1
|
Salvas JP, Moore‐Morris T, Goergen CJ, Sicard P. Left atrial reservoir strain as a predictor of cardiac dysfunction in a murine model of pressure overload. Acta Physiol (Oxf) 2025; 241:e14277. [PMID: 39822162 PMCID: PMC11737473 DOI: 10.1111/apha.14277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/15/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
AIM Left atrial (LA) strain is emerging as a valuable metric for evaluating cardiac function, particularly under pathological conditions such as pressure overload. This preclinical study investigates the predictive utility of LA strain on cardiac function in a murine model subjected to pressure overload, mimicking pathologies such as hypertension and aortic stenosis. METHODS High-resolution ultrasound was performed in a cohort of mice (n = 16) to evaluate left atrial and left ventricular function at baseline and 2 and 4 weeks after transverse aortic constriction (TAC). Acute adaptations in cardiac function were assessed in a subgroup of mice (n = 10) with 3 days post-TAC imaging. RESULTS We report an increase in LA max volume from 11.0 ± 4.3 μL at baseline to 26.7 ± 16.7 μL at 4 weeks (p = 0.002) and a decrease in LA reservoir strain from 20.8 ± 5.4% at baseline to 10.2 ± 6.9% at 4 weeks (p = 0.001). In the acute phase, LA strain dysfunction was present at 3 days (p < 0.001), prior to alterations in LA volume (p = 0.856) or left ventricular (LV) ejection fraction (p = 0.120). LA reservoir strain correlated with key indicators of cardiac performance including left ventricular (LV) ejection fraction (r = 0.541, p < 0.001), longitudinal strain (r = -0.637, p < 0.001), and strain rate (r = 0.378, p = 0.007). Furthermore, markers of atrial structure and function including LA max volume (AUC = 0.813, p = 0.003), ejection fraction (AUC = 0.853, p = 0.001), and strain (AUC = 0.884, p < 0.001) all predicted LV dysfunction. CONCLUSION LA strain and function assessments provide a reliable, non-invasive method for the early detection and prediction of cardiac dysfunction in a model of pressure overload.
Collapse
Affiliation(s)
- John P. Salvas
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
- Indiana University School of MedicineIndianapolisIndianaUSA
| | - Thomas Moore‐Morris
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Craig J. Goergen
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Pierre Sicard
- PhyMedExp, IPAM/Biocampus, University of Montpellier, INSERM, CNRS.MontpellierFrance
| |
Collapse
|
2
|
Henderson DJ, Alqahtani A, Chaudhry B, Cook A, Eley L, Houyel L, Hughes M, Keavney B, de la Pompa JL, Sled J, Spielmann N, Teboul L, Zaffran S, Mill P, Liu KJ. Beyond genomic studies of congenital heart defects through systematic modelling and phenotyping. Dis Model Mech 2024; 17:dmm050913. [PMID: 39575509 PMCID: PMC11603121 DOI: 10.1242/dmm.050913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024] Open
Abstract
Congenital heart defects (CHDs), the most common congenital anomalies, are considered to have a significant genetic component. However, despite considerable efforts to identify pathogenic genes in patients with CHDs, few gene variants have been proven as causal. The complexity of the genetic architecture underlying human CHDs likely contributes to this poor genetic discovery rate. However, several other factors are likely to contribute. For example, the level of patient phenotyping required for clinical care may be insufficient for research studies focused on mechanistic discovery. Although several hundred mouse gene knockouts have been described with CHDs, these are generally not phenotyped and described in the same way as CHDs in patients, and thus are not readily comparable. Moreover, most patients with CHDs carry variants of uncertain significance of crucial cardiac genes, further complicating comparisons between humans and mouse mutants. In spite of major advances in cardiac developmental biology over the past 25 years, these advances have not been well communicated to geneticists and cardiologists. As a consequence, the latest data from developmental biology are not always used in the design and interpretation of studies aimed at discovering the genetic causes of CHDs. In this Special Article, while considering other in vitro and in vivo models, we create a coherent framework for accurately modelling and phenotyping human CHDs in mice, thereby enhancing the translation of genetic and genomic studies into the causes of CHDs in patients.
Collapse
Affiliation(s)
- Deborah J. Henderson
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Ahlam Alqahtani
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Andrew Cook
- University College London, Zayed Centre for Research, London WC1N 1DZ, UK
| | - Lorraine Eley
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Lucile Houyel
- Congenital and Pediatric Cardiology Unit, M3C-Necker, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Cité, 149 Rue de Sèvres, 75015 Paris, France
| | - Marina Hughes
- Cardiology Department, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - John Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto M5G 1XS, Canada. Department of Medical Biophysics, University of Toronto, Toronto M5G 1XS, Canada
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich (GmbH), German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Lydia Teboul
- Mary Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, UK
| | - Stephane Zaffran
- Aix Marseille Université, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France
| | - Pleasantine Mill
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- MRC Human Genetics Unit, Institute for Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen J. Liu
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
3
|
Samani SL, Barlow SC, Freeburg LA, Catherwood GM, Churillo AM, Jones TL, Altomare D, Ji H, Shtutman M, Zile MR, Shazly T, Spinale FG. Heart failure with preserved ejection fraction in pigs causes shifts in posttranscriptional checkpoints. Am J Physiol Heart Circ Physiol 2024; 327:H1272-H1285. [PMID: 39240258 PMCID: PMC11560071 DOI: 10.1152/ajpheart.00551.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024]
Abstract
Left ventricular pressure overload (LVPO) can lead to heart failure with a preserved ejection fraction (HFpEF) and LV chamber stiffness (LV Kc) is a hallmark. This project tested the hypothesis that the development of HFpEF due to an LVPO stimulus will alter posttranscriptional regulation, specifically microRNAs (miRs). LVPO was induced in pigs (n = 9) by sequential ascending aortic cuff and age- and weight-matched pigs (n = 6) served as controls. LV function was measured by echocardiography and LV Kc by speckle tracking. LV myocardial miRs were quantified using an 84-miR array. Treadmill testing and natriuretic peptide-A (NPPA) mRNA levels in controls and LVPO were performed (n = 10, n = 9, respectively). LV samples from LVPO and controls (n = 6, respectively) were subjected to RNA sequencing. LV mass and Kc increased by over 40% with LVPO (P < 0.05). A total of 30 miRs shifted with LVPO of which 11 miRs correlated to LV Kc (P < 0.05) that mapped to functional domains relevant to Kc such as fibrosis and calcium handling. LVPO resulted in reduced exercise tolerance (oxygen saturation, respiratory effort) and NPPA mRNA levels increased by fourfold (P < 0.05). RNA analysis identified several genes that mapped to specific miRs that were altered with LVPO. In conclusion, a specific set of miRs are changed in a large animal model consistent with the HFpEF phenotype, were related to LV stiffness properties, and several miRs mapped to molecular pathways that may hold relevance in terms of prognosis and therapeutic targets.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is an ever-growing cause for the HF burden. HFpEF is particularly difficult to treat as the mechanisms responsible for this specific form of HF are poorly understood. Using a relevant large animal model, this study uncovered a unique molecular signature with the development of HFpEF that regulates specific biological pathways relevant to the progression of this ever-growing cause of HF.
Collapse
Affiliation(s)
- Stephanie L Samani
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Shayne C Barlow
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Lisa A Freeburg
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Grayson M Catherwood
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Traci L Jones
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Diego Altomare
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Hao Ji
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Michael Shtutman
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Michael R Zile
- Division of Cardiology, Ralph H. Johnson Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Tarek Shazly
- College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, United States
| | - Francis G Spinale
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| |
Collapse
|
4
|
Zhan G, Wang X, Wang X, Li J, Tang Y, Bi H, Yang X, Xia Y. Dapagliflozin: A sodium-glucose cotransporter 2 inhibitor, attenuates angiotensin II-induced atrial fibrillation by regulating atrial electrical and structural remodeling. Eur J Pharmacol 2024; 978:176712. [PMID: 38906237 DOI: 10.1016/j.ejphar.2024.176712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024]
Abstract
AIM Atrial fibrillation (AF), the most common arrhythmia, is characterized by atrial electrical and structural remodeling. Previous studies have found that sodium-glucose cotransporter 2 inhibitor (SGLT2i) can protect myocardium in a glucose independent mechanism. But the role of SGLT2i in regulating AF remains largely unknown. This study, we aimed to investigate the effect of Dapagliflozin (DAPA) in reducing AF susceptibility via inhibiting electrical and structural remodeling. METHOD The mouse model was established by Angiotensin II (2000 ng/kg/min) infusion for 3 weeks, and an in vitro model was generated by stimulating HL-1 and primary mouse fibroblast with Ang II (1 μM) for 24 h. Programmed electrical stimulation, ECG and whole-cell patch clamp were used to detect DAPA effect on atrial electrical remodeling induced by Ang II. To observe DAPA effect on atrial structural remodeling induced by Ang II, we used echocardiographic, H&E and Masson staining to evaluate atrial dilation. To further explore the protective mechanism of DAPA, we adopt in silico molecular docking approaches to investigate the binding affinity of Ang II and CaMKII at Met-281 site. Western blot was to detect expression level of CaMKII, ox-CaMKII, Nav1.5, Kv4.3, Kv4.2, Kchip2, Kir2.1 and Cx40. RESULTS Ang II induced AF, atrial dilatation and fibrosis, led to atrial electrical and structural remodeling. However, these effects were markedly abrogated by DAPA treatment, a specific SGLT2i. Our observation of atrial electrical activity in mice revealed that DAPA could rescue the prolonged action potential duration (APD) and the abnormal currents of IK1, Ito and INaL triggered by Ang II infusion. DAPA could reduce the binding affinity of Ang II and CaMKII at Met-281 site, which indicated that DAPA may directly alleviate the activation of CaMKII caused by Ang II. DAPA could reduce the upregulation of ox-CaMKII caused by Ang II infusion in atrial tissues. Moreover, DAPA also ameliorated the aberrant expression levels of electrical activity related proteins (Nav1.5, Kv4.3, Kv4.2, Kchip2, Kir2.1 and Cx40) and fibrosis related signal pathways (TGF-β1, p-smad/smad) caused by Ang II. Furthermore, we confirmed that DAPA, as well as other SGLT2i (EMPA, CANA), could reverse these abnormalities caused by Ang II incubation in HL-1 cells and primary mouse fibroblasts, respectively. CONCLUSION Overall, our study identifies DAPA, a widely used SGLT2i, contributes to inhibiting Ang II-induced ox-CaMKII upregulation and electrical and structural remodeling to reduce AF susceptibility, suggesting that DAPA may be a potential therapy of treating AF.
Collapse
Affiliation(s)
- Ge Zhan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xinying Wang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xin Wang
- Department of Ultrasound, The Affiliated Hospital of Innermongolia Medical University, Huhhot 010050, China; Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiatian Li
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Yuqi Tang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Hailian Bi
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China; Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China.
| | - Yunlong Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China; Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China.
| |
Collapse
|
5
|
Liang D, Zhou L, Zhou H, Zhang F, Fang G, Leng J, Wu Y, Zhang Y, Yang A, Liu Y, Chen YH. A GABAergic system in atrioventricular node pacemaker cells controls electrical conduction between the atria and ventricles. Cell Res 2024; 34:556-571. [PMID: 38849501 PMCID: PMC11291642 DOI: 10.1038/s41422-024-00980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
Physiologically, the atria contract first, followed by the ventricles, which is the prerequisite for normal blood circulation. The above phenomenon of atrioventricular sequential contraction results from the characteristically slow conduction of electrical excitation of the atrioventricular node (AVN) between the atria and the ventricles. However, it is not clear what controls the conduction of electrical excitation within AVNs. Here, we find that AVN pacemaker cells (AVNPCs) possess an intact intrinsic GABAergic system, which plays a key role in electrical conduction from the atria to the ventricles. First, along with the discovery of abundant GABA-containing vesicles under the surface membranes of AVNPCs, key elements of the GABAergic system, including GABA metabolic enzymes, GABA receptors, and GABA transporters, were identified in AVNPCs. Second, GABA synchronously elicited GABA-gated currents in AVNPCs, which significantly weakened the excitability of AVNPCs. Third, the key molecular elements of the GABAergic system markedly modulated the conductivity of electrical excitation in the AVN. Fourth, GABAA receptor deficiency in AVNPCs accelerated atrioventricular conduction, which impaired the AVN's protective potential against rapid ventricular frequency responses, increased susceptibility to lethal ventricular arrhythmias, and decreased the cardiac contractile function. Finally, interventions targeting the GABAergic system effectively prevented the occurrence and development of atrioventricular block. In summary, the endogenous GABAergic system in AVNPCs determines the slow conduction of electrical excitation within AVNs, thereby ensuring sequential atrioventricular contraction. The endogenous GABAergic system shows promise as a novel intervention target for cardiac arrhythmias.
Collapse
Affiliation(s)
- Dandan Liang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, China
| | - Liping Zhou
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huixing Zhou
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fulei Zhang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guojian Fang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junwei Leng
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yahan Wu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuemei Zhang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anqi Yang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Liu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi-Han Chen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China.
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, China.
| |
Collapse
|
6
|
Fang L, Chen Q, Cheng X, Li X, Zou T, Chen J, Xiang G, Xue Q, Li Y, Zhang J. Calcium-mediated DAD in membrane potentials and triggered activity in atrial myocytes of ETV1 f / fMyHC Cre /+ mice. J Cell Mol Med 2024; 28:e70005. [PMID: 39159135 PMCID: PMC11332596 DOI: 10.1111/jcmm.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 08/21/2024] Open
Abstract
The E-twenty-six variant 1 (ETV1)-dependent transcriptome plays an important role in atrial electrical and structural remodelling and the occurrence of atrial fibrillation (AF), but the underlying mechanism of ETV1 in AF is unclear. In this study, cardiomyocyte-specific ETV1 knockout (ETV1f/fMyHCCre/+, ETV1-CKO) mice were constructed to observe the susceptibility to AF and the underlying mechanism in AF associated with ETV1-CKO mice. AF susceptibility was examined by intraesophageal burst pacing, induction of AF was increased obviously in ETV1-CKO mice than WT mice. Electrophysiology experiments indicated shortened APD50 and APD90, increased incidence of DADs, decreased density of ICa,L in ETV1-CKO mice. There was no difference in VINACT,1/2 and VACT,1/2, but a significantly longer duration of the recovery time after inactivation in the ETV1-CKO mice. The recording of intracellular Ca2+ showed that there was significantly increased in the frequency of calcium spark, Ca2+ transient amplitude, and proportion of SCaEs in ETV1-CKO mice. Reduction of Cav1.2 rather than NCX1 and SERCA2a, increase RyR2, p-RyR2 and CaMKII was reflected in ETV1-CKO group. This study demonstrates that the increase in calcium spark and SCaEs corresponding to Ca2+ transient amplitude may trigger DAD in membrane potential in ETV1-CKO mice, thereby increasing the risk of AF.
Collapse
Affiliation(s)
- Li‐Hua Fang
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
| | - Qian Chen
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
- Department of Critical Care Medicine Division FourFujian Provincial HospitalFuzhouFujianPeople's Republic of China
| | - Xian‐Lu Cheng
- Department of CardiologyNanping First Hospital Affiliated to Fujian Medical UniversityNanpingFujianPeople's Republic of China
| | - Xiao‐Qian Li
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
| | - Tian Zou
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
- Department of CardiologyFujian Provincial HospitalFuzhouFujianPeople's Republic of China
| | - Jian‐Quan Chen
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
- Department of CardiologyFujian Provincial HospitalFuzhouFujianPeople's Republic of China
| | - Guo‐Jian Xiang
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
- Department of CardiologyFujian Provincial HospitalFuzhouFujianPeople's Republic of China
| | - Qiao Xue
- Department of Cardiology, the Sixth Medical CenterChinese People's Liberation Army HospitalBeijingPeople's Republic of China
| | - Yang Li
- Department of Cardiology, the Sixth Medical CenterChinese People's Liberation Army HospitalBeijingPeople's Republic of China
| | - Jian‐Cheng Zhang
- Shengli Clinical Medicine College of Fujian Medical UniversityFuzhouFujianChina
- Department of CardiologyFujian Provincial HospitalFuzhouFujianPeople's Republic of China
| |
Collapse
|
7
|
Rao S, Aggarwal S, Mani S, Balasubramanian A, Veluswami K. Uncovering the Role of Direct Oral Anticoagulants in Stroke Prevention for Atrial Fibrillation: A Review of the Literature. Cureus 2024; 16:e63675. [PMID: 39092362 PMCID: PMC11293488 DOI: 10.7759/cureus.63675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Atrial fibrillation (AF) is a predominant contributor to morbidity and mortality, and stroke prevention remains the mainstay for the management of AF. The precise mechanism involved in thrombus formation remains unknown. However, factors such as stretch-induced fibrosis, endothelial dysfunction, disordered atrial contractions, and pro-thrombotic states have been postulated for the development of AF. Various risk assessment strategies have been acknowledged for determining the risk of stroke in AF, of which the congestive heart failure, hypertension, age ≥75, diabetes, stroke, vascular disease, age between 65-74, and female sex (CHA2DS2-VASc) score remains the ultimate risk stratification tool. For the longest time, vitamin K antagonists (VKA) were the only oral anticoagulants available but were associated with an increased risk of bleeding. Recently, direct oral anticoagulants (DOACs) were approved and considered more efficient and safer than or as secure as warfarin in stroke prevention and lowering intra-cranial bleeding events. The pharmacodynamics and pharmacokinetics of DOACs were also clarified in this article. This review article compiles current evidence-based data on the role of DOACs, uncovering their underlying mechanisms, and comparing their efficacy with warfarin in stroke prevention in AF.
Collapse
Affiliation(s)
- Sudipta Rao
- Internal Medicine, JSS Medical College, Mysore, IND
| | | | - Sweatha Mani
- Internal Medicine, K.A.P. Viswanatham Government Medical College, Tiruchirappalli, IND
| | | | | |
Collapse
|
8
|
Berezin OO, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AE. Diagnostic and predictive abilities of myokines in patients with heart failure. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:45-98. [PMID: 39059994 DOI: 10.1016/bs.apcsb.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Myokines are defined as a heterogenic group of numerous cytokines, peptides and metabolic derivates, which are expressed, synthesized, produced, and released by skeletal myocytes and myocardial cells and exert either auto- and paracrine, or endocrine effects. Previous studies revealed that myokines play a pivotal role in mutual communications between skeletal muscles, myocardium and remote organs, such as brain, vasculature, bone, liver, pancreas, white adipose tissue, gut, and skin. Despite several myokines exert complete divorced biological effects mainly in regulation of skeletal muscle hypertrophy, residential cells differentiation, neovascularization/angiogenesis, vascular integrity, endothelial function, inflammation and apoptosis/necrosis, attenuating ischemia/hypoxia and tissue protection, tumor growth and malignance, for other occasions, their predominant effects affect energy homeostasis, glucose and lipid metabolism, adiposity, muscle training adaptation and food behavior. Last decade had been identified 250 more myokines, which have been investigating for many years further as either biomarkers or targets for heart failure management. However, only few myokines have been allocated to a promising tool for monitoring adverse cardiac remodeling, ischemia/hypoxia-related target-organ dysfunction, microvascular inflammation, sarcopenia/myopathy and prediction for poor clinical outcomes among patients with HF. This we concentrate on some most plausible myokines, such as myostatin, myonectin, brain-derived neurotrophic factor, muslin, fibroblast growth factor 21, irisin, leukemia inhibitory factor, developmental endothelial locus-1, interleukin-6, nerve growth factor and insulin-like growth factor-1, which are suggested to be useful biomarkers for HF development and progression.
Collapse
Affiliation(s)
- Oleksandr O Berezin
- Luzerner Psychiatrie AG, Department of Senior Psychiatrie, St. Urban, Switzerland
| | - Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
9
|
Song S, Zhang X, Huang Z, Zhao Y, Lu S, Zeng L, Cai F, Wang T, Pei Z, Weng X, Luo W, Lu H, Wei Z, Wu J, Yu P, Shen L, Zhang X, Sun A, Ge J. TEA domain transcription factor 1(TEAD1) induces cardiac fibroblasts cells remodeling through BRD4/Wnt4 pathway. Signal Transduct Target Ther 2024; 9:45. [PMID: 38374140 PMCID: PMC10876703 DOI: 10.1038/s41392-023-01732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 02/21/2024] Open
Abstract
Cardiac fibroblasts (CFs) are the primary cells tasked with depositing and remodeling collagen and significantly associated with heart failure (HF). TEAD1 has been shown to be essential for heart development and homeostasis. However, fibroblast endogenous TEAD1 in cardiac remodeling remains incompletely understood. Transcriptomic analyses revealed consistently upregulated cardiac TEAD1 expression in mice 4 weeks after transverse aortic constriction (TAC) and Ang-II infusion. Further investigation revealed that CFs were the primary cell type expressing elevated TEAD1 levels in response to pressure overload. Conditional TEAD1 knockout was achieved by crossing TEAD1-floxed mice with CFs- and myofibroblasts-specific Cre mice. Echocardiographic and histological analyses demonstrated that CFs- and myofibroblasts-specific TEAD1 deficiency and treatment with TEAD1 inhibitor, VT103, ameliorated TAC-induced cardiac remodeling. Mechanistically, RNA-seq and ChIP-seq analysis identified Wnt4 as a novel TEAD1 target. TEAD1 has been shown to promote the fibroblast-to-myofibroblast transition through the Wnt signalling pathway, and genetic Wnt4 knockdown inhibited the pro-transformation phenotype in CFs with TEAD1 overexpression. Furthermore, co-immunoprecipitation combined with mass spectrometry, chromatin immunoprecipitation, and luciferase assays demonstrated interaction between TEAD1 and BET protein BRD4, leading to the binding and activation of the Wnt4 promoter. In conclusion, TEAD1 is an essential regulator of the pro-fibrotic CFs phenotype associated with pathological cardiac remodeling via the BRD4/Wnt4 signalling pathway.
Collapse
Affiliation(s)
- Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaokai Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zihang Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yongchao Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shuyang Lu
- Department of cardiac surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linqi Zeng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Fengze Cai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Tongyao Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhiqiang Pei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Wei Luo
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zilun Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jian Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peng Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaochun Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Xiao Z, Pan Y, Kong B, Meng H, Shuai W, Huang H. Ubiquitin-specific protease 38 promotes inflammatory atrial fibrillation induced by pressure overload. Europace 2023; 26:euad366. [PMID: 38288617 PMCID: PMC10823351 DOI: 10.1093/europace/euad366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/05/2023] [Indexed: 02/01/2024] Open
Abstract
AIMS Atrial structural and electrical remodelling is a major reason for the initiation and perpetuation of atrial fibrillation (AF). Ubiquitin-specific protease 38 (USP38) is a deubiquitinating enzyme, but its function in the heart remains unknown. The aim of this study was to investigate the effect of USP38 in pressure overload-induced AF. METHODS AND RESULTS Cardiac-specific knockout USP38 and cardiac-specific transgenic USP38 mice and their corresponding control mice were used in this study. After 4 weeks with or without aortic banding (AB) surgery, atrial echocardiography, atrial histology, electrophysiological study, and molecular analysis were assessed. Ubiquitin-specific protease 38 knockout mice showed a remarkable improvement in vulnerability to AF, atrial weight and diameter, atrial fibrosis, and calcium-handling protein expression after AB surgery. Conversely, USP38 overexpression further increased susceptibility to AF by exacerbating atrial structural and electrical remodelling. Mechanistically, USP38 interacted with and deubiquitinated nuclear factor-kappa B (NF-κB), and USP38 overexpression increased the level of p-NF-κB in vivo and in vitro, accompanied by the upregulation of NOD-like receptor protein 3 (NLRP3) and inflammatory cytokines, suggesting that USP38 contributes to adverse effects by driving NF-κB/NLRP3-mediated inflammatory responses. CONCLUSION Overall, our study indicates that USP38 promotes pressure overload-induced AF through targeting NF-κB/NLRP3-mediated inflammatory responses.
Collapse
Affiliation(s)
- Zheng Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Yucheng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Hong Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| |
Collapse
|
11
|
Hu Z, Ding L, Yao Y. Atrial fibrillation: mechanism and clinical management. Chin Med J (Engl) 2023; 136:2668-2676. [PMID: 37914663 PMCID: PMC10684204 DOI: 10.1097/cm9.0000000000002906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 11/03/2023] Open
Abstract
ABSTRACT Atrial fibrillation (AF), the most common sustained arrhythmia, is associated with a range of symptoms, including palpitations, cognitive impairment, systemic embolism, and increased mortality. It places a significant burden on healthcare systems worldwide. Despite decades of research, the precise mechanisms underlying AF remain elusive. Current understanding suggests that factors like stretch-induced fibrosis, epicardial adipose tissue (EAT), chronic inflammation, autonomic nervous system (ANS) imbalances, and genetic mutations all play significant roles in its development. In recent years, the advent of wearable devices has revolutionized AF diagnosis, enabling timely detection and monitoring. However, balancing early diagnosis with efficient resource utilization presents new challenges for healthcare providers. AF management primarily focuses on stroke prevention and symptom alleviation. Patients at high risk of thromboembolism require anticoagulation therapy, and emerging pipeline drugs, particularly factor XI inhibitors, hold promise for achieving effective anticoagulation with reduced bleeding risks. The scope of indications for catheter ablation in AF has expanded significantly. Pulsed field ablation, as a novel energy source, shows potential for improving success rates while ensuring safety. This review integrates existing knowledge and ongoing research on AF pathophysiology and clinical management, with emphasis on diagnostic devices, next-generation anticoagulants, drugs targeting underlying mechanisms, and interventional therapies. It offers a comprehensive mosaic of AF, providing insights into its complexities.
Collapse
Affiliation(s)
| | | | - Yan Yao
- Cardiac Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
12
|
Yu S, Sun Y, Wang N, Lv X, Si J, Zhang X, Zhang Y, Liu Y. Effect of sodium-glucose cotransporter 2 inhibitors on left atrial remodeling and prognosis in patients with type 2 diabetes and heart failure with reduced ejection fraction. J Cardiovasc Med (Hagerstown) 2023; 24:829-837. [PMID: 37642649 DOI: 10.2459/jcm.0000000000001523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
AIMS Sodium-glucose cotransporter 2 inhibitors (SGLT2is) have been found to minimize hospitalization for heart failure and cardiovascular death. Cardiac reverse remodeling may be a mechanism responsible for the favorable clinical efficacy of SGLT2is on heart failure. To date, few studies have examined their effects on the left atrium. Therefore, the purpose of this study was to explore whether SGLT2is improve left atrial adverse remodeling in patients with type 2 diabetes and heart failure with reduced ejection fraction (HFrEF). METHODS A single-center, retrospective, observational study was conducted. Consecutive patients with type 2 diabetes and HFrEF hospitalized at the First Affiliated Hospital of Dalian Medical University for acute decompensated heart failure between 1 January 2019 and 1 March 2022 were identified. On the basis of their treatment strategies, the enrolled participants were classified into SGLT2i and non-SGLT2i groups. The primary end point was all-cause mortality. Changes in left atrial echocardiographic indices from baseline to follow-up were also assessed. RESULTS A total of 198 patients (mean age: 63.96 ± 12.11 years, 20.71% women) were included. Greater reductions from baseline were seen with SGLT2i in the left atrial diameter ( P < 0.001), left atrial superior-inferior diameter ( P = 0.027), left atrial transverse diameter ( P = 0.020), left atrial volume ( P = 0.005), and left atrial volume index ( P = 0.004). Moreover, 48 cases (48.48%) in the SGLT2i group and 33 (33.33%) in the non-SGLT2i group showed left atrial reverse remodeling ( P = 0.003). Survival analysis demonstrated significantly lower overall mortality in the SGLT2i group compared with the non-SGLT2i group. CONCLUSION This study found that SGLT2i therapy promoted left atrial structure reverse remodeling. This beneficial effect may be a vital mechanism by which SGLT2i improved clinical outcomes in patients with HFrEF.
Collapse
Affiliation(s)
- Songqi Yu
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Yuxi Sun
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ning Wang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Xin Lv
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Jinping Si
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Xinxin Zhang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Yanli Zhang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| | - Ying Liu
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province
| |
Collapse
|
13
|
Xue Z, Zhu J, Liu J, Wang L, Ding J. Research progress of non-coding RNA in atrial fibrillation. Front Cardiovasc Med 2023; 10:1210762. [PMID: 37522088 PMCID: PMC10379658 DOI: 10.3389/fcvm.2023.1210762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Atrial fibrillation (AF) is a common arrhythmia in clinic, and its incidence is increasing year by year. In today's increasingly prevalent society, ageing poses a huge challenge to global healthcare systems. AF not only affects patients' quality of life, but also causes thrombosis, heart failure and other complications in severe cases. Although there are some measures for the diagnosis and treatment of AF, specific serum markers and targeted therapy are still lacking. In recent years, ncRNAs have become a hot topic in cardiovascular disease research. These ncRNAs are not only involved in the occurrence and development of AF, but also in pathophysiological processes such as myocardial infarction and atherosclerosis, and are potential biomarkers of cardiovascular diseases. We believe that the understanding of the pathophysiological mechanism of AF and the study of diagnosis and treatment targets can form a more systematic diagnosis and treatment framework of AF and provide convenience for individuals with AF and the society.
Collapse
|
14
|
Shi W, Li X, Su Y, Liu D, Wu L, Li S, He W, Zhong G, Jiang Z. PILRA is associated with immune cells infiltration in atrial fibrillation based on bioinformatics and experiment validation. Front Cardiovasc Med 2023; 10:1082015. [PMID: 37396579 PMCID: PMC10311564 DOI: 10.3389/fcvm.2023.1082015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Background and aims inflammation plays an important role in atrial fibrillation (AF). In this study, we investigated the significance of immune cell infiltration in AF and identified the potential Hub genes involved in the regulation of immune cell infiltration in AF. Methods we obtained AF datasets from the GEO database and analyzed them for obtaining differentially expressed genes (DEGs) by R software. Then, we performed GO, KEGG, and GSEA enrichment analyses of DEGs. The Hub genes of AF were determined by least absolute shrinkage selection operator (LASSO) regression analysis and weighted gene co-expression network analysis (WGCNA). Their validation was verified by using quantitative polymerase chain reaction (qPCR) in the AF rat model. Finally, we used a single sample GSEA (ssGSEA) to analyze immune cell infiltration and its relationship with hub genes. Results We obtained 298 DGEs from the heatmap and found that DGEs were closely related to inflammation, immunity, and cytokine interactions by enrichment analyses. We obtained 10 co-expression modules by WGCNA. Among them, the module including CLEC4A, COTL1, EVI2B, FCER1G, GAPT, HCST, NCF2, PILRA, TLR8, and TYROBP had the highest correlation with AF. Four Hub genes (PILRA, NCF2, EVI2B, GAPT) were obtained further by LASSO analysis. The results suggested that the expression level of PILRA was significantly elevated in the rats with AF by qPCR, compared to the rats without AF. The results revealed that the infiltration of neutrophils, macrophages, monocytes, mast cells, immature B cells, myeloid-derived suppressor cell (MDSC), dendritic cell, and T cells and their partial subpopulations were closely related to AF by ssGSEA analysis, and PILRA was positively correlated with immature B cell, monocyte, macrophage, mast cell, dendritic cell, and T cells and their partial subpopulations by Spearman correlation analysis. Conclusions PILRA was closely related to multiple types of immune cell infiltration, which may be associated with AF. PILRA may be a novel target of intervention for AF.
Collapse
Affiliation(s)
- Weihua Shi
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xiaoli Li
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yongxing Su
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Dezhao Liu
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Liying Wu
- Department of Pharmacy, Guangxi Zhuang Autonomous Region People’s Hospital, Nanning, China
| | - Shuo Li
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Wenxiu He
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Guoqiang Zhong
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Zhiyuan Jiang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Portes AMO, Paula ABR, Miranda DCD, Resende LT, Coelho BIC, Teles MC, Jardim IABA, Natali AJ, Castrucci AMDL, Isoldi MC. A systematic review of the effects of cold exposure on pathological cardiac remodeling in mice. J Therm Biol 2023; 114:103598. [PMID: 37321023 DOI: 10.1016/j.jtherbio.2023.103598] [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: 01/26/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Exposure to cold promotes cardiac remodeling, characterized by deleterious effects on structure and function, contributing to increased mortality from cardiovascular diseases. The mechanisms associated with these changes are poorly understood. This review gathers the literature data on the main alterations and mechanisms associated with the adverse cardiac structural and functional remodeling induced by cold exposure in mice. Original studies were identified by searching PubMed, Scopus, and Embase databases from January 1990 to June 2022. This systematic review was conducted in accordance with the criteria established by PRISMA and registered in PROSPERO (CRD42022350637). The risk of bias was evaluated by the SYRCLE. Eligible studies included original papers published in English that evaluated cardiac outcomes in mice submitted to short- or long-time cold exposure and had a control group at room temperature. Seventeen original articles were included in this review. Cold exposure induces pathological cardiac remodeling, characterized by detrimental structural and functional parameters, changes in metabolism and autophagy process, and increases in oxidative stress, inflammation, and apoptosis. In addition, Nppa, AT1A, Fbp3, BECN, ETA, and MT, appear to play fundamental roles in regulating cardiac remodeling. We suggest that strategies that seek to minimize the CVD risk and adverse effects of cold exposure should target these agents.
Collapse
Affiliation(s)
- Alexandre Martins Oliveira Portes
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil; Department of Physical Education, Federal University of Viçosa, Viçosa, Brazil.
| | | | - Denise Coutinho de Miranda
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil; Department of Nutrition, Governador Ozanam Coelho University Center, Uba, Brazil
| | | | | | - Maria Cecília Teles
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | | | - Antônio José Natali
- Department of Physical Education, Federal University of Viçosa, Viçosa, Brazil
| | - Ana Maria de Lauro Castrucci
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; Department of Biology, University of Virginia, Charlottesville, United States
| | - Mauro César Isoldi
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
16
|
Yu J, Zhao X, Yan X, Li W, Liu Y, Wang J, Wang J, Yang Y, Hao Y, Liang Z, Tao Y, Yuan Y, Du Z. Aloe-emodin ameliorated MI-induced cardiac remodeling in mice via inhibiting TGF-β/SMAD signaling via up-regulating SMAD7. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154793. [PMID: 37011420 DOI: 10.1016/j.phymed.2023.154793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Aloe-emodin (AE), a natural anthraquinone extract from traditional Chinese medicinal plants, has been certified to protect against acute myocardial ischemia. However, its effect on cardiac remodeling after chronic myocardial infarction (MI) and the possible mechanism remain unclear. PURPOSE This study investigated the effect of AE on cardiac remodeling and oxidative damage induced by myocardial infarction (MI) in vitro and explored the underlying mechanisms. METHODS Echocardiography and Masson staining were used to demonstrate myocardial dysfunction and fibrosis. Cell apoptosis was detected by TUNEL staining. The expressions of fibrosis-related factors such as type I collagen, α-smooth muscle actin (α-SMA) and connective tissue growth factor (CTGF) were detected by Western blot. RESULTS Our data demonstrated that AE treatment significantly improved cardiac function, reduced structural remodeling, and reduced cardiac apoptosis and oxidative stress in mice with myocardial infarction. In vitro, AE could protect neonatal mouse cardiomyocytes (NMCM) from angiotensin II (Ang II)-induced cardiomyocyte hypertrophy and apoptosis, and significantly inhibited (p < 0.05) Ang II-induced reactive oxygen species (ROS) increase. Furthermore, AE treatment significantly reversed the Ang ii-induced upregulation. CONCLUSION In summary, our work reveals for the first time that AE activates the TGF-β signaling pathway by up-regulating Smad7 expression, which in turn regulates the expression of fibrosis-related genes, ultimately improving cardiac function, inhibiting the development of cardiac fibrosis and hypertrophy in rats with chronic MI.
Collapse
Affiliation(s)
- Jie Yu
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiuye Zhao
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiuqing Yan
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wen Li
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yunqi Liu
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiapan Wang
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jia Wang
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yilian Yang
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan Hao
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhen Liang
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yiping Tao
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ye Yuan
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; National key laboratory of frigid cardiovascular disease, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Zhimin Du
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, China; National key laboratory of frigid cardiovascular disease, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
17
|
Hou Z, Lin Y, Yang X, Chen J, Li G. Therapeutics of Extracellular Vesicles in Cardiocerebrovascular and Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:187-205. [PMID: 37603281 DOI: 10.1007/978-981-99-1443-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Extracellular vesicles (EVs) are nanoscale membranous vesicles containing DNA, RNA, lipids, and proteins, which play versatile roles in intercellular communications. EVs are increasingly being recognized as the promising therapeutic agents for many diseases, including cardiocerebrovascular and metabolic diseases, due to their ability to deliver functional and therapeutical molecules. In this chapter, the biological characteristics and functions of EVs are briefly summarized. Importantly, the current state of applying EVs in the prevention and treatment of cardiocerebrovascular and metabolic diseases, including myocardial infarction, atrial fibrillation, myocardial hypertrophy, stroke, diabetes, Alzheimer's disease, fatty liver, obesity, thyroid diseases, and osteoporosis, is discussed. Lastly, the challenges and prospects related to the preclinical and clinical application of EVs receive a particular focus.
Collapse
Affiliation(s)
- Zhitao Hou
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated with Beijing University of Chinese Medicine, Beijing, China
| | - Yiyan Lin
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinyu Yang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated with Beijing University of Chinese Medicine, Beijing, China
- Fangshan Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jing Chen
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Kreimer F, Gotzmann M. Left Atrial Cardiomyopathy - A Challenging Diagnosis. Front Cardiovasc Med 2022; 9:942385. [PMID: 35845077 PMCID: PMC9280085 DOI: 10.3389/fcvm.2022.942385] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 01/08/2023] Open
Abstract
Left atrial cardiomyopathy (LACM) has been an ongoing focus of research for several years. There is evidence that LACM is responsible for atrial fibrillation and embolic strokes of undetermined sources. Therefore, the correct diagnosis of LACM is of clinical importance. Various techniques, including electrocardiography, echocardiography, cardiac magnetic resonance imaging, computed tomography, electroanatomic mapping, genetic testing, and biomarkers, can both identify and quantify structural, mechanical as well as electrical dysfunction in the atria. However, the question arises whether these techniques can reliably diagnose LACM. Because of its heterogeneity, clinical diagnosis is challenging. To date, there are no recommendations for standardized diagnosis of suspected LACM. However, standardization could help to classify LACM more precisely and derive therapeutic directions to improve individual patient management. In addition, uniform diagnostic criteria for LACM could be important for future studies. Combining several parameters and relating them seems beneficial to approach the diagnosis of LACM. This review provides an overview of the current evidence regarding the diagnosis of LACM, in which several potential parameters are discussed and, consequently, a proposal for a diagnostic algorithm is presented.
Collapse
Affiliation(s)
- Fabienne Kreimer
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
| | - Michael Gotzmann
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
19
|
Steimle JD, Grisanti Canozo FJ, Park M, Kadow ZA, Samee MAH, Martin JF. Decoding the PITX2-controlled genetic network in atrial fibrillation. JCI Insight 2022; 7:e158895. [PMID: 35471998 PMCID: PMC9221021 DOI: 10.1172/jci.insight.158895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia and a major risk factor for stroke, often arises through ectopic electrical impulses derived from the pulmonary veins (PVs). Sequence variants in enhancers controlling expression of the transcription factor PITX2, which is expressed in the cardiomyocytes (CMs) of the PV and left atrium (LA), have been implicated in AF predisposition. Single nuclei multiomic profiling of RNA and analysis of chromatin accessibility combined with spectral clustering uncovered distinct PV- and LA-enriched CM cell states. Pitx2-mutant PV and LA CMs exhibited gene expression changes consistent with cardiac dysfunction through cell type-distinct, PITX2-directed, cis-regulatory grammars controlling target gene expression. The perturbed network targets in each CM were enriched in distinct human AF predisposition genes, suggesting combinatorial risk for AF genesis. Our data further reveal that PV and LA Pitx2-mutant CMs signal to endothelial and endocardial cells through BMP10 signaling with pathogenic potential. This work provides a multiomic framework for interrogating the basis of AF predisposition in the PVs of humans.
Collapse
Affiliation(s)
| | | | | | - Zachary A. Kadow
- Program in Developmental Biology, and
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | | | - James F. Martin
- Department of Integrative Physiology
- Texas Heart Institute, Houston, Texas, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
20
|
TRPC1 Contributes to Endotoxemia-induced Myocardial Dysfunction via Mediating Myocardial Apoptosis and Autophagy. Pharmacol Res 2022; 181:106262. [PMID: 35598715 DOI: 10.1016/j.phrs.2022.106262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/16/2022]
Abstract
Cardiac dysfunction is a vital complication of endotoxemia (ETM) with limited therapeutic options. Transient receptor potential canonical channel (TRPC)1 was involved in various heart diseases. While, the role of TRPC1 in ETM-induced cardiac dysfunction remains to be defined. In this study, we found that TRPC1 protein expression was significantly upregulated in hearts of lipopolysaccharide (LPS)-challenged mice. What's more, TRPC1 knockdown significantly alleviated LPS-induced cardiac dysfunction and injury. Further myocardial mRNA-sequencing analysis revealed that TRPC1 might participate in pathogenesis of ETM-induced cardiac dysfunction via mediating myocardial apoptosis and autophagy. Data showed that knockdown of TRPC1 significantly ameliorated LPS-induced myocardial apoptotic injury, cardiomyocytes autophagosome accumulation, and myocardial autophagic flux. Simultaneously, deletion of TRPC1 reversed LPS-induced molecular changes of apoptosis/autophagy signaling pathway in cardiomyocytes. Moreover, TRPC1 could promote LPS-triggered intracellular Ca2+ release, subsequent calpain activation and caveolin-1 degradation. Either blocking calpain by PD150606 or enhancing the amount of caveolin-1 scaffolding domain that interacts with TRPC1 by cell-permeable peptide cavtratin significantly alleviated the LPS-induced cardiac dysfunction and cardiomyocytes apoptosis/autophagy. Furthermore, cavtratin could inhibit LPS-induced calpain activation in cardiomyocytes. caveolin-1 could directly interact with calpain 2 both in vivo and in vitro. Importantly, cecal ligation and puncture-stimulated cardiac dysfunction and mortality were significantly alleviated in Trpc1-/- and cavtratin-treated mice, which further validated the contribution of TRPC1-caveolin-1 signaling axis in sepsis-induced pathological process. Overall, this study indicated that TRPC1 could promote LPS-triggered intracellular Ca2+ release, mediate caveolin-1 reduction, and in turn activates calpain to regulate myocardial apoptosis and autophagy, contributing to ETM-induced cardiac dysfunction of mice.
Collapse
|
21
|
Diaz JR, Martá-Ariza M, Khodadadi-Jamayran A, Heguy A, Tsirigos A, Pankiewicz JE, Sullivan PM, Sadowski MJ. Apolipoprotein E4 Effects a Distinct Transcriptomic Profile and Dendritic Arbor Characteristics in Hippocampal Neurons Cultured in vitro. Front Aging Neurosci 2022; 14:845291. [PMID: 35572125 PMCID: PMC9099260 DOI: 10.3389/fnagi.2022.845291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The APOE gene is diversified by three alleles ε2, ε3, and ε4 encoding corresponding apolipoprotein (apo) E isoforms. Possession of the ε4 allele is signified by increased risks of age-related cognitive decline, Alzheimer's disease (AD), and the rate of AD dementia progression. ApoE is secreted by astrocytes as high-density lipoprotein-like particles and these are internalized by neurons upon binding to neuron-expressed apoE receptors. ApoE isoforms differentially engage neuronal plasticity through poorly understood mechanisms. We examined here the effects of native apoE lipoproteins produced by immortalized astrocytes homozygous for ε2, ε3, and ε4 alleles on the maturation and the transcriptomic profile of primary hippocampal neurons. Control neurons were grown in the presence of conditioned media from Apoe -/- astrocytes. ApoE2 and apoE3 significantly increase the dendritic arbor branching, the combined neurite length, and the total arbor surface of the hippocampal neurons, while apoE4 fails to produce similar effects and even significantly reduces the combined neurite length compared to the control. ApoE lipoproteins show no systemic effect on dendritic spine density, yet apoE2 and apoE3 increase the mature spines fraction, while apoE4 increases the immature spine fraction. This is associated with opposing effects of apoE2 or apoE3 and apoE4 on the expression of NR1 NMDA receptor subunit and PSD95. There are 1,062 genes differentially expressed across neurons cultured in the presence of apoE lipoproteins compared to the control. KEGG enrichment and gene ontology analyses show apoE2 and apoE3 commonly activate expression of genes involved in neurite branching, and synaptic signaling. In contrast, apoE4 cultured neurons show upregulation of genes related to the glycolipid metabolism, which are involved in dendritic spine turnover, and those which are usually silent in neurons and are related to cell cycle and DNA repair. In conclusion, our work reveals that lipoprotein particles comprised of various apoE isoforms differentially regulate various neuronal arbor characteristics through interaction with neuronal transcriptome. ApoE4 produces a functionally distinct transcriptomic profile, which is associated with attenuated neuronal development. Differential regulation of neuronal transcriptome by apoE isoforms is a newly identified biological mechanism, which has both implication in the development and aging of the CNS.
Collapse
Affiliation(s)
- Jenny R. Diaz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Mitchell Martá-Ariza
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | | | - Adriana Heguy
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Joanna E. Pankiewicz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University School of Medicine, Durham, NC, United States
- Durham VA Medical Center’s, Geriatric Research Education and Clinical Center, Durham, NC, United States
| | - Martin J. Sadowski
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
22
|
Prediction of Drug Targets for Specific Diseases Leveraging Gene Perturbation Data: A Machine Learning Approach. Pharmaceutics 2022; 14:pharmaceutics14020234. [PMID: 35213968 PMCID: PMC8878225 DOI: 10.3390/pharmaceutics14020234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/08/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Identification of the correct targets is a key element for successful drug development. However, there are limited approaches for predicting drug targets for specific diseases using omics data, and few have leveraged expression profiles from gene perturbations. We present a novel computational approach for drug target discovery based on machine learning (ML) models. ML models are first trained on drug-induced expression profiles with outcomes defined as whether the drug treats the studied disease. The goal is to “learn” the expression patterns associated with treatment. Then, the fitted ML models were applied to expression profiles from gene perturbations (overexpression (OE)/knockdown (KD)). We prioritized targets based on predicted probabilities from the ML model, which reflects treatment potential. The methodology was applied to predict targets for hypertension, diabetes mellitus (DM), rheumatoid arthritis (RA), and schizophrenia (SCZ). We validated our approach by evaluating whether the identified targets may ‘re-discover’ known drug targets from an external database (OpenTargets). Indeed, we found evidence of significant enrichment across all diseases under study. A further literature search revealed that many candidates were supported by previous studies. For example, we predicted PSMB8 inhibition to be associated with the treatment of RA, which was supported by a study showing that PSMB8 inhibitors (PR-957) ameliorated experimental RA in mice. In conclusion, we propose a new ML approach to integrate the expression profiles from drugs and gene perturbations and validated the framework. Our approach is flexible and may provide an independent source of information when prioritizing drug targets.
Collapse
|
23
|
Prominent atrial fibrosis and its correlation with dilated atrium atrial tachyarrhythmia and long after classic Fontan surgery. J Cardiol 2021; 79:671-677. [DOI: 10.1016/j.jjcc.2021.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/20/2022]
|
24
|
Oh S, Shin S, Janknecht R. Sumoylation of transcription factor ETV1 modulates its oncogenic potential in prostate cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:795-810. [PMID: 34367411 PMCID: PMC8339722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
The transcription factor ETS variant 1 (ETV1) is capable of promoting prostate tumorigenesis. We demonstrate that ETV1 can be posttranslationally modified by covalent attachment of small ubiquitin-like modifier 1 (SUMO1) onto four different lysine residues. In human embryonic kidney 293T cells, mutation of these sumoylation sites stimulated the transactivation potential of ETV1 at the matrix metalloproteinase 1 (MMP1), but not Yes-associated protein 1 gene promoter, while ETV1 protein stability and intracellular localization remained unchanged. In stark contrast, sumoylation-deficient ETV1 was repressed in its ability to stimulate the MMP1 promoter and to cooperate with a histone demethylase, JmjC domain-containing 2A (JMJD2A), in LNCaP prostate cancer cells. Mutation of sumoylation sites enhanced the ability of ETV1 to interact with the histone deacetylase (HDAC) 1, but had basically no impact on complex formation with HDAC3 or JMJD2A. Further, compared to non-sumoylated ETV1, its sumoylated forms were less able to bind to the transcription factor, SMAD family member 4. Lastly, in contrast to wild-type ETV1, sumoylation-deficient ETV1 repressed LNCaP cell growth. Altogether, these data suggest that sumoylation modulates ETV1 function in a cell type-specific manner, possibly by altering the spectrum of transcriptional cofactors being recruited. Notably, SUMO pathway components SUMO1, ubiquitin-like modifier activating enzyme 2 and ubiquitin conjugating enzyme 9 were upregulated in prostate tumors, implying that enhanced sumoylation indeed promotes ETV1's oncogenic activity during prostate cancer formation.
Collapse
Affiliation(s)
- Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| | - Sook Shin
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma City, Oklahoma, USA
| |
Collapse
|