1
|
Chen Z, Cheng Z, Ding C, Cao T, Chen L, Wang H, Li J, Huang X. ROS-Activated TRPM2 Channel: Calcium Homeostasis in Cardiovascular/renal System and Speculation in Cardiorenal Syndrome. Cardiovasc Drugs Ther 2025; 39:615-631. [PMID: 38108918 DOI: 10.1007/s10557-023-07531-3] [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] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
The transient receptor potential melastatin 2 (TRPM2) channel is a nonselective calcium channel that is sensitive to oxidative stress (OS), and is widely expressed in multiple organs, such as the heart, kidney, and brain, which is inextricably related to calcium dyshomeostasis and downstream pathological events. Due to the increasing global burden of kidney or cardiovascular diseases (CVDs), safe and efficient drugs specific to novel targets are imperatively needed. Notably, investigation of the possibility to regard the TRPM2 channel as a new therapeutic target in ROS-related CVDs or renal diseases is urgently required because the roles of the TRPM2 channel in heart or kidney diseases have not received enough attention and thus have not been fully elaborated. Therefore, we aimed to review the involvement of the TRPM2 channel in cardiovascular disorders related to kidney or typical renal diseases and attempted to speculate about TRPM2-mediated mechanisms of cardiorenal syndrome (CRS) to provide representative perspectives for future research about novel and effective therapeutic strategies.
Collapse
Affiliation(s)
- Zihan Chen
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, China
| | - Zaihua Cheng
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Congcong Ding
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tianyu Cao
- Biological anthropology, University of California, Santa Barbara, CA, USA
| | - Ling Chen
- Department of Cardiology, the First People's Hospital of Jiujiang, Jiujiang, China
| | - Hong Wang
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Junpei Li
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Xiao Huang
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
2
|
Thirugnanam K, Rizvi F, Jahangir A, Homar P, Shabnam F, Palecek SP, Kumar SN, Pan A, Bai X, Sekine H, Ramchandran R. SNRK regulates TGFβ levels in atria to control cardiac fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.612951. [PMID: 39386731 PMCID: PMC11463613 DOI: 10.1101/2024.09.24.612951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Atrial fibrosis is central to the pathology of heart failure (HF) and atrial fibrillation (AF). Identifying precise mechanisms underlying atrial fibrosis will provide effective strategies for clinical intervention. This study investigates a metabolic serine threonine kinase gene, sucrose non-fermenting related kinase (SNRK), that we previously reported to control cardiac metabolism and function. Conditional knockout of Snrk in mouse cardiomyocytes ( Snrk cmcKO) leads to atrial fibrosis and subsequently HF. The precise mechanism underlying cardiomyocyte SNRK-driven repression of fibrosis is not known. Here, using mouse, rat, and human tissues, we demonstrate that SNRK expression is high in atria, especially in atrial cardiomyocytes. SNRK expression correlates with lower levels of pro-fibrotic protein transforming growth factor-beta 1 (TGFβ1) in the atrial cardiomyocytes. In HL-1 adult immortalized mouse atrial cells, using siRNA approaches, we show that Snrk knockdown cells show more TGFβ1 secretion, which was also observed in heart lysates from Snrk cardiac-specific knockout mice in vivo. These effects were exacerbated upon infusion of Angiotensin II. Results from Snrk knockdown cardiomyocytes co-cultured with cardiac fibroblasts suggest that SNRK represses TGFβ1 signaling (Smad 2/3) in atrial CMs and prevents paracrine cardiac fibroblast activation (α-SMA marker). In conclusion, high SNRK expression in atria regulates cardiac homeostasis, by preventing the release of TGFβ1 secretion to block cardiac fibrosis. These studies will assist in developing heart chamber-specific fibrosis therapy for non-ischemic HF and AF.
Collapse
|
3
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
4
|
Si W, Teng P, Ma L. Model for predicting the recurrence of atrial fibrillation after monopolar or bipolar radiofrequency ablation in patients with AF and mitral valve disease. J Cardiothorac Surg 2024; 19:290. [PMID: 38750504 PMCID: PMC11097403 DOI: 10.1186/s13019-024-02742-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/29/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVES This study aimed to identify the risk factors for postoperative atrial fibrillation in patients with valvular atrial fibrillation, and establish predictive models of atrial fibrillation recurrence. METHODS Overall, 224 patients who underwent radiofrequency ablation of atrial fibrillation from November 2014 to November 2020 were included. The statistical package for social sciences, X-tile, and R-studio were used for statistical analysis. RESULTS Patients were divided into training and validation sets according to a ratio of 3:1. The training set was analysed using univariate and multivariate Cox regression analysis and showed that preoperative uric acid > 401 μmol/L (P = 0.006), B-type natriuretic peptide > 202 ng/L (P = 0.042), hypersensitivity C-reactive protein > 6.1 mg/L (P = 0.026), erythrocyte sedimentation rate > 7.0 mm/h (P = 0.016), preoperative left atrial diameter > 48 mm (P = 0.031) were significantly correlated with the recurrence of atrial fibrillation after radiofrequency ablation in patients with valvular atrial fibrillation. In the training set, a Cox regression model of the five related factors was established using the R language. The C-index of the model was 0.82, and the area under the receiver operating characteristic curve was 0.831 (P < 0.001). Internal and external verification was performed in the training and validation sets, respectively, and both showed that the fit of the verification curve was relatively good at 3 months, 6 months, 1 year, and 3 years postoperatively. After calculating the weight of each related factor using the nomogram, a new risk predictive model (BLUCE) for postoperative atrial fibrillation was established. CONCLUSIONS In patients with atrial fibrillation, preoperative uric acid, B-type natriuretic peptide, hypersensitivity C-reactive protein, erythrocyte sedimentation rate, and left atrial diameter are risk factors for atrial fibrillation or atrial flutter recurrence after radiofrequency ablation. The BLUCE predictive model can distinguish high-risk groups of postoperative atrial fibrillation. High-risk patients in the BLUCE model were more likely to experience recurrence of atrial fibrillation after radiofrequency ablation and a low possibility of maintaining sinus rhythm.
Collapse
Affiliation(s)
- Wei Si
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Teng
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Ma
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Lai YJ, Chang SH, Tung YC, Chang GJ, Almeida CD, Chen WJ, Yeh YH, Tsai FC. Naringin activates semaphorin 3A to ameliorate TGF-β-induced endothelial-to-mesenchymal transition related to atrial fibrillation. J Cell Physiol 2024; 239:e31248. [PMID: 38501506 DOI: 10.1002/jcp.31248] [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: 07/25/2023] [Revised: 02/25/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
The loss of semaphorin 3A (Sema3A), which is related to endothelial-to-mesenchymal transition (EndMT) in atrial fibrosis, is implicated in the pathogenesis of atrial fibrillation (AF). To explore the mechanisms by which EndMT affects atrial fibrosis and assess the potential of a Sema3A activator (naringin) to prevent atrial fibrosis by targeting transforming growth factor-beta (TGF-β)-induced EndMT, we used human atria, isolated human atrial endocardial endothelial cells (AEECs), and used transgenic mice expressing TGF-β specifically in cardiac tissues (TGF-β transgenic mice). We evaluated an EndMT marker (Twist), a proliferation marker (proliferating cell nuclear antigen; PCNA), and an endothelial cell (EC) marker (CD31) through triple immunohistochemistry and confirmed that both EndMT and EC proliferation contribute to atrial endocardial fibrosis during AF in TGF-β transgenic mice and AF patient tissue sections. Additionally, we investigated the impact of naringin on EndMT and EC proliferation in AEECs and atrial fibroblasts. Naringin exhibited an antiproliferative effect, to which AEECs were more responsive. Subsequently, we downregulated Sema3A in AEECs using small interfering RNA to clarify a correlation between the reduction in Sema3A and the elevation of EndMT markers. Naringin treatment induced the expression of Sema3A and a concurrent decrease in EndMT markers. Furthermore, naringin administration ameliorated AF and endocardial fibrosis in TGF-β transgenic mice by stimulating Sema3A expression, inhibiting EndMT markers, reducing atrial fibrosis, and lowering AF vulnerability. This suggests therapeutic potential for naringin in AF treatment.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Respiratory Care, Chang-Gung University of Science and Technology, Chia-Yi, Puzi, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Ying-Chang Tung
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Celina De Almeida
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Feng-Chun Tsai
- Department of Surgery, Division of Cardiovascular Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Kostin S, Richter M, Ganceva N, Sasko B, Giannakopoulos T, Ritter O, Szalay Z, Pagonas N. Atrial fibrillation in human patients is associated with increased collagen type V and TGFbeta1. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VASCULATURE 2024; 50:101327. [PMID: 38419608 PMCID: PMC10899732 DOI: 10.1016/j.ijcha.2023.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 03/02/2024]
Abstract
Background and aim Atrial fibrosis is an important factor in initiating and maintaining atrial fibrillation (AF). Collagen V belongs to fibrillar collagens. There are, however no data on collagen V in AF. The aim of this work was to study the quantity of collagen V and its relationship with the number of fibroblasts and TGF- b 1 expression in patients in sinus rhythm (SR) and in patients with atrial fibrillation (AF). Methods We used quantitative immuhistochemistry to study collagen V in right and left atrial biopsies obtained from 35 patients in SR, 35 patients with paroxysmal AF (pAF) and 27 patients with chronic, long-standing persistent AF (cAF). In addition, we have quantified the number of vimentin-positive fibroblasts and expression levels of TGF-β1. Results Compared to patients in SR, collagen V was increased 1.8- and 3.1-fold in patients with pAF and cAF, respectively. In comparison with SR patients, the number of vimentin-positive cells increased significantly 1.46- and 1.8-fold in pAF and cAF patients, respectively.Compared to SR patients, expression levels of TGF-ß1, expressed as fluorescence units per tissue area, was significantly increased by 77 % and 300 % in patients with pAF and cAF, respectively. Similar to intensity measurements, the number of TGFß1-positive cells per 1 mm2 atrial tissue increased significantly from 35.5 ± 5.5 cells in SR patients to 61.9 ± 12.4 cells in pAF and 131.5 ± 23.5 cells in cAF. In both types of measurements, there was a statistically significant difference between pAF and cAF groups. Conclusions This is the first study to show that AF is associated with increased expression levels of collagen V and TGF-ß1indicating its role in the pathogenesis of atrial fibrosis. In addition, increases in collagen V correlate with increased number of fibroblasts and TGF-β1 and are more pronounced in cAF patients than those in pAF patients.
Collapse
Affiliation(s)
- Sawa Kostin
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Natalia Ganceva
- Department of Anesthesiology and Intensive Care, Kerckoff-Clinic, Bad Nauheim, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr-University of Bochum, Germany
| | | | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Cardiology, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Zoltan Szalay
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Nikolaos Pagonas
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Internal Medicine, University Hospital Ruppin-Brandenburg, Neuruppin, Germany
| |
Collapse
|
7
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [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: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Chalise U, Hale TM. Fibroblasts under pressure: cardiac fibroblast responses to hypertension and antihypertensive therapies. Am J Physiol Heart Circ Physiol 2024; 326:H223-H237. [PMID: 37999643 PMCID: PMC11219059 DOI: 10.1152/ajpheart.00401.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Approximately 50% of Americans have hypertension, which significantly increases the risk of heart failure. In response to increased peripheral resistance in hypertension, intensified mechanical stretch in the myocardium induces cardiomyocyte hypertrophy and fibroblast activation to withstand increased pressure overload. This changes the structure and function of the heart, leading to pathological cardiac remodeling and eventual progression to heart failure. In the presence of hypertensive stimuli, cardiac fibroblasts activate and differentiate to myofibroblast phenotype capable of enhanced extracellular matrix secretion in coordination with other cell types, mainly cardiomyocytes. Both systemic and local renin-angiotensin-aldosterone system activation lead to increased angiotensin II stimulation of fibroblasts. Angiotensin II directly activates fibrotic signaling such as transforming growth factor β/SMAD and mitogen-activated protein kinase (MAPK) signaling to produce extracellular matrix comprised of collagens and matricellular proteins. With the advent of single-cell RNA sequencing techniques, heterogeneity in fibroblast populations has been identified in the left ventricle in models of hypertension and pressure overload. The various clusters of fibroblasts reveal a range of phenotypes and activation states. Select antihypertensive therapies have been shown to be effective in limiting fibrosis, with some having direct actions on cardiac fibroblasts. The present review focuses on the fibroblast-specific changes that occur in response to hypertension and pressure overload, the knowledge gained from single-cell analyses, and the effect of antihypertensive therapies. Understanding the dynamics of hypertensive fibroblast populations and their similarities and differences by sex is crucial for the advent of new targets and personalized medicine.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix, Arizona, United States
| |
Collapse
|
9
|
Ruf L, Bukowska A, Gardemann A, Goette A. Coagulation Factor Xa Has No Effects on the Expression of PAR1, PAR2, and PAR4 and No Proinflammatory Effects on HL-1 Cells. Cells 2023; 12:2849. [PMID: 38132169 PMCID: PMC10741780 DOI: 10.3390/cells12242849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Atrial fibrillation (AF), characterised by irregular high-frequency contractions of the atria of the heart, is of increasing clinical importance. The reasons are the increasing prevalence and thromboembolic complications caused by AF. So-called atrial remodelling is characterised, among other things, by atrial dilatation and fibrotic remodelling. As a result, AF is self-sustaining and forms a procoagulant state. But hypercoagulation not only appears to be the consequence of AF. Coagulation factors can exert influence on cells via protease-activated receptors (PAR) and thereby the procoagulation state could contribute to the development and maintenance of AF. In this work, the influence of FXa on Heart Like-1 (HL-1) cells, which are murine adult atrial cardiomyocytes (immortalized), was investigated. PAR1, PAR2, and PAR4 expression was detected. After incubations with FXa (5-50 nM; 4-24 h) or PAR1- and PAR2-agonists (20 µM; 4-24 h), no changes occurred in PAR expression or in the inflammatory signalling cascade. There were no time- or concentration-dependent changes in the phosphorylation of the MAP kinases ERK1/2 or the p65 subunit of NF-κB. In addition, there was no change in the mRNA expression of the cell adhesion molecules (ICAM-1, VCAM-1, fibronectin). Thus, FXa has no direct PAR-dependent effects on HL-1 cells. Future studies should investigate the influence of FXa on human cardiomyocytes or on other cardiac cell types like fibroblasts.
Collapse
Affiliation(s)
- Lukas Ruf
- Institute of Clinical Chemistry and Pathobiochemistry, Department of Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Alicja Bukowska
- Institute of Clinical Chemistry and Pathobiochemistry, Department of Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Andreas Gardemann
- Institute of Clinical Chemistry and Pathobiochemistry, Department of Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Andreas Goette
- Institute of Clinical Chemistry and Pathobiochemistry, Department of Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
- Department of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Am Busdorf 2, 33098 Paderborn, Germany
| |
Collapse
|
10
|
Zhang K, Ernst L, Schobert I, Philipp K, Böning G, Heinzel FR, Boldt LH, Gehle P. Is Marfan Syndrome Associated with Primary Structural Changes in the Left Atrium? Diagnostics (Basel) 2023; 13:3278. [PMID: 37892098 PMCID: PMC10606247 DOI: 10.3390/diagnostics13203278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Marfan syndrome (MFS) is an autosomal-dominant multisystem connective tissue disorder that is based on mutations in the FBN1 gene and variably affects different organs, including the heart. In this study, we investigated cardiac function with a focus on the left atrium (LA) in a relatively large cohort of patients with MFS. After screening of 1165 patients that had been examined in our center between 2016 and 2020, 231 adult MFS patients with and without aortic operation were included in our study and compared to a healthy control group (n = 106). Cardiac function was assessed by transthoracic echocardiography and NT-proBNP was used as a secretory marker. Most (94.8%) of the patients received genetic testing. Left ventricular function was within normal ranges and not impaired. Interestingly, we found that LA size and secretory activity were increased in MFS patients, despite normal left ventricular filling pressures. This finding was even more pronounced in MFS patients with prior aortic surgery. A correlation between LA size or NT-proBNP levels and the type of pathogenic FBN1 variant could not be identified. Right ventricular function and right atrial size were increased only in MFS patients that had undergone aortic surgery. In conclusion, these findings suggest that MFS leads to structural changes in the LA that are not solely resulting from left ventricular dysfunction, but probably can be considered a primary pathology of MFS.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
| | - Lucas Ernst
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Isabel Schobert
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Karla Philipp
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Georg Böning
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Frank R. Heinzel
- 2. Medical Clinic, Municipal Clinic Dresden, 01324 Dresden, Germany
| | - Leif-Hendrik Boldt
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Petra Gehle
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
| |
Collapse
|
11
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
12
|
Sweat ME, Cao Y, Zhang X, Burnicka-Turek O, Perez-Cervantes C, Arulsamy K, Lu F, Keating EM, Akerberg BN, Ma Q, Wakimoto H, Gorham JM, Hill LD, Kyoung Song M, Trembley MA, Wang P, Gianeselli M, Prondzynski M, Bortolin RH, Bezzerides VJ, Chen K, Seidman JG, Seidman CE, Moskowitz IP, Pu WT. Tbx5 maintains atrial identity in post-natal cardiomyocytes by regulating an atrial-specific enhancer network. NATURE CARDIOVASCULAR RESEARCH 2023; 2:881-898. [PMID: 38344303 PMCID: PMC10854392 DOI: 10.1038/s44161-023-00334-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/21/2023] [Indexed: 02/15/2024]
Abstract
Understanding how the atrial and ventricular heart chambers maintain distinct identities is a prerequisite for treating chamber-specific diseases. Here, we selectively knocked out (KO) the transcription factor Tbx5 in the atrial working myocardium to evaluate its requirement for atrial identity. Atrial Tbx5 inactivation downregulated atrial cardiomyocyte (aCM) selective gene expression. Using concurrent single nucleus transcriptome and open chromatin profiling, genomic accessibility differences were identified between control and Tbx5 KO aCMs, revealing that 69% of the control-enriched ATAC regions were bound by TBX5. Genes associated with these regions were downregulated in KO aCMs, suggesting they function as TBX5-dependent enhancers. Comparing enhancer chromatin looping using H3K27ac HiChIP identified 510 chromatin loops sensitive to TBX5 dosage, and 74.8% of control-enriched loops contained anchors in control-enriched ATAC regions. Together, these data demonstrate TBX5 maintains the atrial gene expression program by binding to and preserving the tissue-specific chromatin architecture of atrial enhancers.
Collapse
Affiliation(s)
- Mason E. Sweat
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Yangpo Cao
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Ozanna Burnicka-Turek
- Department of Pediatrics, Pathology, and Human Genetics, The University of Chicago, Chicago, IL
| | - Carlos Perez-Cervantes
- Department of Pediatrics, Pathology, and Human Genetics, The University of Chicago, Chicago, IL
| | - Kulandai Arulsamy
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Erin M. Keating
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Brynn N. Akerberg
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua M. Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren D. Hill
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Mi Kyoung Song
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Michael A. Trembley
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Peizhe Wang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Matteo Gianeselli
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | | | - Raul H. Bortolin
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | | | - Kaifu Chen
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| | - Jonathan G. Seidman
- Department of Pediatrics, Pathology, and Human Genetics, The University of Chicago, Chicago, IL
| | - Christine E. Seidman
- Department of Pediatrics, Pathology, and Human Genetics, The University of Chicago, Chicago, IL
| | - Ivan P. Moskowitz
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115
| |
Collapse
|
13
|
Sweeney M, O’Fee K, Villanueva-Hayes C, Rahman E, Lee M, Vanezis K, Andrew I, Lim WW, Widjaja A, Barton PJR, Cook SA. Cardiomyocyte-Restricted Expression of IL11 Causes Cardiac Fibrosis, Inflammation, and Dysfunction. Int J Mol Sci 2023; 24:12989. [PMID: 37629170 PMCID: PMC10455677 DOI: 10.3390/ijms241612989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Cardiac fibrosis is a common pathological process in heart disease, representing a therapeutic target. Transforming growth factor β (TGFβ) is the canonical driver of cardiac fibrosis and was recently shown to be dependent on interleukin 11 (IL11) for its profibrotic effects in fibroblasts. In the opposite direction, recombinant human IL11 has been reported as anti-fibrotic and anti-inflammatory in the mouse heart. In this study, we determined the effects of IL11 expression in cardiomyocytes on cardiac pathobiology and function. We used the Cre-loxP system to generate a tamoxifen-inducible mouse with cardiomyocyte-restricted murine Il11 expression. Using protein assays, bulk RNA-sequencing, and in vivo imaging, we analyzed the effects of IL11 on myocardial fibrosis, inflammation, and cardiac function, challenging previous reports suggesting the cardioprotective potential of IL11. TGFβ stimulation of cardiomyocytes caused Il11 upregulation. Compared to wild-type controls, Il11-expressing hearts demonstrated severe cardiac fibrosis and inflammation that was associated with the upregulation of cytokines, chemokines, complement factors, and increased inflammatory cells. IL11 expression also activated a program of endothelial-to-mesenchymal transition and resulted in left ventricular dysfunction. Our data define species-matched IL11 as strongly profibrotic and proinflammatory when secreted from cardiomyocytes and further establish IL11 as a disease factor.
Collapse
Affiliation(s)
- Mark Sweeney
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
- Wellcome Trust/NIHR 4i Clinical Research Fellow, Imperial College, London W12 0NN, UK
| | - Katie O’Fee
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
| | - Chelsie Villanueva-Hayes
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
| | - Ekhlas Rahman
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
| | - Michael Lee
- National Heart and Lung Institute, Imperial College, London W12 0NN, UK
| | - Konstantinos Vanezis
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
- National Heart and Lung Institute, Imperial College, London W12 0NN, UK
| | - Ivan Andrew
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Anissa Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Paul J. R. Barton
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
- National Heart and Lung Institute, Imperial College, London W12 0NN, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Stuart A. Cook
- 1MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London W12 0NN, UK
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| |
Collapse
|
14
|
Kmieć P, Rosenkranz S, Odenthal M, Caglayan E. Differential Role of Aldosterone and Transforming Growth Factor Beta-1 in Cardiac Remodeling. Int J Mol Sci 2023; 24:12237. [PMID: 37569619 PMCID: PMC10419155 DOI: 10.3390/ijms241512237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Angiotensin II, a major culprit in cardiovascular disease, activates mediators that are also involved in pathological cardiac remodeling. In this context, we aimed at investigating the effects of two of them: aldosterone (Ald) and transforming growth factor beta-1 (TGF-β1) in an in vivo model. Six-week-old male wild-type (WT) and TGF-β1-overexpressing transgenic (TGF-β1-TG) mice were infused with subhypertensive doses of Ald for 2 weeks and/or treated orally with eplerenone from postnatal day 21. Thehearts' ventricles were examined by morphometry, immunoblotting to assess the intracellular signaling pathways and RT qPCR to determine hypertrophy and fibrosis marker genes. The TGF-β1-TG mice spontaneously developed cardiac hypertrophy and interstitial fibrosis and exhibited a higher baseline phosphorylation of p44/42 and p38 kinases, fibronectin and ANP mRNA expression. Ald induced a comparable increase in the ventricular-heart-weight-to-body-weight ratio and cardiomyocyte diameter in both strains, but a less pronounced increase in interstitial fibrosis in the transgenic compared to the WT mice (23.6% vs. 80.9%, p < 0.005). Ald increased the phosphorylation of p44/42 and p38 in the WT but not the TGF-β1-TG mice. While the eplerenone-enriched chow partially prevented Ald-induced cardiac hypertrophy in both genotypes and interstitial fibrosis in the WT controls, it completely protected against additional fibrosis in transgenic mice. Ald appears to induce cardiac hypertrophy independently of TGF-β1, while in the case of fibrosis, the downstream signaling pathways of these two factors probably converge.
Collapse
Affiliation(s)
- Piotr Kmieć
- Department of Endocrinology and Internal Medicine, Medical University of Gdańsk, 80214 Gdańsk, Poland;
| | - Stephan Rosenkranz
- Clinic for Internal Medicine III and Cologne Cardiovascular Research Center, Cologne University Heart Center, 50937 Köln, Germany;
| | - Margarete Odenthal
- Institute of Pathology, University Hospital of Cologne and Center for Molecular Medicine, University of Cologne, 50937 Köln, Germany;
| | - Evren Caglayan
- Department of Cardiology, University-Medicine Rostock, 18057 Rostock, Germany
| |
Collapse
|
15
|
Hall C, Law JP, Reyat JS, Cumberland MJ, Hang S, Vo NTN, Raniga K, Weston CJ, O'Shea C, Townend JN, Gehmlich K, Ferro CJ, Denning C, Pavlovic D. Chronic activation of human cardiac fibroblasts in vitro attenuates the reversibility of the myofibroblast phenotype. Sci Rep 2023; 13:12137. [PMID: 37495732 PMCID: PMC10372150 DOI: 10.1038/s41598-023-39369-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 07/24/2023] [Indexed: 07/28/2023] Open
Abstract
Activation of cardiac fibroblasts and differentiation to myofibroblasts underlies development of pathological cardiac fibrosis, leading to arrhythmias and heart failure. Myofibroblasts are characterised by increased α-smooth muscle actin (α-SMA) fibre expression, secretion of collagens and changes in proliferation. Transforming growth factor-beta (TGF-β) and increased mechanical stress can initiate myofibroblast activation. Reversibility of the myofibroblast phenotype has been observed in murine cells but has not been explored in human cardiac fibroblasts. In this study, chronically activated adult primary human ventricular cardiac fibroblasts and human induced pluripotent stem cell derived cFbs (hiPSC-cFbs) were used to investigate the potential for reversal of the myofibroblast phenotype using either subculture on soft substrates or TGF-β receptor inhibition. Culture on softer plates (25 or 2 kPa Young's modulus) did not alter proliferation or reduce expression of α-SMA and collagen 1. Similarly, culture of myofibroblasts in the presence of TGF-β inhibitor did not reverse myofibroblasts back to a quiescent phenotype. Chronically activated hiPSC-cFbs also showed attenuated response to TGF-β receptor inhibition and inability to reverse to quiescent fibroblast phenotype. Our data demonstrate substantial loss of TGF-β signalling plasticity as well as a loss of feedback from the surrounding mechanical environment in chronically activated human myofibroblasts.
Collapse
Affiliation(s)
- Caitlin Hall
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jonathan P Law
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jasmeet S Reyat
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Max J Cumberland
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Shaun Hang
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nguyen T N Vo
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Kavita Raniga
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Chris J Weston
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jonathan N Townend
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Department of Cardiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, B15 2GW, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Charles J Ferro
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Department of Renal Medicine, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, B15 2GW, UK
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
16
|
Ziegler KA, Ahles A, Dueck A, Esfandyari D, Pichler P, Weber K, Kotschi S, Bartelt A, Sinicina I, Graw M, Leonhardt H, Weckbach LT, Massberg S, Schifferer M, Simons M, Hoeher L, Luo J, Ertürk A, Schiattarella GG, Sassi Y, Misgeld T, Engelhardt S. Immune-mediated denervation of the pineal gland underlies sleep disturbance in cardiac disease. Science 2023; 381:285-290. [PMID: 37471539 DOI: 10.1126/science.abn6366] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/01/2023] [Indexed: 07/22/2023]
Abstract
Disruption of the physiologic sleep-wake cycle and low melatonin levels frequently accompany cardiac disease, yet the underlying mechanism has remained enigmatic. Immunostaining of sympathetic axons in optically cleared pineal glands from humans and mice with cardiac disease revealed their substantial denervation compared with controls. Spatial, single-cell, nuclear, and bulk RNA sequencing traced this defect back to the superior cervical ganglia (SCG), which responded to cardiac disease with accumulation of inflammatory macrophages, fibrosis, and the selective loss of pineal gland-innervating neurons. Depletion of macrophages in the SCG prevented disease-associated denervation of the pineal gland and restored physiological melatonin secretion. Our data identify the mechanism by which diurnal rhythmicity in cardiac disease is disturbed and suggest a target for therapeutic intervention.
Collapse
Affiliation(s)
- Karin A Ziegler
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Andrea Ahles
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Anne Dueck
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Dena Esfandyari
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Pauline Pichler
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
| | - Karolin Weber
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Alexander Bartelt
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard. T.H. Chan School of Public Health, Boston, MA, USA
| | - Inga Sinicina
- Institute of Legal Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Matthias Graw
- Institute of Legal Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Heinrich Leonhardt
- Human Biology & Bioimaging, Faculty of Biology, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Ludwig T Weckbach
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
| | - Steffen Massberg
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
| | - Martina Schifferer
- DZNE (German Center for Neurodegenerative Diseases), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- DZNE (German Center for Neurodegenerative Diseases), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich (TUM), Munich, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
| | - Jie Luo
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Ali Ertürk
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Gabriele G Schiattarella
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Yassine Sassi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Thomas Misgeld
- DZNE (German Center for Neurodegenerative Diseases), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich (TUM), Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
17
|
Zang X, Zhao Z, Chen K, Song W, Ma J, Fu H, Wang X, Zhao Y. SHP-1 alleviates atrial fibrosis in atrial fibrillation by modulating STAT3 activation. Exp Biol Med (Maywood) 2023; 248:979-990. [PMID: 37226737 PMCID: PMC10525403 DOI: 10.1177/15353702231165717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/04/2023] [Indexed: 05/26/2023] Open
Abstract
Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) has a well-established role in myocardial infarction, yet its involvement in atrial fibrosis and atrial fibrillation (AF) has not been elucidated. As cardiac arrhythmias caused by AF are a major global health concern, we investigated whether SHP-1 modulates AF development. The degree of atrial fibrosis was examined using Masson's trichrome staining, and SHP-1 expression in the human atrium was assessed using quantitative polymerase chain reaction (qPCR), immunohistochemistry (IHC), and western blotting (WB). We also examined SHP-1 expression in cardiac tissue from an AF mouse model, as well as in angiotensin II (Ang II)-treated mouse atrial myocytes and fibroblasts. We found that SHP-1 expression was reduced with the aggravation of atrial fibrosis in clinical samples of patients with AF. SHP-1 was also downregulated in the heart tissue of AF mice and Ang II-treated myocytes and fibroblasts, compared with that in the control groups. Next, we demonstrated that SHP-1 overexpression alleviated AF severity in mice by injecting a lentiviral vector into the pericardial space. In Ang II-treated myocytes and fibroblasts, we observed excessive extracellular matrix (ECM) deposition, reactive oxygen species (ROS) generation, and transforming growth factor beta 1 (TGF-β1)/mothers against decapentaplegic homolog 2 (SMAD2) pathway activation, all of which were counteracted by the overexpression of SHP-1. Our WB data showed that STAT3 activation was inversely correlated with SHP-1 expression in samples from patients with AF, AF mice, and Ang II-treated cells. Furthermore, administration of colivelin, a STAT3 agonist, in SHP-1-overexpressing, Ang II-treated myocytes and fibroblasts resulted in higher levels of ECM deposition, ROS generation, and TGF-β1/SMAD2 activation. These findings indicate that SHP-1 regulates AF fibrosis progression by modulating STAT3 activation and is thus a potential treatment target for atrial fibrosis and AF.
Collapse
Affiliation(s)
- Xiaobiao Zang
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Zhihan Zhao
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Ke Chen
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Weifeng Song
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Jifang Ma
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Haixia Fu
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Xianqing Wang
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| | - Yonghui Zhao
- Department of Cardiology, Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou 451460, China
| |
Collapse
|
18
|
Ock S, Choi SW, Choi SH, Kang H, Kim SJ, Lee WS, Kim J. Insulin signaling is critical for sinoatrial node maintenance and function. Exp Mol Med 2023; 55:965-973. [PMID: 37121973 PMCID: PMC10238478 DOI: 10.1038/s12276-023-00988-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/12/2023] [Accepted: 02/12/2023] [Indexed: 05/02/2023] Open
Abstract
Insulin and insulin-like growth factor 1 (IGF-1) signaling regulate cellular growth and glucose metabolism in the myocardium. However, their physiological role in the cells of the cardiac conduction system has never been explored. Therefore, we sought to determine the spatiotemporal function of insulin/IGF-1 receptors in the sinoatrial node (SAN). We generated cardiac conduction cell-specific inducible IGF-1 receptor (IGF-1R) knockout (KO) (CSIGF1RKO), insulin receptor (IR) KO (CSIRKO), and IR/IGF-1R double-KO (CSDIRKO) mice and evaluated their phenotypes. Telemetric electrocardiography revealed regular sinus rhythm in CSIGF1RKO mice, indicating that IGF-1R is dispensable for normal pacemaking. In contrast, CSIRKO and CSDIRKO mice exhibited profound sinus bradycardia. CSDIRKO mice showed typical sinus node dysfunction characterized by junctional rhythm and sinus pauses on electrocardiography. Interestingly, the lack of an insulin receptor in the SAN cells of CSIRKO and CSDIRKO mice caused sinus nodal fibrosis. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) protein expression significantly decreased in the CSIRKO and CSDIRKO mice relative to the controls. A patch-clamp study of the SAN cells of CSIRKO mice revealed a significant decrease in the funny current, which is responsible for spontaneous diastolic depolarization in the SAN. This result suggested that insulin receptor loss reduces the heart rate via downregulation of the HCN4 channel. Additionally, HCN1 expression was decreased in CSDIRKO mice, explaining their sinus node dysfunction. Our results reveal a previously unrecognized role of insulin/IGF-1 signaling in sinus node structural maintenance and pacemaker function.
Collapse
Affiliation(s)
- Sangmi Ock
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Seong Woo Choi
- Departments of Physiology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea
- Department of Physiology, Dongguk University College of Medicine, Gyeongju, Korea
| | - Seung Hee Choi
- Division of Endocrinology and Metabolism, Departments of Internal Medicine and Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hyun Kang
- Department of Anesthesiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Sung Joon Kim
- Departments of Physiology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea
| | - Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea.
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea.
| |
Collapse
|
19
|
Al-U'datt DGF, Tranchant CC, Alu'datt M, Abusara S, Al-Dwairi A, AlQudah M, Al-Shboul O, Hiram R, Altuntas Y, Jaradat S, Alzoubi KH. Inhibition of transglutaminase 2 (TG2) ameliorates ventricular fibrosis in isoproterenol-induced heart failure in rats. Life Sci 2023; 321:121564. [PMID: 36931499 DOI: 10.1016/j.lfs.2023.121564] [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: 12/03/2022] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
AIMS Transglutaminase (TG) inhibitors represent promising therapeutic interventions in cardiac fibrosis and related dysfunctions. However, it remains unknown how TG inhibition, TG2 in particular, affects the signaling systems that drive pathological fibrosis. This study aimed to examine the effect TG inhibition by cystamine on the progression of isoproterenol (ISO)-induced cardiac fibrosis and dysfunction in rats. MATERIALS AND METHODS Cardiac fibrosis was established by intraperitoneal injection of ISO to rats (ISO group), followed by 6 weeks of cystamine injection (ISO + Cys group). The control groups were administered normal saline alone or with cystamine. Hemodynamics, lipid profile, liver enzymes, urea, and creatinine were assessed in conjunction with heart failure markers (serum NT-proANP and cTnI). Left ventricular (LV) and atrial (LA) fibrosis, total collagen content, and mRNA expression of profibrotic markers including TG2 were quantified by Masson's trichrome staining, LC-MS/MS and quantitative PCR, respectively. KEY FINDINGS Cystamine administration to ISO rats significantly decreased diastolic and mean arterial pressures, total cholesterol, triglycerides, LDL, liver enzymes, urea, and creatinine levels, while increasing HDL. NT-proANP and cTnI serum levels remained unchanged. In LV tissues, significant reductions in ISO-induced fibrosis and elevated total collagen content were achieved after cystamine treatment, together with a reduction in TG2 concentration. Reduced mRNA expression of several profibrotic genes (COL1A1, FN1, MMP-2, CTGF, periostin, CX43) was also evidenced in LV tissues of ISO rats upon cystamine administration, whereas TGF-β1 expression was depressed in LA tissues. Cystamine decreased TG2 mRNA expression in the LV of control rats, while LV expression of TG2 was relatively low in ISO rats irrespective of cystamine treatment. SIGNIFICANCE TG2 inhibition by cystamine in vivo exerted cardioprotective effects against ISO-induced cardiac fibrosis in rats decreasing the LV abundance of several profibrotic markers and the content of TG2 and collagen, suggesting that TG2 pharmacological inhibition could be beneficial to alleviate cardiac fibrosis.
Collapse
Affiliation(s)
- Doa'a G F Al-U'datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Carole C Tranchant
- School of Food Science, Nutrition and Family Studies, Faculty of Health Sciences and Community Services, Université de Moncton, New Brunswick, Canada
| | - Muhammad Alu'datt
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Sara Abusara
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad AlQudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; Physiology Department, Arabian Gulf University, Manama, Bahrain
| | - Othman Al-Shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Roddy Hiram
- Montreal Heart Institute and Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Yasemin Altuntas
- Montreal Heart Institute and Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Saied Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
20
|
Manoj P, Kim JA, Kim S, Li T, Sewani M, Chelu MG, Li N. Sinus node dysfunction: current understanding and future directions. Am J Physiol Heart Circ Physiol 2023; 324:H259-H278. [PMID: 36563014 PMCID: PMC9886352 DOI: 10.1152/ajpheart.00618.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. Normal SAN function is crucial in maintaining proper cardiac rhythm and contraction. Sinus node dysfunction (SND) is due to abnormalities within the SAN, which can affect the heartbeat frequency, regularity, and the propagation of electrical pulses through the cardiac conduction system. As a result, SND often increases the risk of cardiac arrhythmias. SND is most commonly seen as a disease of the elderly given the role of degenerative fibrosis as well as other age-dependent changes in its pathogenesis. Despite the prevalence of SND, current treatment is limited to pacemaker implantation, which is associated with substantial medical costs and complications. Emerging evidence has identified various genetic abnormalities that can cause SND, shedding light on the molecular underpinnings of SND. Identification of these molecular mechanisms and pathways implicated in the pathogenesis of SND is hoped to identify novel therapeutic targets for the development of more effective therapies for this disease. In this review article, we examine the anatomy of the SAN and the pathophysiology and epidemiology of SND. We then discuss in detail the most common genetic mutations correlated with SND and provide our perspectives on future research and therapeutic opportunities in this field.
Collapse
Affiliation(s)
- Pavan Manoj
- School of Public Health, Texas A&M University, College Station, Texas
| | - Jitae A Kim
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kim
- Department of BioSciences, Rice University, Houston, Texas
| | - Tingting Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Maham Sewani
- Department of BioSciences, Rice University, Houston, Texas
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Na Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
21
|
Wang R, Qi YF, Ding CH, Zhang D. Sulfur-containing amino acids and their metabolites in atrial fibrosis. Front Pharmacol 2022; 13:1063241. [DOI: 10.3389/fphar.2022.1063241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Atrial fibrosis, a symbol of atrial structural remodelling, is a complex process involved in the occurrence and maintenance of atrial fibrillation (AF). Atrial fibrosis is regulated by multiple factors. Sulfur containing amino acids and their metabolites, such as hydrogen sulfide (H2S) and taurine, can inhibit the process of atrial fibrosis and alleviate atrial remodeling. However, homocysteine can promote the activation of atrial fibroblasts and further promote atrial fibrosis. In this review, we will focus on the recent progress in atrial structural changes and molecular mechanisms of atrial fibrosis, as well as the regulatory roles and possible mechanisms of sulfur containing amino acids and their metabolites in atrial fibrosis. It is expected to provide new ideas for clarifying the mechanism of atrial fibrosis and finding targets to inhibit the progress of atrial fibrosis.
Collapse
|
22
|
Wu M, Xing Q, Duan H, Qin G, Sang N. Suppression of NADPH oxidase 4 inhibits PM 2.5-induced cardiac fibrosis through ROS-P38 MAPK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155558. [PMID: 35504386 DOI: 10.1016/j.scitotenv.2022.155558] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) has been consistently linked to cardiovascular diseases, and cardiac fibrosis plays a crucial role in the occurrence and development of heart diseases. It is reported that NOX4-dependent redox signaling are responsible for TGFβ-mediated profibrotic responses. The current study was designed to explore the possible mechanisms of cardiac fibrosis by PM2.5 both in vitro and in vivo. Female C57BL/6 mice received PM2.5 (3 mg/kg b.w.) exposure with/without NOX4 inhibitor (apocynin, 25 mg/kg b.w.) or ROS scavenger (NALC, 50 mg/kg b.w.), every other day, for 4 weeks. H9C2 cells were incubated with PM2.5 (3 μg/mL) with/without 5 mM NALC, TGFβ inhibitor (SB431542, 10 μM), or siRNA-NOX4 for 24 h. The results demonstrated that PM2.5 induced evident collagen deposition and elevated expression of fibrosis biomarkers (Col1a1 & Col3a1). Significant systemic inflammatory response and cardiac oxidative stress were triggered by PM2.5. PM2.5 increased the protein expression of TGFβ1, NOX4, and P38 MAPK. Notably, the increased effects of PM2.5 could be suppressed by SB431542, siRNA-NOX4 in vitro or apocynin in vivo, and NALC. The reverse verification experiments further supported the involvement of the TGFβ/NOX4/ROS/P38 MAPK signaling pathway in the myocardial fibrosis induced by PM2.5. In summary, the current study provided evidence that PM2.5 challenge led to cardiac fibrosis through oxidative stress, systemic inflammation, and subsequent TGFβ/NOX4/ROS/P38 MAPK pathway and may offer new therapeutic targets in cardiac fibrosis.
Collapse
Affiliation(s)
- Meiqiong Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China; Department of Children and Adolescences Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China.
| | - Qisong Xing
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huiling Duan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
23
|
New Insights into the Functions of MicroRNAs in Cardiac Fibrosis: From Mechanisms to Therapeutic Strategies. Genes (Basel) 2022; 13:genes13081390. [PMID: 36011301 PMCID: PMC9407613 DOI: 10.3390/genes13081390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/16/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a significant global health problem associated with almost all types of heart disease. Extensive cardiac fibrosis reduces tissue compliance and contributes to adverse outcomes, such as cardiomyocyte hypertrophy, cardiomyocyte apoptosis, and even heart failure. It is mainly associated with pathological myocardial remodeling, characterized by the excessive deposition of extracellular matrix (ECM) proteins in cardiac parenchymal tissues. In recent years, a growing body of evidence demonstrated that microRNAs (miRNAs) have a crucial role in the pathological development of cardiac fibrosis. More than sixty miRNAs have been associated with the progression of cardiac fibrosis. In this review, we summarized potential miRNAs and miRNAs-related regulatory mechanisms for cardiac fibrosis and discussed the potential clinical application of miRNAs in cardiac fibrosis.
Collapse
|
24
|
Lai YJ, Tsai FC, Chang GJ, Chang SH, Huang CC, Chen WJ, Yeh YH. miR-181b targets semaphorin 3A to mediate TGF-β-induced endothelial-mesenchymal transition related to atrial fibrillation. J Clin Invest 2022; 132:142548. [PMID: 35775491 PMCID: PMC9246393 DOI: 10.1172/jci142548] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrosis is an essential contributor to atrial fibrillation (AF). It remains unclear whether atrial endocardial endothelial cells (AEECs) that undergo endothelial-mesenchymal transition (EndMT) are among the sources of atrial fibroblasts. We studied human atria, TGF-β-treated human AEECs, cardiac-specific TGF-β-transgenic mice, and heart failure rabbits to identify the underlying mechanism of EndMT in atrial fibrosis. Using isolated AEECs, we found that miR-181b was induced in TGF-β-treated AEECs, which decreased semaphorin 3A (Sema3A) and increased EndMT markers, and these effects could be reversed by a miR-181b antagomir. Experiments in which Sema3A was increased by a peptide or decreased by a siRNA in AEECs revealed a mechanistic link between Sema3A and LIM-kinase 1/phosphorylated cofilin (LIMK/p-cofilin) signaling and suggested that Sema3A is upstream of LIMK in regulating actin remodeling through p-cofilin. Administration of the miR-181b antagomir or recombinant Sema3A to TGF-β-transgenic mice evoked increased Sema3A, reduced EndMT markers, and significantly decreased atrial fibrosis and AF vulnerability. Our study provides a mechanistic link between the induction of EndMT by TGF-β via miR-181b/Sema3A/LIMK/p-cofilin signaling to atrial fibrosis. Blocking miR-181b and increasing Sema3A are potential strategies for AF therapeutic intervention.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chia Yi, Taiwan
| | - Feng-Chun Tsai
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao Yuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Tao Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| |
Collapse
|
25
|
Chen X, Yang Q, Bai W, Yao W, Liu L, Xing Y, Meng C, Qi P, Dang Y, Qi X. Dapagliflozin Attenuates Myocardial Fibrosis by Inhibiting the TGF-β1/Smad Signaling Pathway in a Normoglycemic Rabbit Model of Chronic Heart Failure. Front Pharmacol 2022; 13:873108. [PMID: 35645838 PMCID: PMC9136228 DOI: 10.3389/fphar.2022.873108] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/13/2022] [Indexed: 11/14/2022] Open
Abstract
Recent studies have shown that sodium-glucose cotransporter-2 (SGLT2) inhibitors play a beneficial role for normoglycemic patients with heart failure (HF). However, the underlying mechanism remains largely unexplored. In the present study, we aimed to investigate the cardioprotective effect of SGLT2 inhibitors in a normoglycemic rabbit model of chronic heart failure (CHF) and its potential mechanism was also explored. A total of 24 male New Zealand white rabbits were randomly divided into the sham group, HF group, perindopril group, and dapagliflozin (DAPA) group. The normoglycemic CHF model was established by aortic constriction for 12 weeks. In the 13th week, DAPA (1 mg/kg/day) or perindopril (0.5 mg/kg/day) was administered by oral gavage daily for 10 weeks. Both the sham group and HF group were given normal saline via gavage. After 10 weeks, the heart structure and function were evaluated by echocardiography and plasma NT-proBNP. Moreover, cardiac fibrosis was analyzed using immunohistochemistry, Masson’s trichrome staining, and Western blotting analysis. The results showed that DAPA improved the myocardial structure and function of normoglycemic CHF rabbits and ameliorated myocardial fibrosis. Further study indicated that DAPA suppressed cardiac fibrosis by inhibiting the transforming growth factor β1 (TGF-β1)/Smad signaling pathway. Collectively, our findings showed that DAPA could ameliorate cardiac fibrosis in normoglycemic CHF rabbits by inhibiting the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xuefeng Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Qian Yang
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Wenlou Bai
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Wenjing Yao
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Litian Liu
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Yuanyuan Xing
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Cunliang Meng
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Peng Qi
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Yi Dang
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoyong Qi
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
- *Correspondence: Xiaoyong Qi,
| |
Collapse
|
26
|
Su KN, Ma Y, Cacheux M, Ilkan Z, Raad N, Muller GK, Wu X, Guerrera N, Thorn SL, Sinusas AJ, Foretz M, Viollet B, Akar JG, Akar FG, Young LH. Atrial AMP-activated protein kinase is critical for prevention of dysregulation of electrical excitability and atrial fibrillation. JCI Insight 2022; 7:141213. [PMID: 35451373 PMCID: PMC9089788 DOI: 10.1172/jci.insight.141213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/23/2022] [Indexed: 12/03/2022] Open
Abstract
Metabolic stress is an important cause of pathological atrial remodeling and atrial fibrillation. AMPK is a ubiquitous master metabolic regulator, yet its biological function in the atria is poorly understood in both health and disease. We investigated the impact of atrium-selective cardiac AMPK deletion on electrophysiological and structural remodeling in mice. Loss of atrial AMPK expression caused atrial changes in electrophysiological properties and atrial ectopic activity prior to the onset of spontaneous atrial fibrillation. Concomitant transcriptional downregulation of connexins and atrial ion channel subunits manifested with delayed left atrial activation and repolarization. The early molecular and electrophysiological abnormalities preceded left atrial structural remodeling and interstitial fibrosis. AMPK inactivation induced downregulation of transcription factors (Mef2c and Pitx2c) linked to connexin and ion channel transcriptional reprogramming. Thus, AMPK plays an essential homeostatic role in atria, protecting against adverse remodeling potentially by regulating key transcription factors that control the expression of atrial ion channels and gap junction proteins.
Collapse
Affiliation(s)
- Kevin N Su
- Department of Cellular & Molecular Physiology and
| | - Yina Ma
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marine Cacheux
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zeki Ilkan
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nour Raad
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Xiaohong Wu
- Department of Cellular & Molecular Physiology and
| | - Nicole Guerrera
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie L Thorn
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert J Sinusas
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marc Foretz
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Benoit Viollet
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Joseph G Akar
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Fadi G Akar
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lawrence H Young
- Department of Cellular & Molecular Physiology and.,Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
27
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
28
|
A Review of the Molecular Mechanisms Underlying Cardiac Fibrosis and Atrial Fibrillation. J Clin Med 2021; 10:jcm10194430. [PMID: 34640448 PMCID: PMC8509789 DOI: 10.3390/jcm10194430] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023] Open
Abstract
The cellular and molecular mechanism involved in the pathogenesis of atrial fibrosis are highly complex. We have reviewed the literature that covers the effectors, signal transduction and physiopathogenesis concerning extracellular matrix (ECM) dysregulation and atrial fibrosis in atrial fibrillation (AF). At the molecular level: angiotensin II, transforming growth factor-β1, inflammation, and oxidative stress are particularly important for ECM dysregulation and atrial fibrotic remodelling in AF. We conclude that the Ang-II-MAPK and TGF-β1-Smad signalling pathways play a major, central role in regulating atrial fibrotic remodelling in AF. The above signalling pathways induce the expression of genes encoding profibrotic molecules (MMP, CTGF, TGF-β1). An important mechanism is also the generation of reactive oxygen species. This pathway induced by the interaction of Ang II with the AT2R receptor and the activation of NADPH oxidase. Additionally, the interplay between cardiac MMPs and their endogenous tissue inhibitors of MMPs, is thought to be critical in atrial ECM metabolism and fibrosis. We also review recent evidence about the role of changes in the miRNAs expression in AF pathophysiology and their potential as therapeutic targets. Furthermore, keeping the balance between miRNA molecules exerting anti-/profibrotic effects is of key importance for the control of atrial fibrosis in AF.
Collapse
|
29
|
Plikus MV, Wang X, Sinha S, Forte E, Thompson SM, Herzog EL, Driskell RR, Rosenthal N, Biernaskie J, Horsley V. Fibroblasts: Origins, definitions, and functions in health and disease. Cell 2021; 184:3852-3872. [PMID: 34297930 PMCID: PMC8566693 DOI: 10.1016/j.cell.2021.06.024] [Citation(s) in RCA: 541] [Impact Index Per Article: 135.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling niches through biophysical and biochemical cues. Transcriptionally and functionally heterogeneous across and within organs, fibroblasts encode regional positional information and maintain distinct cellular progeny. We summarize their development, lineages, functions, and contributions to fibrosis in four fibroblast-rich organs: skin, lung, skeletal muscle, and heart. We propose that fibroblasts are uniquely poised for tissue repair by easily reentering the cell cycle and exhibiting a reversible plasticity in phenotype and cell fate. These properties, when activated aberrantly, drive fibrotic disorders in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Sean M Thompson
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Erica L Herzog
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Ryan R Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA.
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK.
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Valerie Horsley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
30
|
Hall C, Gehmlich K, Denning C, Pavlovic D. Complex Relationship Between Cardiac Fibroblasts and Cardiomyocytes in Health and Disease. J Am Heart Assoc 2021; 10:e019338. [PMID: 33586463 PMCID: PMC8174279 DOI: 10.1161/jaha.120.019338] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts are the primary cell type responsible for deposition of extracellular matrix in the heart, providing support to the contracting myocardium and contributing to a myriad of physiological signaling processes. Despite the importance of fibrosis in processes of wound healing, excessive fibroblast proliferation and activation can lead to pathological remodeling, driving heart failure and the onset of arrhythmias. Our understanding of the mechanisms driving the cardiac fibroblast activation and proliferation is expanding, and evidence for their direct and indirect effects on cardiac myocyte function is accumulating. In this review, we focus on the importance of the fibroblast-to-myofibroblast transition and the cross talk of cardiac fibroblasts with cardiac myocytes. We also consider the current use of models used to explore these questions.
Collapse
Affiliation(s)
- Caitlin Hall
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom.,Division of Cardiovascular Medicine Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford University of Oxford United Kingdom
| | - Chris Denning
- Biodiscovery Institute University of Nottingham United Kingdom
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| |
Collapse
|
31
|
Kakinuma Y. Characteristic Effects of the Cardiac Non-Neuronal Acetylcholine System Augmentation on Brain Functions. Int J Mol Sci 2021; 22:ijms22020545. [PMID: 33430415 PMCID: PMC7826949 DOI: 10.3390/ijms22020545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Since the discovery of non-neuronal acetylcholine in the heart, this specific system has drawn scientific interest from many research fields, including cardiology, immunology, and pharmacology. This system, acquired by cardiomyocytes independent of the parasympathetic nervous system of the autonomic nervous system, helps us to understand unsolved issues in cardiac physiology and to realize that the system may be more pivotal for cardiac homeostasis than expected. However, it has been shown that the effects of this system may not be restricted to the heart, but rather extended to cover extra-cardiac organs. To this end, this system intriguingly influences brain function, specifically potentiating blood brain barrier function. Although the results reported appear to be unusual, this novel characteristic can provide us with another research interest and therapeutic application mode for central nervous system diseases. In this review, we discuss our recent studies and raise the possibility of application of this system as an adjunctive therapeutic modality.
Collapse
Affiliation(s)
- Yoshihiko Kakinuma
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
32
|
Chung H, Kim Y, Park CH, Kim IS, Kim JY, Min PK, Yoon YW, Kim TH, Lee BK, Hong BK, Rim SJ, Kwon HM, Lee KA, Choi EY. Contribution of sarcomere gene mutations to left atrial function in patients with hypertrophic cardiomyopathy. Cardiovasc Ultrasound 2021; 19:4. [PMID: 33407484 PMCID: PMC7789152 DOI: 10.1186/s12947-020-00233-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/14/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Left atrial (LA) enlargement and dysfunction are related to clinical course in patients with hypertrophic cardiomyopathy (HCM). We aimed to investigate genetic contribution to LA structural and functional remodeling. METHODS Two hundred twelve patients were consecutively enrolled, and echocardiography and extensive genetic analysis were performed. Cardiac magnetic resonance (CMR) was performed in 135 patients. Echocardiography was also performed in controls (n = 30). RESULTS Patients with HCM had lower late-diastolic mitral annular velocity (a') and higher LA volume index (LAVI) than controls. Patients with pathogenic or likely pathogenic sarcomere gene mutations (PSM, n = 67, 32%) had higher LAVI and lower CMR-derived LA total emptying fraction (37.0 ± 18.5 vs. 44.2 ± 12.4%, p = 0.025). In patients without AF (n = 187), the PSM had lower a' (6.9 ± 2.0 vs. 7.8 ± 1.9 cm/s, p = 0.004) than others. The PSM had higher prevalence and amount of late gadolinium enhancement (LGE) in the left ventricle (LV). In multivariate analysis, PSM was significantly related to lower a' independent of E/e', LV mass index, and LAVI. However, the relation significantly attenuated after adjustment for the extent of LGE in the LV, suggesting common myopathy in the LV and LA. In addition, PSM was significantly related to lower LA total emptying fraction independent of age, E/e', s', LV ejection fraction, LV myocardial global longitudinal strain and %LGE mass. CONCLUSIONS PSM was related to LA dysfunction independent of LV filling pressure and LAVI, suggesting its contribution to atrial myopathy in HCM.
Collapse
Affiliation(s)
- Hyemoon Chung
- Department of Internal Medicine, Division of Cardiology, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Yoonjung Kim
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Chul Hwan Park
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - In-Soo Kim
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Jong-Youn Kim
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Pil-Ki Min
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Young Won Yoon
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Tae Hoon Kim
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Byoung Kwon Lee
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Bum-Kee Hong
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Se-Joong Rim
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Hyuck Moon Kwon
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Kyung-A Lee
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea.
| | - Eui-Young Choi
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
33
|
Babapoor-Farrokhran S, Tarighati Rasekhi R, Gill D, Alzubi J, Mainigi SK. How transforming growth factor contributes to atrial fibrillation? Life Sci 2020; 266:118823. [PMID: 33309721 DOI: 10.1016/j.lfs.2020.118823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022]
Abstract
Atrial fibrillation (AF) is the most common clinically significant arrhythmia. There are four fundamental pathophysiological mechanisms of AF including: electrical remodeling, structural remodeling, autonomic nervous system changes, and Ca2+ handling abnormalities. The transforming growth factor-β (TGF-β) superfamily are cytokines that have the ability to regulate numerous cell functions including proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and production of extracellular matrix. During the last decade numerous studies have demonstrated that TGF-β affects the architecture of the heart. TGF-β1 has been shown to be involved in the development and propagation of atrial fibrillation (AF). Investigators have studied TGF-β signaling in AF with the aim of discovering potential therapeutic agents. In this review we discuss the role of TGF-β in atrial fibrillation and specifically its role in atrial structural and electrical remodeling.
Collapse
Affiliation(s)
| | - Roozbeh Tarighati Rasekhi
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Deanna Gill
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jafar Alzubi
- Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA
| | - Sumeet K Mainigi
- Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
34
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
35
|
Simard C, Magaud C, Adjlane R, Dupas Q, Sallé L, Manrique A, Bois P, Faivre JF, Guinamard R. TRPM4 non-selective cation channel in human atrial fibroblast growth. Pflugers Arch 2020; 472:1719-1732. [PMID: 33047172 DOI: 10.1007/s00424-020-02476-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
Cardiac fibroblasts play an important role in cardiac matrix turnover and are involved in cardiac fibrosis development. Ca2+ is a driving belt in this phenomenon. This study evaluates the functional expression and contribution of the Ca2+-activated channel TRPM4 in atrial fibroblast phenotype. Molecular and electrophysiological investigations were conducted in human atrial fibroblasts in primary culture and in atrial fibroblasts obtained from wild-type and transgenic mice with disrupted Trpm4 gene (Trpm4-/-). A typical TRPM4 current was recorded on human cells (equal selectivity for Na+ and K+, activation by internal Ca2+, voltage sensitivity, conductance of 23.2 pS, inhibition by 9-phenanthrol (IC50 = 6.1 × 10-6 mol L-1)). Its detection rate was 13% on patches at days 2-4 in culture but raised to 100% on patches at day 28. By the same time, a cell growth was observed. This growth was smaller when cells were maintained in the presence of 9-phenanthrol. Similar cell growth was measured on wild-type mice atrial fibroblasts during culture. However, this growth was minimized on Trpm4-/- mice fibroblasts compared to control animals. In addition, the expression of alpha smooth muscle actin increased during culture of atrial fibroblasts from wild-type mice. This was not observed in Trpm4-/- mice fibroblasts. It is concluded that TRPM4 participates in fibroblast growth and could thus be involved in cardiac fibrosis.
Collapse
Affiliation(s)
- Christophe Simard
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Christophe Magaud
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Racim Adjlane
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Quentin Dupas
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Laurent Sallé
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Alain Manrique
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Jean-François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Romain Guinamard
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France.
| |
Collapse
|
36
|
Sweeney M, Corden B, Cook SA. Targeting cardiac fibrosis in heart failure with preserved ejection fraction: mirage or miracle? EMBO Mol Med 2020; 12:e10865. [PMID: 32955172 PMCID: PMC7539225 DOI: 10.15252/emmm.201910865] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is central to the pathology of heart failure, particularly heart failure with preserved ejection fraction (HFpEF). Irrespective of the underlying profibrotic condition (e.g. ageing, diabetes, hypertension), maladaptive cardiac fibrosis is defined by the transformation of resident fibroblasts to matrix-secreting myofibroblasts. Numerous profibrotic factors have been identified at the molecular level (e.g. TGFβ, IL11, AngII), which activate gene expression programs for myofibroblast activation. A number of existing HF therapies indirectly target fibrotic pathways; however, despite multiple clinical trials in HFpEF, a specific clinically effective antifibrotic therapy remains elusive. Therapeutic inhibition of TGFβ, the master-regulator of fibrosis, has unfortunately proven toxic and ineffective in clinical trials to date, and new approaches are needed. In this review, we discuss the pathophysiology and clinical implications of interstitial fibrosis in HFpEF. We provide an overview of trials targeting fibrosis in HFpEF to date and discuss the promise of potential new therapeutic approaches and targets in the context of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Mark Sweeney
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- Wellcome Trust 4i/NIHR Clinical Research FellowImperial CollegeLondonUK
| | - Ben Corden
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Stuart A Cook
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
37
|
Ding WY, Gupta D, Wong CF, Lip GYH. Pathophysiology of atrial fibrillation and chronic kidney disease. Cardiovasc Res 2020; 117:1046-1059. [PMID: 32871005 DOI: 10.1093/cvr/cvaa258] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/28/2020] [Accepted: 08/25/2020] [Indexed: 01/06/2023] Open
Abstract
Atrial fibrillation (AF) and chronic kidney disease (CKD) are closely related conditions with shared risk factors. The growing prevalence of both AF and CKD indicates that more patients will suffer from concurrent conditions. There are various complex interlinking mechanisms with important implications for the management of these patients. Furthermore, there is uncertainty regarding the use of oral anticoagulation (OAC) in AF and CKD that is reflected by a lack of consensus between international guidelines. Therefore, the importance of understanding the implications of co-existing AF and CKD should not be underestimated. In this review, we discuss the pathophysiology and association between AF and CKD, including the underlying mechanisms, risk of thrombo-embolic and bleeding complications, influence on stroke management, and evidence surrounding the use of OAC for stroke prevention.
Collapse
Affiliation(s)
- Wern Yew Ding
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Dhiraj Gupta
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Christopher F Wong
- Department of Renal Medicine, Liverpool University Hospital, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
38
|
Hou J, Huang S, Long Y, Huang J, Yang S, Yao J, Chen G, Yue Y, Liang M, Mei B, Li J, Wu Z. DACT2 regulates structural and electrical atrial remodeling in atrial fibrillation. J Thorac Dis 2020; 12:2039-2048. [PMID: 32642106 PMCID: PMC7330378 DOI: 10.21037/jtd-19-4206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Atrial fibrillation (AF) is the most common sustained arrhythmia. DACT2 is a novel and important mediator of signaling pathways. The aim of this study was to investigate the clinical significance and functions of DACT2 expression in AF. Methods Immunohistochemistry was used to detect the DACT2 expression pattern in valvular disease patients. DACT2 was overexpressed in HL-1 cells and primary atrial fibroblasts. The expression levels of the potassium channel, the L-type calcium current channel, sodium ion channel proteins and collagen proteins were detected by real-time polymerase chain reaction (RT-PCR). The proteins involved in the Wnt and TGF-β signaling pathways were detected after DACT2 overexpression by western blotting. Results DACT2 expression was significantly associated with AF (P=0.016). The fibrosis ratio in the strong DACT2 expression group was significantly lower than that in the weak DACT2 expression group (weak: 0.198±0.091, strong: 0.129±0.064, P=0.048), and a negative correlation between DACT2 expression levels and fibrosis severity was observed (Spearman rho =−0.476, P=0.010). DACT2 significantly increased the expression levels of KCNE5 and decreased the levels of KCNH2 and SCN5A. Overexpression of DACT2 significantly inhibited the expression of collagen I and collagen III in primary rat atrial fibroblasts. DACT2 could facilitate β-catenin accumulation by reducing its phosphorylation at Thr41/Ser45 in HL-1 cells and inhibit the TGF-β signaling pathway in primary atrial fibroblasts. Conclusions DACT2 played a role in AF by regulating both structural and electrical atrial remodeling and by affecting β-catenin accumulation and TGF-β signaling, and it could serve as a protective factor against AF in valvular heart disease.
Collapse
Affiliation(s)
- Jian Hou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shaojie Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yan Long
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jiaxing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Song Yang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jianping Yao
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Guangxian Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan Yue
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Mengya Liang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Bo Mei
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jiawen Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
39
|
Reese-Petersen AL, Olesen MS, Karsdal MA, Svendsen JH, Genovese F. Atrial fibrillation and cardiac fibrosis: A review on the potential of extracellular matrix proteins as biomarkers. Matrix Biol 2020; 91-92:188-203. [PMID: 32205152 DOI: 10.1016/j.matbio.2020.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 01/06/2023]
Abstract
The involvement of fibrosis as an underlying pathology in heart diseases is becoming increasingly clear. In recent years, fibrosis has been granted a causative role in heart diseases and is now emerging as a major contributor to Atrial Fibrillation (AF) pathogenesis. AF is the most common arrhythmia encountered in the clinic, but the substrate for AF is still being debated. Consensus in the field is a combination of cardiac tissue remodeling, inflammation and genetic predisposition. The extracellular matrix (ECM) is subject of growing investigation, since measuring circulatory biomarkers of ECM formation and degradation provides both diagnostic and prognostic information. However, fibrosis is not just fibrosis. Each specific collagen biomarker holds information on regulatory mechanisms, as well as information about which section of the ECM is being remodeled, providing a detailed description of cardiac tissue homeostasis. This review entails an overview of the implication of fibrosis in AF, the different collagens and their significance, and the potential of using biomarkers of ECM remodeling as tools for understanding AF pathogenesis and identifying patients at risk for further disease progression.
Collapse
Affiliation(s)
| | - Morten S Olesen
- Labratory of Molecular Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | - Jesper H Svendsen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
40
|
Rahmutula D, Zhang H, Wilson EE, Olgin JE. Absence of natriuretic peptide clearance receptor attenuates TGF-β1-induced selective atrial fibrosis and atrial fibrillation. Cardiovasc Res 2020; 115:357-372. [PMID: 30239604 DOI: 10.1093/cvr/cvy224] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 09/14/2018] [Indexed: 01/04/2023] Open
Abstract
Aims TGF-β1 plays an important role in atrial fibrosis and atrial fibrillation (AF); previous studies have shown that the atria are more susceptible to TGF-β1 mediated fibrosis than the ventricles. Natriuretic peptides (NPs) play an important role in cardiac remodelling and fibrosis, but the role of natriuretic peptide clearance (NPR-C) receptor is largely unknown. We investigated the role of NPR-C in modulating TGF-β1 signalling in the atria. Methods and results MHC-TGF-β1 transgenic (TGF-β1-Tx) mice, which develop isolated atrial fibrosis and AF, were cross-bred with NPR-C knock-out mice (NPR-C-KO). Transverse aortic constriction (TAC) was performed in wild type (Wt) and NPR-C knockout mice to study. Atrial fibrosis and AF inducibility in a pathophysiologic model. Electrophysiology, molecular, and histologic studies were performed in adult mice. siRNA was used to interrogate the interaction between TGF-β1 and NP signalling pathways in isolated atrial and ventricular fibroblasts/myofibroblasts. NPR-C expression level was 17 ± 5.8-fold higher in the atria compared with the ventricle in Wt mice (P = 0.009). Cross-bred mice demonstrated markedly decreased pSmad2 and collagen expression, atrial fibrosis, and AF compared with TGF-β1-Tx mice with intact NPR-C. There was a marked reduction in atrial fibrosis gene expression and AF inducibility in the NPR-C-KO-TAC mice compared with Wt-TAC. In isolated fibroblasts, knockdown of NPR-C resulted in a marked reduction of pSmad2 (56 ± 4% and 24 ± 14% reduction in atrial and ventricular fibroblasts, respectively) and collagen (76 ± 15% and 35 ± 23% reduction in atrial and ventricular fibroblasts/myofibroblasts, respectively) in response to TGF-β1 stimulation. This effect was reversed by simultaneously knocking down NPR-A but not with simultaneous knock down of PKG-1. Conclusion The differential response to TGF-β1 stimulated fibrosis between the atria and ventricle are in part mediated by the abundance of NPR-C receptors in the atria.
Collapse
Affiliation(s)
- Dolkun Rahmutula
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Hao Zhang
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Emily E Wilson
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Jeffrey E Olgin
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| |
Collapse
|
41
|
Mou F, Mou C. Necrostatin-1 Alleviates Bleomycin-Induced Pulmonary Fibrosis and Extracellular Matrix Expression in Interstitial Pulmonary Fibrosis. Med Sci Monit 2020; 26:e919739. [PMID: 32019905 PMCID: PMC7020761 DOI: 10.12659/msm.919739] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Interstitial pulmonary fibrosis (IPF) is harmful for patients’ life and health. The effective treatment of IPF is lacking because of unclear pathogenesis. Necrostatin-1 has protective effects on lung injury and can suppress the fibrosis development. I this study we investigated whether necrostatin-1 could decrease the proliferation of pulmonary fibroblasts, pulmonary fibrosis and expression of extracellular matrix (ECM) in IPF. Material/Methods The IPF mice model was conducted by intra-tracheal injection of bleomycin (BLM) (2 mg/kg) for C57BL/6N mice. Necrostatin-1 treatment was performed with 1 mg/kg necrostatin-1 by an intravenous injection for C57BL/6N mice. Lung tissue structures and collagen deposition were observed by hematoxylin and eosin staining and Masson staining. IPF in vitro model was constructed by MRC-5 cells induced by transforming growth factor beta 1 (TGF-β1). And, 20 μM necrostatin-1 was used to treat the TGF-β1 induced MRC-5 cells. Cell Counting Kit-8 (CCK-8) assay detected the viability of MRC-5 cells. The expression of receptor-interacting protein kinase-1 and -3 (RIPK1 and RIPK3), α smooth muscle actin (α-SMA), collagen IV, collagen I, fibronectin (FN), and transforming growth factor-β (TGF-β) in lung tissues and MRC-5 cells was measured by western blot analysis. The α-SMA expression in lung tissues was also analyzed by immunohistochemistry. Results The expression of RIPK1 and RIPK3 in lung tissues of BLM induced mice was increased. The degree of pulmonary fibrosis and expression of α-SMA, collagen IV, collagen I, FN, and TGF-β in lung tissues of BLM induced mice was enhanced. The proliferation of MRC-5 cells was increased when MRC-5 cells were induced by TGF-β. The expression of RIPK1, RIPK3, α-SMA, collagen IV, collagen I, and FN was increased in TGF-β induced MRC-5 cells. And, necrostatin-1 could effectively reverse the changes of pulmonary fibrosis, RIPK1, RIPK3, and ECM in vivo and in vitro experiments. Conclusions Necrostatin-1 attenuated pulmonary fibrosis in lung tissues of BLM induced mice and inhibited the fibroblast proliferation. And, necrostatin-1 also decreased the expression of RIPK1, RIPK3, and ECM in lung tissues of BLM induced mice and TGF-β induced fibroblasts. Necrostatin-1 could be a new effective drug for the treatment of IPF.
Collapse
Affiliation(s)
- Fanghong Mou
- Department of Respiration, The People's Hospital of Kaizhou District, Chongqing, China (mainland)
| | - Canglang Mou
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
42
|
Mayyas F, Alsaheb A, Alzoubi KH. The role of fish oil in attenuating cardiac oxidative stress, inflammation and fibrosis in rat model of thyrotoxicosis. Heliyon 2019; 5:e02976. [PMID: 31872133 PMCID: PMC6909073 DOI: 10.1016/j.heliyon.2019.e02976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
Hyperthyroidism is associated with cardiovascular complications. Fish oil reduces risk of cardiovascular diseases. This study aims to evaluate the impact of fish oil on myocardial oxidative stress, inflammation and fibrosis in rat model of thyrotoxicosis. Rats were randomized into four groups; control rats, fish oil treated rats (FO, 100mg omega-3/100g body weight/day), hyperthyroid rats (Hyper, i.p levothyroxine 3 mg/kg/day), and hyperthyroid rats treated with fish oil (Hyper + FO) for 8 weeks. Changes in oxidants/antioxidants, inflammatory and fibrotic markers were measured. Thyrotoxicosis increased serum endothelin-1, thiobarbituric acid reactive substances (TBARS) and reduced activities of cardiac catalase and super oxide dismutase (SOD). Cardiac fibrosis paralleled with a decrease of matrix metalloproteinase -2 (MMP2) levels were observed in Hyper group. Use of FO increased activities of SOD and catalase, increased TBARS levels, and attenuated cardiac fibrosis by normalizing MMP-2 levels. Use of FO may attenuate cardiac oxidative stress and fibrosis in hyperthyroid states.
Collapse
Affiliation(s)
- F Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - A Alsaheb
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - K H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
43
|
Eghbali A, Dukes A, Toischer K, Hasenfuss G, Field LJ. Cell Cycle-Mediated Cardiac Regeneration in the Mouse Heart. Curr Cardiol Rep 2019; 21:131. [PMID: 31529165 DOI: 10.1007/s11886-019-1206-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE OF REVIEW Many forms of heart disease result in the essentially irreversible loss of cardiomyocytes. The ability to promote cardiomyocyte renewal may be a promising approach to reverse injury in diseased hearts. The purpose of this review is to describe the impact of cardiomyocyte cell cycle activation on cardiac function and structure in several different models of myocardial disease. RECENT FINDINGS Transgenic mice expressing cyclin D2 (D2 mice) exhibit sustained cardiomyocyte renewal in the adult heart. Earlier studies demonstrated that D2 mice exhibited progressive myocardial regeneration in experimental models of myocardial infarction, and that cardiac function was normalized to values seen in sham-operated litter mates by 180 days post-injury. D2 mice also exhibited markedly improved atrial structure in a genetic model of atrial fibrosis. More recent studies revealed that D2 mice were remarkably resistant to heart failure induced by chronic elevated afterload as compared with their wild type (WT siblings), with a 6-fold increase in median survival as well as retention of relatively normal cardiac function. Finally, D2 mice exhibited a progressive recovery in cardiac function to normal levels and a concomitant reduction in adverse myocardial remodeling in an anthracycline cardiotoxicity model. The studies reviewed here make a strong case for the potential utility of inducing cardiomyocyte renewal as a means to treat injured hearts. Several challenges which must be met to develop a viable therapeutic intervention based on these observations are discussed.
Collapse
Affiliation(s)
- Arash Eghbali
- Krannert Institute of Cardiology and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Austin Dukes
- Krannert Institute of Cardiology and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Karl Toischer
- Department of Cardiology and Pneumology, Heart Center, Georg-August-University, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Goettingen, Goettingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Heart Center, Georg-August-University, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Goettingen, Goettingen, Germany
| | - Loren J Field
- Krannert Institute of Cardiology and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut Street, Indianapolis, IN, 46202, USA.
| |
Collapse
|
44
|
Choi JI, Baek YS, Roh SY, Piccini JP, Kim YH. Chromosome 4q25 variants and biomarkers of myocardial fibrosis in patients with atrial fibrillation. J Cardiovasc Electrophysiol 2019; 30:1904-1913. [PMID: 31393025 DOI: 10.1111/jce.14104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/16/2019] [Accepted: 08/03/2019] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Little is known about how genetic predisposition and fibrosis relate in atrial fibrillation (AF). Hence, we sought to determine whether the genetic variants and biomarkers for fibrosis enhance prediction of outcomes after catheter ablation. METHODS AND RESULTS Consecutive patients who underwent catheter ablation of AF (paroxysmal, 158; nonparoxysmal, 137) or supraventricular tachycardia without AF (n = 70) were studied retrospectively. Plasma levels of transforming growth factor β1 (TGF-β1), tissue inhibitor of metalloproteinase 1 (TIMP-1), and 4q25 single-nucleotide polymorphisms (SNPs) (rs10033464 and rs220073) were measured. Mean plasma levels of both TGF-β1 and TIMP-1 were higher in patients with AF than in the control (all P < .001). Plasma levels of TIMP-1 were higher in patients with recurrence compared with those without recurrence (P = .039). Patients with variant alleles of rs10033464 showed increased recurrence after catheter ablation in patients with paroxysmal AF including after adjustment (P = .027). Patients with TIMP-1 < 107 ng/mL and no variant allele (GG) at rs10033464 had lower recurrence rates compared with other groups in those with paroxysmal AF (logrank; P = .007), whereas there was no significant difference among those patients with persistent forms of AF. Inclusion of biomarkers and genotype improved discrimination of AF recurrence in patients with paroxysmal AF (C-statistic .499 vs .600). CONCLUSIONS The combination of plasma TIMP-1 concentrations less than 107 ng/mL and the absence of a variant allele at rs10033464 was associated with lower recurrence rates in patients with paroxysmal AF. This study suggests that 4q25 SNPs and biomarkers for fibrosis may provide additive value in risk stratification for AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Center, Seoul, Republic of Korea
| | - Yong Soo Baek
- Division of Cardiology, Department of Internal Medicine, Inha University Hospital, Incheon, Republic of Korea
| | - Seung Young Roh
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Center, Seoul, Republic of Korea
| | - Jonathan P Piccini
- Duke Center for Atrial Fibrillation, Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina
| | - Young-Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Center, Seoul, Republic of Korea
| |
Collapse
|
45
|
Tsai YT, Lin FY, Lin CS, Loh SH, Li CY, Lin CY, Lin YW, Tsai CS. B-type natriuretic peptide enhances fibrotic effects via matrix metalloproteinase-2 expression in the mouse atrium in vivo and in human atrial myofibroblasts in vitro. Transl Res 2019; 208:30-46. [PMID: 30857762 DOI: 10.1016/j.trsl.2019.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/04/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
B-type natriuretic peptide (BNP) was approved by the US Food and Drug Administration in 2001 for the treatment of heart failure. However, the effects of BNP in clinical applications are controversial and uncertain. Recently, study indicated that high BNP levels are associated with an increased risk of developing atrial fibrillation. In this study, we investigated the direct effects of BNP on TNF-α-induced atrial fibrosis mice, as well as its effects on human atrial myofibroblasts. We found that injecting TNF-α-induced mice with recombinant human BNP enhanced atrial fibrosis via matrix metalloproteinase-2 (MMP-2) expression and collagen accumulation. Furthermore, we found that BNP stimulated MMP-2 expression in human atrial myofibroblasts. Treatment of human atrial myofibroblasts with cycloheximide had no effect on this outcome; however, treatment of cells with MG132 enhanced BNP-induced MMP-2 expression, indicating that protein stability and inhibition of proteasome-mediated protein degradation pathways are potentially involved. Inhibition of SIRT1 significantly decreased BNP-induced MMP-2 expression. Additionally, confocal and coimmunoprecipitation data indicated that BNP-regulated MMP-2 expression are likely to be mediated through direct interaction with SIRT1, which is thought to deacetylate MMP-2 and to increase its protein stability in human atrial myofibroblasts. Finally, we confirmed that SIRT1 is expressed and cytoplasmically redistributed as well as colocalized with MMP-2 in mouse fibrotic atrial tissue. We suggest a possible fibrosis-promoting role of BNP in the atrium, although the antifibrotic properties of BNP in the ventricle have been reported in previous studies, and that the coordination between MMP-2 and SIRT1 in BNP-induced atrial myofibroblasts participates in atrial fibrosis.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology and Cardiovascular Research Center, TaipeiMedical University Hospital, Taipei, Taiwan; Departments of Internal Medicine, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hurng Loh
- Department of Pharmacology & Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Yuan Li
- Department of Anesthesiology, China Medical University and Hospital, Taichung, Taiwan
| | - Cheng-Yen Lin
- Departments of Internal Medicine, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan; Healthcare Information and Management Department, Ming Chuan University, Taipei, Taiwan
| | - Yi-Wen Lin
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.
| | - Chien-Sung Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Pharmacology & Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
46
|
Ziegler KA, Ahles A, Wille T, Kerler J, Ramanujam D, Engelhardt S. Local sympathetic denervation attenuates myocardial inflammation and improves cardiac function after myocardial infarction in mice. Cardiovasc Res 2019; 114:291-299. [PMID: 29186414 PMCID: PMC5852629 DOI: 10.1093/cvr/cvx227] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/23/2017] [Indexed: 01/09/2023] Open
Abstract
Aims Cardiac inflammation has been suggested to be regulated by the sympathetic nervous system (SNS). However, due to the lack of methodology to surgically eliminate the myocardial SNS in mice, neuronal control of cardiac inflammation remains ill-defined. Here, we report a procedure for local cardiac sympathetic denervation in mice and tested its effect in a mouse model of heart failure post-myocardial infarction. Methods and results Upon preparation of the carotid bifurcation, the right and the left superior cervical ganglia were localized and their pre- and postganglionic branches dissected before removal of the ganglion. Ganglionectomy led to an almost entire loss of myocardial sympathetic innervation in the left ventricular anterior wall. When applied at the time of myocardial infarction (MI), cardiac sympathetic denervation did not affect acute myocardial damage and infarct size. In contrast, cardiac sympathetic denervation significantly attenuated chronic consequences of MI, including myocardial inflammation, myocyte hypertrophy, and overall cardiac dysfunction. Conclusion These data suggest a critical role for local sympathetic control of cardiac inflammation. Our model of myocardial sympathetic denervation in mice should prove useful to further dissect the molecular mechanisms underlying cardiac neural control.
Collapse
Affiliation(s)
- Karin A Ziegler
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany
| | - Andrea Ahles
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, Munich 80937, Germany
| | - Julia Kerler
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| |
Collapse
|
47
|
Low Left Atrial Strain Is Associated With Adverse Outcomes in Hypertrophic Cardiomyopathy Patients. J Am Soc Echocardiogr 2019; 32:593-603.e1. [PMID: 30904367 DOI: 10.1016/j.echo.2019.01.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Paroxysmal atrial fibrillation (PAF) and left atrial (LA) structural remodeling are common in hypertrophic cardiomyopathy (HCM) patients, who are also at risk for adverse cardiovascular outcomes. OBJECTIVE We assessed whether PAF and/or LA remodeling was associated with adverse outcomes in HCM. METHODS We retrospectively studied 45 HCM patients with PAF (PAF group) and 59 HCM patients without atrial fibrillation (AF; no-AF group). LA/left ventricular (LV) function and mechanics were assessed by echocardiography. Patients were followed for development of the composite endpoint comprising heart failure, stroke, and death. RESULTS Clinical/demographic characteristics, degree of LV hypertrophy, and E/e' were similar in the two groups The PAF group had significantly higher LA volume, but lower LA ejection fraction (LAEF), LA contractile, and reservoir strain/strain rate than the no-AF group. During follow-up, 27 patients developed the composite endpoint. Incidence of the composite endpoint was similar in the two groups. Absolute values of 23.8% for reservoir strain and 10.2% for conduit strain were the best cutoffs for the composite endpoint, using receiver operating characteristic analysis. Kaplan-Meier survival analysis showed lower event-free survival in patients with reservoir strain ≤23.8% or conduit strain ≤10.2%. Univariate Cox analysis revealed an association between female sex, LAEF, LA reservoir/conduit strain, and LV global longitudinal strain with the composite endpoint. The association between LA reservoir/conduit strain and the composite endpoint persisted after controlling for age, sex, LAEF, and LV global longitudinal strain. CONCLUSIONS In this pilot HCM patient study, PAF was associated with a greater degree of LA myopathy, and low LA reservoir and conduit strain were associated with higher risk for adverse cardiovascular outcomes.
Collapse
|
48
|
Sivalokanathan S, Zghaib T, Greenland GV, Vasquez N, Kudchadkar SM, Kontari E, Lu DY, Dolores-Cerna K, van der Geest RJ, Kamel IR, Olgin JE, Abraham TP, Nazarian S, Zimmerman SL, Abraham MR. Hypertrophic Cardiomyopathy Patients With Paroxysmal Atrial Fibrillation Have a High Burden of Left Atrial Fibrosis by Cardiac Magnetic Resonance Imaging. JACC Clin Electrophysiol 2019; 5:364-375. [DOI: 10.1016/j.jacep.2018.10.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/30/2022]
|
49
|
Latif SR, Nguyen VQ, Peters DC, Soufer A, Henry ML, Grunseich K, Testani J, Hur DJ, Huber S, Mojibian H, Dicks D, Sinusas AJ, Meadows JL, Papoutsidakis N, Jacoby D, Baldassarre LA. Left atrial fibrosis correlates with extent of left ventricular myocardial delayed enhancement and left ventricular strain in hypertrophic cardiomyopathy. Int J Cardiovasc Imaging 2019; 35:1309-1318. [PMID: 30790116 DOI: 10.1007/s10554-019-01551-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/02/2019] [Indexed: 12/18/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is associated with increased left ventricular (LV) mass, decreased myocardial strain, and the presence of LV fibrosis and scar. The relationship between LV scar and fibrosis with left atrial (LA) fibrosis in the setting of HCM has not been examined. The purpose of this study is to demonstrate a correlation between the degree of LA fibrosis and LV parameters in subjects with HCM. Twenty-eight subjects with HCM were imaged on a 1.5T MRI scanner with cine, LV and LA late gadolinium enhancement (LGE) sequences. LA LGE and LA measurements were correlated with LV measurements of volumes, mass, strain, and LGE. Other clinical conditions and medication usage were also examined and evaluated for correlation with LA and LV parameters. LV LGE was identified in 24 (86%) of the cases and LA LGE was identified in all of the cases. Extent of LA fibrosis significantly correlated with percent LV LGE (r = 0.64, p = 0.001), but not with indexed LV mass or maximum wall thickness. Extent of LA fibrosis also moderately correlated with decreased LV global strain (radial, r = - 0.50, p = 0.013; circumferential, r = 0.47, p = 0.02; longitudinal, r = 0.52, p = 0.013). Increased LA systolic volume correlated moderately with LV end diastolic volume (r = 0.50, p = 0.006). Patients on therapy with Renin-Angiotensin-Aldosterone System (RAAS) Inhibition had significantly less LA LGE compared to those without (18.6% vs 10.8%, p = 0.023). LA fibrosis, as measured by LGE, is prevalent in HCM and is correlated with LV LGE. The correlation between LA and LV LGE might suggest either that LA fibrosis is a consequence of LV remodeling, or that LA and LV fibrosis are both manifestations of the same cardiomyopathic process. Further study is warranted to determine the causality of LA scar in this population.
Collapse
Affiliation(s)
- Syed R Latif
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Vinh Q Nguyen
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA.,Department of Cardiovascular Medicine, Allegheny General Hospital, 320 E North Ave, CVI 3, Pittsburgh, PA, 15212, USA
| | - Dana C Peters
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA
| | - Aaron Soufer
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Mariana L Henry
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Karl Grunseich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 505 Parnassus Avenue, M-391, San Francisco, CA, 94143-0628, USA
| | - Jeffrey Testani
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - David J Hur
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Steffen Huber
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA
| | - Hamid Mojibian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA
| | - Demetrius Dicks
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA
| | - Albert J Sinusas
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA
| | - Judith L Meadows
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Nikolaos Papoutsidakis
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Daniel Jacoby
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA
| | - Lauren A Baldassarre
- Department of Internal Medicine (Cardiology), Yale School of Medicine, 330 Cedar St, Boardman 110, P.O. Box 208056, New Haven, CT, 06520-8056, USA. .,Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208042, New Haven, CT, 06520-8042, USA. .,, New Haven, CT, USA.
| |
Collapse
|
50
|
Tingting H, Guangzhong L, Yanxiang Z, DongDong Y, Li S, Li W. Qiliqiangxin attenuates atrial structural remodeling in prolonged pacing-induced atrial fibrillation in rabbits. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:585-592. [PMID: 30627756 DOI: 10.1007/s00210-018-01611-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/27/2018] [Indexed: 12/27/2022]
Abstract
Qiliqiangxin (QL) can attenuate myocardial remodeling and improve cardiac function in some cardiac diseases, including heart failure and hypertension. This study was to explore the effects and mechanism of QL on atrial structural remodeling in atrial fibrillation (AF). Twenty-one rabbits were randomly divided into a sham-operation group, pacing group (pacing with 600 beats per minute for 4 weeks), and treatment group (2.5 g/kg/day). Before pacing, the rabbits received QL-administered p.o. for 1 week. We measured atrial electrophysiological parameters in all groups to evaluate AF inducibility and the atrial effective refractory period (AERP). Echocardiography evaluated cardiac function and structure. TUNEL detection, hematoxylin and eosin (HE) staining, and Masson's trichrome staining were performed. Immunohistochemistry and western blotting (WB) were used to detect alterations in calcium channel L-type dihydropyridine receptor α2 subunit (DHPR) and fibrosis-related regulatory factors. AF inducibility was markedly decreased after QL treatment. Furthermore, we found that AERP and DHPR were reduced significantly in pacing rabbits compared with sham rabbits; treatment with QL increased DHPR and AERP compared to the pacing group. The QL group showed significantly decreased mast cell density and improved atrial ejection fraction values compared with the pacing group. Moreover, QL decreased interventricular septum thickness (IVSd) and left ventricular end-diastolic diameter (LVEDD). Compared with the sham group, the levels of TGFβ1 and P-smad2/3 were significantly upregulated in the pacing group. QL reduced TGF-β1 and P-smad2/3 levels and downstream fibrosis-related factors. Our study demonstrated that QL treatment attenuates atrial structural remodeling potentially by inhibiting TGF-β1/P-smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Hou Tingting
- Department of Respiratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, Bengbu, China
| | - Liu Guangzhong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zang Yanxiang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yu DongDong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Sun Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Weimin Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China.
- Department of Cardiology, The First Hospital of Harbin, No. 151 Diduan Street, DaoLi District, Harbin, 150056, Heilongjiang Province, China.
| |
Collapse
|