1
|
Mo D, Wang M, Zhang P, Dai H, Guan J. Factors predicting the recurrence of atrial fibrillation after catheter ablation: A review. Heliyon 2024; 10:e34205. [PMID: 39071658 PMCID: PMC11277434 DOI: 10.1016/j.heliyon.2024.e34205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/26/2024] [Accepted: 07/04/2024] [Indexed: 07/30/2024] Open
Abstract
Atrial fibrillation (AF) is the most common and clinically significant type of cardiac arrhythmia. Although catheter ablation (CA) can restore sinus rhythm in patients with AF, some patients experience recurrence after the procedure. This requires us to find a simple and effective way to identify patients at a high risk of recurrence and to intervene early in the high-risk population to improve patient prognosis. The mechanism of AF recurrence is unclear, but it involves several aspects including patient history, inflammation, myocardial fibrosis, and genes. This article summarizes the current predictors of AF recurrence after CA, including myocardial fibrosis markers, inflammatory markers, MicroRNAs, Circular RNAs, AF recurrence scores, and imaging indicators. Each predictor has its own scope of application, and the predictive capacity and joint application of multiple predictors may improve the predictive power. In addition, we summarize the mechanisms involved in AF recurrence. We hope that this review will assist researchers understand the current predictors of AF recurrence and help them conduct further related studies.
Collapse
Affiliation(s)
- Degang Mo
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Mengmeng Wang
- Department of Cardiology, Qingdao University, Qingdao, 266011, China
| | - Peng Zhang
- Department of Cardiology, Qingdao University, Qingdao, 266011, China
| | - Hongyan Dai
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Jun Guan
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, 266011, China
| |
Collapse
|
2
|
Zeng Q, Wen BB, Liu X, Luo YY, Hu ZG, Huang L, Zhang XH, Huang XT, Zhou TT, Sang XX, Luo YY, Xiong DY, Luo ZQ, Liu W, Tang SY. NBR1-p62-Nrf2 mediates the anti-pulmonary fibrosis effects of protodioscin. Chin Med 2024; 19:60. [PMID: 38589903 PMCID: PMC11003024 DOI: 10.1186/s13020-024-00930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/31/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis is a persistent disease of the lung interstitium for which there is no efficacious pharmacological therapy. Protodioscin, a steroidal saponin, possesses diverse pharmacological properties; however, its function in pulmonary fibrosis is yet to be established. Hence, in this investigation, it was attempted to figure out the anti-pulmonary fibrosis influences of protodioscin and its pharmacological properties related to oxidative stress. METHODS A mouse lung fibrosis model was generated using tracheal injections of bleomycin, followed by intraperitoneal injection of different concentrations of protodioscin, and the levels of oxidative stress and fibrosis were detected in the lungs. Multiple fibroblasts were treated with TGF-β to induce their transition to myofibroblasts. It was attempted to quantify myofibroblast markers' expression levels and reactive oxygen species levels as well as Nrf2 activation after co-incubation of TGF-β with fibroblasts and different concentrations of protodioscin. The influence of protodioscin on the expression and phosphorylation of p62, which is associated with Nrf2 activation, were detected, and p62 related genes were predicted by STRING database. The effects of Nrf2 inhibitor or silencing of the Nrf2, p62 and NBR1 genes, respectively, on the activation of Nrf2 by protodioscin were examined. The associations between p62, NBR1, and Keap1 in the activation of Nrf2 by protodioscin was demonstrated using a co-IP assay. Nrf2 inhibitor were used when protodioscin was treated in mice with pulmonary fibrosis and lung tissue fibrosis and oxidative stress levels were detected. RESULTS In vivo, protodioscin decreased the levels of fibrosis markers and oxidative stress markers and activated Nrf2 in mice with pulmonary fibrosis, and these effects were inhibited by Nrf2 inhibitor. In vitro, protodioscin decreased the levels of myofibroblast markers and oxidative stress markers during myofibroblast transition and promoted Nrf2 downstream gene expression, with reversal of these effects after Nrf2, p62 and NBR1 genes were silenced or Nrf2 inhibitors were used, respectively. Protodioscin promoted the binding of NBR1 to p62 and Keap1, thereby reducing Keap1-Nrf2 binding. CONCLUSION The NBR1-p62-Nrf2 axis is targeted by protodioscin to reduce oxidative stress and inhibit pulmonary fibrosis.
Collapse
Affiliation(s)
- Qian Zeng
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Bin-Bin Wen
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xin Liu
- The Orthopedics Hospital of Traditional Chinese Medicine Zhuzhou City, Zhuzhou, Hunan, China
| | - Yong-Yu Luo
- Guiyang Second People's Hospital, Guiyang, Guizhou, China
| | - Zhen-Gang Hu
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Hua Zhang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ting-Ting Zhou
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiao-Xue Sang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Yang Luo
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Da-Yan Xiong
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Zi-Qiang Luo
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
3
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
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
|
5
|
Demirel O, Berezin AE, Mirna M, Boxhammer E, Gharibeh SX, Hoppe UC, Lichtenauer M. Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion. Biomedicines 2023; 11:1452. [PMID: 37239123 PMCID: PMC10216298 DOI: 10.3390/biomedicines11051452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Atrial fibrillation (AF) is associated with atrial remodeling, cardiac dysfunction, and poor clinical outcomes. External direct current electrical cardioversion is a well-developed urgent treatment strategy for patients presenting with recent-onset AF. However, there is a lack of accurate predictive serum biomarkers to identify the risks of AF relapse after electrical cardioversion. We reviewed the currently available data and interpreted the findings of several studies revealing biomarkers for crucial elements in the pathogenesis of AF and affecting cardiac remodeling, fibrosis, inflammation, endothelial dysfunction, oxidative stress, adipose tissue dysfunction, myopathy, and mitochondrial dysfunction. Although there is ample strong evidence that elevated levels of numerous biomarkers (such as natriuretic peptides, C-reactive protein, galectin-3, soluble suppressor tumorigenicity-2, fibroblast growth factor-23, turn-over collagen biomarkers, growth differential factor-15) are associated with AF occurrence, the data obtained in clinical studies seem to be controversial in terms of their predictive ability for post-cardioversion outcomes. Novel circulating biomarkers are needed to elucidate the modality of this approach compared with conventional predictive tools. Conclusions: Biomarker-based strategies for predicting events after AF treatment require extensive investigation in the future, especially in the presence of different gender and variable comorbidity profiles. Perhaps, a multiple biomarker approach exerts more utilization for patients with different forms of AF than single biomarker use.
Collapse
Affiliation(s)
- Ozan Demirel
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Alexander E. Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
- Internal Medicine Department, Zaporozhye State Medical University, 69035 Zaporozhye, Ukraine
| | - Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Elke Boxhammer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Sarah X. Gharibeh
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Uta C. Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| |
Collapse
|
6
|
Hofer F, Hammer A, Pailer U, Koller L, Kazem N, Steinacher E, Steinlechner B, Andreas M, Laufer G, Wojta J, Zelniker TA, Hengstenberg C, Niessner A, Sulzgruber P. Relationship of Fibroblast Growth Factor 23 With Hospitalization for Heart Failure and Cardiovascular Outcomes in Patients Undergoing Cardiac Surgery. J Am Heart Assoc 2023; 12:e027875. [PMID: 36802737 PMCID: PMC10111457 DOI: 10.1161/jaha.122.027875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Background Fibroblast growth factor 23 (FGF-23) is crucial in regulating phosphate and vitamin D metabolism and is moreover associated with an increased cardiovascular risk. The specific objective of this study was to investigate the influence of FGF-23 on cardiovascular outcomes, including hospitalization for heart failure (HHF), postoperative atrial fibrillation, and cardiovascular death, in an unselected patient population after cardiac surgery. Methods and Results Patients undergoing elective coronary artery bypass graft and/or cardiac valve surgery were prospectively enrolled. FGF-23 blood plasma concentrations were assessed before surgery. A composite of cardiovascular death/HHF was chosen as primary end point. A total of 451 patients (median age 70 years; 28.8% female) were included in the present analysis and followed over a median of 3.9 years. Individuals with higher FGF-23 quartiles showed elevated incidence rates of the composite of cardiovascular death/HHF (quartile 1, 7.1%; quartile 2, 8.6%; quartile 3, 15.1%; and quartile 4, 34.3%). After multivariable adjustment, FGF-23 modeled as a continuous variable (adjusted hazard ratio for a 1-unit increase in standardized log-transformed biomarker, 1.82 [95% CI, 1.34-2.46]) as well as using predefined risk groups and quartiles remained independently associated with the risk of cardiovascular death/HHF and the secondary outcomes, including postoperative atrial fibrillation. Reclassification analysis indicated that the addition of FGF-23 to N-terminal pro-B-type natriuretic peptide provides a significant improvement in risk discrimination (net reclassification improvement at the event rate, 0.58 [95% CI, 0.34-0.81]; P<0.001; integrated discrimination increment, 0.03 [95% CI, 0.01-0.05]; P<0.001). Conclusions FGF-23 is an independent predictor of cardiovascular death/HHF and postoperative atrial fibrillation in individuals undergoing cardiac surgery. Considering an individualized risk assessment, routine preoperative FGF-23 evaluation may improve detection of high-risk patients.
Collapse
Affiliation(s)
- Felix Hofer
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Andreas Hammer
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | | | - Lorenz Koller
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Niema Kazem
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Eva Steinacher
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | | | - Martin Andreas
- Department of Cardiac Surgery Medical University of Vienna Vienna Austria
| | - Günther Laufer
- Department of Cardiac Surgery Medical University of Vienna Vienna Austria
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Thomas A Zelniker
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Patrick Sulzgruber
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| |
Collapse
|
7
|
Fan X, Feng K, Liu Y, Yang L, Zhao Y, Tian L, Tang Y, Wang X. miR-135a Regulates Atrial Fibrillation by Targeting Smad3. Cardiovasc Ther 2023; 2023:8811996. [PMID: 37187923 PMCID: PMC10181910 DOI: 10.1155/2023/8811996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/12/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Background Atrial fibrillation (AF) is the most common arrhythmia in clinical. Atrial fibrosis is a hallmark feature of atrial structural remodeling in AF, which is regulated by the TGF-β1/Smad3 pathway. Recent studies have implicated that miRNAs are involved in the process of AF. However, the regulatory mechanisms of miRNAs remain largely unknown. This study is aimed at investigating the function and regulatory network of miR-135a in AF. Methods In vivo, the plasma was collected from patients with AF and non-AF subjects. Adult SD rats were induced by acetylcholine (ACh) (66 μg/ml)-CaCl2 (10 mg/ml) to establish an AF rat model. In vitro, atrial fibroblasts (AFs), isolated from adult SD rats, were treated with high-frequency electrical stimulation (HES) (12 h) and hypoxia (24 h) to mimic the AF and atrial fibrosis, respectively. miR-135a expression was detected through quantitative real-time polymerase chain reaction (qRT-PCR). The association between miR-135a and Smad3 was speculated by the TargetScan database and confirmed by the luciferase reporter assay. Fibrosis-related genes, Smad3, and TRPM7 were all assessed. Results The expression of miR-135a was markedly decreased in the plasma of AF patients and AF rats, which was consistent with that in HES-treated and hypoxia-treated AFs. Smad3 was identified as a target of miR-135a. the downregulation of miR-135a was associated with the enhancement of Smad3/TRPM7 expressions in AFs. Additionally, the knockdown of Smad3 significantly reduced the expression of TRPM7 and further inhibited atrial fibrosis. Conclusions Our study demonstrates that miR-135a regulates AF via Smad3/TRPM7, which is a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Xueting Fan
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmacy, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Kai Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yonghui Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Leixi Yang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yizhuo Zhao
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmacy, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, Ningbo 315010, China
| | - Liping Tian
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaozhi Wang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
8
|
Schnabel RB, Marinelli EA, Arbelo E, Boriani G, Boveda S, Buckley CM, Camm AJ, Casadei B, Chua W, Dagres N, de Melis M, Desteghe L, Diederichsen SZ, Duncker D, Eckardt L, Eisert C, Engler D, Fabritz L, Freedman B, Gillet L, Goette A, Guasch E, Svendsen JH, Hatem SN, Haeusler KG, Healey JS, Heidbuchel H, Hindricks G, Hobbs FDR, Hübner T, Kotecha D, Krekler M, Leclercq C, Lewalter T, Lin H, Linz D, Lip GYH, Løchen ML, Lucassen W, Malaczynska-Rajpold K, Massberg S, Merino JL, Meyer R, Mont L, Myers MC, Neubeck L, Niiranen T, Oeff M, Oldgren J, Potpara TS, Psaroudakis G, Pürerfellner H, Ravens U, Rienstra M, Rivard L, Scherr D, Schotten U, Shah D, Sinner MF, Smolnik R, Steinbeck G, Steven D, Svennberg E, Thomas D, True Hills M, van Gelder IC, Vardar B, Palà E, Wakili R, Wegscheider K, Wieloch M, Willems S, Witt H, Ziegler A, Daniel Zink M, Kirchhof P. Early diagnosis and better rhythm management to improve outcomes in patients with atrial fibrillation: the 8th AFNET/EHRA consensus conference. Europace 2022; 25:6-27. [PMID: 35894842 PMCID: PMC9907557 DOI: 10.1093/europace/euac062] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite marked progress in the management of atrial fibrillation (AF), detecting AF remains difficult and AF-related complications cause unacceptable morbidity and mortality even on optimal current therapy. This document summarizes the key outcomes of the 8th AFNET/EHRA Consensus Conference of the Atrial Fibrillation NETwork (AFNET) and the European Heart Rhythm Association (EHRA). Eighty-three international experts met in Hamburg for 2 days in October 2021. Results of the interdisciplinary, hybrid discussions in breakout groups and the plenary based on recently published and unpublished observations are summarized in this consensus paper to support improved care for patients with AF by guiding prevention, individualized management, and research strategies. The main outcomes are (i) new evidence supports a simple, scalable, and pragmatic population-based AF screening pathway; (ii) rhythm management is evolving from therapy aimed at improving symptoms to an integrated domain in the prevention of AF-related outcomes, especially in patients with recently diagnosed AF; (iii) improved characterization of atrial cardiomyopathy may help to identify patients in need for therapy; (iv) standardized assessment of cognitive function in patients with AF could lead to improvement in patient outcomes; and (v) artificial intelligence (AI) can support all of the above aims, but requires advanced interdisciplinary knowledge and collaboration as well as a better medico-legal framework. Implementation of new evidence-based approaches to AF screening and rhythm management can improve outcomes in patients with AF. Additional benefits are possible with further efforts to identify and target atrial cardiomyopathy and cognitive impairment, which can be facilitated by AI.
Collapse
Affiliation(s)
- Renate B Schnabel
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | | | - Elena Arbelo
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Giuseppe Boriani
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Polyclinic of Modena, Modena, Italy
| | - Serge Boveda
- Cardiology—Heart Rhythm Management Department, Clinique Pasteur, 45 Avenue de Lombez, 31076 Toulouse, France,Universiteit Ziekenhuis, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - A John Camm
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Institute, St. George's University of London, London, UK
| | - Barbara Casadei
- RDM, Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Winnie Chua
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nikolaos Dagres
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Mirko de Melis
- Medtronic Bakken Research Center, Maastricht, The Netherlands
| | - Lien Desteghe
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium,Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium,Heart Centre Hasselt, Jessa Hospital, Hasselt, Belgium
| | - Søren Zöga Diederichsen
- Department of Cardiology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - David Duncker
- Hannover Heart Rhythm Center, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Lars Eckardt
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Division of Electrophysiology, Department of Cardiology and Angiology, Münster, Germany
| | | | - Daniel Engler
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK,University Center of Cardiovascular Science Hamburg, Hamburg, Germany
| | - Ben Freedman
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | | | - Andreas Goette
- Atrial Fibrillation Network (AFNET), Muenster, Germany,St Vincenz Hospital, Paderborn, Germany
| | - Eduard Guasch
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Jesper Hastrup Svendsen
- Department of Cardiology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Karl Georg Haeusler
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Neurology, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Jeff S Healey
- Population Health Research Institute, McMaster University Hamilton, ON, Canada
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Gerhard Hindricks
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | | | | | - Dipak Kotecha
- University of Birmingham & University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | - Thorsten Lewalter
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Hospital Munich South, Department of Cardiology, Munich, Germany,Department of Cardiology, University of Bonn, Bonn, Germany
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Center and Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Maja Lisa Løchen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Wim Lucassen
- Amsterdam UMC (location AMC), Department General Practice, Amsterdam, The Netherlands
| | | | - Steffen Massberg
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany,German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, Germany
| | - Jose L Merino
- Arrhythmia & Robotic EP Unit, La Paz University Hospital, IDIPAZ, Madrid, Spain
| | | | - Lluıs Mont
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | | | - Lis Neubeck
- Arrhythmia & Robotic EP Unit, La Paz University Hospital, IDIPAZ, Madrid, Spain
| | - Teemu Niiranen
- Medtronic, Dublin, Ireland,Centre for Cardiovascular Health Edinburgh Napier University, Edinburgh, UK
| | - Michael Oeff
- Atrial Fibrillation Network (AFNET), Muenster, Germany
| | - Jonas Oldgren
- University of Turku and Turku University Hospital, Turku, Finland
| | | | - George Psaroudakis
- Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Helmut Pürerfellner
- School of Medicine, Belgrade University, Cardiology Clinic, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Ursula Ravens
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Bayer AG, Leverkusen, Germany
| | - Michiel Rienstra
- Ordensklinikum Linz, Elisabethinen, Cardiological Department, Linz, Austria
| | - Lena Rivard
- Institute of Experimental Cardiovascular Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Daniel Scherr
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ulrich Schotten
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Montreal Heart Institute, University of Montreal, Montreal, Canada
| | - Dipen Shah
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Moritz F Sinner
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Amsterdam UMC (location AMC), Department General Practice, Amsterdam, The Netherlands,Royal Brompton Hospital, London, UK
| | | | - Gerhard Steinbeck
- Atrial Fibrillation Network (AFNET), Muenster, Germany,MUMC+, Maastricht, The Netherlands
| | - Daniel Steven
- Atrial Fibrillation Network (AFNET), Muenster, Germany,University Hospital of Geneva, Cardiac Electrophysiology Unit, Geneva, Switzerland
| | - Emma Svennberg
- Center for Cardiology at Clinic Starnberg, Starnberg, Germany
| | - Dierk Thomas
- Atrial Fibrillation Network (AFNET), Muenster, Germany,University Hospital Cologne, Heart Center, Department of Electrophysiology, Cologne, Germany,Karolinska Institutet, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden,Department of Cardiology, Medical University Hospital, Heidelberg, Germany
| | - Mellanie True Hills
- HCR (Heidelberg Center for Heart Rhythm Disorders), Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Isabelle C van Gelder
- DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Burcu Vardar
- Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Elena Palà
- StopAfib.org, American Foundation for Women’s Health, Decatur, TX, USA
| | - Reza Wakili
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Karl Wegscheider
- Atrial Fibrillation Network (AFNET), Muenster, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Neurovascular Research Laboratory, Vall d’Hebron Institute of Research (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Mattias Wieloch
- Department of Cardiology and Vascular Medicine, Westgerman Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany,Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Germany
| | - Stephan Willems
- Atrial Fibrillation Network (AFNET), Muenster, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Department of Coagulation Disorders, Skane University Hospital, Lund University, Malmö, Sweden
| | | | | | - Matthias Daniel Zink
- Asklepios Hospital St Georg, Department of Cardiology and Internal Intensive Care Medicine, Faculty of Medicine, Semmelweis University Campus Hamburg, Hamburg, Germany
| | - Paulus Kirchhof
- Corresponding author. Tel: +49 40 7410 52438; Fax: +49 40 7410 55862. E-mail address:
| |
Collapse
|
9
|
Lu W, Zhu H, Wu J, Liao S, Cheng G, Li X. Rhein attenuates angiotensin II-induced cardiac remodeling by modulating AMPK–FGF23 signaling. J Transl Med 2022; 20:305. [PMID: 35794561 PMCID: PMC9258170 DOI: 10.1186/s12967-022-03482-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/11/2022] [Indexed: 12/07/2022] Open
Abstract
Background Increasing evidence indicates that myocardial oxidative injury plays a crucial role in the pathophysiology of cardiac hypertrophy (CH) and heart failure (HF). The active component of rhubarb, rhein exerts significant actions on oxidative stress and inflammation. Nonetheless, its role in cardiac remodeling remains unclear. Methods CH was induced by angiotensin II (Ang II, 1.4 mg/kg/d for 4 weeks) in male C57BL/6 J mice. Then, rhein (50 and 100 mg/kg) was injected intraperitoneally for 28 days. CH, fibrosis, oxidative stress, and cardiac function in the mice were examined. In vitro, neonatal rat cardiomyocytes (CMs) and cardiac fibroblasts (CFs) pre-treated with rhein (5 and 25 μM) were challenged with Ang II. We performed RNA sequencing to determine the mechanistic role of rhein in the heart. Results Rhein significantly suppressed Ang II-induced CH, fibrosis, and reactive oxygen species production and improved cardiac systolic dysfunction in vivo. In vitro, rhein significantly attenuated Ang II-induced CM hypertrophy and CF collagen expression. In addition, rhein obviously alleviated the increased production of superoxide induced by Ang II. Mechanistically, rhein inhibited FGF23 expression significantly. Furthermore, FGF23 overexpression abolished the protective effects of rhein on CMs, CFs, and cardiac remodeling. Rhein reduced FGF23 expression, mostly through the activation of AMPK (AMP-activated protein kinase). AMPK activity inhibition suppressed Ang II-induced CM hypertrophy and CF phenotypic transformation. Conclusion Rhein inhibited Ang II-induced CH, fibrosis, and oxidative stress during cardiac remodeling through the AMPK–FGF23 axis. These findings suggested that rhein could serve as a potential therapy in cardiac remodeling and HF. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03482-9.
Collapse
|
10
|
Inflammasome Signaling in Atrial Fibrillation. J Am Coll Cardiol 2022; 79:2349-2366. [DOI: 10.1016/j.jacc.2022.03.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/26/2022]
|
11
|
Scarano Pereira JP, Owen E, Martinino A, Akmal K, Abouelazayem M, Graham Y, Weiner S, Sakran N, Dekker LR, Parmar C, Pouwels S. Epicardial adipose tissue, obesity and the occurrence of atrial fibrillation: an overview of pathophysiology and treatment methods. Expert Rev Cardiovasc Ther 2022; 20:307-322. [PMID: 35443854 DOI: 10.1080/14779072.2022.2067144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Obesity is a chronic disease, which has significant health consequences and is a staggering burden to health care systems. Obesity can have harmful effects on the cardiovascular system, including heart failure, hypertension, coronary heart disease, and atrial fibrillation (AF). One of the possible substrates might be epicardial adipose tissue (EAT), which can be the link between AF and obesity. EAT is a fat deposit located between the myocardium and the visceral pericardium. Numerous studies have demonstrated that EAT plays a pivotal role in this relationship regarding atrial fibrillation. AREAS COVERED This review will focus on the role of obesity and the occurrence of atrial fibrillation (AF) and examine the connection between these and epicardial adipose tissue (EAT). The first part of this review will explain the pathophysiology of EAT and its association with the occurrence of AF. Secondly, we will review bariatric and metabolic surgery and its effects on EAT and AF. EXPERT COMMENTARY In this review, the epidemiology, pathophysiology, and treatments methods of AF are explained. Secondly the effects on EAT were elucidated. Due to the complex pathophysiological link between EAT, AF, and obesity, it is still uncertain which treatment strategy is superior.
Collapse
Affiliation(s)
| | - Eloise Owen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | | | - Kiran Akmal
- Faculty of Medicine, Brighton and Sussex Medical School, Brighton, United Kingdom
| | - Mohamed Abouelazayem
- Department of Surgery, Royal Free London Hospitals NHS Foundation, London, United Kingdom
| | - Yitka Graham
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom.,Facultad de Psucologia, Universidad Anahuac Mexico, Mexico City, Mexico
| | - Sylvia Weiner
- Department of Bariatric and Metabolic Surgery, Krankenhaus Nordwest, Frankfurt am Main, Germany
| | - Nasser Sakran
- Department of Surgery, Holy Family Hospital, Nazareth, Israel.,Azrieli, Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Lukas R Dekker
- Department of Cardiology, Catharina Hospital, Eindhoven, The Netherlands
| | - Chetan Parmar
- Department of Surgery, Whittington Health NHS Trust, London, United Kingdom
| | - Sjaak Pouwels
- Department of Intensive Care Medicine, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | | |
Collapse
|
12
|
Wang Y, Yang Y, He F. Insights into Concomitant Atrial Fibrillation and Chronic Kidney Disease. Rev Cardiovasc Med 2022; 23:105. [PMID: 35345272 DOI: 10.31083/j.rcm2303105] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 01/03/2025] Open
Abstract
Chronic kidney disease (CKD) shows a high prevalence and is characterized by progressive and irreversible loss of renal function. It is also associated with a high risk of cardiovascular disease. The CKD population often suffers from atrial fibrillation (AF), which is associated with cardiovascular and all-cause mortality. There is a pernicious bidirectional relationship between CKD and AF: renal dysfunction can help promote AF initiation and maintenance, while unmanageable AF often accelerates kidney function deterioration. Therefore, it is necessary to determine the interactive mechanisms between CKD and AF for optimal management of patients. However, due to renal function impairment and changes in the pharmacokinetics of anticoagulants, it is still elusive to formulate a normative therapeutic schedule for the AF population concomitant with CKD especially those with end-stage kidney failure. This review describes the possible molecular mechanisms linking CKD to AF and existing therapeutic options.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| |
Collapse
|
13
|
Comparison of Yield, Purity, and Functional Properties of Large-Volume Exosome Isolation Using Ultrafiltration and Polymer-Based Precipitation. Plast Reconstr Surg 2022; 149:638-649. [PMID: 35196679 DOI: 10.1097/prs.0000000000008830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mesenchymal stem cell-derived exosomes are known to produce effects similar to those of source cells and therefore represent a new approach in cell-free regenerative medicine. Their potential clinical application demands efficient isolation of stable and functional exosomes from a large volume of biological fluid. METHODS Exosomes from adipose-tissue conditioned medium of the same volume were isolated using either (1) ultrafiltration with size exclusion or (2) ExoQuick-TC. The isolated exosomes were characterized by protein concentration, particle size, exosomal marker expression, RNA expression profiles, and roles in dermal fibroblast proliferation and migration. RESULTS Both isolation methods produced exosomes within the size range defined for exosomes (50 to 200 nm) and common markers were enriched. Compared to the ExoQuick-TC precipitation method, the ultrafiltration method produced a significantly higher protein yield (p < 0.001) but a lower particle-to-protein ratio (p < 0.05); it also yielded higher RNA contents from the same fat tissue indicated by housekeeping genes, but with overall lower purity. The expression of several mRNAs and miRNAs related to tissue regeneration showed that there was no statistical difference between both methods, except miR-155 and miR-223 (p < 0.05). However, there was no difference in overall fibroblast proliferation and migration between exosomes isolated by these two methods. CONCLUSIONS Ultrafiltration with size exclusion demonstrated higher yields, acceptable purity, and comparable biophysical properties and biological functions to the more expensive commercial precipitation method. Therefore, it may conceivably be translated into yield-efficient and cost-effective modalities for therapeutic purposes. CLINICAL RELEVANCE STATEMENT Ultrafiltration with size exclusion may be amenable for exosome isolation from large-volume complex fluids such as tissue conditioned media for clinical application in future regenerative medicine.
Collapse
|
14
|
p62-Nrf2 Regulatory Loop Mediates the Anti-Pulmonary Fibrosis Effect of Bergenin. Antioxidants (Basel) 2022; 11:antiox11020307. [PMID: 35204190 PMCID: PMC8868171 DOI: 10.3390/antiox11020307] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) can severely disrupt lung function, leading to fatal consequences, and there is currently a lack of specific therapeutic drugs. Bergenin is an isocoumarin compound with lots of biological functions including antioxidant activity. This study evaluated the potential beneficial effects of bergenin on pulmonary fibrosis and investigated the possible mechanisms. We found that bergenin alleviated bleomycin-induced pulmonary fibrosis by relieving oxidative stress, reducing the deposition of the extracellular matrix (ECM) and inhibiting the formation of myofibroblasts. Furthermore, we showed that bergenin could induce phosphorylation and expression of p62 and activation of Nrf2, Nrf2 was required for bergenin-induced p62 upregulation, and p62 knockdown reduced bergenin-induced Nrf2 activity. More importantly, knockdown of Nrf2 or p62 could abrogate the antioxidant activity of bergenin and the inhibition effect of bergenin on TGF-β-induced ECM deposition and myofibroblast differentiation. Thereby, a regulatory loop is formed between p62 and Nrf2, which is an important target for bergenin aimed at treating pulmonary fibrosis.
Collapse
|
15
|
Meyre PB, Aeschbacher S, Blum S, Voellmin G, Kastner PM, Hennings E, Kaufmann BA, Kühne M, Osswald S, Conen D. Biomarkers associated with rhythm status after cardioversion in patients with atrial fibrillation. Sci Rep 2022; 12:1680. [PMID: 35102265 PMCID: PMC8803959 DOI: 10.1038/s41598-022-05769-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/13/2022] [Indexed: 01/13/2023] Open
Abstract
Biomarkers may help to improve our knowledge about the complex pathophysiology of atrial fibrillation (AF). In this study we sought to identify significant changes in biomarkers and clinical measures in patients with and without AF recurrence after electrical cardioversion. We measured 21 conventional and new biomarkers before and 30 days after electrical cardioversion and assessed the associations of changes in biomarker levels with rhythm status at follow-up. Significant between-group changes were observed for bone morphogenetic protein 10 (BMP10), N-terminal pro-B-type natriuretic peptide (NT-proBNP) and total bilirubin. Their respective changes were - 10.4%, - 62.0% and - 25.6% in patients with sinus rhythm, and 3.1%, 1.1% and - 9.4% in patients with recurrent AF, for a between-group difference of - 13.5% (95% confidence interval [CI] - 19.3% to - 7.6%; P < 0.001), - 63.1% (95% CI - 76.6% to - 49.6%; P < 0.001) and - 16.3% (95% CI - 27.9% to - 4.7%; P = 0.007). In multivariable models, the reductions of BMP10 and NT-proBNP were significantly associated with follow-up rhythm status (β coefficient per 1 - SD decrease, - 3.85; 95% CI - 6.34 to - 1.35; P = 0.003 for BMP10 and - 5.84; 95% CI - 10.22 to - 1.47; P = 0.009 for NT-proBNP. In conclusion, changes in BMP10 und NT-proBNP levels were independently associated with rhythm status after cardioversion, suggesting that these markers may be dependent on the actual heart rhythm.
Collapse
Affiliation(s)
- Pascal B Meyre
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland.
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland.
| | - Stefanie Aeschbacher
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - Steffen Blum
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - Gian Voellmin
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | | | - Elisa Hennings
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - Beat A Kaufmann
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - Michael Kühne
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - Stefan Osswald
- Division of Cardiology, Department of Medicine, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University Hospital Basel, Spitalstrasse 2, 4031, Basel, Switzerland
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
Sunderraj A, Rivera A, Gaddam M, Kim S, McCook J, O'Neal J, Lomasney J, Lloyd-Jones DM, Baumer Y, Powell-Wiley TM, Feinstein MJ. Associations of Social Vulnerability Index With Pathologic Myocardial Findings at Autopsy. Front Cardiovasc Med 2022; 8:805278. [PMID: 35004916 PMCID: PMC8733155 DOI: 10.3389/fcvm.2021.805278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Social vulnerability is an important determinant of cardiovascular health. Prior investigations have shown strong associations of social determinants of health with cardiovascular risk factors, imaging findings, and clinical events. However, limited data exist regarding the potential role of social vulnerability and related physiologic stressors on tissue-level pathology. Methods: We analyzed clinical data and linked autopsy reports from 853 decedent individuals who underwent autopsy from 4/6/2002 to 4/1/2021 at a large urban medical center. The mean age at death was 62.9 (SD = 15.6) and 49% of decedent individuals were men. The primary exposure was census-tract level composite social vulnerability index based on the Centers for Disease Control and Prevention Social Vulnerability Index (SVI). Individuals were geocoded to census tracts and assigned SVI accordingly. Four myocardial tissue-level outcomes from autopsy were recorded as present or absent: any coronary atherosclerosis, severe/obstructive coronary atherosclerosis, myocardial fibrosis, and/or myopericardial inflammation. Multivariable-adjusted logistic regression models were constructed with SVI as the primary exposure and covariates including age, sex, race, body mass index (BMI), diabetes, and hypertension. Additional analyses were performed stratified by clinical diagnoses of heart failure (HF) and coronary artery disease (CAD). Results: In the overall cohort, SVI was not associated with outcomes on cardiac pathology in multivariable-adjusted models. However, in stratified multivariable-adjusted analyses, higher SVI (higher social vulnerability) was associated with a higher odds of myocardial fibrosis among individuals without clinical diagnoses of HF. Conclusions: Higher indices of social vulnerability are associated with a higher odds of myocardial fibrosis at autopsy among individuals without known clinical diagnoses of HF. Potential pathophysiological mechanisms and implications for prevention/treatment of myocardial dysfunction require further study.
Collapse
Affiliation(s)
- Ashwin Sunderraj
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Adovich Rivera
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Meghna Gaddam
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Sarah Kim
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Juan McCook
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Janelle O'Neal
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jon Lomasney
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Donald M Lloyd-Jones
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States.,Intramural Research Program, National Institute on Minority Health and Health Disparities, Bethesda, MD, United States
| | - Matthew J Feinstein
- Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
17
|
Assessing and counteracting fibrosis is a cornerstone of the treatment of CKD secondary to systemic and renal limited autoimmune disorders. Autoimmun Rev 2021; 21:103014. [PMID: 34896651 DOI: 10.1016/j.autrev.2021.103014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an increasing cause of morbidity and mortality worldwide. Besides the higher prevalence of diabetes, hypertension and aging worldwide, immune mediated disorders remain an important cause of kidney disease and are especially prevalent in young adults. Regardless of the initial insult, final pathway to CKD and kidney failure is always the loss of normal tissue and fibrosis development, in which the dynamic equilibrium between extracellular matrix synthesis and degradation is disturbed, leading to excessive production and accumulation. During fibrosis, a multitude of cell types intervene at different levels, but myofibroblasts and inflammatory cells are considered critical in the process. They exert their effects through different molecular pathways, of which transforming growth factor β (TGF-β) has demonstrated to be of particular importance. Additionally, CKD itself promotes fibrosis due to the accumulation of toxins and hormonal changes, and proteinuria is simultaneously a manifestation of CKD and a specific driver of renal fibrosis. Pathways involved in renal fibrosis and CKD are closely interrelated, and although important advances have been made in our knowledge of them, it is still necessary to translate them into clinical practice. Given the complexity of this process, it is highly likely that its treatment will require a multi-target strategy to control the origin of the damage but also the mechanisms that perpetuate it. Fortunately, rapid technology development over the last years and new available drugs in the nephrologist's armamentarium give reasons for optimism that more personalized assistance for CKD and renal fibrosis will appear in the future.
Collapse
|
18
|
Zhu J, Zhu N, Xu J. miR‑101a‑3p overexpression prevents acetylcholine‑CaCl 2‑induced atrial fibrillation in rats via reduction of atrial tissue fibrosis, involving inhibition of EZH2. Mol Med Rep 2021; 24:740. [PMID: 34435649 PMCID: PMC8404104 DOI: 10.3892/mmr.2021.12380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/09/2021] [Indexed: 11/23/2022] Open
Abstract
Atrial fibrillation (AF), a clinically common heart arrhythmia, can result in left ventricular hypofunction, embolism and infarction. MicroRNA (miR)‑101a‑3p is lowly expressed in atrial tissues of patients with AF, but its role in AF remains unknown. In the present study, an AF model in rats was established via intravenous injection of acetylcholine (Ach)‑CaCl2. The downregulation of miR‑101a‑3p and upregulation of enhancer of zeste 2 homolog 2 (EZH2) were observed in AF model rats, indicating the involvement of miR‑101a‑3p and EZH2 in AF development. To study the effect of miR‑101a‑3p on AF in vivo, AF model rats were intramyocardially injected with lentivirus expressing miR‑101a‑3p. Electrocardiogram analysis identified that miR‑101a‑3p overexpression restored disappeared P wave and R‑R interphase changes in Ach‑CaCl2‑induced rats. Overexpression of miR‑101a‑3p also increased the atrial effective refractory period, reduced AF incidence and shortened duration of AF. Histological changes in atrial tissues were observed after H&E and Masson staining, which demonstrated that miR‑101a‑3p reduced atrial remodeling and fibrosis in AF model rats. Moreover, EZH2 expression was downregulated in atrial tissues by miR‑101a‑3p induction. Immunohistochemistry for collagen Ⅰ and collagen III revealed a reduction in atrial collagen synthesis following miR‑101a‑3p overexpression in AF model rats. Additionally, miR‑101a‑3p lowered the expression of pro‑fibrotic biomarkers, including TGF‑β1, connective tissue growth factor, fibronectin and α‑smooth muscle actin. The luciferase reporter assay results also indicated that EZH2 was a target gene of miR‑101a‑3p. Taken together, it was found that miR‑101a‑3p prevented AF in rats possibly via inhibition of collagen synthesis and atrial fibrosis by targeting EZH2, which provided a potential target for preventing AF.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Cardiology, The First Affiliated Hospital of USTC, Hefei, Anhui 230001, P.R. China
| | - Ning Zhu
- Department of Respiratory Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Jian Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
19
|
Liu L, Su J, Li R, Luo F. Changes in Intestinal Flora Structure and Metabolites Are Associated With Myocardial Fibrosis in Patients With Persistent Atrial Fibrillation. Front Nutr 2021; 8:702085. [PMID: 34497820 PMCID: PMC8419273 DOI: 10.3389/fnut.2021.702085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The occurrence of atrial fibrillation is often accompanied by myocardial fibrosis. An increasing number of studies have shown that intestinal flora is involved in the occurrence and development of a variety of cardiovascular diseases. This study explores the relationship between changes in the structure and function of intestinal flora and the progression of myocardial fibrosis in patients with persistent atrial fibrillation. Methods: Serum and stool samples were collected from 10 healthy people and 10 patients with persistent atrial fibrillation (PeAF), and statistical analyses were performed on the subjects' clinical baseline conditions. ELISA was used to measure the levels of carboxy-terminal telopeptide of type I collagen (CTX-I), propeptide of type I procollagen (PICP), procollagen III N-terminal propeptide (PIIINP), fibroblast growth factor-23 (FGF-23), and transforming growth factor-beta 1 (TGF-β1) in serum. Through 16S rRNA sequencing technology, the structural composition of the intestinal flora was detected and analyzed. In addition, metabolomics data were analyzed to determine the differences in the metabolites produced by the intestinal flora of the subjects. Results: By comparing the baseline data of the subjects, it was found that compared with those of the control group, the levels of creatinine (CRE) and serum uric acid (SUA) in the serum of PeAF patients were significantly increased. In addition, we found that the levels of CTX-I, PICP, PIIINP, and TGF-β1 in the serum of PeAF patients were significantly higher than those of the control group subjects. Although the control and PeAF groups exhibited no significant differences in the α diversity index, there were significant differences in the β diversity indexes (Bray-Curtis, weighted UniFrac and Anosim). At the phylum, family and species levels, the community structure and composition of the intestinal flora of the control group and those of the PeAF group showed significant differences. In addition, the compositions of the intestinal metabolites in the two different groups of people were significantly different. They were correlated considerably with PIIINP and specific communities in the intestinal flora. Conclusion: Pathologically, PeAF patients may have a higher risk of myocardial fibrosis. Systematically, abnormal changes in the structure and composition of the intestinal flora in PeAF patients may lead to differences in intestinal metabolites, which are involved in the process of myocardial fibrosis through metabolite pathways.
Collapse
Affiliation(s)
- Langsha Liu
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Su
- Department of Medical Administration, Zhuzhou Central Hospital, Zhuzhou, China
| | - Rui Li
- Operating Theatre, Zhuzhou Central Hospital, Zhuzhou, China
| | - Fanyan Luo
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Couselo-Seijas M, Rodríguez-Mañero M, González-Juanatey JR, Eiras S. Updates on epicardial adipose tissue mechanisms on atrial fibrillation. Obes Rev 2021; 22:e13277. [PMID: 34002458 DOI: 10.1111/obr.13277] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/19/2021] [Indexed: 02/06/2023]
Abstract
Obesity is a well-known risk factor for atrial fibrillation (AF). Local epi-myocardial or intra-myocardial adiposity caused by aging, obesity, or cardiovascular disease (CVD) is considered to be a better predictor of the risk of AF than general adiposity. Some of the described mechanisms suggest that epicardial adipose tissue (EAT) participates in structural remodeling owing to its endocrine activity or its infiltration between cardiomyocytes. Epicardial fat also wraps up the ganglionated plexi that reach the myocardium. Although the increment of volume/thickness and activity of EAT might modify autonomic activity, autonomic system dysfunction might also change the endocrine activity of epicardial fat in a feedback response. As a result, new preventive therapeutic strategies are focused on reducing adiposity and weight loss before AF ablation or inhibiting autonomic neurotransmitter secretion on fat pads during open-heart surgery to reduce the recurrence or postoperative risk of AF. In this manuscript, we review some of the novel findings regarding the pathophysiology and associated risk factors of AF, with special emphasis on the role of EAT in the electrical, structural, and molecular mechanisms of AF initiation and maintenance. In addition, we have included a brief note provided on epicardial fat preclinical models that could be useful for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Marinela Couselo-Seijas
- Translational Cardiology group, Health Research Institute, Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Moisés Rodríguez-Mañero
- Translational Cardiology group, Health Research Institute, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain.,Cardiovascular Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - José R González-Juanatey
- University of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain.,Cardiovascular Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiology group, Health Research Institute, Santiago de Compostela, Spain
| | - Sonia Eiras
- Translational Cardiology group, Health Research Institute, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
21
|
Papathanasiou KA, Giotaki SG, Vrachatis DA, Siasos G, Lambadiari V, Iliodromitis KE, Kossyvakis C, Kaoukis A, Raisakis K, Deftereos G, Papaioannou TG, Giannopoulos G, Avramides D, Deftereos SG. Molecular Insights in Atrial Fibrillation Pathogenesis and Therapeutics: A Narrative Review. Diagnostics (Basel) 2021; 11:diagnostics11091584. [PMID: 34573926 PMCID: PMC8470040 DOI: 10.3390/diagnostics11091584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The prevalence of atrial fibrillation (AF) is bound to increase globally in the following years, affecting the quality of life of millions of people, increasing mortality and morbidity, and beleaguering health care systems. Increasingly effective therapeutic options against AF are the constantly evolving electroanatomic substrate mapping systems of the left atrium (LA) and ablation catheter technologies. Yet, a prerequisite for better long-term success rates is the understanding of AF pathogenesis and maintenance. LA electrical and anatomical remodeling remains in the epicenter of current research for novel diagnostic and treatment modalities. On a molecular level, electrical remodeling lies on impaired calcium handling, enhanced inwardly rectifying potassium currents, and gap junction perturbations. In addition, a wide array of profibrotic stimuli activates fibroblast to an increased extracellular matrix turnover via various intermediaries. Concomitant dysregulation of the autonomic nervous system and the humoral function of increased epicardial adipose tissue (EAT) are established mediators in the pathophysiology of AF. Local atrial lymphomononuclear cells infiltrate and increased inflammasome activity accelerate and perpetuate arrhythmia substrate. Finally, impaired intracellular protein metabolism, excessive oxidative stress, and mitochondrial dysfunction deplete atrial cardiomyocyte ATP and promote arrhythmogenesis. These overlapping cellular and molecular alterations hinder us from distinguishing the cause from the effect in AF pathogenesis. Yet, a plethora of therapeutic modalities target these molecular perturbations and hold promise in combating the AF burden. Namely, atrial selective ion channel inhibitors, AF gene therapy, anti-fibrotic agents, AF drug repurposing, immunomodulators, and indirect cardiac neuromodulation are discussed here.
Collapse
Affiliation(s)
- Konstantinos A. Papathanasiou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Sotiria G. Giotaki
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Dimitrios A. Vrachatis
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Gerasimos Siasos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Vaia Lambadiari
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Charalampos Kossyvakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Andreas Kaoukis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Konstantinos Raisakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Gerasimos Deftereos
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Theodore G. Papaioannou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Dimitrios Avramides
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Spyridon G. Deftereos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
- Correspondence: ; Tel.: +30-21-0583-2355
| |
Collapse
|
22
|
Li W, Qi N, Wang S, Jiang W, Liu T. miR-455-5p regulates atrial fibrillation by targeting suppressor of cytokines signaling 3. J Physiol Biochem 2021; 77:481-490. [PMID: 33792885 DOI: 10.1007/s13105-021-00808-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/08/2021] [Indexed: 01/02/2023]
Abstract
Atrial fibrillation (AF) is a condition that heart beats quaveringly or irregularly, which causes blood clots, heart failure, stroke, and other heart-related complications. Therefore, early diagnosis and timely preventions are necessary for AF treatment. Compelling evidence indicated that microRNAs (miRNAs) become emerging biomarkers of AF; thus, we aimed to investigate the possibility of miR-455-5p as an AF marker to provide a new strategy for early diagnosis of AF. A minipump containing angiotensin II was implanted into mice to induce AF, and adeno-associated virus (AAV) carrying anti-miR-negative control (NC) or anti-miR-455-5p was injected into the pericardial space of mice respectively. Next, myocytes isolated from wild-type newborn mice were stimulated with angiotensin II and anti-miR-NC or anti-miR-455-5p mimic. The results showed that the expression of miR-455-5p was positively correlated with the severity of AF, and miR-455-5p mimic accelerated the progression of AF by directly binding to its target gene suppressor of cytokines signaling 3 (SOCS3), leading to the activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway. On the contrary, inhibition of miR-455-5p expression effectively ameliorated AF. In conclusion, miR-455-5p might serve as a biomarker of AF.
Collapse
Affiliation(s)
- Weiling Li
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei, China
| | - Na Qi
- Department of Respiratory and Critical Care Medicine, Hengshui People Hospital, Hengshui, 053000, China
| | - Shuo Wang
- Department of Cardiology, Shijiazhuang General Hospital, No. 9 Fangbei Road, Shijiazhuang, 050000, Hebei, China
| | - Wenyan Jiang
- Department of Cardiology, Tangshan People's Hospital, Tangshan, 063000, Hebei, China
| | - Tao Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
23
|
Vázquez-Sánchez S, Poveda J, Navarro-García JA, González-Lafuente L, Rodríguez-Sánchez E, Ruilope LM, Ruiz-Hurtado G. An Overview of FGF-23 as a Novel Candidate Biomarker of Cardiovascular Risk. Front Physiol 2021; 12:632260. [PMID: 33767635 PMCID: PMC7985069 DOI: 10.3389/fphys.2021.632260] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor-23 (FGF)-23 is a phosphaturic hormone involved in mineral bone metabolism that helps control phosphate homeostasis and reduces 1,25-dihydroxyvitamin D synthesis. Recent data have highlighted the relevant direct FGF-23 effects on the myocardium, and high plasma levels of FGF-23 have been associated with adverse cardiovascular outcomes in humans, such as heart failure and arrhythmias. Therefore, FGF-23 has emerged as a novel biomarker of cardiovascular risk in the last decade. Indeed, experimental data suggest FGF-23 as a direct mediator of cardiac hypertrophy development, cardiac fibrosis and cardiac dysfunction via specific myocardial FGF receptor (FGFR) activation. Therefore, the FGF-23/FGFR pathway might be a suitable therapeutic target for reducing the deleterious effects of FGF-23 on the cardiovascular system. More research is needed to fully understand the intracellular FGF-23-dependent mechanisms, clarify the downstream pathways and identify which could be the most appropriate targets for better therapeutic intervention. This review updates the current knowledge on both clinical and experimental studies and highlights the evidence linking FGF-23 to cardiovascular events. The aim of this review is to establish the specific role of FGF-23 in the heart, its detrimental effects on cardiac tissue and the possible new therapeutic opportunities to block these effects.
Collapse
Affiliation(s)
- Sara Vázquez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jonay Poveda
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Rodríguez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Luis M. Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
24
|
Zhou C, Shi Z, Ouyang N, Ruan X. Hyperphosphatemia and Cardiovascular Disease. Front Cell Dev Biol 2021; 9:644363. [PMID: 33748139 PMCID: PMC7970112 DOI: 10.3389/fcell.2021.644363] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hyperphosphatemia or even serum phosphate levels within the “normal laboratory range” are highly associated with increased cardiovascular disease risk and mortality in the general population and patients suffering from chronic kidney disease (CKD). As the kidney function declines, serum phosphate levels rise and subsequently induce the development of hypertension, vascular calcification, cardiac valvular calcification, atherosclerosis, left ventricular hypertrophy and myocardial fibrosis by distinct mechanisms. Therefore, phosphate is considered as a promising therapeutic target to improve the cardiovascular outcome in CKD patients. The current therapeutic strategies are based on dietary and pharmacological reduction of serum phosphate levels to prevent hyperphosphatemia in CKD patients. Large randomized clinical trials with hard endpoints are urgently needed to establish a causal relationship between phosphate excess and cardiovascular disease (CVD) and to determine if lowering serum phosphate constitutes an effective intervention for the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengyu Shi
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Ouyang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiongzhong Ruan
- John Moorhead Research Laboratory, Centre for Nephrology, University College London (UCL) Medical School, London, United Kingdom.,Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Noubiap JJ, Sanders P, Nattel S, Lau DH. Biomarkers in Atrial Fibrillation: Pathogenesis and Clinical Implications. Card Electrophysiol Clin 2021; 13:221-233. [PMID: 33516400 DOI: 10.1016/j.ccep.2020.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Biomarkers derived from the key components of the pathophysiology of atrial fibrillation (AF) and its complications have the potential to play an important role in earlier characterization of AF phenotype and in risk prediction of adverse clinical events, which may translate into improved management strategies. C-reactive protein, natriuretic peptides, cardiac troponins, growth differentiation factor-15, and fibroblast growth factor-23 have been shown to be the most promising biomarkers in AF. Some biomarkers have already been included in clinical risk scores to predict postoperative AF, thromboembolism, major bleeding, and death. Considerably more work is needed to bring these novel biomarkers into routine clinical management of patients with AF.
Collapse
Affiliation(s)
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, University of Adelaide, Adelaide, Australia; Department of Cardiology, Royal Adelaide Hospital, Adelaide, Australia
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Dennis H Lau
- Centre for Heart Rhythm Disorders, University of Adelaide, Adelaide, Australia; Department of Cardiology, Royal Adelaide Hospital, Adelaide, Australia.
| |
Collapse
|
26
|
Roy C, Lejeune S, Slimani A, de Meester C, Ahn As SA, Rousseau MF, Mihaela A, Ginion A, Ferracin B, Pasquet A, Vancraeynest D, Beauloye C, Vanoverschelde JL, Horman S, Gruson D, Gerber BL, Pouleur AC. Fibroblast growth factor 23: a biomarker of fibrosis and prognosis in heart failure with preserved ejection fraction. ESC Heart Fail 2020; 7:2494-2507. [PMID: 32578967 PMCID: PMC7524237 DOI: 10.1002/ehf2.12816] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/05/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
AIMS Besides regulating calcium-phosphate metabolism, fibroblast growth factor 23 (FGF-23) has been associated with incident heart failure (HF) and left ventricular hypertrophy. However, data about FGF-23 in HF and preserved ejection fraction (HFpEF) remain limited. The aim of this study was to assess the association between FGF-23 levels, clinical and imaging characteristics, particularly diffuse myocardial fibrosis, and prognosis in HFpEF patients. METHODS AND RESULTS We prospectively included 143 consecutive HFpEF patients (78 ± 8 years, 61% female patients) and 31 controls of similar age and gender (75 ± 6 years, 61% female patients). All subjects underwent a complete two-dimensional echocardiography and cardiac magnetic resonance with extracellular volume (ECV) assessment by T1 mapping. FGF-23 was measured at baseline. Among the patients, differences in clinical and imaging characteristics across tertiles of FGF-23 levels were analysed with a trend test across the ordered groups. Patients were followed over time for a primary endpoint of all-cause mortality and first HF hospitalization and a secondary endpoint of all-cause mortality. Median FGF-23 was significantly higher in HFpEF patients compared with controls of similar age and gender (247 [115; 548] RU/mL vs. 61 [51; 68] RU/mL, P < 0.001). Among HFpEF patients, higher FGF-23 levels were associated with female sex, higher incidence of atrial fibrillation, lower haemoglobin, worse renal function, and higher N terminal pro brain natriuretic peptide levels (P for trend < 0.05 for all). Regarding imaging characteristics, patients with higher FGF-23 levels had greater left atrial volumes, worse right ventricular systolic function, and more fibrosis estimated by ECV (P for trend < 0.05 for all). FGF-23 was moderately correlated with ECV (r = 0.46, P < 0.001). Over a mean follow-up of 30 ± 8 months, 43 patients (31%) died and 69 patients (49%) were hospitalized for HF. A total of 87 patients (62%) reached the primary composite endpoint of all-cause mortality and/or first HF hospitalization. In multivariate Cox regression analysis for the primary endpoint, FGF-23 (HR: 3.44 [2.01; 5.90], P < 0.001) and E wave velocities (HR: 1.01 [1.00; 1.02], P = 0.034) were independent predictors of the primary composite endpoint. In multivariate Cox regression analysis for the secondary endpoint, ferritin (HR: 1.02 [1.01; 1.03], P < 0.001), FGF-23 (HR: 2.85 [1.26; 6.44], P = 0.012), and ECV (HR: 1.26 [1.03; 1.23], P = 0.008) were independent predictors of all-cause mortality. CONCLUSIONS Fibroblast growth factor 23 (FGF-23) levels were significantly higher in HFpEF patients compared with controls of similar age and gender. FGF-23 was correlated with fibrosis evaluated by ECV. High levels of FGF-23 were significantly associated with signs of disease severity such as worse renal function, larger left atrial volumes, and right ventricular dysfunction. Moreover, FGF-23 was a strong predictor of poor outcome (mortality and first HF hospitalization).
Collapse
Affiliation(s)
- Clotilde Roy
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Sibille Lejeune
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Alisson Slimani
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Christophe de Meester
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Sylvie A Ahn As
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Michel F Rousseau
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Amzulescu Mihaela
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Audrey Ginion
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Benjamin Ferracin
- Clinical Biology Department, Cliniques Universitaires St Luc, Université catholique de Louvain, Brussels, Belgium
| | - Agnès Pasquet
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - David Vancraeynest
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Beauloye
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Louis Vanoverschelde
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Damien Gruson
- Clinical Biology Department, Cliniques Universitaires St Luc, Université catholique de Louvain, Brussels, Belgium
| | - Bernhard L Gerber
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Anne-Catherine Pouleur
- Division of Cardiology, Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, Avenue Hippocrate, 10, Brussels, 1200, Belgium.,Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
27
|
Law JP, Price AM, Pickup L, Radhakrishnan A, Weston C, Jones AM, McGettrick HM, Chua W, Steeds RP, Fabritz L, Kirchhof P, Pavlovic D, Townend JN, Ferro CJ. Clinical Potential of Targeting Fibroblast Growth Factor-23 and αKlotho in the Treatment of Uremic Cardiomyopathy. J Am Heart Assoc 2020; 9:e016041. [PMID: 32212912 PMCID: PMC7428638 DOI: 10.1161/jaha.120.016041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease is highly prevalent, affecting 10% to 15% of the adult population worldwide and is associated with increased cardiovascular morbidity and mortality. As chronic kidney disease worsens, a unique cardiovascular phenotype develops characterized by heart muscle disease, increased arterial stiffness, atherosclerosis, and hypertension. Cardiovascular risk is multifaceted, but most cardiovascular deaths in patients with advanced chronic kidney disease are caused by heart failure and sudden cardiac death. While the exact drivers of these deaths are unknown, they are believed to be caused by uremic cardiomyopathy: a specific pattern of myocardial hypertrophy, fibrosis, with both diastolic and systolic dysfunction. Although the pathogenesis of uremic cardiomyopathy is likely to be multifactorial, accumulating evidence suggests increased production of fibroblast growth factor-23 and αKlotho deficiency as potential major drivers of cardiac remodeling in patients with uremic cardiomyopathy. In this article we review the increasing understanding of the physiology and clinical aspects of uremic cardiomyopathy and the rapidly increasing knowledge of the biology of both fibroblast growth factor-23 and αKlotho. Finally, we discuss how dissection of these pathological processes is aiding the development of therapeutic options, including small molecules and antibodies, directly aimed at improving the cardiovascular outcomes of patients with chronic kidney disease and end-stage renal disease.
Collapse
Affiliation(s)
- Jonathan P. Law
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Anna M. Price
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Luke Pickup
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Ashwin Radhakrishnan
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
| | - Chris Weston
- Institute of Immunology and ImmunotherapyUniversity of BirminghamUnited Kingdom
- NIHR Birmingham Biomedical Research CentreUniversity Hospitals Birmingham NHS Foundation Trust and University of BirminghamUnited Kingdom
| | - Alan M. Jones
- School of PharmacyUniversity of BirminghamUnited Kingdom
| | | | - Winnie Chua
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Richard P. Steeds
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Larissa Fabritz
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Paulus Kirchhof
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Davor Pavlovic
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Jonathan N. Townend
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Charles J. Ferro
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| |
Collapse
|
28
|
Su L, Yao Y, Song W. Downregulation of miR-96 suppresses the profibrogenic functions of cardiac fibroblasts induced by angiotensin II and attenuates atrial fibrosis by upregulating KLF13. Hum Cell 2020; 33:337-346. [PMID: 32034721 DOI: 10.1007/s13577-020-00326-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/28/2020] [Indexed: 01/05/2023]
Abstract
Atrial fibrosis is a hallmark of structural remodeling in atrial fibrillation (AF). MicroRNA-96 (miR-96) has been reported to be associated with pulmonary fibrosis and hepatic fibrosis. Nevertheless, the role of miR-96 in atrial fibrosis is still unclear. In our study, we showed that miR-96 is upregulated in human atrial tissues from AF patients and positively correlates with collagen I and collagen III levels. Knockdown of miR-96 reduced angiotensin II (Ang-II)-induced cardiac-fibroblast proliferation, migration, and collagen production, whereas ectopic expression of miR-96 yielded opposite results. Furthermore, we demonstrated that miR-96 represses KLF13 expression, subsequently promoting Ang-II-induced proliferation, migration, and collagen production in murine cardiac fibroblasts. Moreover, we observed that the knockdown of miR-96 attenuated the Ang-II-induced atrial fibrosis in a mouse model of AF. All the findings point to a potential target for the prevention or treatment of atrial fibrosis.
Collapse
Affiliation(s)
- Lijie Su
- Department of Cardiovascular, Shu Guang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
| | - Yili Yao
- Department of Cardiovascular, Shu Guang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
| | - Wei Song
- Department of Cardiovascular, Shu Guang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China.
| |
Collapse
|
29
|
MFGE8 attenuates Ang-II-induced atrial fibrosis and vulnerability to atrial fibrillation through inhibition of TGF-β1/Smad2/3 pathway. J Mol Cell Cardiol 2020; 139:164-175. [PMID: 31958465 DOI: 10.1016/j.yjmcc.2020.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 12/12/2019] [Accepted: 01/10/2020] [Indexed: 12/23/2022]
Abstract
Atrial fibrillation (AF) is characterized by potentiated growth of atrial fibroblasts and excessive deposition of the extracellular matrix. Atrial fibrosis has emerged as a hallmark of atrial structural remodeling linked to AF. Nonetheless, the specific mechanism underlying the progression of atrial fibrosis to AF is still largely unknown. MFGE8 (milk fat globule-EGF factor 8) is a soluble glycoprotein associated with many human diseases. Recently, a number of studies revealed that MFGE8 plays a crucial role in heart disease. Yet, MFGE8 regulation and function in the process of atrial fibrosis and vulnerability to AF remain unexplored. In this study, we found that the expression of MFGE8 was downregulated in the atriums of patients with AF compared with individuals without AF. In addition, the expression of MFGE8 was lower in atriums of angiotensin II (Ang-II)-stimulated rats as compared with the sham group. In vitro, silencing of MFGE8 by small interfering RNA significantly increased Ang-II-induced atrial fibrosis, whereas administration of recombinant human MFGE8 (rhMFGE8) attenuated the atrial fibrosis. Moreover, we found that the activated TGF-β1/Smad2/3 pathway after Ang-II treatment was significantly potentiated by the MFGE8 knockdown but inhibited by rhMFGE8 in vitro. Inhibition of integrin β3 which is the receptor for MFGE8, suppressed the TGF-β1/Smad2/3 activating effects of the MFGE8 knockdown in Ang-II-treated rat atrial fibroblasts. Finally, we administered rhMFGE8 to rats; it attenuated atrial fibrosis and remodeling and further reduced AF vulnerability induced by Ang-II, indicating that MFGE8 might have the potential both as a novel biomarker and as a therapeutic target in atrial fibrosis and AF.
Collapse
|
30
|
Uncovering Synergistic Mechanism of Chinese Herbal Medicine in the Treatment of Atrial Fibrillation with Obstructive Sleep Apnea Hypopnea Syndrome by Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8691608. [PMID: 31949472 PMCID: PMC6948354 DOI: 10.1155/2019/8691608] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/18/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Paroxysmal atrial fibrillation (AF) combined with obstructive sleep apnea hypopnea syndrome (OSAHS) is very common in clinical practice. Traditional Chinese medicine (TCM) rule of regulating the liver based on psycho-cardiology shows satisfactory effectiveness in the treatment of paroxysmal AF combined with OSAHS. However, its underlying pharmacological mechanism has not yet been elucidated. This study applied network pharmacology to identify 94 active components in the six TCM liver-regulating herbs and 182 corresponding targets from several databases and comprehensive literature studies, as well as retrieved AF combined with OSAHS-related targets. Cytoscape software was adopted to construct the component-component target network and component-putative target-AF combined with OSAHS target network. Then, we obtained 38 putative therapeutic targets against AF combined with OSAHS. After the production of a putative therapeutic target interaction network, topological analysis was adopted to determine the core targets of TCM liver-regulating herbs in the treatment of paroxysmal AF combined with OSAHS. For all putative therapeutic targets, biological process analysis and pathway enrichment analysis were utilized to investigate the possible mechanism of TCM liver-regulating herbs in the treatment of paroxysmal AF combined with OSAHS. Mechanistically, it included positive regulation of nitric oxide biosynthetic process, aging, response to hypoxia, TNF signaling pathway, HIF-1 signaling pathway, PI3K-Akt signaling pathway, neuroactive ligand-receptor interaction, and calcium signaling pathway. Especially, six core targets of TCM liver-regulating herbs, namely, TNF, STAT3, AKT1, IL-6, TP53, and INS, were significant in the regulation of the above biological processes and pathways. This study demonstrates the multicomponent, multitarget, and multipathway feature of TCM liver-regulating herbs, provides an extensional foundation for further research, and facilitates the reasonable application of TCM liver-regulating herbs in treating paroxysmal AF combined with OSAHS.
Collapse
|
31
|
FGF23-Mediated Activation of Local RAAS Promotes Cardiac Hypertrophy and Fibrosis. Int J Mol Sci 2019; 20:ijms20184634. [PMID: 31540546 PMCID: PMC6770314 DOI: 10.3390/ijms20184634] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are prone to developing cardiac hypertrophy and fibrosis, which is associated with increased fibroblast growth factor 23 (FGF23) serum levels. Elevated circulating FGF23 was shown to induce left ventricular hypertrophy (LVH) via the calcineurin/NFAT pathway and contributed to cardiac fibrosis by stimulation of profibrotic factors. We hypothesized that FGF23 may also stimulate the local renin–angiotensin–aldosterone system (RAAS) in the heart, thereby further promoting the progression of FGF23-mediated cardiac pathologies. We evaluated LVH and fibrosis in association with cardiac FGF23 and activation of RAAS in heart tissue of 5/6 nephrectomized (5/6Nx) rats compared to sham-operated animals followed by in vitro studies with isolated neonatal rat ventricular myocytes and fibroblast (NRVM, NRCF), respectively. Uremic rats showed enhanced cardiomyocyte size and cardiac fibrosis compared with sham. The cardiac expression of Fgf23 and RAAS genes were increased in 5/6Nx rats and correlated with the degree of cardiac fibrosis. In NRVM and NRCF, FGF23 stimulated the expression of RAAS genes and induced Ngal indicating mineralocorticoid receptor activation. The FGF23-mediated hypertrophic growth of NRVM and induction of NFAT target genes were attenuated by cyclosporine A, losartan and spironolactone. In NRCF, FGF23 induced Tgfb and Ctgf, which were suppressed by losartan and spironolactone, only. Our data suggest that FGF23-mediated activation of local RAAS in the heart promotes cardiac hypertrophy and fibrosis.
Collapse
|