Hospital
| | - Taishi Yonetsu
- Department of Cardiovascular Medicine, Tokyo Medical Dental University Hospital
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Tokyo Medical Dental University Hospital
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital
| |
Collapse
|
3
|
van Drie RWA, van de Wouw J, Zandbergen LM, Dehairs J, Swinnen JV, Mulder MT, Verhaar MC, MaassenVanDenBrink A, Duncker DJ, Sorop O, Merkus D. Vasodilator reactive oxygen species ameliorate perturbed myocardial oxygen delivery in exercising swine with multiple comorbidities. Basic Res Cardiol 2024; 119:869-887. [PMID: 38796544 PMCID: PMC11461570 DOI: 10.1007/s00395-024-01055-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/28/2024]
Abstract
Multiple common cardiovascular comorbidities produce coronary microvascular dysfunction. We previously observed in swine that a combination of diabetes mellitus (DM), high fat diet (HFD) and chronic kidney disease (CKD) induced systemic inflammation, increased oxidative stress and produced coronary endothelial dysfunction, altering control of coronary microvascular tone via loss of NO bioavailability, which was associated with an increase in circulating endothelin (ET). In the present study, we tested the hypotheses that (1) ROS scavenging and (2) ETA+B-receptor blockade improve myocardial oxygen delivery in the same female swine model. Healthy female swine on normal pig chow served as controls (Normal). Five months after induction of DM (streptozotocin, 3 × 50 mg kg-1 i.v.), hypercholesterolemia (HFD) and CKD (renal embolization), swine were chronically instrumented and studied at rest and during exercise. Sustained hyperglycemia, hypercholesterolemia and renal dysfunction were accompanied by systemic inflammation and oxidative stress. In vivo ROS scavenging (TEMPOL + MPG) reduced myocardial oxygen delivery in DM + HFD + CKD swine, suggestive of a vasodilator influence of endogenous ROS, while it had no effect in Normal swine. In vitro wire myography revealed a vasodilator role for hydrogen peroxide (H2O2) in isolated small coronary artery segments from DM + HFD + CKD, but not Normal swine. Increased catalase activity and ceramide production in left ventricular myocardial tissue of DM + HFD + CKD swine further suggest that increased H2O2 acts as vasodilator ROS in the coronary microvasculature. Despite elevated ET-1 plasma levels in DM + HFD + CKD swine, ETA+B blockade did not affect myocardial oxygen delivery in Normal or DM + HFD + CKD swine. In conclusion, loss of NO bioavailability due to 5 months exposure to multiple comorbidities is partially compensated by increased H2O2-mediated coronary vasodilation.
Collapse
Affiliation(s)
- R W A van Drie
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - L M Zandbergen
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377 LMU, Munich, Germany
| | - J Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
| | - J V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
| | - M T Mulder
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A MaassenVanDenBrink
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - D J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - O Sorop
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - D Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377 LMU, Munich, Germany.
- Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, 81377, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), University Clinic Munich, LMU, Munich, Germany.
| |
Collapse
|
4
|
Zhang M, Xu B, Li W, Yu B, Peng H, Gui F, Ai F, Chen Z. lncRNA CCAT2 Protects Against Cardiomyocyte Injury After Myocardial Ischemia/Reperfusion by Regulating BMI1 Expression. Int Heart J 2024; 65:279-291. [PMID: 38556336 DOI: 10.1536/ihj.23-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Myocardial ischemia/reperfusion (I/R) decreases cardiac function and efficiency. Accumulating evidence suggests that long noncoding RNAs (lncRNAs) have been linked to the cellular processes of myocardial I/R injury. The present investigation elucidated the function of lncRNA colon cancer-associated transcript 2 (CCAT2) in myocardial I/R injury and the related mechanisms.AC16 cardiomyocytes were exposed to hypoxia (16 hours) /reoxygenation (6 hours) (H/R) to mimic myocardial I/R models in vitro. CCAT2 and microRNA (miR) -539-3p expressions in AC16 cardiomyocytes were measured using real-time quantitative polymerase chain reaction. B-cell-specific Moloney murine leukemia virus insertion region 1 (BMI1) protein levels in AC16 cardiomyocytes were determined by western blotting. Cell viability, lactate dehydrogenase (LDH) leakage, reactive oxygen species (ROS) levels, mitochondrial membrane potential, and apoptosis were detected using Counting Kit-8, LDH Assay Kit, dihydroethidium assay, 5,5',6,6'-tetrachloro1,1',3,3'-tetramethylbenzimidazolylcarbocyanine iodide staining, flow cytometry, and western blotting, respectively. The interactions between the molecules were confirmed using the dual-luciferase gene reporter. The wingless/integrated/beta-catenin (Wnt/β-catenin) pathway under the H/R condition was detected by western blotting.CCAT2 and BMI1 mRNA expressions were reduced in H/R-exposed AC16 cardiomyocytes. CCAT2 overexpression exerted protective effects against H/R-induced cardiomyocyte injury, as demonstrated by increased cell viability and mitochondrial membrane potential and decreased LDH leakage, ROS levels, and apoptosis. In addition, CCAT2 positively regulated BMI1 expression by binding to miR-539-3p. CCAT2 knockdown or miR-539-3p overexpression restrained the protective effects of BMI1 against H/R-induced cardiomyocyte injury. In addition, miR-539-3p overexpression reversed the protective effects of CCAT2. Furthermore, CCAT2 activated the Wnt/β-catenin pathway under the H/R condition via the miR-539-3p/BMI1 axis.Overall, this investigation showed the protective effects of the CCAT2/miR-539-3p/BMI1/Wnt/β-catenin regulatory axis against cardiomyocyte injury induced by H/R.
Collapse
Affiliation(s)
- Mengli Zhang
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Bei Xu
- Department of Cardiovasology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Wei Li
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Bo Yu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Huan Peng
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
5
|
Yang N, Zhang R, Zhao C, Sun B, Wang B, Song Y, Qi S, Liu J, Sun Y, Liu H, An H, Zhang X, Xiang F, Yang Q, Han W. Association between Troponin Elevation and Decreased Myocardial Blood Flow Reserve in Patients without Obstructive Coronary Artery Disease. Cardiology 2023; 149:40-50. [PMID: 37944497 PMCID: PMC11251656 DOI: 10.1159/000534867] [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: 01/15/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION To study the prognostic factors of patients with chest pain and without obstructive coronary artery disease is of great significance for the management of such patients. We assessed whether a high-sensitivity troponin I (hs-TnI) is associated with prognosis in patients with chest pain and without obstructive coronary artery disease. METHODS From 2011 to 2017, 489 consecutively hospitalized patients with chest pain and without significant coronary artery stenosis (<50%) were tested for hs-TnI and underwent stress myocardial contrast echocardiography (MCE). Myocardial blood flow reserve (MBFR) was measured by stress MCE. Patients were followed (median, 41 months) for composite endpoints, including cardiovascular death and non-fatal myocardial infarction. Cox proportional hazards models were performed to determine associations between hs-TnI and the composite endpoints. RESULTS Among 489 patients with chest pain and without significant coronary artery stenosis, 257 patients (52.6%) had elevated hs-TnI. Compared to patients with normal hs-TnI, patients with elevated hs-TnI were older (p = 0.013) and had a higher prevalence of atrial fibrillation (p = 0.003), higher left ventricular mass index (p = 0.002) and E/e' septal (p < 0.001), and a lower MBFR (p < 0.001). After adjustment, there was still a significant association between hs-TnI and MBFR (odds ratio = 1.145; 95% confidence interval [CI], 1.079-1.214; p < 0.001). Compared with patients with normal hs-TnI, patients with elevated hs-TnI had a greater cumulative event rate (log-rank p = 0.002). Males (hazard ratio [HR], 4.770; 95% CI, 1.175-19.363; p = 0.029) and reduced MBFR (HR, 2.496; 95% CI, 1.446-4.311; p = 0.001) were risk factors associated with composite endpoints in patients with elevated hs-TnI. CONCLUSIONS In patients with chest pain and without obstructive coronary artery disease, elevated hs-TnI is associated with decreased myocardial perfusion by contrast echocardiography as well as a higher incidence of cardiovascular events.
Collapse
Affiliation(s)
- Ning Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Rongzhen Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Chaoqun Zhao
- Department of Cardiology, Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao, PR China
| | - Bochen Sun
- Department of Cardiology, Qingdao Third People’s Hospital, Qingdao, PR China
| | - Biyu Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Yuwei Song
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Shuhan Qi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Jianan Liu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yujia Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Hui Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Huanhuan An
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Xingyue Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Fei Xiang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Qinghui Yang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Wei Han
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, PR China
| |
Collapse
|
6
|
Bennett J, van Dinther M, Voorter P, Backes W, Barnes J, Barkhof F, Captur G, Hughes AD, Sudre C, Treibel TA. Assessment of Microvascular Disease in Heart and Brain by MRI: Application in Heart Failure with Preserved Ejection Fraction and Cerebral Small Vessel Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1596. [PMID: 37763715 PMCID: PMC10534635 DOI: 10.3390/medicina59091596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
The objective of this review is to investigate the commonalities of microvascular (small vessel) disease in heart failure with preserved ejection fraction (HFpEF) and cerebral small vessel disease (CSVD). Furthermore, the review aims to evaluate the current magnetic resonance imaging (MRI) diagnostic techniques for both conditions. By comparing the two conditions, this review seeks to identify potential opportunities to improve the understanding of both HFpEF and CSVD.
Collapse
Affiliation(s)
- Jonathan Bennett
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| | - Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
| | - Paulien Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Walter Backes
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Josephine Barnes
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
| | - Frederick Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije University, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
- Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
| | - Gabriella Captur
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- Centre for Inherited Heart Muscle Conditions, Cardiology Department, The Royal Free Hospital, London NW3 2QG, UK
| | - Alun D. Hughes
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
| | - Carole Sudre
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London WC2R 2LS, UK
| | - Thomas A. Treibel
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| |
Collapse
|
7
|
Tah S, Valderrama M, Afzal M, Iqbal J, Farooq A, Lak MA, Gostomczyk K, Jami E, Kumar M, Sundaram A, Sharifa M, Arain M. Heart Failure With Preserved Ejection Fraction: An Evolving Understanding. Cureus 2023; 15:e46152. [PMID: 37900404 PMCID: PMC10613100 DOI: 10.7759/cureus.46152] [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: 08/23/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a clinical syndrome in which patients have signs and symptoms of HF due to high left ventricular (LV) filling pressure despite normal or near normal LV ejection fraction. It is more common than HF with reduced ejection fraction (HFrEF), and its diagnosis and treatment are more challenging than HFrEF. Although hypertension is the primary risk factor, coronary artery disease and other comorbidities, such as atrial fibrillation (AF), diabetes, chronic kidney disease (CKD), and obesity, also play an essential role in its formation. This review summarizes current knowledge about HFpEF, its pathophysiology, clinical presentation, diagnostic challenges, current treatments, and promising novel treatments. It is essential to continue to be updated on the latest treatments for HFpEF so that patients always receive the most therapeutic treatments. The use of GnRH agonists in the management of HFpEF, infusion of Apo a-I nanoparticle, low-level transcutaneous vagal stimulation (LLTS), and estrogen only in post-menopausal women are promising strategies to prevent diastolic dysfunction and HFpEF; however, there is still no proven curative treatment for HFpEF yet.
Collapse
Affiliation(s)
- Sunanda Tah
- Surgery, Beckley Appalachian Regional Healthcare (ARH) Hospital, Beckley, USA
- Surgery, Saint James School of Medicine, Arnos Vale, VCT
| | | | - Maham Afzal
- Medicine, Fatima Jinnah Medical University, Lahore, PAK
| | | | - Aisha Farooq
- Internal Medicine, Dr. Ruth Pfau Hospital, Karachi, PAK
| | | | - Karol Gostomczyk
- Medicine, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, POL
| | - Elhama Jami
- Internal Medicine, Herat Regional Hospital, Herat, AFG
| | | | | | | | - Mustafa Arain
- Internal Medicine, Civil Hospital Karachi, Karachi, PAK
| |
Collapse
|
8
|
Toya T, Nagatomo Y, Ikegami Y, Masaki N, Adachi T. Coronary microvascular dysfunction in heart failure patients. Front Cardiovasc Med 2023; 10:1153994. [PMID: 37332583 PMCID: PMC10272355 DOI: 10.3389/fcvm.2023.1153994] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Coronary microcirculation has multiple layers of autoregulatory function to maintain resting flow and augment hyperemic flow in response to myocardial demands. Functional or structural alterations in the coronary microvascular function are frequently observed in patients with heart failure with preserved or reduced ejection fraction, which may lead to myocardial ischemic injury and resultant worsening of clinical outcomes. In this review, we describe our current understanding of coronary microvascular dysfunction in the pathogenesis of heart failure with preserved and reduced ejection fraction.
Collapse
|
9
|
Saavedra-Alvarez A, Pereyra KV, Toledo C, Iturriaga R, Del Rio R. Vascular dysfunction in HFpEF: Potential role in the development, maintenance, and progression of the disease. Front Cardiovasc Med 2022; 9:1070935. [PMID: 36620616 PMCID: PMC9810809 DOI: 10.3389/fcvm.2022.1070935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex, heterogeneous disease characterized by autonomic imbalance, cardiac remodeling, and diastolic dysfunction. One feature that has recently been linked to the pathology is the presence of macrovascular and microvascular dysfunction. Indeed, vascular dysfunction directly affects the functionality of cardiomyocytes, leading to decreased dilatation capacity and increased cell rigidity, which are the outcomes of the progressive decline in myocardial function. The presence of an inflammatory condition in HFpEF produced by an increase in proinflammatory molecules and activation of immune cells (i.e., chronic low-grade inflammation) has been proposed to play a pivotal role in vascular remodeling and endothelial cell death, which may ultimately lead to increased arterial elastance, decreased myocardium perfusion, and decreased oxygen supply to the tissue. Despite this, the precise mechanism linking low-grade inflammation to vascular alterations in the setting of HFpEF is not completely known. However, the enhanced sympathetic vasomotor tone in HFpEF, which may result from inflammatory activation of the sympathetic nervous system, could contribute to orchestrate vascular dysfunction in the setting of HFpEF due to the exquisite sympathetic innervation of both the macro and microvasculature. Accordingly, the present brief review aims to discuss the main mechanisms that may be involved in the macro- and microvascular function impairment in HFpEF and the potential role of the sympathetic nervous system in vascular dysfunction.
Collapse
Affiliation(s)
- Andrea Saavedra-Alvarez
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine V. Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile,*Correspondence: Rodrigo Del Rio
| |
Collapse
|
10
|
Tu Y, Li Q, Zhou Y, Ye Z, Wu C, Xie E, Li Y, Li P, Wu Y, Guo Z, Yu C, Zheng J, Gao Y. Empagliflozin inhibits coronary microvascular dysfunction and reduces cardiac pericyte loss in db/db mice. Front Cardiovasc Med 2022; 9:995216. [PMID: 36588571 PMCID: PMC9800791 DOI: 10.3389/fcvm.2022.995216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Background Coronary microvascular dysfunction (CMD) is a pathophysiological feature of diabetic heart disease. However, whether sodium-glucose cotransporter 2 (SGLT2) inhibitors protect the cardiovascular system by alleviating CMD is not known. Objective We observed the protective effects of empagliflozin (EMPA) on diabetic CMD. Materials and methods The mice were randomly divided into a db/db group and a db/db + EMPA group, and db/m mice served as controls. At 8 weeks of age, the db/db + EMPA group was given empagliflozin 10 mg/(kg⋅d) by gavage for 8 weeks. Body weight, fasting blood glucose and blood pressure were dynamically observed. Cardiac systolic and diastolic function and coronary flow reserve (CFR) were detected using echocardiography. The coronary microvascular structure and distribution of cardiac pericytes were observed using immunofluorescence staining. Picrosirius red staining was performed to evaluate cardiac fibrosis. Results Empagliflozin lowered the increased fasting blood glucose levels of the db/db group. The left ventricular ejection fraction, left ventricular fractional shortening, E/A ratio and E/e' ratio were not significantly different between the three groups. CFR was decreased in the db/db group, but EMPA significantly improved CFR. In contrast to the sparse and abnormal expansion of coronary microvessels observed in the db/db group, the number of coronary microvessels was increased, and the capillary diameter was decreased in the db/db + EMPA group. The number and microvascular coverage of cardiac pericytes were reduced in the db/db mice but were improved by EMPA. The cardiac fibrosis was increased in db/db group and may alleviate by EMPA. Conclusion Empagliflozin inhibited CMD and reduced cardiac pericyte loss in diabetic mice.
Collapse
Affiliation(s)
- Yimin Tu
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qing Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuanchen Zhou
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Zixiang Ye
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Chao Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Enmin Xie
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yike Li
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peizhao Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yaxin Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Ziyu Guo
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Changan Yu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
11
|
Huang S, Wang L, Li J, Ma L, He X, Zhang Q, Li Y. Oxygen pulse variation in symptomatic patients with suspected coronary artery disease: a diagnostic analysis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1225. [PMID: 36544671 PMCID: PMC9761160 DOI: 10.21037/atm-22-5279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Abstract
Background Cardiopulmonary exercise testing (CPET) has been found high sensitivity and specificity in cardiac ischemia. However, the role of CPET in coronary artery disease (CAD) is unclear. This study was to explore the diagnostic value of CPET indicators in CAD. Methods A total of 138 symptomatic patients with suspected CAD who underwent a CPET were included in this cross-sectional study. CPET indicators of all individuals were collected. ΔVO2/HR(Peak-AT) defined as the difference between the value of the oxygen consumption/heart rate (VO2/HR) at anaerobic threshold and peak exercise. The synergy between percutaneous coronary intervention with taxus and cardiac surgery (SYNTAX) score of all the CAD patients was calculated based on the complexity of the coronary lesions. The diagnostic performance of the CPET indicators was assessed by the area under the curve (AUC), sensitivity, and specificity. Results No significant differences in the CPET indicators were observed among the patients with or without CAD. The high SNYTAX score (≥22) group showed a significant reduction in the ΔVO2/HR(Peak-AT) compared to the low SNYTX score (<22) group (P=0.004). The AUC of the ΔVO2/HR(Peak-AT) was 0.804 (P=0.005), with the sensitivity of 95.7% and the specificity of 62.5%. The other CPET indicators did not differ significantly between the 2 groups. Oxygen pulse variation after the anaerobic threshold (AT) is superior to other CPET-derived variables in detecting intermediate to severe stenosis of the coronary artery in CAD patients. Conclusions The ΔVO2/HR(Peak-AT) is a quantitative indicator of the variation of the oxygen pulse response after the AT during incremental exercise. However, due to sample limitations, our results need to be interpreted with caution.
Collapse
Affiliation(s)
- Shangwei Huang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lijie Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jimin Li
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Ma
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan He
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Li
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Tune JD, Goodwill AG, Baker HE, Dick GM, Warne CM, Tucker SM, Essajee SI, Bailey CA, Klasing JA, Russell JJ, McCallinhart PE, Trask AJ, Bender SB. Chronic high-rate pacing induces heart failure with preserved ejection fraction-like phenotype in Ossabaw swine. Basic Res Cardiol 2022; 117:50. [PMID: 36222894 DOI: 10.1007/s00395-022-00958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 01/31/2023]
Abstract
The lack of pre-clinical large animal models of heart failure with preserved ejection fraction (HFpEF) remains a growing, yet unmet obstacle to improving understanding of this complex condition. We examined whether chronic cardiometabolic stress in Ossabaw swine, which possess a genetic propensity for obesity and cardiovascular complications, produces an HFpEF-like phenotype. Swine were fed standard chow (lean; n = 13) or an excess calorie, high-fat, high-fructose diet (obese; n = 16) for ~ 18 weeks with lean (n = 5) and obese (n = 8) swine subjected to right ventricular pacing (180 beats/min for ~ 4 weeks) to induce heart failure (HF). Baseline blood pressure, heart rate, LV end-diastolic volume, and ejection fraction were similar between groups. High-rate pacing increased LV end-diastolic pressure from ~ 11 ± 1 mmHg in lean and obese swine to ~ 26 ± 2 mmHg in lean HF and obese HF swine. Regression analyses revealed an upward shift in LV diastolic pressure vs. diastolic volume in paced swine that was associated with an ~ twofold increase in myocardial fibrosis and an ~ 50% reduction in myocardial capillary density. Hemodynamic responses to graded hemorrhage revealed an ~ 40% decrease in the chronotropic response to reductions in blood pressure in lean HF and obese HF swine without appreciable changes in myocardial oxygen delivery or transmural perfusion. These findings support that high-rate ventricular pacing of lean and obese Ossabaw swine initiates underlying cardiac remodeling accompanied by elevated LV filling pressures with normal ejection fraction. This distinct pre-clinical tool provides a unique platform for further mechanistic and therapeutic studies of this highly complex syndrome.
Collapse
Affiliation(s)
- Johnathan D Tune
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| | - Adam G Goodwill
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Hana E Baker
- Diabetes and Complications Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Gregory M Dick
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Cooper M Warne
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Selina M Tucker
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Salman I Essajee
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Chastidy A Bailey
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Jessica A Klasing
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Jacob J Russell
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Patricia E McCallinhart
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
13
|
Sechtem U, Seitz A, McChord J, Ong P. Ist die weibliche koronare Mikrovaskulatur anders? AKTUELLE KARDIOLOGIE 2022. [DOI: 10.1055/a-1631-4314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungViele Frauen mit Angina pectoris haben keine stenosierende koronare Herzerkrankung. Oft
liegen der Symptomatik koronare Funktionsstörungen zugrunde, die wiederum häufig die
koronare Mikrovaskulatur betreffen. Zu unterscheiden ist zwischen mikrovaskulären
Störungen der Vasodilatation bzw. der Vasokonstriktion, wobei letztere häufiger sind. Die
genaue Diagnose erfordert in aller Regel invasive Funktionstests mit Applikation von
Adenosin bzw. Acetylcholin. Die Testergebnisse erlauben die Einteilung in Endotypen, die
wiederum die Basis für eine angepasste und daher erfolgreiche medikamentöse Therapie sind.
Es wurde spekuliert, dass die weibliche Mikrovaskulatur anders aufgebaut sein bzw. anders
funktionieren könne als die männliche. Wahrscheinlich ist der wesentliche Unterschied aber
die schnellere und daher frühere Entwicklung epikardialer stenosierender Läsionen bei
Männern, bei denen dann nicht weiter nach dem Zustand der Mikrovaskulatur geschaut wird.
Dafür spricht die hohe Zahl von Patienten, die auch nach Koronarintervention weiter Angina
pectoris haben.
Collapse
Affiliation(s)
- Udo Sechtem
- Cardiologicum, Stuttgart, Deutschland
- Abteilung für Kardiologie und Angiologie,
Robert-Bosch-Krankenhaus GmbH, Stuttgart, Deutschland
| | - Andreas Seitz
- Abteilung für Kardiologie und Angiologie,
Robert-Bosch-Krankenhaus GmbH, Stuttgart, Deutschland
| | - Johanna McChord
- Abteilung für Kardiologie und Angiologie,
Robert-Bosch-Krankenhaus GmbH, Stuttgart, Deutschland
| | - Peter Ong
- Abteilung für Kardiologie und Angiologie,
Robert-Bosch-Krankenhaus GmbH, Stuttgart, Deutschland
| |
Collapse
|
14
|
Pahimi N, Rasool AHG, Sanip Z, Bokti NA, Yusof Z, W. Isa WYH. An Evaluation of the Role of Oxidative Stress in Non-Obstructive Coronary Artery Disease. J Cardiovasc Dev Dis 2022; 9:jcdd9020051. [PMID: 35200704 PMCID: PMC8878238 DOI: 10.3390/jcdd9020051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Approximately half of all women presenting to the emergency department with angina chest pain do not have obstructive coronary artery disease (CAD) on coronary angiography. This condition is termed non-obstructive coronary artery disease (NOCAD), and includes ischemia with no obstructive coronary artery disease (INOCA) and myocardial infarction with non-obstructive coronary arteries (MINOCA). Oxidative stress has been reported to be involved in the development and progression of CAD. However, a scarcity of studies has assessed a correlation between oxidative stress and NOCAD. Thus, a literature review was performed of available reports on the role of oxidative stress in NOCAD. Possible mechanisms involved in oxidative stress that may contribute to NOCAD were identified and evaluated. A key finding of this literature review was that oxidative stress caused vasoconstriction and endothelial damage, and this results in coronary microvascular dysfunction and vasospasm, which, in turn, lead to the pathogenesis of NOCAD.
Collapse
Affiliation(s)
- Nurnajwa Pahimi
- Pharmacology Vascular Laboratory, Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia; (N.P.); (A.H.G.R.)
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
| | - Aida Hanum Ghulam Rasool
- Pharmacology Vascular Laboratory, Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia; (N.P.); (A.H.G.R.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Zulkefli Sanip
- Central Research Laboratory, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Nur Adilah Bokti
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Zurkurnai Yusof
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - W. Yus Haniff W. Isa
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: ; Tel.: +60-9767-3987
| |
Collapse
|
15
|
Mechanism of the switch from NO to H 2O 2 in endothelium-dependent vasodilation in diabetes. Basic Res Cardiol 2022; 117:2. [PMID: 35024970 PMCID: PMC8886611 DOI: 10.1007/s00395-022-00910-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
Coronary microvascular dysfunction is prevalent among people with diabetes and is correlated with cardiac mortality. Compromised endothelial-dependent dilation (EDD) is an early event in the progression of diabetes, but its mechanisms remain incompletely understood. Nitric oxide (NO) is the major endothelium-dependent vasodilatory metabolite in the healthy coronary circulation, but this switches to hydrogen peroxide (H2O2) in coronary artery disease (CAD) patients. Because diabetes is a significant risk factor for CAD, we hypothesized that a similar NO-to-H2O2 switch would occur in diabetes. Vasodilation was measured ex vivo in isolated coronary arteries from wild type (WT) and microRNA-21 (miR-21) null mice on a chow or high-fat/high-sugar diet, and B6.BKS(D)-Leprdb/J (db/db) mice using myography. Myocardial blood flow (MBF), blood pressure, and heart rate were measured in vivo using contrast echocardiography and a solid-state pressure sensor catheter. RNA from coronary arteries, endothelial cells, and cardiac tissues was analyzed via quantitative real-time PCR for gene expression, and cardiac protein expression was assessed via western blot analyses. Superoxide was detected via electron paramagnetic resonance. (1) Ex vivo coronary EDD and in vivo MBF were impaired in diabetic mice. (2) Nω-Nitro-L-arginine methyl ester, an NO synthase inhibitor (L-NAME), inhibited ex vivo coronary EDD and in vivo MBF in WT. In contrast, polyethylene glycol-catalase, an H2O2 scavenger (Peg-Cat), inhibited diabetic mouse EDD ex vivo and MBF in vivo. (3) miR-21 was upregulated in diabetic mouse endothelial cells, and the deficiency of miR-21 prevented the NO-to-H2O2 switch and ameliorated diabetic mouse vasodilation impairments. (4) Diabetic mice displayed increased serum NO and H2O2, upregulated mRNA expression of Sod1, Sod2, iNos, and Cav1, and downregulated Pgc-1α in coronary arteries, but the deficiency of miR-21 reversed these changes. (5) miR-21-deficient mice exhibited increased cardiac PGC-1α, PPARα and eNOS protein and reduced endothelial superoxide. (6) Inhibition of PGC-1α changed the mRNA expression of genes regulated by miR-21, and overexpression of PGC-1α decreased the expression of miR-21 in high (25.5 mM) glucose treated coronary endothelial cells. Diabetic mice exhibit a NO-to-H2O2 switch in the mediator of coronary EDD, which contributes to microvascular dysfunction and is mediated by miR-21. This study represents the first mouse model recapitulating the NO-to-H2O2 switch seen in CAD patients in diabetes.
Collapse
|
16
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
17
|
Hu WH, Khoo MCK. Treatment of Cheyne-Stokes Respiration in Heart Failure with Adaptive Servo-Ventilation: An Integrative Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1384:79-103. [PMID: 36217080 DOI: 10.1007/978-3-031-06413-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The SERVE-HF (Treatment of Predominant Central Sleep Apnea by Adaptive Servo Ventilation in Patients with Heart Failure) multicenter trial found a small but significant increase in all-cause and cardiovascular mortality in patients assigned to adaptive servo-ventilation (ASV) versus guideline-based medical treatment. To better understand the physiological underpinnings of this clinical outcome, we employ an integrative computer model to simulate congestive heart failure with Cheyne-Stokes respiration (CHF-CSR) in subjects with a broad spectrum of underlying pathogenetic mechanisms, as well as to determine the in silico changes in cardiopulmonary and autonomic physiology resulting from ASV. Our simulation results demonstrate that while the elimination of CSR through ASV can partially restore cardiorespiratory and autonomic physiology toward normality in the vast majority of CHF phenotypes, the degree of restoration can be highly variable, depending on the combination of CHF mechanisms in play. The group with the lowest left ventricular ejection fraction (LVEF) appears to be most vulnerable to the potentially adverse effects of ASV, but the level of pulmonary capillary wedge pressure (PCWP) plays an important role in determining the nature of these effects.
Collapse
|
18
|
Sinha A, Rahman H, Webb A, Shah AM, Perera D. Untangling the pathophysiologic link between coronary microvascular dysfunction and heart failure with preserved ejection fraction. Eur Heart J 2021; 42:4431-4441. [PMID: 34529791 PMCID: PMC8599060 DOI: 10.1093/eurheartj/ehab653] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/10/2021] [Accepted: 09/03/2021] [Indexed: 01/03/2023] Open
Abstract
Coronary microvascular disease (CMD), characterized by impaired coronary flow reserve (CFR), is a common finding in patients with stable angina. Impaired CFR, in the absence of obstructive coronary artery disease, is also present in up to 75% of patients with heart failure with preserved ejection fraction (HFpEF). Heart failure with preserved ejection fraction is a heterogeneous syndrome comprising distinct endotypes and it has been hypothesized that CMD lies at the centre of the pathogenesis of one such entity: the CMD–HFpEF endotype. This article provides a contemporary review of the pathophysiology underlying CMD, with a focus on the mechanistic link between CMD and HFpEF. We discuss the central role played by subendocardial ischaemia and impaired lusitropy in the development of CMD–HFpEF, as well as the clinical and research implications of the CMD–HFpEF mechanistic link. Future prospective follow-up studies detailing outcomes in patients with CMD and HFpEF are much needed to enhance our understanding of the pathological processes driving these conditions, which may lead to the development of physiology-stratified therapy to improve the quality of life and prognosis in these patients.
Collapse
Affiliation(s)
- Aish Sinha
- British Heart Foundation Centre of Excellence and National Institute for Health Research Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, St. Thomas' Hospital, Westminster bridge road, London SE1 7EH, UK
| | - Haseeb Rahman
- British Heart Foundation Centre of Excellence and National Institute for Health Research Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, St. Thomas' Hospital, Westminster bridge road, London SE1 7EH, UK
| | - Andrew Webb
- British Heart Foundation Centre of Excellence and National Institute for Health Research Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, St. Thomas' Hospital, Westminster bridge road, London SE1 7EH, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Excellence and National Institute for Health Research Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, St. Thomas' Hospital, Westminster bridge road, London SE1 7EH, UK
| | - Divaka Perera
- British Heart Foundation Centre of Excellence and National Institute for Health Research Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, St. Thomas' Hospital, Westminster bridge road, London SE1 7EH, UK
| |
Collapse
|
19
|
van de Wouw J, Steenhorst JJ, Sorop O, van Drie RWA, Wielopolski PA, Kleinjan A, Hirsch A, Duncker DJ, Merkus D. Impaired pulmonary vasomotor control in exercising swine with multiple comorbidities. Basic Res Cardiol 2021; 116:51. [PMID: 34510273 PMCID: PMC8435524 DOI: 10.1007/s00395-021-00891-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/29/2021] [Indexed: 12/15/2022]
Abstract
Pulmonary hypertension is common in heart failure with preserved ejection fraction (HFpEF). Here, we tested the hypothesis that comorbidities [diabetes mellitus (DM, streptozotocin), hypercholesterolemia (HC, high-fat diet) and chronic kidney disease (CKD, renal microembolization)] directly impair pulmonary vasomotor control in a DM + HC + CKD swine model. 6 months after induction of DM + HC + CKD, pulmonary arterial pressure was similar in chronically instrumented female DM + HC + CKD (n = 19) and Healthy swine (n = 18). However, cardiac output was lower both at rest and during exercise, implying an elevated pulmonary vascular resistance (PVR) in DM + HC + CKD swine (153 ± 10 vs. 122 ± 9 mmHg∙L-1∙min∙kg). Phosphodiesterase 5 inhibition and endothelin receptor antagonism decreased PVR in DM + HC + CKD (- 12 ± 12 and - 22 ± 7 mmHg∙L-1∙min∙kg) but not in Healthy swine (- 1 ± 12 and 2 ± 14 mmHg∙L-1∙min∙kg), indicating increased vasoconstrictor influences of phosphodiesterase 5 and endothelin. Inhibition of nitric oxide synthase produced pulmonary vasoconstriction that was similar in Healthy and DM + HC + CKD swine, but unmasked a pulmonary vasodilator effect of endothelin receptor antagonism in Healthy (- 56 ± 26 mmHg∙L-1∙min∙kg), whereas it failed to significantly decrease PVR in DM + HC + CKD, indicating loss of nitric oxide mediated inhibition of endothelin in DM + HC + CKD. Scavenging of reactive oxygen species (ROS) had no effect on PVR in either Healthy or DM + HC + CKD swine. Cardiovascular magnetic resonance imaging, under anesthesia, showed no right ventricular changes. Finally, despite an increased contribution of endogenous nitric oxide to vasomotor tone regulation in the systemic vasculature, systemic vascular resistance at rest was higher in DM + HC + CKD compared to Healthy swine (824 ± 41 vs. 698 ± 35 mmHg∙L-1∙min∙kg). ROS scavenging induced systemic vasodilation in DM + HC + CKD, but not Healthy swine. In conclusion, common comorbidities directly alter pulmonary vascular control, by enhanced PDE5 and endothelin-mediated vasoconstrictor influences, well before overt left ventricular backward failure or pulmonary hypertension develop.
Collapse
Affiliation(s)
- Jens van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Jarno J Steenhorst
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Ruben W A van Drie
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Piotr A Wielopolski
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alex Kleinjan
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alexander Hirsch
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Institute for Surgical Research, Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, LMU Munich, Munich, Germany.
- German Center for Cardiovascular Research, Partner Site Munich, Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
20
|
Hou X, Li L, Chen S, Ge C, Shen M, Fu Z. MKP-1 Overexpression Reduces Postischemic Myocardial Damage through Attenuation of ER Stress and Mitochondrial Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8905578. [PMID: 34512872 PMCID: PMC8433005 DOI: 10.1155/2021/8905578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022]
Abstract
Mitochondrial dysfunction and endoplasmic reticulum (ER) stress contribute to postischemic myocardial damage, but the upstream regulatory mechanisms have not been identified. In this study, we analyzed the role of mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) in the regulation of mitochondrial function and ER stress in hypoxic cardiomyocytes. Our results show that MKP-1 overexpression sustains viability and reduces hypoxia-induced apoptosis among H9C2 cardiomyocytes. MKP-1 overexpression attenuates ER stress and expression of ER stress genes and improves mitochondrial function in hypoxia-treated H9C2 cells. MKP-1 overexpression also increases ATP production and mitochondrial respiration and attenuates mitochondrial oxidative damage in hypoxic cardiomyocytes. Moreover, our results demonstrate that ERK and JNK are the downstream signaling targets of MKP-1 and that MKP-1 overexpression activates ERK, while it inhibits JNK. Inhibition of ERK reduces the ability of MKP-1 to preserve mitochondrial function and ER homeostasis in hypoxic cardiomyocytes. These results show that MKP-1 plays an essential role in the regulation of mitochondrial function and ER stress in hypoxic H9C2 cardiomyocytes through normalization of the ERK pathway and suggest that MKP-1 may serve as a novel target for the treatment of postischemic myocardial injury.
Collapse
Affiliation(s)
- Xiaoling Hou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Lijun Li
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Si Chen
- Department of Cardiology, The First Medical Center of People's Liberation Army General Hospital, China
| | - Cheng Ge
- Department of Cardiology, The First Medical Center of People's Liberation Army General Hospital, China
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan 572013, China
| | - Zhenhong Fu
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
21
|
van de Wouw J, Sorop O, van Drie RWA, Joles JA, Danser AHJ, Verhaar MC, Merkus D, Duncker DJ. Reduced nitric oxide bioavailability impairs myocardial oxygen balance during exercise in swine with multiple risk factors. Basic Res Cardiol 2021; 116:50. [PMID: 34435256 PMCID: PMC8387273 DOI: 10.1007/s00395-021-00890-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/20/2021] [Indexed: 10/28/2022]
Abstract
In the present study, we tested the hypothesis that multiple risk factors, including diabetes mellitus (DM), dyslipidaemia and chronic kidney disease (CKD) result in a loss of nitric oxide (NO) signalling, thereby contributing to coronary microvascular dysfunction. Risk factors were induced in 12 female swine by intravenous streptozotocin injections (DM), a high fat diet (HFD) and renal artery embolization (CKD). Female healthy swine (n = 13) on normal diet served as controls (Normal). After 5 months, swine were chronically instrumented and studied at rest and during exercise. DM + HFD + CKD swine demonstrated significant hyperglycaemia, dyslipidaemia and impaired kidney function compared to Normal swine. These risk factors were accompanied by coronary microvascular endothelial dysfunction both in vivo and in isolated small arteries, due to a reduced NO bioavailability, associated with perturbations in myocardial oxygen balance at rest and during exercise. NO synthase inhibition caused coronary microvascular constriction in exercising Normal swine, but had no effect in DM + HFD + CKD animals, while inhibition of phosphodiesterase 5 produced similar vasodilator responses in both groups, indicating that loss of NO bioavailability was principally responsible for the observed coronary microvascular dysfunction. This was associated with an increase in myocardial 8-isoprostane levels and a decrease in antioxidant capacity, while antioxidants restored the vasodilation to bradykinin in isolated coronary small arteries, suggesting that oxidative stress was principally responsible for the reduced NO bioavailability. In conclusion, five months of combined exposure to DM + HFD + CKD produces coronary endothelial dysfunction due to impaired NO bioavailability, resulting in impaired myocardial perfusion at rest and during exercise.
Collapse
Affiliation(s)
- Jens van de Wouw
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Oana Sorop
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Ruben W A van Drie
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377, LMU Munich, Germany.,German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, 81377, Munich, Germany
| | - Dirk J Duncker
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands.
| |
Collapse
|
22
|
LATS2 Deletion Attenuates Myocardial Ischemia-Reperfusion Injury by Promoting Mitochondrial Biogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1058872. [PMID: 34457109 PMCID: PMC8390173 DOI: 10.1155/2021/1058872] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/25/2021] [Accepted: 08/07/2021] [Indexed: 02/03/2023]
Abstract
Reperfusion therapy is the most effective treatment for acute myocardial infarction, but it can damage cardiomyocytes through a mechanism known as myocardial ischemia/reperfusion injury (MIRI). In this study, we investigated whether the large tumor suppressor kinase 2 (LATS2) contributes to the development of myocardial MIRI by disrupting mitochondrial biogenesis. Our in vitro data demonstrate that cardiomyocyte viability was reduced and apoptosis was increased in response to hypoxia/reoxygenation (H/R) injury. However, suppression of LATS2 by shRNA sustained cardiomyocyte viability by maintaining mitochondrial function. Compared to H/R-treated control cardiomyocytes, cardiomyocytes transfected with LATS2 shRNA exhibited increased mitochondrial respiration, improved mitochondrial ATP generation, and more stable mitochondrial membrane potential. LATS2 suppression increased cardiomyocyte viability and mitochondrial biogenesis in a manner dependent on PGC1α, a key regulator of mitochondrial metabolism. These results identify LATS2 as a new inducer of mitochondrial damage and myocardial MIRI and suggest that approaches targeting LATS2 or mitochondrial biogenesis may be beneficial in the clinical management of cardiac MIRI.
Collapse
|
23
|
Myocardial Tissue Characterization in Heart Failure with Preserved Ejection Fraction: From Histopathology and Cardiac Magnetic Resonance Findings to Therapeutic Targets. Int J Mol Sci 2021; 22:ijms22147650. [PMID: 34299270 PMCID: PMC8304780 DOI: 10.3390/ijms22147650] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome responsible for high mortality and morbidity rates. It has an ever growing social and economic impact and a deeper knowledge of molecular and pathophysiological basis is essential for the ideal management of HFpEF patients. The association between HFpEF and traditional cardiovascular risk factors is known. However, myocardial alterations, as well as pathophysiological mechanisms involved are not completely defined. Under the definition of HFpEF there is a wide spectrum of different myocardial structural alterations. Myocardial hypertrophy and fibrosis, coronary microvascular dysfunction, oxidative stress and inflammation are only some of the main pathological detectable processes. Furthermore, there is a lack of effective pharmacological targets to improve HFpEF patients' outcomes and risk factors control is the primary and unique approach to treat those patients. Myocardial tissue characterization, through invasive and non-invasive techniques, such as endomyocardial biopsy and cardiac magnetic resonance respectively, may represent the starting point to understand the genetic, molecular and pathophysiological mechanisms underlying this complex syndrome. The correlation between histopathological findings and imaging aspects may be the future challenge for the earlier and large-scale HFpEF diagnosis, in order to plan a specific and effective treatment able to modify the disease's natural course.
Collapse
|
24
|
Jiang X, Wu D, Jiang Z, Ling W, Qian G. Protective Effect of Nicorandil on Cardiac Microvascular Injury: Role of Mitochondrial Integrity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4665632. [PMID: 34285763 PMCID: PMC8275446 DOI: 10.1155/2021/4665632] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/28/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023]
Abstract
A major shortcoming of postischemic therapy for myocardial infarction is the no-reflow phenomenon due to impaired cardiac microvascular function including microcirculatory barrier function, loss of endothelial activity, local inflammatory cell accumulation, and increased oxidative stress. Consequently, inadequate reperfusion of the microcirculation causes secondary ischemia, aggravating the myocardial reperfusion injury. ATP-sensitive potassium ion (KATP) channels regulate the coronary blood flow and protect cardiomyocytes from ischemia-reperfusion injury. Studies in animal models of myocardial ischemia-reperfusion have illustrated that the opening of mitochondrial KATP (mito-KATP) channels alleviates endothelial dysfunction and reduces myocardial necrosis. By contrast, blocking mito-KATP channels aggravates microvascular necrosis and no-reflow phenomenon following ischemia-reperfusion injury. Nicorandil, as an antianginal drug, has been used for ischemic preconditioning (IPC) due to its mito-KATP channel-opening effect, thereby limiting infarct size and subsequent severe ischemic insult. In this review, we analyze the protective actions of nicorandil against microcirculation reperfusion injury with a focus on improving mitochondrial integrity. In addition, we discuss the function of mitochondria in the pathogenesis of myocardial ischemia.
Collapse
Affiliation(s)
- Xiaosi Jiang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Dan Wu
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zichao Jiang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Weiwei Ling
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Ji H, Wu D, Kimberlee O, Li R, Qian G. Molecular Perspectives of Mitophagy in Myocardial Stress: Pathophysiology and Therapeutic Targets. Front Physiol 2021; 12:700585. [PMID: 34276422 PMCID: PMC8279814 DOI: 10.3389/fphys.2021.700585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
A variety of complex risk factors and pathological mechanisms contribute to myocardial stress, which ultimately promotes the development of cardiovascular diseases, including acute cardiac insufficiency, myocardial ischemia, myocardial infarction, high-glycemic myocardial injury, and acute alcoholic cardiotoxicity. Myocardial stress is characterized by abnormal metabolism, excessive reactive oxygen species production, an insufficient energy supply, endoplasmic reticulum stress, mitochondrial damage, and apoptosis. Mitochondria, the main organelles contributing to the energy supply of cardiomyocytes, are key determinants of cell survival and death. Mitophagy is important for cardiomyocyte function and metabolism because it removes damaged and aged mitochondria in a timely manner, thereby maintaining the proper number of normal mitochondria. In this review, we first introduce the general characteristics and regulatory mechanisms of mitophagy. We then describe the three classic mitophagy regulatory pathways and their involvement in myocardial stress. Finally, we discuss the two completely opposite effects of mitophagy on the fate of cardiomyocytes. Our summary of the molecular pathways underlying mitophagy in myocardial stress may provide therapeutic targets for myocardial protection interventions.
Collapse
Affiliation(s)
- Haizhe Ji
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - O'Maley Kimberlee
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Ruibing Li
- Department of Clinical Laboratory Medicine, The First Medical Center, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
26
|
Wang Y, Jasper H, Toan S, Muid D, Chang X, Zhou H. Mitophagy coordinates the mitochondrial unfolded protein response to attenuate inflammation-mediated myocardial injury. Redox Biol 2021; 45:102049. [PMID: 34174558 PMCID: PMC8246635 DOI: 10.1016/j.redox.2021.102049] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction is a fundamental challenge in septic cardiomyopathy. Mitophagy and the mitochondrial unfolded protein response (UPRmt) are the predominant stress-responsive and protective mechanisms involved in repairing damaged mitochondria. Although mitochondrial homeostasis requires the coordinated actions of mitophagy and UPRmt, their molecular basis and interactive actions are poorly understood in sepsis-induced myocardial injury. Our investigations showed that lipopolysaccharide (LPS)-induced sepsis contributed to cardiac dysfunction and mitochondrial damage. Although both mitophagy and UPRmt were slightly activated by LPS in cardiomyocytes, their endogenous activation failed to prevent sepsis-mediated myocardial injury. However, administration of urolithin A, an inducer of mitophagy, obviously reduced sepsis-mediated cardiac depression by normalizing mitochondrial function. Interestingly, this beneficial action was undetectable in cardiomyocyte-specific FUNDC1 knockout (FUNDC1CKO) mice. Notably, supplementation with a mitophagy inducer had no impact on UPRmt, whereas genetic ablation of FUNDC1 significantly upregulated the expression of genes related to UPRmt in LPS-treated hearts. In contrast, enhancement of endogenous UPRmt through oligomycin administration reduced sepsis-mediated mitochondrial injury and myocardial dysfunction; this cardioprotective effect was imperceptible in FUNDC1CKO mice. Lastly, once UPRmt was inhibited, mitophagy-mediated protection of mitochondria and cardiomyocytes was partly blunted. Taken together, it is plausible that endogenous UPRmt and mitophagy are slightly activated by myocardial stress and they work together to sustain mitochondrial performance and cardiac function. Endogenous UPRmt, a downstream signal of mitophagy, played a compensatory role in maintaining mitochondrial homeostasis in the case of mitophagy inhibition. Although UPRmt activation had no negative impact on mitophagy, UPRmt inhibition compromised the partial cardioprotective actions of mitophagy. This study shows how mitophagy modulates UPRmt to attenuate inflammation-related myocardial injury and suggests the potential application of mitophagy and UPRmt targeting in the treatment of myocardial stress. Mitochondrial dysfunction is a fundamental challenge in septic cardiomyopathy. LPS-induced sepsis contributes to cardiac dysfunction and mitochondrial damage. Endogenous UPRmt and mitophagy could be slightly activated by myocardial stress. Mitophagy modulates UPRmt to attenuate inflammation-related myocardial injury. Mitophagy and UPRmt targeting can be applied in treatment of myocardial stress.
Collapse
Affiliation(s)
- Yue Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Heinrich Jasper
- Center for Molecular Medicine, Tarrant County College, TX, 76102, USA
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN, 55812, USA
| | - David Muid
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xing Chang
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, 100053, China.
| | - Hao Zhou
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, 100853, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
| |
Collapse
|
27
|
Atiq F, van de Wouw J, Sorop O, Heinonen I, de Maat MPM, Merkus D, Duncker DJ, Leebeek FWG. Endothelial Dysfunction, Atherosclerosis, and Increase of von Willebrand Factor and Factor VIII: A Randomized Controlled Trial in Swine. Thromb Haemost 2021; 121:676-686. [PMID: 33506473 DOI: 10.1055/s-0040-1722185] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is well known that high von Willebrand factor (VWF) and factor VIII (FVIII) levels are associated with an increased risk of cardiovascular disease. It is still debated whether VWF and FVIII are biomarkers of endothelial dysfunction and atherosclerosis or whether they have a direct causative role. Therefore, we aimed to unravel the pathophysiological pathways of increased VWF and FVIII levels associated with cardiovascular risk factors. First, we performed a randomized controlled trial in 34 Göttingen miniswine. Diabetes mellitus (DM) was induced with streptozotocin and hypercholesterolemia (HC) via a high-fat diet in 18 swine (DM + HC), while 16 healthy swine served as controls. After 5 months of follow-up, FVIII activity (FVIII:C) was significantly higher in DM + HC swine (5.85 IU/mL [5.00-6.81]) compared with controls (4.57 [3.76-5.40], p = 0.010), whereas VWF antigen (VWF:Ag) was similar (respectively 0.34 IU/mL [0.28-0.39] vs. 0.34 [0.31-0.38], p = 0.644). DM + HC swine had no endothelial dysfunction or atherosclerosis during this short-term follow-up. Subsequently, we performed a long-term (15 months) longitudinal cohort study in 10 Landrace-Yorkshire swine, in five of which HC and in five combined DM + HC were induced. VWF:Ag was higher at 15 months compared with 9 months in HC (0.37 [0.32-0.42] vs. 0.27 [0.23-0.40], p = 0.042) and DM + HC (0.33 [0.32-0.37] vs. 0.25 [0.24-0.33], p = 0.042). Both long-term groups had endothelial dysfunction compared with controls and atherosclerosis after 15 months. In conclusion, short-term hyperglycemia and dyslipidemia increase FVIII, independent of VWF. Long-term DM and HC increase VWF via endothelial dysfunction and atherosclerosis. Therefore, VWF seems to be a biomarker for advanced cardiovascular disease.
Collapse
Affiliation(s)
- Ferdows Atiq
- Department of Hematology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jens van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ilkka Heinonen
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Rydberg Laboratory of Applied Sciences, University of Halmstad, Halmstad, Sweden
| | - Moniek P M de Maat
- Department of Hematology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
Pigs represent a potentially attractive model for medical research. Similar body size and physiological patterns of kidney injury that more closely mimic those described in humans make larger animals attractive for experimentation. Using larger animals, including pigs, to investigate the pathogenesis of acute kidney injury (AKI) also serves as an experimental bridge, narrowing the gap between clinical disease and preclinical discoveries. This article compares the advantages and disadvantages of large versus small AKI animal models and provides a comprehensive overview of the development and application of porcine models of AKI induced by clinically relevant insults, including ischemia-reperfusion, sepsis, and nephrotoxin exposure. The primary focus of this review is to evaluate the use of pigs for AKI studies by current investigators, including areas where more information is needed.
Collapse
Affiliation(s)
- Jianni Huang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - George Bayliss
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| |
Collapse
|
29
|
Sinha A, Rahman H, Perera D. Coronary microvascular disease: current concepts of pathophysiology, diagnosis and management. Cardiovasc Endocrinol Metab 2021; 10:22-30. [PMID: 33634252 PMCID: PMC7901821 DOI: 10.1097/xce.0000000000000223] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022]
Abstract
Coronary microvascular disease (CMD) is present in 30% of patients with angina and is associated with increased morbidity and mortality. We now have an improved understanding of the pathophysiology of CMD and the invasive and noninvasive tests that can be used to make the diagnosis. Recent studies have shown that management of CMD guided by physiological testing yields better results than empirical treatment. Despite major advances in diagnosing and stratifying this condition, therapeutic strategies remain limited and poorly defined. This review article discusses recent advances in understanding the pathophysiology of CMD, the modalities that are available to diagnose it clinically, current management options and a look at what is in store for the future.
Collapse
Affiliation(s)
- Aish Sinha
- The BHF Centre of Excellence and the NIHR Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Haseeb Rahman
- The BHF Centre of Excellence and the NIHR Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Divaka Perera
- The BHF Centre of Excellence and the NIHR Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| |
Collapse
|
30
|
Beck S, Pereyra VM, Seitz A, McChord J, Hubert A, Bekeredjian R, Sechtem U, Ong P. Invasive Diagnosis of Coronary Functional Disorders Causing Angina Pectoris. Eur Cardiol 2021; 16:e27. [PMID: 34276812 PMCID: PMC8280748 DOI: 10.15420/ecr.2021.06] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/26/2021] [Indexed: 01/16/2023] Open
Abstract
Coronary vasomotion disorders represent a frequent cause of angina and/or dyspnoea in patients with non-obstructed coronary arteries. The highly sophisticated interplay of vasodilatation and vasoconstriction can be assessed in an interventional diagnostic procedure. Established parameters characterising adequate vasodilatation are coronary blood flow at rest, and, after drug-induced vasodilation, coronary flow reserve, and microvascular resistance (hyperaemic microvascular resistance, index of microcirculatory resistance). An increased vasoconstrictive potential is diagnosed by provocation testing with acetylcholine or ergonovine. This enables a diagnosis of coronary epicardial and/or microvascular spasm. Ischaemia associated with microvascular spasm can be confirmed by ischaemic ECG changes and the measurement of lactate concentrations in the coronary sinus. Although interventional diagnostic procedures are helpful for determining the mechanism of the angina, which may be the key to successful medical treatment, they are still neither widely accepted nor applied in many medical centres. This article summarises currently well-established invasive methods for the diagnosis of coronary functional disorders causing angina pectoris.
Collapse
Affiliation(s)
- Sascha Beck
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | | | - Andreas Seitz
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Johanna McChord
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Astrid Hubert
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Raffi Bekeredjian
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Udo Sechtem
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| | - Peter Ong
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus Stuttgart, Germany
| |
Collapse
|
31
|
Sinha A, Rahman H, Perera D. Ischaemia without obstructive coronary artery disease: the pathophysiology of microvascular dysfunction. Curr Opin Cardiol 2020; 35:720-725. [PMID: 32852345 DOI: 10.1097/hco.0000000000000788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE OF REVIEW Nearly one-third of patients presenting with angina have unobstructed epicardial coronary arteries and evidence of coronary microvascular disease. Up until recently, the pathophysiology of coronary microvascular disease has been poorly understood, resulting in limited effective therapeutic options in these patients. As a result, patients with coronary microvascular disease continue to suffer from a poor quality of life and adverse cardiovascular outcomes. RECENT FINDINGS Recent mechanistic studies have improved our understanding of the pathophysiology underlying coronary microvascular dysfunction; these studies have implicated the nitric oxide and endothelin pathways as the main drivers. The aim of this article is to review our current understanding of the pathophysiology of ischaemia in patients with coronary microvascular disease. SUMMARY Patients with angina who have coronary microvascular disease, but no obstructive coronary artery disease, are unable to augment their coronary blood flow in response to physiological stress, thereby predisposing them to myocardial ischaemia as a result of supply:demand mismatch in the myocardium. In addition to abnormalities of vascular resistance, perturbations in cardiac-coronary coupling also contribute to ischaemia in these patients. Although impaired flow reserve is the diagnostic hallmark, mechanistic studies have demonstrated that the underlying pathophysiology is heterogeneous. At present, two main endotypes have been identified, which can be readily differentiated on the basis of minimal microvascular resistance. A better understanding of the pathophysiology and mechanisms driving ischaemia in coronary microvascular dysfunction may stimulate the development of individualised therapies that may lead to an improvement in patients' quality of life and prognosis.
Collapse
Affiliation(s)
- Aish Sinha
- The BHF Centre of Excellence and the NIHR Biomedical Research Centre at the School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | | | | |
Collapse
|
32
|
Keijzer LBH, Caenen A, Voorneveld J, Strachinaru M, Bowen DJ, van de Wouw J, Sorop O, Merkus D, Duncker DJ, van der Steen AFW, de Jong N, Bosch JG, Vos HJ. A direct comparison of natural and acoustic-radiation-force-induced cardiac mechanical waves. Sci Rep 2020; 10:18431. [PMID: 33116234 PMCID: PMC7595170 DOI: 10.1038/s41598-020-75401-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
Natural and active shear wave elastography (SWE) are potential ultrasound-based techniques to non-invasively assess myocardial stiffness, which could improve current diagnosis of heart failure. This study aims to bridge the knowledge gap between both techniques and discuss their respective impacts on cardiac stiffness evaluation. We recorded the mechanical waves occurring after aortic and mitral valve closure (AVC, MVC) and those induced by acoustic radiation force throughout the cardiac cycle in four pigs after sternotomy. Natural SWE showed a higher feasibility than active SWE, which is an advantage for clinical application. Median propagation speeds of 2.5-4.0 m/s and 1.6-4.0 m/s were obtained after AVC and MVC, whereas ARF-based median speeds of 0.9-1.2 m/s and 2.1-3.8 m/s were reported for diastole and systole, respectively. The different wave characteristics in both methods, such as the frequency content, complicate the direct comparison of waves. Nevertheless, a good match was found in propagation speeds between natural and active SWE at the moment of valve closure, and the natural waves showed higher propagation speeds than in diastole. Furthermore, the results demonstrated that the natural waves occur in between diastole and systole identified with active SWE, and thus represent a myocardial stiffness in between relaxation and contraction.
Collapse
Affiliation(s)
- Lana B H Keijzer
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.
| | - Annette Caenen
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.
- IBiTech-bioMMeda, Ghent University, Ghent, Belgium.
- Cardiovascular Imaging and Dynamics Lab, Catholic University of Leuven, Leuven, Belgium.
| | - Jason Voorneveld
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Daniel J Bowen
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jens van de Wouw
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Oana Sorop
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Daphne Merkus
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Antonius F W van der Steen
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Nico de Jong
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Johan G Bosch
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Hendrik J Vos
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
33
|
Zou H, Liu G. Inhibition of endoplasmic reticulum stress through activation of MAPK/ERK signaling pathway attenuates hypoxia-mediated cardiomyocyte damage. J Recept Signal Transduct Res 2020; 41:532-537. [PMID: 33023351 DOI: 10.1080/10799893.2020.1831534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Huanxue Zou
- Department of Cardiology, Yuyao People’s Hospital, Yuyao, China
| | - Gang Liu
- Department of Cardiology, Yuyao People’s Hospital, Yuyao, China
| |
Collapse
|
34
|
The risk of cardiovascular complications in inflammatory bowel disease. Clin Exp Med 2020; 20:481-491. [PMID: 32785793 PMCID: PMC7568702 DOI: 10.1007/s10238-020-00639-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing disease of unknown etiology involving gastrointestinal tract. IBD comprises two main entities: ulcerative colitis and Crohn's disease. Several studies showed increased risk of cardiovascular complications in chronic inflammatory disorders, especially during IBD relapses. Endothelium plays a role in physiologic regulation of vascular tone, cell adhesion, migration and resistance to thrombosis. Also, its dysfunction is associated with increased risk of atherosclerosis development. There are several potential links between chronic IBD-related inflammatory processes and the risk of cardiovascular disease, but insight into pathogenetic pathways remains unclear. We present the current concepts and review of adult and pediatric studies on the risk of CVD in IBD.
Collapse
|
35
|
Microvascular and lymphatic dysfunction in HFpEF and its associated comorbidities. Basic Res Cardiol 2020; 115:39. [PMID: 32451732 PMCID: PMC7248044 DOI: 10.1007/s00395-020-0798-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex heterogeneous disease for which our pathophysiological understanding is still limited and specific prevention and treatment strategies are lacking. HFpEF is characterised by diastolic dysfunction and cardiac remodelling (fibrosis, inflammation, and hypertrophy). Recently, microvascular dysfunction and chronic low-grade inflammation have been proposed to participate in HFpEF development. Furthermore, several recent studies demonstrated the occurrence of generalized lymphatic dysfunction in experimental models of risk factors for HFpEF, including obesity, hypercholesterolaemia, type 2 diabetes mellitus (T2DM), hypertension, and aging. Here, we review the evidence for a combined role of coronary (micro)vascular dysfunction and lymphatic vessel alterations in mediating key pathological steps in HFpEF, including reduced cardiac perfusion, chronic low-grade inflammation, and myocardial oedema, and their impact on cardiac metabolic alterations (oxygen and nutrient supply/demand imbalance), fibrosis, and cardiomyocyte stiffness. We focus primarily on HFpEF caused by metabolic risk factors, such as obesity, T2DM, hypertension, and aging.
Collapse
|
36
|
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy -
| |
Collapse
|