1
|
Maiorov I, Bagrov K, Efraim R, Ankri Eliyahu G, Livneh A, Landesberg A. MMP-8 causes leftward shift in end-diastolic pressure-volume relationship and may explain the development of diastolic dysfunction in septic cardiomyopathy. Am J Physiol Heart Circ Physiol 2024; 327:H1098-H1111. [PMID: 39178029 DOI: 10.1152/ajpheart.00240.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Septic cardiomyopathy (SCM) with diastolic dysfunction carries a poor prognosis, and the mechanisms underlying the development of diastolic dysfunction remain unclear. Matrix metalloproteinase-8 (MMP-8) is released from neutrophils and degrades collagen I. MMP-8 levels correlate with SCM severity. We scrutinized, for the first time, the direct impact of MMP-8 on cardiac systolic and diastolic functions. Isolated rat hearts were perfused with Krebs-Henseleit solution in a Langendorff setup with computer-controlled filling pressures of both ventricles in an isovolumetric regime. The end-diastolic pressure (EDP) varied periodically between 3 and 20 mmHg. After baseline recordings, MMP-8 (100 µg/mL) was added to the perfusion. Short-axis views of both ventricles were continuously acquired by echocardiography. MMP-8 perfusion resulted in a progressive decline in peak systolic pressures (Psys) in both ventricles, but without significant changes in their end-systolic pressure-area relationships (ESPARs). Counterintuitively, conspicuous leftward shifts of the end-diastolic pressure-area relationships (EDPARs) were observed in both ventricles. The left ventricle (LV) end-diastolic area (EDA) decreased by 32.8 ± 5.7% (P = 0.008) at an EDP of 10.5 ± 0.4 mmHg, when LV Psys dropped by 20%. The decline of Psys was primarily due to the decrease in EDA, and restoring the baseline EDA by increasing EDP recovered 81.33 ± 5.87% of the pressure drop. Collagen I generates tensile (eccentric) stress, and its degradation by MMP-8 causes end-diastolic pressure-volume relationship (EDPVR) leftward shift, resulting in diastolic and systolic dysfunctions. The diastolic dysfunction explains the clinically observed fluid unresponsiveness, whereas the decrease in end-diastolic volume (EDV) diminishes the systolic functions. MMP-8 can explain the development of SCM with diastolic dysfunction.NEW & NOTEWORTHY MMP-8, released from activated neutrophils and macrophages, is markedly elevated in sepsis, correlating with sepsis severity and mortality. MMP-8 targets collagen I of the cardiac ECM and induces diastolic dysfunction with fluid unresponsiveness, associated with decreased EDV, reduced sarcomere length, and diminished systolic function. Unlike other MMPs that predominantly cleave collagen-III and contribute to cardiac dilatation, thereby increasing sarcomere length, MMP-8 leads to a leftward shift in the EDPVR, resulting in diastolic and systolic dysfunctions.
Collapse
Affiliation(s)
- Ida Maiorov
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Konstantin Bagrov
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Roy Efraim
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| | - Galit Ankri Eliyahu
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Amit Livneh
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Amir Landesberg
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| |
Collapse
|
2
|
Nakajima T, Haruyama A, Fukuda T, Minami K, Hirose S, Yazawa H, Nakajima T, Hasegawa T, Kitagawa Y, Obi S, Inami S, Oguri G, Shibasaki I, Amano H, Arikawa T, Sakuma M, Abe S, Fukuda H, Toyoda S. Left atrial reservoir strain is a marker of atrial fibrotic remodeling in patients undergoing cardiovascular surgery: Analysis of gene expression. PLoS One 2024; 19:e0306323. [PMID: 38976680 PMCID: PMC11230549 DOI: 10.1371/journal.pone.0306323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024] Open
Abstract
Left atrial strain (LAS) measured by two-dimensional speckle tracking echocardiography (2DSTE) is considered to be a marker of LA structural remodeling, but it remains unsettled. We investigated the potential usefulness and clinical relevance of LAS to detect atrial remodeling including fibrosis by analyzing gene expression in cardiovascular surgery patients. Preoperative 2DSTE was performed in 131 patients (92 patients with sinus rhythm [SR] patients including paroxysmal AF [PAF], 39 atrial fibrillation [AF]) undergoing cardiovascular surgery. Atrial samples were obtained from the left atrial appendages, and mRNA expression level was analyzed by real-time reverse transcription polymerase chain reaction (RT-PCR) in 59 cases (24 PAF, 35 AF). Mean value of left atrial reservoir strain (mLASr) correlated with left atrial volume index (LAVI), and left atrial conduit strain (mLAScd). mLASr also correlated with left atrial contractile strain (mLASct) in SR patients including PAF. mLASr was significantly lower, and LAVI was higher, in the AF group, compared with SR patients including PAF. The expression of COL1A1 mRNA encoding collagen type I α1 significantly increased in AF patients (p = 0.031). mLASr negatively correlated with COL1A1 expression level, and multivariate regression analysis showed that mLASr was an independent predictor of atrial COL1A1 expression level, even after adjusting for age, sex, and BMI. But, neither mLAScd / mLASct nor LAVI (bp) correlated with COL1A1 gene expression. The expression level of COL1A1 mRNA strongly correlated with ECM-related genes (COL3A1, FN1). It also correlated ECM degradation-related genes (MMP2, TIMP1, and TIMP2), pro-fibrogenic cytokines (TGFB1 encoding TGFβ1, END1, PDGFD, CTGF), oxidant stress-related genes (NOX2, NOX4), ACE, inflammation-related genes (NLRP, IL1B, MCP-1), and apoptosis (BAX). Among the fibrosis-related genes examined, univariable regression analysis showed that log (COL1A1) was associated with log (TGFB1) (adjusted R2 = 0.685, p<0.001), log (NOX4) (adjusted R2 = 0.622, p<0.001), log (NOX2) (adjusted R2 = 0.611, p<0.001), suggesting that TGFB1 and NOX4 was the potent independent determinants of COL1A1 expression level. mLASr negatively correlated with the ECM-related genes, and fibrosis-related gene expression level including TGFB1, NOX2, and NLRP3 in PAF patients. PAF patients with low mLASr had higher expression of the fibrosis-related gene expression, compared with those with high mLASr. These results suggest that LASr correlates with atrial COL1A1 gene expression associated with fibrosis-related gene expression. Patients with low LASr exhibit increased atrial fibrosis-related gene expression, even those with PAF, highlighting the utility of LAS as a marker for LA fibrosis in cardiovascular surgery patients.
Collapse
Affiliation(s)
- Toshiaki Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Akiko Haruyama
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Taira Fukuda
- Department of Liberal Arts and Human Development, Kanagawa University of Human Services, Kanagawa, Japan
| | - Kentaro Minami
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Suguru Hirose
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Hiroko Yazawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takafumi Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takaaki Hasegawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Yoshiyuki Kitagawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Syotaro Obi
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Shu Inami
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Gaku Oguri
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Ikuko Shibasaki
- Department of Cardiovascular Surgery, Dokkyo Medical University Hospital, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Hirohisa Amano
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takuo Arikawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Shichiro Abe
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Hirotsugu Fukuda
- Department of Cardiovascular Surgery, Dokkyo Medical University Hospital, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| |
Collapse
|
3
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [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: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Liu W, Long L, Wang Z, He S, Han Y, Yang L, Hu C, Wang Y. A Whole-Course-Repair System Based on Stimulus-Responsive Multifunctional Hydrogels for Myocardial Tissue Regeneration. SMALL METHODS 2024:e2400121. [PMID: 38923800 DOI: 10.1002/smtd.202400121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/10/2024] [Indexed: 06/28/2024]
Abstract
Myocardial infarction (MI) has emerged as the predominant cause of cardiovascular morbidity globally. The pathogenesis of MI unfolds as a progressive process encompassing three pivotal phases: inflammation, proliferation, and remodeling. Smart stimulus-responsive hydrogels have garnered considerable attention for their capacity to deliver therapeutic drugs precisely and controllably at the MI site. Here, a smart stimulus-responsive hydrogel with a dual-crosslinked network structure is designed, which enables the precise and controlled release of therapeutic drugs in different pathological stages for the treatment of MI. The hydrogel can rapidly release curcumin (Cur) in the inflammatory phase of MI to exert anti-apoptotic/anti-inflammatory effects. Recombinant humanized collagen type III (rhCol III) is loaded in the hydrogel and released as the hydrogel swelled/degraded during the proliferative phase to promote neovascularization. RepSox (a selective TGF-β inhibitor) releases from Pluronic F-127 grafted with aldehyde nanoparticles (PF127-CHO@RepSox NPs) in the remodeling phase to against fibrosis. The results in vitro and in vivo suggest that the hydrogel improves cardiac function and alleviates cardiac remodeling by suppressing inflammation and apoptosis, promoting neovascularization, and inhibiting myocardial fibrosis. A whole-course-repair system, leveraging stimulus-responsive multifunctional hydrogels, demonstrates notable effectiveness in enhancing post-MI cardiac function and facilitating the restoration of damaged myocardial tissue.
Collapse
Affiliation(s)
- Wenqi Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Linyu Long
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Zhicun Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Shuyi He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yaling Han
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
- Department of Cardiology, General Hospital of Northern Theater Command Shenyang, Shenyang, 110016, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Med-X Center for Materials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
5
|
Alvarez-Araos P, Jiménez S, Salazar-Ardiles C, Núñez-Espinosa C, Paez V, Rodriguez-Fernandez M, Raberin A, Millet GP, Iturriaga R, Andrade DC. Baroreflex and chemoreflex interaction in high-altitude exposure: possible role on exercise performance. Front Physiol 2024; 15:1422927. [PMID: 38895516 PMCID: PMC11184637 DOI: 10.3389/fphys.2024.1422927] [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: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
The hypoxic chemoreflex and the arterial baroreflex are implicated in the ventilatory response to exercise. It is well known that long-term exercise training increases parasympathetic and decreases sympathetic tone, both processes influenced by the arterial baroreflex and hypoxic chemoreflex function. Hypobaric hypoxia (i.e., high altitude [HA]) markedly reduces exercise capacity associated with autonomic reflexes. Indeed, a reduced exercise capacity has been found, paralleled by a baroreflex-related parasympathetic withdrawal and a pronounced chemoreflex potentiation. Additionally, it is well known that the baroreflex and chemoreflex interact, and during activation by hypoxia, the chemoreflex is predominant over the baroreflex. Thus, the baroreflex function impairment may likely facilitate the exercise deterioration through the reduction of parasympathetic tone following acute HA exposure, secondary to the chemoreflex activation. Therefore, the main goal of this review is to describe the main physiological mechanisms controlling baro- and chemoreflex function and their role in exercise capacity during HA exposure.
Collapse
Affiliation(s)
- Pablo Alvarez-Araos
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Departamento de Kinesiología, Facultad de Ciencias de la Salud, Universidad de Atacama, Copiapó, Chile
| | - Sergio Jiménez
- Departamento de Kinesiología, Facultad de Ciencias de la Salud, Universidad de Atacama, Copiapó, Chile
| | - Camila Salazar-Ardiles
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristian Núñez-Espinosa
- Escuela de Medicina de la Universidad de Magallanes, Punta Arenas, Chile
- Centro Asistencial de Docencia e Investigación (CADI-UMAG), Santiago, Chile
| | - Valeria Paez
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maria Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antoine Raberin
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gregoire P. Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Rodrigo Iturriaga
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - David C. Andrade
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura, Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
6
|
Matta A, Ohlmann P, Nader V, Moussallem N, Carrié D, Roncalli J. A review of therapeutic approaches for post-infarction left ventricular remodeling. Curr Probl Cardiol 2024; 49:102562. [PMID: 38599556 DOI: 10.1016/j.cpcardiol.2024.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Left ventricular remodeling is an adaptive process initially developed in response to acute myocardial infarction (AMI), but it ends up with negative adverse outcomes such as infarcted wall thinning, ventricular dilation, and cardiac dysfunction. A prolonged excessive inflammatory reaction to cardiomyocytes death and necrosis plays the crucial role in the pathophysiological mechanisms. The pharmacological treatment includes nitroglycerine, β-blockers, ACEi/ARBs, SGLT2i, mineralocorticoid receptor antagonists, and some miscellaneous aspects. Stem cells therapy, CD34+ cells transplantation and gene therapy constitute the promissing therapeutic approaches for post AMI cardiac remodeling, thereby enhancing angiogenesis, cardiomyocytes differenciation and left ventricular function on top of inhibiting apoptosis, inflammation, and collagen deposition. All these lead to reduce infarct size, scar formation and myocardial fibrosis.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France; School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon.
| | - Patrick Ohlmann
- Department of Cardiology, Strasbourg University Hospital, Strasbourg, France
| | - Vanessa Nader
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France
| | - Nicolas Moussallem
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon
| | - Didier Carrié
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| | - Jerome Roncalli
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
7
|
Guedes MR, de Noronha SISR, Chírico MTT, da Costa GDC, de Freitas Castro T, de Brito RCF, Vieira LG, Reis TO, Ribeiro MC, Reis AB, Carneiro CM, Bezerra FS, Montano N, da Silva VJD, de Menezes RCA, Chianca-Jr DA, Silva FCDS. Ivabradine restores tonic cardiovascular autonomic control and reduces tachycardia, hypertension and left ventricular inflammation in post-weaning protein malnourished rats. Life Sci 2024; 346:122636. [PMID: 38614307 DOI: 10.1016/j.lfs.2024.122636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/22/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Malnutrition results in autonomic imbalance and heart hypertrophy. Overexpression of hyperpolarization-activated cyclic nucleotide-gated channels (HCN) in the left ventricles (LV) is linked to hypertrophied hearts and abnormal myocardium automaticity. Given that ivabradine (IVA) has emerging pleiotropic effects, in addition to the widely known bradycardic response, this study evaluated if IVA treatment could repair the autonomic control and cardiac damages in malnourished rats. AIM Assess the impact of IVA on tonic cardiovascular autonomic control and its relationship with hemodynamics regulation, LV inflammation, and HCN gene expression in post-weaning protein malnutrition condition. MAIN METHODS After weaning, male rats were divided into control (CG; 22 % protein) and malnourished (MG; 6 % protein) groups. At 35 days, groups were subdivided into CG-PBS, CG-IVA, MG-PBS and MG-IVA (PBS 1 ml/kg or IVA 1 mg/kg) received during 8 days. We performed jugular vein cannulation and electrode implant for drug delivery and ECG registration to assess tonic cardiovascular autonomic control; femoral cannulation for blood pressure (BP) and heart rate (HR) assessment; and LV collection to evaluate ventricular remodeling and HCN gene expression investigation. KEY FINDINGS Malnutrition induced BP and HR increases, sympathetic system dominance, and LV remodeling without affecting HCN gene expression. IVA reversed the cardiovascular autonomic imbalance; prevented hypertension and tachycardia; and inhibited the LV inflammatory process and fiber thickening caused by malnutrition. SIGNIFICANCE Our findings suggest that ivabradine protects against malnutrition-mediated cardiovascular damage. Moreover, our results propose these effects were not attributed to HCN expression changes, but rather to IVA pleiotropic effects on autonomic control and inflammation.
Collapse
Affiliation(s)
- Mariana Reis Guedes
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Sylvana Izaura Salyba Rendeiro de Noronha
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Máira Tereza Talma Chírico
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Gabriela Dias Carvalho da Costa
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Thalles de Freitas Castro
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Rory Cristiane Fortes de Brito
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Lucas Gabriel Vieira
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Thayane Oliveira Reis
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Marcelo Carlos Ribeiro
- Statistics Department, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Alexandre Barbosa Reis
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Cláudia Martins Carneiro
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Frank Silva Bezerra
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Nicola Montano
- Department of Clinical Sciences and Community Health, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.
| | - Valdo José Dias da Silva
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil; Graduate Program in Physiological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil.
| | - Rodrigo Cunha Alvim de Menezes
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Deoclécio Alves Chianca-Jr
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| | - Fernanda Cacilda Dos Santos Silva
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil.
| |
Collapse
|
8
|
Bonet F, Hernandez-Torres F, Ramos-Sánchez M, Quezada-Feijoo M, Bermúdez-García A, Daroca T, Alonso-Villa E, García-Padilla C, Mangas A, Toro R. Unraveling the Etiology of Dilated Cardiomyopathy through Differential miRNA-mRNA Interactome. Biomolecules 2024; 14:524. [PMID: 38785931 PMCID: PMC11117812 DOI: 10.3390/biom14050524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Dilated cardiomyopathy (DCM) encompasses various acquired or genetic diseases sharing a common phenotype. The understanding of pathogenetic mechanisms and the determination of the functional effects of each etiology may allow for tailoring different therapeutic strategies. MicroRNAs (miRNAs) have emerged as key regulators in cardiovascular diseases, including DCM. However, their specific roles in different DCM etiologies remain elusive. Here, we applied mRNA-seq and miRNA-seq to identify the gene and miRNA signature from myocardial biopsies from four patients with DCM caused by volume overload (VCM) and four with ischemic DCM (ICM). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used for differentially expressed genes (DEGs). The miRNA-mRNA interactions were identified by Pearson correlation analysis and miRNA target-prediction programs. mRNA-seq and miRNA-seq were validated by qRT-PCR and miRNA-mRNA interactions were validated by luciferase assays. We found 112 mRNAs and five miRNAs dysregulated in VCM vs. ICM. DEGs were positively enriched for pathways related to the extracellular matrix (ECM), mitochondrial respiration, cardiac muscle contraction, and fatty acid metabolism in VCM vs. ICM and negatively enriched for immune-response-related pathways, JAK-STAT, and NF-kappa B signaling. We identified four pairs of negatively correlated miRNA-mRNA: miR-218-5p-DDX6, miR-218-5p-TTC39C, miR-218-5p-SEMA4A, and miR-494-3p-SGMS2. Our study revealed novel miRNA-mRNA interaction networks and signaling pathways for VCM and ICM, providing novel insights into the development of these DCM etiologies.
Collapse
Affiliation(s)
- Fernando Bonet
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | - Francisco Hernandez-Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Mónica Ramos-Sánchez
- Cardiology Department, Central de la Cruz Roja Hospital, 28003 Madrid, Spain; (M.R.-S.); (M.Q.-F.)
- Medicine Department, School of Medicine, Alfonso X EL Sabio University, 28691 Madrid, Spain
| | - Maribel Quezada-Feijoo
- Cardiology Department, Central de la Cruz Roja Hospital, 28003 Madrid, Spain; (M.R.-S.); (M.Q.-F.)
- Medicine Department, School of Medicine, Alfonso X EL Sabio University, 28691 Madrid, Spain
| | - Aníbal Bermúdez-García
- Cardiovascular Surgery Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain (T.D.)
| | - Tomás Daroca
- Cardiovascular Surgery Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain (T.D.)
| | - Elena Alonso-Villa
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | | | - Alipio Mangas
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
- Internal Medicine Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | - Rocio Toro
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| |
Collapse
|
9
|
Siebers P, Gembruch U, Merz WM, Recker F, Müller A, Strizek B, Geipel A, Berg C, Weber EC. Fetal NT-proBNP levels and their course in severe anemia during intrauterine treatment. Arch Gynecol Obstet 2024; 309:1341-1351. [PMID: 36966429 PMCID: PMC10894143 DOI: 10.1007/s00404-023-07006-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/07/2023] [Indexed: 03/27/2023]
Abstract
PURPOSE In adults and fetuses, N-terminal pro-B-type natriuretic peptide (NT-proBNP) is a marker of cardiac failure and myocardial remodelling. We examined the effect of anemia and intrauterine transfusion (IUT) on NT-proBNP concentrations in fetuses with anemia and established gestational age-dependent reference values of a control group. METHODS We analyzed NT-proBNP levels in anemic fetuses that underwent serial intrauterine transfusions (IUT), focusing on different causes and severity of anemia and comparing the results to a non-anemic control group. RESULTS In the control group, the average NT-proBNP concentration was 1339 ± 639 pg/ml, decreasing significantly with increasing gestational age (R = - 74.04, T = - 3.65, p = 0.001). Subjects had significantly higher NT-proBNP concentrations before initiation of IUT therapy (p < 0.001), showing fetuses with parvovirus B19 (PVB19) infection having the highest concentrations. Hydropic fetuses also showed an increased NT-proBNP concentration compared to non-hydropic fetuses (p < 0.001). During the course of therapy, NT-proBNP concentration before subsequent IUT decreased significantly from pathologically high levels, while MoM-Hb and MoM-MCA-PSV remained pathological. CONCLUSION NT-pro BNP levels in non-anemic fetuses are higher than in postnatal life, decreasing with ongoing pregnancy. Anemia is a hyperdynamic state and its severity correlates with circulating NT-proBNP levels. Highest concentrations occur in fetuses with hydrops and with PVB19 infection, respectively. Treatment by IUT leads to a normalisation of NT-proBNP concentrations, so the measurement of its levels may be useful in therapy monitoring.
Collapse
Affiliation(s)
- Pauline Siebers
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Ulrich Gembruch
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Waltraut Maria Merz
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Florian Recker
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Andreas Müller
- Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, Bonn, Germany
| | - Brigitte Strizek
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Annegret Geipel
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Christoph Berg
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
- Division of Prenatal Medicine, Gynecological Ultrasound and Fetal Surgery, Department of Obstetrics and Gynecology, University Hospital Cologne, Cologne, Germany
| | - Eva Christin Weber
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany.
- Division of Prenatal Medicine, Gynecological Ultrasound and Fetal Surgery, Department of Obstetrics and Gynecology, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Dias LG, Reis CHO, Dos Santos L, Krause Neto W, Lima-Leopoldo AP, Baker JS, Leopoldo AS, Bocalini DS. Strength training improves heart function, collagen and strength in rats with heart failure. J Physiol Sci 2024; 74:10. [PMID: 38365576 PMCID: PMC10873996 DOI: 10.1186/s12576-024-00899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/20/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND/OBJECTIVES Myocardial infarction (MI) frequently leads to cardiac remodeling and failure with impaired life quality, playing an important role in cardiovascular deaths. Although physical exercise is a well-recognized effective non-pharmacological therapy for cardiovascular diseases, the effects of strength training (ST) on the structural and functional aspects of cardiac remodeling need to be further documented. In this study, we aimed to investigate the role of a linear block ST protocol in the rat model of MI. METHODS AND RESULTS After 6 weeks of MI induction or sham surgery, male adult rats performed ST for the following 12 weeks. The ladder-based ST program was organized in three mesocycles of 4 weeks, with one load increment for each block according to the maximal carrying load test. After 12 weeks, the infarcted-trained rats exhibited an increase in performance, associated with reduced cardiac hypertrophy and pulmonary congestion compared with the untrained group. Despite not changing MI size, the ST program partially prevented cardiac dilatation and ventricular dysfunction assessed by echocardiography and hemodynamics, and interstitial fibrosis evaluated by histology. In addition, isolated cardiac muscles from infarcted-trained rats had improved contractility parameters in a steady state, and in response to calcium or stimuli pauses. CONCLUSIONS The ST in infarcted rats increased the capacity to carry mass, associated with attenuation of cardiac remodeling and pulmonary congestion with improving cardiac function that could be attributed, at least in part, to the improvement of myocardial contractility.
Collapse
Affiliation(s)
- Leisiane G Dias
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Carlos H O Reis
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Walter Krause Neto
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Paula Lima-Leopoldo
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Julien S Baker
- Department of Sport, Physical Education and Health, Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - André S Leopoldo
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Danilo S Bocalini
- Experimental Physiology and Biochemistry Laboratory. Physical Education and Sport Center, Federal University of Espirito Santo, Vitoria, Brazil.
| |
Collapse
|
11
|
Luo Q, Wei Y, Lv X, Chen W, Yang D, Tuo Q. The Effect and Mechanism of Oleanolic Acid in the Treatment of Metabolic Syndrome and Related Cardiovascular Diseases. Molecules 2024; 29:758. [PMID: 38398510 PMCID: PMC10892503 DOI: 10.3390/molecules29040758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Metabolic syndromes (MetS) and related cardiovascular diseases (CVDs) pose a serious threat to human health. MetS are metabolic disorders characterized by obesity, dyslipidemia, and hypertension, which increase the risk of CVDs' initiation and development. Although there are many availabile drugs for treating MetS and related CVDs, some side effects also occur. Considering the low-level side effects, many natural products have been tried to treat MetS and CVDs. A five-cyclic triterpenoid natural product, oleanolic acid (OA), has been reported to have many pharmacologic actions such as anti-hypertension, anti-hyperlipidemia, and liver protection. OA has specific advantages in the treatment of MetS and CVDs. OA achieves therapeutic effects through a variety of pathways, attracting great interest and playing a vital role in the treatment of MetS and CVDs. Consequently, in this article, we aim to review the pharmacological actions and potential mechanisms of OA in treating MetS and related CVDs.
Collapse
Affiliation(s)
- Quanye Luo
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Yu Wei
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Xuzhen Lv
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, The School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China;
| | - Wen Chen
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Dongmei Yang
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Qinhui Tuo
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| |
Collapse
|
12
|
Jurado MR, Tombor LS, Arsalan M, Holubec T, Emrich F, Walther T, Abplanalp W, Fischer A, Zeiher AM, Schulz MH, Dimmeler S, John D. Improved integration of single-cell transcriptome data demonstrates common and unique signatures of heart failure in mice and humans. Gigascience 2024; 13:giae011. [PMID: 38573186 PMCID: PMC10993718 DOI: 10.1093/gigascience/giae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/17/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Cardiovascular research heavily relies on mouse (Mus musculus) models to study disease mechanisms and to test novel biomarkers and medications. Yet, applying these results to patients remains a major challenge and often results in noneffective drugs. Therefore, it is an open challenge of translational science to develop models with high similarities and predictive value. This requires a comparison of disease models in mice with diseased tissue derived from humans. RESULTS To compare the transcriptional signatures at single-cell resolution, we implemented an integration pipeline called OrthoIntegrate, which uniquely assigns orthologs and therewith merges single-cell RNA sequencing (scRNA-seq) RNA of different species. The pipeline has been designed to be as easy to use and is fully integrable in the standard Seurat workflow.We applied OrthoIntegrate on scRNA-seq from cardiac tissue of heart failure patients with reduced ejection fraction (HFrEF) and scRNA-seq from the mice after chronic infarction, which is a commonly used mouse model to mimic HFrEF. We discovered shared and distinct regulatory pathways between human HFrEF patients and the corresponding mouse model. Overall, 54% of genes were commonly regulated, including major changes in cardiomyocyte energy metabolism. However, several regulatory pathways (e.g., angiogenesis) were specifically regulated in humans. CONCLUSIONS The demonstration of unique pathways occurring in humans indicates limitations on the comparability between mice models and human HFrEF and shows that results from the mice model should be validated carefully. OrthoIntegrate is publicly accessible (https://github.com/MarianoRuzJurado/OrthoIntegrate) and can be used to integrate other large datasets to provide a general comparison of models with patient data.
Collapse
Affiliation(s)
- Mariano Ruz Jurado
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Lukas S Tombor
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
| | - Mani Arsalan
- Department of Cardiovascular Surgery, Goethe University Hospital, 60590 Frankfurt am Main, Germany
| | - Tomas Holubec
- Department of Cardiovascular Surgery, Goethe University Hospital, 60590 Frankfurt am Main, Germany
| | - Fabian Emrich
- Department of Cardiovascular Surgery, Goethe University Hospital, 60590 Frankfurt am Main, Germany
| | - Thomas Walther
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Department of Cardiovascular Surgery, Goethe University Hospital, 60590 Frankfurt am Main, Germany
| | - Wesley Abplanalp
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ariane Fischer
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andreas M Zeiher
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Marcel H Schulz
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - David John
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), 60590 Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
13
|
Liu W, Hu C, Long L, He S, Zhang W, Wang Z, Yang L, Wang Y. An injectable carrier for spatiotemporal and sequential release of therapeutic substances to treat myocardial infarction. J Control Release 2024; 365:29-42. [PMID: 37931807 DOI: 10.1016/j.jconrel.2023.10.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Myocardial infarction (MI) has become the primary cause of cardiovascular mortality, while the current treatment methods in clinical all have their shortcomings. Injectable biomaterials have emerged as a promising solution for cardiac tissue repair after MI. In this study, we designed a smart multifunctional carrier that could meet the treatment needs of different MI pathological processes by programmatically releasing different therapeutic substances. The carrier could respond to inflammatory microenvironment in the early stage of MI with rapid release of curcumin (Cur), and then sustained release recombinant humanized collagen type III (rhCol III) to treat MI. The rapid release of Cur reduced inflammation and apoptosis in the early stages, while the sustained release of rhCol III promoted angiogenesis and cardiac repair in the later stages. In vitro and in vivo results suggested that the multifunctional carrier could effectively improve cardiac function, promote the repair of infarcted tissue, and inhibit ventricular remodeling by reducing cell apoptosis and inflammation, and promoting angiogenesis in the different pathological processes of MI. Therefore, this programmed-release carrier provides a promising protocol for MI therapy.
Collapse
Affiliation(s)
- Wenqi Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Linyu Long
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Shuyi He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Wen Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Zhicun Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Li Yang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Chuanda-Jinbo Joint Research Center, Sichuan University, Chengdu 610064, People's Republic of China
| |
Collapse
|
14
|
Pradhan G, Kulkarni YA. Diabetes and its Complications: Role of Luteolin, A Wonder Chemical from the Natural Source. Curr Diabetes Rev 2024; 21:e290224227537. [PMID: 38425118 DOI: 10.2174/0115733998285798240217084632] [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: 10/06/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
Flavonoids have been reported to be vital in treating various chronic disorders. Luteolin (3',4',5,7-tetrahydroxyflavone) is a flavonoid present in a variety of plant sources such as celery, green pepper, olive oil, peppermint, thyme, rosemary, oregano, etc. It has been reported to have various pharmacological activities such as antioxidant, anti-inflammatory, anticancer, antidiabetic, anti-Alzheimer, antimicrobial, etc. Many scientific studies have been carried out on luteolin for its possible effects on diabetes and its associated complications. The present review focuses on the role of luteolin in diabetes mellitus and the associated complications. The antidiabetic impact of luteolin is linked with the increased expression of PPARγ and GLUT. Various in vitro and in vivo studies have been performed to explore the effects of luteolin on diabetic complications, and it has shown a significant impact in the management of the same.
Collapse
Affiliation(s)
- Gandhar Pradhan
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| |
Collapse
|
15
|
Tian L, Jarrah M, Herz H, Chu Y, Xu Y, Tang Y, Yuan J, Mokadem M. Toll-like Receptor 4 Differentially Modulates Cardiac Function in Response to Chronic Exposure to High-Fat Diet and Pressure Overload. Nutrients 2023; 15:5139. [PMID: 38140398 PMCID: PMC10747341 DOI: 10.3390/nu15245139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND/AIM The impact of myocardial stressors such as high-fat diet (HFD) and pressure overload has been extensively studied. Toll-like receptor 4 (TLR4) deficiency has been suggested to have a protective role in response to these stressors, although some conflicting data exist. Furthermore, there is limited information about the role of TLR4 on cardiac remodeling in response to long-term exposure to stressors. This study aims to investigate the effects of TLR4 deficiency on cardiac histology and physiology in response to chronic stressors. METHODS TLR4-deficient (TLR4-/-) and wild-type (WT) mice were subjected to either HFD or a normal diet (ND) for 28 weeks. Another group underwent abdominal aortic constriction (AAC) or a sham procedure and was monitored for 12 weeks. Inflammatory markers, histology, and echocardiography were used to assess the effects of these interventions. RESULTS TLR4-/- mice exhibited reduced cardiac hypertrophy and fibrosis after long-term HFD exposure compared to ND without affecting cardiac function. On the other hand, TLR4 deficiency worsened cardiac function in response to AAC, leading to decreased ejection fraction (EF%) and increased end-systolic volume (ESV). CONCLUSIONS TLR4 deficiency provided protection against HFD-induced myocardial inflammation but impaired hemodynamic cardiac function under pressure overload conditions. These findings highlight the crucial role of TLR4 and its downstream signaling pathway in maintaining cardiac output during physiologic cardiac hypertrophy in response to pressure overload.
Collapse
Affiliation(s)
- Liping Tian
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Mohammad Jarrah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Hussein Herz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Yi Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Ying Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining 272067, China
| | - Mohamad Mokadem
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
- Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA 52242, USA
- Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA 52242, USA
- Iowa City Veterans Affairs Health Care System, Iowa City, IA 52242, USA
| |
Collapse
|
16
|
Coronel R. Cross talk: Fat and the arrhythmogenic substrate. Heart Rhythm 2023; 20:1706-1707. [PMID: 37709107 DOI: 10.1016/j.hrthm.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Affiliation(s)
- Ruben Coronel
- Department of Experimental Cardiology, Amsterdam University Medical Centers (location AMC), Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Winsløw U, Sakthivel T, Zheng C, Philbert B, Vinther M, Frandsen E, Iversen K, Bundgaard H, Jøns C, Risum N. The effect of increased plasma potassium on myocardial function; a randomized POTCAST substudy. Int J Cardiovasc Imaging 2023; 39:2097-2106. [PMID: 37470856 PMCID: PMC10673982 DOI: 10.1007/s10554-023-02914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/30/2023] [Indexed: 07/21/2023]
Abstract
Plasma potassium (p-K) in the high-normal range has been suggested to reduce risk of cardiovascular arrythmias and mortality through electrophysiological and mechanical effects on the myocardium. In this study, it was to investigated if increasing p-K to high-normal levels improves systolic- and diastolic myocardial function in patients with low-normal to moderately reduced left ventricular ejection fraction (LVEF). The study included 50 patients (mean age 58 years (SD 14), 81% men), with a mean p-K 3.95 mmol/l (SD 0.19), mean LVEF 48% (SD 7), and mean Global Longitudinal Strain (GLS) -14.6% (SD 3.1) patients with LVEF 35-55% from "Targeted potassium levels to decrease arrhythmia burden in high-risk patients with cardiovascular diseases trial" (POTCAST). Patients were given standard therapy and randomized (1:1) to an intervention that included guidance on potassium-rich diets, potassium supplements, and mineralocorticoid receptor antagonists targeting high-normal p-K levels (4.5-5.0 mmol/l). Echocardiography was done at baseline and after a mean follow-up of 44 days (SD 18) and the echocardiograms were analyzed for changes in GLS, mechanical dispersion, E/A, e', and E/e'. At follow-up, mean difference in changes in p-K was 0.52 mmol/l (95%CI 0.35;0.69), P<0.001 in the intervention group compared to controls. GLS was improved with a mean difference in changes of -1.0% (-2;-0.02), P<0.05 and e' and E/e' were improved with a mean difference in changes of 0.9 cm/s (0.02;1.7), P = 0.04 and ? 1.5 (-2.9;-0.14), P = 0.03, respectively. Thus, induced increase in p-K to the high-normal range improved indices of systolic and diastolic function in patients with low-normal to moderately reduced LVEF.
Collapse
Affiliation(s)
- Ulrik Winsløw
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark.
| | - Tharsika Sakthivel
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Chaoqun Zheng
- Department of Cardiology, Zealand University Hospital, Roskilde, Denmark
| | - Berit Philbert
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Michael Vinther
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Emil Frandsen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Kasper Iversen
- Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian Jøns
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Niels Risum
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark
| |
Collapse
|
19
|
Chu L, Xie D, Xu D. Epigenetic Regulation of Fibroblasts and Crosstalk between Cardiomyocytes and Non-Myocyte Cells in Cardiac Fibrosis. Biomolecules 2023; 13:1382. [PMID: 37759781 PMCID: PMC10526373 DOI: 10.3390/biom13091382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/10/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Epigenetic mechanisms and cell crosstalk have been shown to play important roles in the initiation and progression of cardiac fibrosis. This review article aims to provide a thorough overview of the epigenetic mechanisms involved in fibroblast regulation. During fibrosis, fibroblast epigenetic regulation encompasses a multitude of mechanisms, including DNA methylation, histone acetylation and methylation, and chromatin remodeling. These mechanisms regulate the phenotype of fibroblasts and the extracellular matrix composition by modulating gene expression, thereby orchestrating the progression of cardiac fibrosis. Moreover, cardiac fibrosis disrupts normal cardiac function by imposing myocardial mechanical stress and compromising cardiac electrical conduction. This review article also delves into the intricate crosstalk between cardiomyocytes and non-cardiomyocytes in the heart. A comprehensive understanding of the mechanisms governing epigenetic regulation and cell crosstalk in cardiac fibrosis is critical for the development of effective therapeutic strategies. Further research is warranted to unravel the precise molecular mechanisms underpinning these processes and to identify potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Dachun Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 315 Yanchang Middle Road, Shanghai 200072, China; (L.C.); (D.X.)
| |
Collapse
|
20
|
Zhang K, Zheng Y, Bao G, Ma W, Han B, Shi H, Zhao Z. Flt3 Activation Mitigates Mitochondrial Fragmentation and Heart Dysfunction through Rebalanced L-OPA1 Processing by Hindering the Interaction between Acetylated p53 and PHB2 in Cardiac Remodeling. Antioxidants (Basel) 2023; 12:1657. [PMID: 37759959 PMCID: PMC10525215 DOI: 10.3390/antiox12091657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023] Open
Abstract
Recent studies have shown that FMS-like receptor tyrosine kinase 3 (Flt3) has a beneficial effect on cardiac maladaptive remodeling. However, the role and mechanism of Flt3 in mitochondrial dynamic imbalance under cardiac stress remains poorly understood. This study aims to investigate how Flt3 regulates p53-mediated optic atrophy 1 (OPA1) processing and mitochondrial fragmentation to improve cardiac remodeling. Mitochondrial fragmentation in cardiomyocytes was induced by isoprenaline (ISO) and H2O2 challenge, respectively, in vitro. Cardiac remodeling in mice was established by ligating the left anterior descending coronary artery or by chronic ISO challenge, respectively, in vivo. Our results demonstrated that the protein expression of acetylated-p53 (ac-p53) in mitochondria was significantly increased under cell stress conditions, facilitating the dissociation of PHB2-OPA1 complex by binding to prohibitin 2 (PHB2), a molecular chaperone that stabilizes OPA1 in mitochondria. This led to the degradation of the long isoform of OPA1 (L-OPA1) that facilitates mitochondrial fusion and resultant mitochondrial network fragmentation. This effect was abolished by a p53 K371R mutant that failed to bind to PHB2 and impeded the formation of the ac-p53-PHB2 complex. The activation of Flt3 significantly reduced ac-p53 expression in mitochondria via SIRT1, thereby hindering the formation of the ac-p53-PHB2 complex and potentiating the stability of the PHB2-OPA1 complex. This ultimately inhibits L-OPA1 processing and leads to the balancing of mitochondrial dynamics. These findings highlight a novel mechanism by which Flt3 activation mitigates mitochondrial fragmentation and dysfunction through the reduction of L-OPA1 processing by dampening the interaction between ac-p53 and PHB2 in cardiac maladaptive remodeling.
Collapse
Affiliation(s)
- Kaina Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yeqing Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Gaowa Bao
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Wenzhuo Ma
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Bing Han
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Hongwen Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Zhenghang Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| |
Collapse
|
21
|
Vasanthan V, Hassanabad AF, Belke D, Teng G, Isidoro CA, Dutta D, Turnbull J, Deniset JF, Fedak PW. Micronized Acellular Matrix Biomaterial Leverages Eosinophils for Postinfarct Cardiac Repair. JACC Basic Transl Sci 2023; 8:939-954. [PMID: 37719429 PMCID: PMC10504403 DOI: 10.1016/j.jacbts.2023.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 09/19/2023]
Abstract
After ischemic injury, immune cells mediate maladaptive cardiac remodeling. Extracellular matrix biomaterials may redirect inflammation toward repair. Pericardial fluid contains pro-reparative immune cells, potentially leverageable by biomaterials. Herein, we explore how pericardial delivery of a micronized extracellular matrix biomaterial affects cardiac healing. In noninfarcted mice, pericardial delivery increases pericardial and myocardial eosinophil counts. This response is sustained after myocardial infarction, stimulating an interleukin 4 rich milieu. Ultimately, the biomaterial improves postinfarct vascularization and cardiac function; and eosinophil-knockout negates these benefits. For the first time, to our knowledge, we demonstrate the therapeutic potential of pericardial biomaterial delivery and the eosinophil's critical role in biomaterial-mediated postinfarct repair.
Collapse
Affiliation(s)
- Vishnu Vasanthan
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ali Fatehi Hassanabad
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Darrell Belke
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Guoqi Teng
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carmina Albertine Isidoro
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Devjyoti Dutta
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeannine Turnbull
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin F. Deniset
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Paul W.M. Fedak
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
22
|
Farag A, Mandour AS, Kaneda M, Elfadadny A, Elhaieg A, Shimada K, Tanaka R. Effect of trehalose on heart functions in rats model after myocardial infarction: assessment of novel intraventricular pressure and heart rate variability. Front Cardiovasc Med 2023; 10:1182628. [PMID: 37469485 PMCID: PMC10353053 DOI: 10.3389/fcvm.2023.1182628] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/09/2023] [Indexed: 07/21/2023] Open
Abstract
Background Myocardial infarctions remain a leading cause of global deaths. Developing novel drugs to target cardiac remodeling after myocardial injury is challenging. There is an increasing interest in exploring natural cardioprotective agents and non-invasive tools like intraventricular pressure gradients (IVPG) and heart rate variability (HRV) analysis in myocardial infarctions. Trehalose (TRE), a natural disaccharide, shows promise in treating atherosclerosis, myocardial infarction, and neurodegenerative disorders. Objectives The objective of this study was to investigate the effectiveness of TRE in improving cardiac functions measured by IVPG and HRV and reducing myocardial remodeling following myocardial infarction in rat model. Methods Rats were divided into three groups: sham, myocardial infarction (MI), and trehalose-treated MI (TRE) groups. The animals in the MI and TRE groups underwent permanent ligation of the left anterior descending artery. The TRE group received 2% trehalose in their drinking water for four weeks after the surgery. At the end of the experiment, heart function was assessed using conventional echocardiography, novel color M-mode echocardiography for IVPG evaluation, and HRV analysis. After euthanasia, gross image scoring, histopathology, immunohistochemistry, and quantitative real-time PCR were performed to evaluate inflammatory reactions, oxidative stress, and apoptosis. Results The MI group exhibited significantly lower values in multiple IVPG parameters. In contrast, TRE administration showed an ameliorative effect on IVPG changes, with results comparable to the sham group. Additionally, TRE improved HRV parameters, mitigated morphological changes induced by myocardial infarction, reduced histological alterations in wall mass, and suppressed inflammatory reactions within the infarcted heart tissues. Furthermore, TRE demonstrated antioxidant, anti-apoptotic and anti-fibrotic properties. Conclusion The investigation into the effect of trehalose on a myocardial infarction rat model has yielded promising outcomes, as evidenced by improvements observed through conventional echocardiography, histological analysis, and immunohistochemical analysis. While minor trends were noticed in IVPG and HRV measurements. However, our findings offer valuable insights and demonstrate a correlation between IVPG, HRV, and other traditional markers of echo assessment in the myocardial infarction vs. sham groups. This alignment suggests the potential of IVPG and HRV as additional indicators for future research in this field.
Collapse
Affiliation(s)
- Ahmed Farag
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ahmed Elfadadny
- Department of Animal Internal Medicine, Faculty of Veterinary Medicine, Damanhur University, Damanhur El-Beheira, Egypt
| | - Asmaa Elhaieg
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kazumi Shimada
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ryou Tanaka
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
23
|
Mantzouratou P, Malaxianaki E, Cerullo D, Lavecchia AM, Pantos C, Xinaris C, Mourouzis I. Thyroid Hormone and Heart Failure: Charting Known Pathways for Cardiac Repair/Regeneration. Biomedicines 2023; 11:975. [PMID: 36979954 PMCID: PMC10046827 DOI: 10.3390/biomedicines11030975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Heart failure affects more than 64 million people worldwide, having a serious impact on their survival and quality of life. Exploring its pathophysiology and molecular bases is an urgent need in order to develop new therapeutic approaches. Thyroid hormone signaling, evolutionarily conserved, controls fundamental biological processes and has a crucial role in development and metabolism. Its active form is L-triiodothyronine, which not only regulates important gene expression by binding to its nuclear receptors, but also has nongenomic actions, controlling crucial intracellular signalings. Stressful stimuli, such as acute myocardial infarction, lead to changes in thyroid hormone signaling, and especially in the relation of the thyroid hormone and its nuclear receptor, which are associated with the reactivation of fetal development programmes, with structural remodeling and phenotypical changes in the cardiomyocytes. The recapitulation of fetal-like features of the signaling may be partially an incomplete effort of the myocardium to recapitulate its developmental program and enable cardiomyocytes to proliferate and finally to regenerate. In this review, we will discuss the experimental and clinical evidence about the role of the thyroid hormone in the recovery of the myocardium in the setting of heart failure with reduced and preserved ejection fraction and its future therapeutic implications.
Collapse
Affiliation(s)
| | | | - Domenico Cerullo
- Centro Anna Maria Astori, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Angelo Michele Lavecchia
- Centro Anna Maria Astori, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | | | - Christodoulos Xinaris
- Centro Anna Maria Astori, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | | |
Collapse
|
24
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
25
|
Li J, Wang Y, Wang L, Qu Y, Zhou W, Zhong H, Xi D, Tang N, He F. Low expression of miR-1929-3p mediates murine cytomegalovirus-induced fibrosis in cardiac fibroblasts via targeting endothelin a receptor/NLRP3 inflammasome pathway. In Vitro Cell Dev Biol Anim 2023; 59:179-192. [PMID: 37002490 DOI: 10.1007/s11626-022-00742-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/22/2022] [Indexed: 04/03/2023]
Abstract
MicroRNAs are crucial in the development of myocardial remodeling in hypertension. Low miR-1929-3p expression induced by murine cytomegalovirus (MCMV) infection is closely related to hypertensive myocardial remodeling. This study investigated the molecular mechanism of miR-1929-3p-induced myocardial remodeling after MCMV infection. We modeled MCMV-infected mouse cardiac fibroblasts (MMCFs) as the primary cell model. First, MCMV infection reduced the expression of miR-1929-3p and increased the mRNA and protein expression of its target gene endothelin receptor type A (ETAR) in mouse cardiac fibroblasts (MCFs), which demonstrated an internal relationship with myocardial fibrosis (MF) based on high proliferation, phenotypic transformation (α-SMA), and collagen expression in MMCFs. The transfection of the miR-1929-3p mimic downregulated the high expression of ETAR and alleviated these adverse effects in MMCFs. Inversely, these effects were exacerbated by the miR-1929-3p inhibitor. Second, the transfection of endothelin receptor type A over-expressed adenovirus (adETAR) reversed these positive effects of the miR-1929-3p mimic on MF improvement. Third, the transfection of adETAR exhibited a strong inflammatory response in MMCFs with increased expression of NOD-like receptors pyrin domain containing 3 (NLRP3) and increased secretion of interleukin-18. However, we found that the ETAR antagonist BQ123 and the selected NLRP3 inflammasome inhibitor MCC950 effectively eliminated the inflammatory response induced by both MCMV infection and miR-1929-3p inhibitor. Moreover, the MCF supernatant was related to cardiomyocyte hypertrophy. Our findings suggest that MCMV infection promotes MF by inducing the downregulation of miR-1929-3p and the high expression of ETAR, which activates NLRP3 inflammasomes in MCFs.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Yongjia Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - LaMei Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Yuanyuan Qu
- Department of Respiratory Medicine, the First Affiliated Hospital of Medical College of Shihezi University, Shihezi, China
| | - Wei Zhou
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Hua Zhong
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - DongMei Xi
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Na Tang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China.
| | - Fang He
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China.
| |
Collapse
|
26
|
Vasanthan V, Shim HB, Teng G, Belke D, Svystonyuk D, Deniset JF, Fedak PWM. Acellular biomaterial modulates myocardial inflammation and promotes endogenous mechanisms of postinfarct cardiac repair. J Thorac Cardiovasc Surg 2023; 165:e122-e140. [PMID: 35058062 DOI: 10.1016/j.jtcvs.2021.12.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/17/2021] [Accepted: 12/15/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE After myocardial infarction, we previously showed that epicardial implantation of porcine small intestinal submucosal extracellular matrix (SIS-ECM) improves postinfarct cardiac function through fibroblast-mediated angiogenic and antifibrotic pathways. Herein, we characterize how SIS-ECM also coordinates a reparative cardiac inflammatory response. METHODS RNA sequencing and multiplex characterized modulation of fibroblast transcriptional and paracrine activity by SIS-ECM. Inhibitors of fibroblast growth factor 2 and toll-like receptor 9 elucidated mechanism. Mice received coronary ligation (infarction) and either SIS-ECM implantation (treatment) or sham surgery (control). Flow cytometry of SIS-ECM and the murine myocardium quantified monocytes, neutrophils, and proangiogenic subtypes. Microscopy tracked fibroblasts and immune cells, and characterized myocardial angiogenesis. RESULTS SIS-ECM increased fibroblast transcription of inflammatory pathways and production of angiogenic vascular endothelial growth factor and inflammatory cytokines via fibroblast growth factor 2 and toll-like receptor 9-dependent pathways. Two-photon microscopy showed that SIS-ECM became engrafted by native fibroblasts and leukocytes, subsequently increasing release of inflammatory cytokines and angiogenic vascular endothelial growth factor. On flow cytometry, SIS-ECM implantation increased day-7 myocardial counts of neutrophils, inflammatory monocytes, and proangiogenic vascular endothelial growth factor recptor 1 subtypes. SIS-ECM has a higher proportion of proangiogenic leukocytes compared with the myocardium. Resonant confocal microscopy showed neovascularization near SIS-ECM. CONCLUSIONS SIS-ECM promotes engraftment by native fibroblasts and leukocytes, and modulates fibroblast activity via fibroblast growth factor 2 and toll-like receptor 9 to potentiate a proangiogenic inflammatory response. Subsequently, the material increases myocardial counts of reparative proangiogenic leukocytes that can induce neovascularization. This reparative inflammatory response may explain previously reported functional improvements. Fibroblast growth factor 2 and toll-like receptor 9 mechanisms can be leveraged to design next-generation materials for postinfarct cardiac repair.
Collapse
Affiliation(s)
- Vishnu Vasanthan
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hanjoo B Shim
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Guoqi Teng
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Darrell Belke
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniyil Svystonyuk
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin F Deniset
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
27
|
Xing C, Bao L, Li W, Fan H. Progress on role of ion channels of cardiac fibroblasts in fibrosis. Front Physiol 2023; 14:1138306. [PMID: 36969589 PMCID: PMC10033868 DOI: 10.3389/fphys.2023.1138306] [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: 01/05/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Cardiac fibrosis is defined as excessive deposition of extracellular matrix (ECM) in pathological conditions. Cardiac fibroblasts (CFs) activated by injury or inflammation differentiate into myofibroblasts (MFs) with secretory and contractile functions. In the fibrotic heart, MFs produce ECM which is composed mainly of collagen and is initially involved in maintaining tissue integrity. However, persistent fibrosis disrupts the coordination of excitatory contractile coupling, leading to systolic and diastolic dysfunction, and ultimately heart failure. Numerous studies have demonstrated that both voltage- and non-voltage-gated ion channels alter intracellular ion levels and cellular activity, contributing to myofibroblast proliferation, contraction, and secretory function. However, an effective treatment strategy for myocardial fibrosis has not been established. Therefore, this review describes the progress made in research related to transient receptor potential (TRP) channels, Piezo1, Ca2+ release-activated Ca2+ (CRAC) channels, voltage-gated Ca2+ channels (VGCCs), sodium channels, and potassium channels in myocardial fibroblasts with the aim of providing new ideas for treating myocardial fibrosis.
Collapse
|
28
|
Shi S, Jiang P. Therapeutic potentials of modulating autophagy in pathological cardiac hypertrophy. Biomed Pharmacother 2022; 156:113967. [DOI: 10.1016/j.biopha.2022.113967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|
29
|
Fatty Acid Amide Hydrolase Deficiency Is Associated with Deleterious Cardiac Effects after Myocardial Ischemia and Reperfusion in Mice. Int J Mol Sci 2022; 23:ijms232012690. [PMID: 36293543 PMCID: PMC9604059 DOI: 10.3390/ijms232012690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Ischemic cardiomyopathy leads to inflammation and left ventricular (LV) dysfunction. Animal studies provided evidence for cardioprotective effects of the endocannabinoid system, including cardiomyocyte adaptation, inflammation, and remodeling. Cannabinoid type-2 receptor (CB2) deficiency led to increased apoptosis and infarctions with worsened LV function in ischemic cardiomyopathy. The aim of our study was to investigate a possible cardioprotective effect of endocannabinoid anandamide (AEA) after ischemia and reperfusion (I/R). Therefore, fatty acid amide hydrolase deficient (FAAH)−/− mice were subjected to repetitive, daily, 15 min, left anterior descending artery (LAD) occlusion over 3 and 7 consecutive days. Interestingly, FAAH−/− mice showed stigmata such as enhanced inflammation, cardiomyocyte loss, stronger remodeling, and persistent scar with deteriorated LV function compared to wild-type (WT) littermates. As endocannabinoids also activate PPAR-α (peroxisome proliferator-activated receptor), PPAR-α mediated effects of AEA were eliminated with PPAR-α antagonist GW6471 i.v. in FAAH−/− mice. LV function was assessed using M-mode echocardiography. Immunohistochemical analysis revealed apoptosis, macrophage accumulation, collagen deposition, and remodeling. Hypertrophy was determined by cardiomyocyte area and heart weight/tibia length. Molecular analyses involved Taqman® RT-qPCR and immune cells were analyzed with fluorescence-activated cell sorting (FACS). Most importantly, collagen deposition was reduced to WT levels when FAAH−/− mice were treated with GW6471. Chemokine ligand-2 (CCL2) expression was significantly higher in FAAH−/− mice compared to WT, followed by higher macrophage infiltration in infarcted areas, both being reversed by GW6471 treatment. Besides restoring antioxidative properties and contractile elements, PPAR-α antagonism also reversed hypertrophy and remodeling in FAAH−/− mice. Finally, FAAH−/−-mice showed more substantial downregulation of PPAR-α compared to WT, suggesting a compensatory mechanism as endocannabinoids are also ligands for PPAR-α, and its activation causes lipotoxicity leading to cardiomyocyte apoptosis. Our study gives novel insights into the role of endocannabinoids acting via PPAR-α. We hypothesize that the increase in endocannabinoids may have partially detrimental effects on cardiomyocyte survival due to PPAR-α activation.
Collapse
|
30
|
Dong Y, Huang T, Zhai Z, Dong Q, Xia Z, Xia Z, Yu J, Jiang X, Hong K, Wu Y, Cheng X, Li J. Lowering serum homocysteine in H-type hypertensive patients with atrial fibrillation after radiofrequency catheter ablation to prevent atrial fibrillation recurrence. Front Nutr 2022; 9:995838. [PMID: 36176636 PMCID: PMC9514121 DOI: 10.3389/fnut.2022.995838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/25/2022] [Indexed: 11/21/2022] Open
Abstract
Background Prior investigation revealed that elevated serum total homocysteine (tHcy) are strongly correlated with atrial fibrillation (AF) recurrence. Herein, the goal of this study was to elucidate whether folic acid (FA) treatment reduced AF recurrence following radiofrequency catheter ablation (RFCA). Methods To conduct this retrospective research, we included consecutive H-type hypertensive AF patients, who were treated with first RFCA, between January 2010 and January 2022. We assessed the AF recurrence risk between patients who were taking 10 mg enalapril and 0.8 mg FA in a single-pill combination (enalapril–FA) daily and those who were taking a pill of 10 mg enalapril only. Outcomes were compared using the propensity-score matched analysis. Cox regression model was employed for the evaluation of AF recurrence events. Results Out of 2,714 patients, 645 patients receiving enalapril and 282 patients receiving enalapril-FA were included for analysis. Following propensity score matching, 239 patients remained in each group. These patients were followed-up for a median of 379 (137–596) days, and revealed that the enalapril-FA patients had drastically reduced AF recurrence, compared to the enalapril patients [adjusted hazard ratio (HR), 0.68; 95% confidence interval (CI), 0.48–0.97; P = 0.029]. Apart from this, no interactions were detected in the subgroup analysis. Conclusion In H-type hypertensive AF patients who were treated with first RFCA, FA supplementation was correlated with a reduced AF recurrence risk.
Collapse
Affiliation(s)
- Youzheng Dong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenyu Zhai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Quanbin Dong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Xia
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zirong Xia
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianhua Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinghua Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kui Hong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, China
| | - Yanqing Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoshu Cheng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Juxiang Li
| |
Collapse
|
31
|
Yan J, Honglei Y, Yun W, Sheng D, Yun H, Anhua Z, Na F, Min L, Dandan S, Jing W, Junming T, Wenjun Z, Xiju H. Puerarin ameliorates myocardial remodeling of spontaneously hypertensive rats through inhibiting TRPC6-CaN-NFATc3 pathway. Eur J Pharmacol 2022; 933:175254. [PMID: 36087696 DOI: 10.1016/j.ejphar.2022.175254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
Puerarin (Pue) has been widely used in the treatment of hypertension and cardiovascular diseases, but the basic mechanism of Pue on myocardial remodeling (MR) of hypertension is not clear. The purpose of this study was to investigate the effect and mechanism of Pue on MR and provide the basis for the clinical application. Thirty male spontaneously hypertensive rats (SHR) and six male Wistar Kyoto rats (WKY) aged 3 months were used in this study, SHR rats were randomly divided into 5 groups, Pue (40 or 80 mg/kg/d, ip) and telmisartan (TELMI) (30 mg/kg/d, ig) were administrated for 12 weeks. We used Echocardiography to detect the cardiac function. Morphology and structure of myocardium were observed. H9C2 cells were subjected to 1 μM Ang Ⅱ in vitro, 100 μM Pue, 0.5 μM Calmodulin-dependent calcineurin (CaN) inhibitor Cyclosporin A (CsA) and 1 μM specific transient receptor potential channel 6 (TRPC6) inhibitor SAR7334 were used in H9C2 cells. Long-term administration of Pue could significantly improve cardiac function, improve morphology and structure of myocardium in vivo. Pue could reduce MR related proteins expression (ACTC1, TGF-β1, CTGF, β-MHC and BNP), attenuate ROS, restore MMP and decrease Ca2+-overload in vitro. Further study indicated that Pue could decrease TRPC6 expression and inhibit nuclear factor of activated T cells 3 (NFATc3) nuclear translocation in vitro. These results suggested that puerarin could ameliorate myocardial remodeling through inhibiting TRPC6-CaN-NFATc3 in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Jiang Yan
- Department of Ultrasound, Taihe Hospital, Jinzhou Medicical University Union Training Base, Shiyan, 442000, China
| | - Yu Honglei
- Department of Ultrasound, Taihe Hospital, Jinzhou Medicical University Union Training Base, Shiyan, 442000, China
| | - Wu Yun
- Department of Ultrasound, Wuhan Asia General Hospital, Wuhan, 430000, China
| | - Dong Sheng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - He Yun
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhang Anhua
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Feng Na
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Lu Min
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Shi Dandan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Wang Jing
- School of Public Health and Health, Hubei University of Medicine, Shiyan, 442000, China
| | - Tang Junming
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhang Wenjun
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - He Xiju
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China; Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
32
|
Liu Q, Sun J, Dong Y, Li P, Wang J, Wang Y, Xu Y, Tian X, Wu B, He P, Yu Q, Lu X, Cao J. Tetramisole is a new I K1 channel agonist and exerts I K1 -dependent cardioprotective effects in rats. Pharmacol Res Perspect 2022; 10:e00992. [PMID: 35880674 PMCID: PMC9316008 DOI: 10.1002/prp2.992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac ischemia, hypoxia, arrhythmias, and heart failure share the common electrophysiological changes featured by the elevation of intracellular Ca2+ (Ca2+ overload) and inhibition of the inward rectifier potassium (IK1 ) channel. IK1 channel agonists have been considered a new type of anti-arrhythmia and cardioprotective agents. We predicted using a drug repurposing strategy that tetramisole (Tet), a known anthelminthic agent, was a new IK1 channel agonist. The present study aimed to experimentally identify the above prediction and further demonstrate that Tet has cardioprotective effects. Results of the whole-cell patch clamp technique showed that Tet at 1-100 μmol/L enhanced IK1 current, hyperpolarized resting potential (RP), and shortened action potential duration (APD) in isolated rat cardiomyocytes, while without effects on other ion channels or transporters. In adult Sprague-Dawley (SD) rats in vivo, Tet showed anti-arrhythmia and anticardiac remodeling effects, respectively, in the coronary ligation-induced myocardial infarction model and isoproterenol (Iso, i.p., 3 mg/kg/day, 10 days) infusion-induced cardiac remodeling model. Tet also showed anticardiomyocyte remodeling effect in Iso (1 μmol/L) infused adult rat ventricular myocytes or cultured H9c2 (2-1) cardiomyocytes. Tet at 0.54 mg/kg in vivo or 30 μmol/L in vitro showed promising protections on acute ischemic arrhythmias, myocardial hypertrophy, and fibrosis. Molecular docking was performed and identified the selective binding of Tet with Kir2.1. The cardioprotection of Tet was associated with the facilitation of IK1 channel forward trafficking, deactivation of PKA signaling, and inhibition of intracellular calcium overload. Enhancing IK1 may play dual roles in anti-arrhythmia and antiventricular remodeling mediated by restoration of Ca2+ homeostasis.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jiaxing Sun
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Yangdou Dong
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Pan Li
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jin Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yulan Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yanwu Xu
- Department of BiochemistryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xinrui Tian
- Department of Respiratory and Critical Care MedicineSecond Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Bowei Wu
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Peifeng He
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Qi Yu
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Xuechun Lu
- Department of Hematology, The Second Medical Center, Chinese PLA General HospitalNational clinical research center for geriatric diseaseBeijingChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
33
|
Li G, Yang J, Zhang D, Wang X, Han J, Guo X. Research Progress of Myocardial Fibrosis and Atrial Fibrillation. Front Cardiovasc Med 2022; 9:889706. [PMID: 35958428 PMCID: PMC9357935 DOI: 10.3389/fcvm.2022.889706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
With the aging population and the increasing incidence of basic illnesses such as hypertension and diabetes (DM), the incidence of atrial fibrillation (AF) has increased significantly. AF is the most common arrhythmia in clinical practice, which can cause heart failure (HF) and ischemic stroke (IS), increasing disability and mortality. Current studies point out that myocardial fibrosis (MF) is one of the most critical substrates for the occurrence and maintenance of AF. Although myocardial biopsy is the gold standard for evaluating MF, it is rarely used in clinical practice because it is an invasive procedure. In addition, serological indicators and imaging methods have also been used to evaluate MF. Nevertheless, the accuracy of serological markers in evaluating MF is controversial. This review focuses on the pathogenesis of MF, serological evaluation, imaging evaluation, and anti-fibrosis treatment to discuss the existing problems and provide new ideas for MF and AF evaluation and treatment.
Collapse
Affiliation(s)
- Guangling Li
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jing Yang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou, China
| | - Demei Zhang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xiaomei Wang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jingjing Han
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xueya Guo
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- *Correspondence: Xueya Guo,
| |
Collapse
|
34
|
Liu Q, Li HY, Wang SJ, Huang SQ, Yue Y, Maihemuti A, Zhang Y, Huang L, Luo L, Feng KN, Wu ZK. Belumosudil, ROCK2-Specific Inhibitor, alleviates cardiac fibrosis by inhibiting cardiac fibroblasts activation. Am J Physiol Heart Circ Physiol 2022; 323:H235-H247. [PMID: 35657612 DOI: 10.1152/ajpheart.00014.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac fibrosis is thought to be the hallmark of pathological hypertrophic remodeling, of which the myofibroblasts transdifferentiation is the key cell biological event. However, there is still no specific and effective therapeutic agent approved for cardiac fibrosis. To investigate the effects of Belumosudil, the first ROCK2-specific inhibitor, on cardiac hypertrophy, fibrosis and dysfunction induced by pressure overload, the transverse aortic constriction (TAC) or sham operation was carried out on wild-type C57BL/6 mice (male, 6-8 week old) under pentobarbital anesthesia. After that, mice were randomly divided into three groups: sham operation + vehicle, TAC + vehicle, TAC + 50 mg·kg-1·d-1 Belumosudil. We found that Belumosudil effectively ameliorated cardiac hypertrophy, fibrosis and dysfunction in TAC mice. To elucidate the underlying mechanism, we inhibited the expression of ROCK2 in vitro by either Belumosudil or siRNA. We showed that the inhibition of ROCK2 by either Belumosudil or knockdown suppressed cardiac fibroblasts activation and proliferation significantly induced by Transforming Growth Factor-β1 (TGF-β1). Furthermore, our study confirmed ROCK2 mediates cardiac fibrosis by interacting with Transforming Growth Factor-β1 (TGF-β1)/mothers against decapentaplegic homolog (Smad2) pathway. Taken together, we demonstrated that Belumosudil ameliorates cardiac hypertrophy and fibrosis induced by TAC via inhibiting cardiac fibroblasts activation. In conclusion, Belumosudil may be a promising therapeutic drug for cardiac hypertrophy and fibrosis induced by myocardial pressure overload.
Collapse
Affiliation(s)
- Quan Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Hua-Yang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Shun-Jun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Sui-Qing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yuan Yue
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Adilai Maihemuti
- Department of Operating Room, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yi Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Lin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li Luo
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Kang-Ni Feng
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhong-Kai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
35
|
Dörr K, Kammer M, Reindl-Schwaighofer R, Lorenz M, Marculescu R, Poglitsch M, Beitzke D, Oberbauer R. The Effect of FGF23 on Cardiac Hypertrophy Is Not Mediated by Systemic Renin-Angiotensin- Aldosterone System in Hemodialysis. Front Med (Lausanne) 2022; 9:878730. [PMID: 35559350 PMCID: PMC9086596 DOI: 10.3389/fmed.2022.878730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/05/2022] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is elevated in patients with chronic kidney disease and contributes to left ventricular hypertrophy (LVH). The aim of the analysis was to determine whether this effect is mediated by the renin-angiotensin-aldosterone system (RAAS) in hemodialysis. Serum samples from 62 randomized hemodialysis patients with LVH were analyzed for plasma renin activity (PRA-S), angiotensin II (AngII), and metabolites, angiotensin-converting enzyme-2 (ACE2) and aldosterone using a high throughput mass spectrometry assay. Compared to healthy individuals, levels of the RAAS parameters PRA-S, AngII and aldosterone were generally lower [median (IQR) PRA-S 130 (46–269) vs. 196 (98, 238) pmol/L; AngII 70 (28–157) vs. 137 (76, 201) pmol/L; Aldosterone 130 (54, 278) vs. 196 (98, 238) pmol/L]. We did not find an indication that the effect of FGF23 on LVH was mediated by RAAS parameters, with all estimated indirect effects virtually zero. Furthermore, FGF23 was not associated with RAAS parameter levels throughout the study. While there was a clear association between FGF23 levels and left ventricular mass index (LVMI) at the end of the study and in the FGF23 fold change and LVMI change analysis, no association between RAAS and LVMI was observed. Serum concentrations of PRA-S, AngII, and aldosterone were below the ranges measured in healthy controls suggesting that RAAS is not systemically activated in hemodialysis patients. The effect of FGF23 on LVMI was not mediated by systemic RAAS activity. These findings challenge the current paradigm of LVH progression and treatment with RAAS blockers in dialysis.
Collapse
Affiliation(s)
- Katharina Dörr
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | - Michael Kammer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria.,Center for Medical Statistics, Informatics, and Intelligent Systems, Section for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | | | | | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Cardiovascular and Interventional Radiology, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Huang S, Li Z, Jiang S, Xu M. Metabolomic study on the protective effect of isoorientin against myocardial infarction. Biochem Biophys Res Commun 2022; 598:81-88. [PMID: 35151208 DOI: 10.1016/j.bbrc.2022.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/03/2022] [Indexed: 12/28/2022]
Abstract
Myocardial infarction has become one of the largest threats to human life. Myocardial ischemia and hypoxia caused by myocardial infarction are important causes of myocardial cell injury. Compared with chemical drugs, botanical drugs that are natural antioxidants have relatively few toxic side effects. Isoorientin (ISO), a C-glucosyl flavone with a chemical nomenclature, exists in the human diet and has antioxidant and anti-inflammatory effects in other diseases. However, its role in myocardial infarction has not been reported. In this study, we investigated the effects of ISO administration on cardiac function in mice after myocardial infarction, on ROS levels in H9C2 myocardial cells after hypoxia in vitro, and on metabolomic changes in mice after myocardial infarction. We found that ISO improved cardiac function in mice after myocardial infarction and inhibited hypoxia-induced oxidative stress injury in H9C2 cells in vitro. We also found through metabolomic analysis and KEGG enrichment analysis that ISO significantly changed metabolic pathways in mice after myocardial infarction, including histidine metabolism, arachidonic acid metabolism, renin secretion and other pathways. These results lay a foundation for further exploration of the protective effect of ISO against myocardial infarction and the development of related drugs.
Collapse
Affiliation(s)
- Shaoman Huang
- Department of Cardiovascular Medicine, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang, 522000, China
| | - Zexiong Li
- Department of Cardiovascular Medicine, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang, 522000, China
| | - Shaoru Jiang
- Department of Cardiovascular Medicine, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang, 522000, China
| | - Mingwei Xu
- Department of Cardiovascular Medicine, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang, 522000, China.
| |
Collapse
|
37
|
Oomen PJA, Phung TKN, Weinberg SH, Bilchick KC, Holmes JW. A rapid electromechanical model to predict reverse remodeling following cardiac resynchronization therapy. Biomech Model Mechanobiol 2022; 21:231-247. [PMID: 34816336 PMCID: PMC9241386 DOI: 10.1007/s10237-021-01532-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
Abstract
Cardiac resynchronization therapy (CRT) is an effective therapy for patients who suffer from heart failure and ventricular dyssynchrony such as left bundle branch block (LBBB). When it works, it reverses adverse left ventricular (LV) remodeling and the progression of heart failure. However, CRT response rate is currently as low as 50-65%. In theory, CRT outcome could be improved by allowing clinicians to tailor the therapy through patient-specific lead locations, timing, and/or pacing protocol. However, this also presents a dilemma: there are far too many possible strategies to test during the implantation surgery. Computational models could address this dilemma by predicting remodeling outcomes for each patient before the surgery takes place. Therefore, the goal of this study was to develop a rapid computational model to predict reverse LV remodeling following CRT. We adapted our recently developed computational model of LV remodeling to simulate the mechanics of ventricular dyssynchrony and added a rapid electrical model to predict electrical activation timing. The model was calibrated to quantitatively match changes in hemodynamics and global and local LV wall mass from a canine study of LBBB and CRT. The calibrated model was used to investigate the influence of LV lead location and ischemia on CRT remodeling outcome. Our model results suggest that remodeling outcome varies with both lead location and ischemia location, and does not always correlate with short-term improvement in QRS duration. The results and time frame required to customize and run this model suggest promise for this approach in a clinical setting.
Collapse
Affiliation(s)
- Pim J. A. Oomen
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22903, USA
- Department of Medicine, University of Virginia, Box 800158, Health System, Charlottesville, VA 22903, USA
| | - Thien-Khoi N. Phung
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA 02115, USA
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Ohio State University, 140 W 19th Ave Columbus, Columbus, OH 43210, USA
| | - Kenneth C. Bilchick
- Department of Medicine, University of Virginia, Box 800158, Health System, Charlottesville, VA 22903, USA
| | - Jeffrey W. Holmes
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22903, USA
- School of Engineering, University of Alabama at Birmingham, 1075 13th St S, Birmingham, AL 35233, USA
| |
Collapse
|
38
|
Kato Y, Nishiyama K, Nishimura A, Nishida M. [Eco-pharma research aimed at developing COVID-19 therapeutic agent]. Nihon Yakurigaku Zasshi 2022; 157:119-123. [PMID: 35228443 DOI: 10.1254/fpj.21070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Novel coronavirus infection disease 2019 (COVID-19) is an emerging infectious disease that has been rampant worldwide since its onset was confirmed in Wuhan, China in 2019. An effective therapy has not yet been established, and there is an urgent need to establish a breakthrough therapeutic strategy for the prevention and treatment of COVID-19 aggravation. The main route of infection is that the Spike protein (S protein) on the surface of SARS-CoV-2 binds to its recognition receptor, angiotensin converting enzyme (ACE) 2, on the host cell surface. Then, SARS-CoV-2 invades the cell via endocytosis-dependent pathway. Although the major symptom of COVID-19 is lung inflammation, ACE2 is expressed not only in the lungs but also in various tissues including heart and digestive organs. We focused on the molecular mechanism underlying the development of heart failure, a pathology involved in COVID-19 aggravation risk factors and COVID-19 squeals. We revealed that cardiac ACE2 receptors were upregulated by exposure to various environmental stresses reported as COVID-19 aggravation risk factors, and the formation of membrane protein complex between TRPC3 and NADPH oxidase (Nox) 2 that participates in myocardial remodeling underlies pathological ACE2 upregulation. Furthermore, we utilized the already approved drugs that inhibit TRPC3-Nox2 protein complex formation, and identified that clomipramine, a tricyclic antidepressant, has the best potency to suppress ACE2 internalization induced by S protein exposure. This review introduces the mechanism of pathological ACE2 receptor upregulation through TRPC3-Nox2 complex formation in the heart, and the identification of a breakthrough drug candidate using in vitro pseudo-infection screening system.
Collapse
Affiliation(s)
- Yuri Kato
- Graduate School of Pharmaceutical Science, Kyushu University
| | | | - Akiyuki Nishimura
- National Institute for Physiological Sciences, National Institutes of Natural Sciences
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Science, Kyushu University
- National Institute for Physiological Sciences, National Institutes of Natural Sciences
| |
Collapse
|
39
|
Proteomic analysis of temperature-dependent developmental plasticity within the ventricle of juvenile Atlantic salmon (Salmo salar). Curr Res Physiol 2022; 5:344-354. [PMID: 36035983 PMCID: PMC9403292 DOI: 10.1016/j.crphys.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022] Open
|
40
|
Sun Y, Zhang Y, Xu N, Bi C, Liu X, Song W, Jiang Y. Assessing the causal role of hypertension on left atrial and left ventricular structure and function: A two-sample Mendelian randomization study. Front Cardiovasc Med 2022; 9:1006380. [PMID: 36407459 PMCID: PMC9666890 DOI: 10.3389/fcvm.2022.1006380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/18/2022] [Indexed: 11/22/2022] Open
Abstract
Aim The aim of this study was to investigate whether hypertension may be causally linked to left atrial (LA) and left ventricular (LV) structure and function. Methods and results We performed a two-Mendelian randomization (MR) analysis implementing the results from the FinnGen large-scale, genome-wide association study for hypertension (N = 218,754), and LV (N = 16,923) and LA studies (N = 35,648) by the UK Biobank to identify genetic instruments. The MR analysis was implemented using an inverse-variance weighted (IVW) approach. We identified a positive potential causal relationship between hypertension and indices for the LA maximum (LAmax with causal estimates of 0.126 [95% CI, (0.093 to 0.160)]); LA minimum (LAmin with causal estimates of 0.122 [95% CI, (0.089 to 0.156)]); LV function (causal estimates are LV end-diastolic volume (LVEDV), 0.078 [95% CI, (0.003 to 0.153)]; LV end-systolic volume (LVESV), 0.102 [95% CI, (0.030 to 0.173)]; LV mass (LVM), 0.171 [95% CI, (0.108 to 0.233)]; and LV mass to end-diastolic volume ratio (LVMVR at 0.098 [95% CI, (0.048 to 0.149)], respectively), which was directionally concordant with other robust MR methods. Other than this, we observed a significantly negative causal relationship between hypertension and the LA active emptying fraction (LAAEF), the LA passive emptying fraction (LAPEF), and the LA total emptying fraction (LATEF). Conclusion Our genetic analyses demonstrated a potential causal relationship between hypertension and the left atrium and left ventricle's structures and functions.
Collapse
Affiliation(s)
- Yancui Sun
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ying Zhang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Nan Xu
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Cheng Bi
- Department of Cardiology, Tieling Central Hospital, Tieling, Liaoning, China
| | - Xiaojie Liu
- Department of Cardiology, The Liaoyu Hospital of Dalian, Dalian, Liaoning, China
| | - Wei Song
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yinong Jiang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
41
|
From dissection of fibrotic pathways to assessment of drug interactions to reduce cardiac fibrosis and heart failure. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100036. [PMID: 34909666 PMCID: PMC8663973 DOI: 10.1016/j.crphar.2021.100036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac fibrosis is characterized by extracellular matrix deposition in the cardiac interstitium, and this contributes to cardiac contractile dysfunction and progression of heart failure. The main players involved in this process are the cardiac fibroblasts, which, in the presence of pro-inflammatory/pro-fibrotic stimuli, undergo a complete transformation acquiring a more proliferative, a pro-inflammatory and a secretory phenotype. This review discusses the cellular effectors and molecular pathways implicated in the pathogenesis of cardiac fibrosis and suggests potential strategies to monitor the effects of specific drugs designed to slow down the progression of this disease by specifically targeting the fibroblasts.
Collapse
|
42
|
Shi Y, Zhao L, Zhang Y, Qin Q, Cong H, Guo Z. Homocysteine promotes cardiac fibrosis by regulating the Akt/FoxO3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1732. [PMID: 35071426 PMCID: PMC8743705 DOI: 10.21037/atm-21-5602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Background Evaluated plasma homocysteine (Hcy) is an independent risk factor for cardiac fibrosis which is a common feature of cardiovascular disease, although the mechanisms are still unclear. This study aims to explore the mechanism of Hcy-induced cardiac fibrosis. Methods The mRNA and protein levels of Forkhead box O3 (FoxO3) and differentiation markers were detected in primary cardiac fibroblasts (CFs) after 300 µM Hcy treatment. Scratch and transwell migration assay were used to determine the effect of Hcy on proliferation and migration in CFs. The protein levels involved in the fibrotic processes in mice fed with high methionine diet (HMD) for 4 or 8 weeks were investigated by western blot. CFs were infected with FoxO3 recombinant adenovirus to explore the potential role of FoxO3 in Hcy-induced cardiac dysfunction. Results Hcy treatment significantly promoted the differentiation, proliferation and migration of CFs, while FoxO3 activity were decreased in CFs. In HMD hearts, the protein levels of TIMP1, Fibronectin and α-SMA were increased after 4 or 8 weeks, but the FoxO3 activity was decreased. Moreover, the HMD hearts had a higher level of Bcl2 but lower of Bax and LC3II protein. In addition, FoxO3 overexpression attenuates Hcy-induced dysfunction in CFs. Conclusions Hcy promotes myofibroblast activation and resistance to autophagy and apoptosis in CFs, and eventually results in cardiac fibrosis by regulating the Akt/FoxO3 pathway. Thus, FoxO3 is a promising therapeutic target to prevent cardiac remodeling.
Collapse
Affiliation(s)
- Ying Shi
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China.,Key Laboratory of Immune Microenvironment and Disease (Tianjin Medical University), Ministry of Education, Tianjin, China
| | - Lili Zhao
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Yifei Zhang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Qin Qin
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Hongliang Cong
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Zhigang Guo
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
43
|
Kessler EL, Wang JW, Kok B, Brans MA, Nederlof A, van Stuijvenberg L, Huang C, Vink A, Arslan F, Efimov IR, Lam CSP, Vos MA, de Kleijn DPV, Fontes MSC, van Veen TAB. Ventricular TLR4 Levels Abrogate TLR2-Mediated Adverse Cardiac Remodeling upon Pressure Overload in Mice. Int J Mol Sci 2021; 22:ijms222111823. [PMID: 34769252 PMCID: PMC8583975 DOI: 10.3390/ijms222111823] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Involvement of the Toll-like receptor 4 (TLR4) in maladaptive cardiac remodeling and heart failure (HF) upon pressure overload has been studied extensively, but less is known about the role of TLR2. Interplay and redundancy of TLR4 with TLR2 have been reported in other organs but were not investigated during cardiac dysfunction. We explored whether TLR2 deficiency leads to less adverse cardiac remodeling upon chronic pressure overload and whether TLR2 and TLR4 additively contribute to this. We subjected 35 male C57BL/6J mice (wildtype (WT) or TLR2 knockout (KO)) to sham or transverse aortic constriction (TAC) surgery. After 12 weeks, echocardiography and electrocardiography were performed, and hearts were extracted for molecular and histological analysis. TLR2 deficiency (n = 14) was confirmed in all KO mice by PCR and resulted in less hypertrophy (heart weight to tibia length ratio (HW/TL), smaller cross-sectional cardiomyocyte area and decreased brain natriuretic peptide (BNP) mRNA expression, p < 0.05), increased contractility (QRS and QTc, p < 0.05), and less inflammation (e.g., interleukins 6 and 1β, p < 0.05) after TAC compared to WT animals (n = 11). Even though TLR2 KO TAC animals presented with lower levels of ventricular TLR4 mRNA than WT TAC animals (13.2 ± 0.8 vs. 16.6 ± 0.7 mg/mm, p < 0.01), TLR4 mRNA expression was increased in animals with the largest ventricular mass, highest hypertrophy, and lowest ejection fraction, leading to two distinct groups of TLR2 KO TAC animals with variations in cardiac remodeling. This variation, however, was not seen in WT TAC animals even though heart weight/tibia length correlated with expression of TLR4 in these animals (r = 0.078, p = 0.005). Our data suggest that TLR2 deficiency ameliorates adverse cardiac remodeling and that ventricular TLR2 and TLR4 additively contribute to adverse cardiac remodeling during chronic pressure overload. Therefore, both TLRs may be therapeutic targets to prevent or interfere in the underlying molecular processes.
Collapse
Affiliation(s)
- Elise L. Kessler
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- Correspondence: ; Tel.: +31-628706156
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore; (J.-W.W.); (C.H.)
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore 117599, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Bart Kok
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Maike A. Brans
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
| | - Angelique Nederlof
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Leonie van Stuijvenberg
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore; (J.-W.W.); (C.H.)
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands;
| | - Fatih Arslan
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands
| | - Igor R. Efimov
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA;
| | - Carolyn S. P. Lam
- National Heart Centre Singapore and Duke-National University of Singapore, 5 Hospital Dr, Singapore 169609, Singapore;
- UMC Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Marc A. Vos
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Dominique P. V. de Kleijn
- Department of Vascular Surgery, The Netherlands & Netherlands Heart Institute, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- The Netherlands Heart Institute, Moreelsepark 1, 3511EP Utrecht, The Netherlands
| | - Magda S. C. Fontes
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| |
Collapse
|
44
|
Sankarankutty AC, Greiner J, Bragard J, Visker JR, Shankar TS, Kyriakopoulos CP, Drakos SG, Sachse FB. Etiology-Specific Remodeling in Ventricular Tissue of Heart Failure Patients and Its Implications for Computational Modeling of Electrical Conduction. Front Physiol 2021; 12:730933. [PMID: 34675817 PMCID: PMC8523803 DOI: 10.3389/fphys.2021.730933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
With an estimated 64.3 million cases worldwide, heart failure (HF) imposes an enormous burden on healthcare systems. Sudden death from arrhythmia is the major cause of mortality in HF patients. Computational modeling of the failing heart provides insights into mechanisms of arrhythmogenesis, risk stratification of patients, and clinical treatment. However, the lack of a clinically informed approach to model cardiac tissues in HF hinders progress in developing patient-specific strategies. Here, we provide a microscopy-based foundation for modeling conduction in HF tissues. We acquired 2D images of left ventricular tissues from HF patients (n = 16) and donors (n = 5). The composition and heterogeneity of fibrosis were quantified at a sub-micrometer resolution over an area of 1 mm2. From the images, we constructed computational bidomain models of tissue electrophysiology. We computed local upstroke velocities of the membrane voltage and anisotropic conduction velocities (CV). The non-myocyte volume fraction was higher in HF than donors (39.68 ± 14.23 vs. 22.09 ± 2.72%, p < 0.01), and higher in ischemic (IC) than nonischemic (NIC) cardiomyopathy (47.2 ± 16.18 vs. 32.16 ± 6.55%, p < 0.05). The heterogeneity of fibrosis within each subject was highest for IC (27.1 ± 6.03%) and lowest for donors (7.47 ± 1.37%) with NIC (15.69 ± 5.76%) in between. K-means clustering of this heterogeneity discriminated IC and NIC with an accuracy of 81.25%. The heterogeneity in CV increased from donor to NIC to IC tissues. CV decreased with increasing fibrosis for longitudinal (R 2 = 0.28, p < 0.05) and transverse conduction (R 2 = 0.46, p < 0.01). The tilt angle of the CV vectors increased 2.1° for longitudinal and 0.91° for transverse conduction per 1% increase in fibrosis. Our study suggests that conduction fundamentally differs in the two etiologies due to the characteristics of fibrosis. Our study highlights the importance of the etiology-specific modeling of HF tissues and integration of medical history into electrophysiology models for personalized risk stratification and treatment planning.
Collapse
Affiliation(s)
- Aparna C Sankarankutty
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Joachim Greiner
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg⋅Bad Krozingen, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jean Bragard
- Department of Physics and Applied Mathematics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Joseph R Visker
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Christos P Kyriakopoulos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
45
|
Tsuda T. Clinical Assessment of Ventricular Wall Stress in Understanding Compensatory Hypertrophic Response and Maladaptive Ventricular Remodeling. J Cardiovasc Dev Dis 2021; 8:122. [PMID: 34677191 PMCID: PMC8538325 DOI: 10.3390/jcdd8100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/12/2023] Open
Abstract
Ventricular wall stress (WS) is an important hemodynamic parameter to represent myocardial oxygen demand and ventricular workload. The normalization of WS is regarded as a physiological feedback signal that regulates the rate and extent of ventricular hypertrophy to maintain myocardial homeostasis. Although hypertrophy is an adaptive response to increased biomechanical stress, persistent hypertrophic stimulation forces the stressed myocardium into a progressive maladaptive process called ventricular remodeling, consisting of ventricular dilatation and dysfunction in conjunction with the development of myocyte hypertrophy, apoptosis, and fibrosis. The critical determinant of this pathological transition is not fully understood, but an energetic mismatch due to uncontrolled WS is thought to be a central mechanism. Despite extensive basic investigations conducted to understand the complex signaling pathways involved in this maladaptive process, clinical diagnostic studies that translate these molecular and cellular changes are relatively limited. Echocardiographic assessment with or without direct measurement of left ventricular pressure used to be a mainstay in estimating ventricular WS in clinical medicine, but in recent years more and more noninvasive applications with magnetic resonance imaging have been studied. In this review article, basic clinical applications of WS assessment are discussed to help understand the progression of ventricular remodeling.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours Children’s Hospital Delaware, 1600 Rockland Rd, Wilmington, DE 19803, USA; ; Tel.: +1-(302)-651-6677; Fax: +1-(302)-651-6601
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, 11th and Walnut Street, Philadelphia, PA 19107, USA
| |
Collapse
|
46
|
Stoeck CT, von Deuster C, Fuetterer M, Polacin M, Waschkies CF, van Gorkum RJH, Kron M, Fleischmann T, Cesarovic N, Weisskopf M, Kozerke S. Cardiovascular magnetic resonance imaging of functional and microstructural changes of the heart in a longitudinal pig model of acute to chronic myocardial infarction. J Cardiovasc Magn Reson 2021; 23:103. [PMID: 34538266 PMCID: PMC8451129 DOI: 10.1186/s12968-021-00794-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND We examined the dynamic response of the myocardium to infarction in a longitudinal porcine study using relaxometry, functional as well as diffusion cardiovascular magnetic resonance (CMR). We sought to compare non contrast CMR methods like relaxometry and in-vivo diffusion to contrast enhanced imaging and investigate the link of microstructural and functional changes in the acute and chronically infarcted heart. METHODS CMR was performed on five myocardial infarction pigs and four healthy controls. In the infarction group, measurements were obtained 2 weeks before 90 min occlusion of the left circumflex artery, 6 days after ischemia and at 5 as well as 9 weeks as chronic follow-up. The timing of measurements was replicated in the control cohort. Imaging consisted of functional cine imaging, 3D tagging, T2 mapping, native as well as gadolinium enhanced T1 mapping, cardiac diffusion tensor imaging, and late gadolinium enhancement imaging. RESULTS Native T1, extracellular volume (ECV) and mean diffusivity (MD) were significantly elevated in the infarcted region while fractional anisotropy (FA) was significantly reduced. During the transition from acute to chronic stages, native T1 presented minor changes (< 3%). ECV as well as MD increased from acute to the chronic stages compared to baseline: ECV: 125 ± 24% (day 6) 157 ± 24% (week 5) 146 ± 60% (week 9), MD: 17 ± 7% (day 6) 33 ± 14% (week 5) 29 ± 15% (week 9) and FA was further reduced: - 31 ± 10% (day 6) - 38 ± 8% (week 5) - 36 ± 14% (week 9). T2 as marker for myocardial edema was significantly increased in the ischemic area only during the acute stage (83 ± 3 ms infarction vs. 58 ± 2 ms control p < 0.001 and 61 ± 2 ms in the remote area p < 0.001). The analysis of functional imaging revealed reduced left ventricular ejection fraction, global longitudinal strain and torsion in the infarct group. At the same time the transmural helix angle (HA) gradient was steeper in the chronic follow-up and a correlation between longitudinal strain and transmural HA gradient was detected (r = 0.59 with p < 0.05). Comparing non-gadolinium enhanced data T2 mapping showed the largest relative change between infarct and remote during the acute stage (+ 33 ± 4% day 6, with p = 0.013 T2 vs. MD, p = 0.009 T2 vs. FA and p = 0.01 T2 vs. T1) while FA exhibited the largest relative change between infarct and remote during the chronic follow-up (+ 31 ± 2% week 5, with p = N.S. FA vs. MD, p = 0.03 FA vs. T2 and p = 0.003 FA vs. T1). Overall, diffusion parameters provided a higher contrast (> 23% for MD and > 27% for FA) during follow-up compared to relaxometry (T1 17-18%/T2 10-20%). CONCLUSION During chronic follow-up after myocardial infarction, cardiac diffusion tensor imaging provides a higher sensitivity for mapping microstructural alterations when compared to non-contrast enhanced relaxometry with the added benefit of providing directional tensor information to assess remodelling of myocyte aggregate orientations, which cannot be otherwise assessed.
Collapse
Affiliation(s)
- Christian T. Stoeck
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Constantin von Deuster
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Maximilian Fuetterer
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Malgorzata Polacin
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Conny F. Waschkies
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Robbert J. H. van Gorkum
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Mareike Kron
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Thea Fleischmann
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
- Institute of Translational Cardiovascular Technologies, ETH Zurich, Zurich, Switzerland
| | - Miriam Weisskopf
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Sebastian Kozerke
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| |
Collapse
|
47
|
Bhullar SK, Shah AK, Dhalla NS. Role of angiotensin II in the development of subcellular remodeling
in heart failure. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of heart failure under various pathological conditions such as myocardial infarction (MI), hypertension and diabetes are accompanied by adverse cardiac remodeling and cardiac dysfunction. Since heart function is mainly determined by coordinated activities of different subcellular organelles including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils for regulating the intracellular concentration of Ca2+, it has been suggested that the occurrence of heart failure is a consequence of subcellular remodeling, metabolic alterations and Ca2+-handling abnormalities in cardiomyocytes. Because of the elevated plasma levels of angiotensin II (ANG II) due to activation of the renin-angiotensin system (RAS) in heart failure, we have evaluated the effectiveness of treatments with angiotensin converting enzyme (ACE) inhibitors and ANG II type 1 receptor (AT1R) antagonists in different experimental models of heart failure. Attenuation of marked alterations in subcellular activities, protein content and gene expression were associated with improvement in cardiac function in MI-induced heart failure by treatment with enalapril (an ACE inhibitor) or losartan (an AT1R antagonist). Similar beneficial effects of ANG II blockade on subcellular remodeling and cardiac performance were also observed in failing hearts due to pressure overload, volume overload or chronic diabetes. Treatments with enalapril and losartan were seen to reduce the degree of RAS activation as well as the level of oxidative stress in failing hearts. These observations provide evidence which further substantiate to support the view that activation of RAS and high level of plasma ANG II play a critical role in inducing subcellular defects and cardiac dys-function during the progression of heart failure.
Collapse
Affiliation(s)
- Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada
| | - Anureet K. Shah
- School of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada; Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 3P5, Canada
| |
Collapse
|
48
|
Abstract
Cardiac hypertrophy, characterized by the enlargement of cardiomyocytes, is initially an adaptive response to physiological and pathological stimuli. Decompensated cardiac hypertrophy is related to fibrosis, inflammatory cytokine, maladaptive remodeling, and heart failure. Although pathological myocardial hypertrophy is the main cause of hypertrophy-related morbidity and mortality, our understanding of its mechanism is still poor. Long noncoding RNAs (lncRNAs) are noncoding RNAs that regulate various physiological and pathological processes through multiple molecular mechanisms. Recently, accumulating evidence has indicated that lncRNA-H19 is a potent regulator of the progression of cardiac hypertrophy. For the first time, this review summarizes the current studies about the role of lncRNA-H19 in cardiac hypertrophy, including its pathophysiological processes and underlying pathological mechanism, including calcium regulation, fibrosis, apoptosis, angiogenesis, inflammation, and methylation. The context within which lncRNA-H19 might be developed as a target for cardiac hypertrophy treatment is then discussed to gain better insight into the possible biological functions of lncRNA-H19 in cardiac hypertrophy.
Collapse
|
49
|
A small-molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase improves obesity, nephropathy and cardiomyopathy in obese ZSF1 rats. PLoS One 2021; 16:e0255022. [PMID: 34339435 PMCID: PMC8328318 DOI: 10.1371/journal.pone.0255022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Prolyl hydroxylase (PH) enzymes control the degradation of hypoxia-inducible factor (HIF), a transcription factor known to regulate erythropoiesis, angiogenesis, glucose metabolism, cell proliferation, and apoptosis. HIF-PH inhibitors (HIF-PHIs) correct anemia in patients with renal disease and in animal models of anemia and kidney disease. However, the effects of HIF-PHIs on comorbidities associated with kidney disease remain largely unknown. We evaluated the effects of the HIF-PHI FG-2216 in obese ZSF1 (Ob-ZSF1) rats, an established model of kidney failure with metabolic syndrome. Following unilateral nephrectomy (Nx) at 8 weeks of age, rats were treated with 40 mg/kg FG-2216 or vehicle by oral gavage three times per week for up to 18 weeks. FG-2216 corrected blood hemoglobin levels and improved kidney function and histopathology in Nx-Ob-ZSF1 rats by increasing the glomerular filtration rate, decreasing proteinuria, and reducing peritubular fibrosis, tubular damage, glomerulosclerosis and mesangial expansion. FG-2216 increased renal glucose excretion and decreased body weight, fat pad weight, and serum cholesterol in Nx-Ob-ZSF1 rats. Additionally, FG-2216 corrected hypertension, improved diastolic and systolic heart function, and reduced cardiac hypertrophy and fibrosis. In conclusion, the HIF-PHI FG-2216 improved renal and cardiovascular outcomes, and reduced obesity in a rat model of kidney disease with metabolic syndrome. Thus, in addition to correcting anemia, HIF-PHIs may provide renal and cardiac protection to patients suffering from kidney disease with metabolic syndrome.
Collapse
|
50
|
Zhou Y, Murugan DD, Khan H, Huang Y, Cheang WS. Roles and Therapeutic Implications of Endoplasmic Reticulum Stress and Oxidative Stress in Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:antiox10081167. [PMID: 34439415 PMCID: PMC8388996 DOI: 10.3390/antiox10081167] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
In different pathological states that cause endoplasmic reticulum (ER) calcium depletion, altered glycosylation, nutrient deprivation, oxidative stress, DNA damage or energy perturbation/fluctuations, the protein folding process is disrupted and the ER becomes stressed. Studies in the past decade have demonstrated that ER stress is closely associated with pathogenesis of obesity, insulin resistance and type 2 diabetes. Excess nutrients and inflammatory cytokines associated with metabolic diseases can trigger or worsen ER stress. ER stress plays a critical role in the induction of endothelial dysfunction and atherosclerosis. Signaling pathways including AMP-activated protein kinase and peroxisome proliferator-activated receptor have been identified to regulate ER stress, whilst ER stress contributes to the imbalanced production between nitric oxide (NO) and reactive oxygen species (ROS) causing oxidative stress. Several drugs or herbs have been proved to protect against cardiovascular diseases (CVD) through inhibition of ER stress and oxidative stress. The present article reviews the involvement of ER stress and oxidative stress in cardiovascular dysfunction and the potential therapeutic implications.
Collapse
Affiliation(s)
- Yan Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China;
| | - Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China;
- Correspondence: ; Tel.: +853-8822-4914
| |
Collapse
|