1
|
Sun S, Lai C, Huang C, Wang R, Fu G, Shang M. Harnessing integrated bioinformatics to identify new diagnostic and therapeutic strategies for heart failure. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167909. [PMID: 40398828 DOI: 10.1016/j.bbadis.2025.167909] [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: 11/09/2024] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
Heart failure (HF) is a life-threatening condition that poses a significant challenge on public health, particularly among the elder populations. To develop new diagnostic and therapeutic strategies for HF, we analyzed large-scale transcriptome sequencing data from HF patients as an exploratory approach. We identified 18 HF-related genes and developed a robust scoring model for HF diagnosis, by applying two machine learning algorithms for data analysis. Meanwhile, we evaluated and compared the predictive abilities of three bioinformatics methods in identifying potential HF treatment drugs. Significantly, an unconventional network-based proximity analysis, integrating multidimensional drug target information, outperformed other methods in the assessment of predictive ability. To validate these findings, we tested several candidate drugs in a mouse model transitioning from acute myocardial infarction (MI) to chronic HF. Among the candidates, mirtazapine exhibited cardioprotective effects in both early (1-week) post-MI and chronic HF (4-week post-MI) settings, while cabergoline showed potential efficacy primarily in the early post-MI phase. Additionally, the screened triamterene, used as a positive control, exhibited protective effects in both early post-MI and chronic HF stages. Mechanistic studies revealed that growth factor receptor-bound protein 14 and Ras-related protein Rab-3A were critical to the observed cardioprotection. These findings provide valuable evidence and insights for exploring potential therapeutic agents for HF treatment.
Collapse
Affiliation(s)
- Shuo Sun
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Jining Medical University, Jining 272067, China; School of Life Sciences, Jining Medical University, Rizhao 276826, China
| | - Chaojie Lai
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Chengchen Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Ruilin Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Min Shang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
2
|
Khattab E, Myrianthefs MM, Sakellaropoulos S, Alexandrou K, Mitsis A. Precision medicine applications in dilated cardiomyopathy: Advancing personalized care. Curr Probl Cardiol 2025; 50:103076. [PMID: 40381754 DOI: 10.1016/j.cpcardiol.2025.103076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Dilated cardiomyopathy (DCM) is a prevalent cardiac disorder affecting 1 in 250-500 individuals, characterized by ventricular dilation and impaired systolic function, leading to heart failure and increased mortality, including sudden cardiac death. DCM arises from genetic and environmental factors, such as drug-induced, inflammatory, and viral causes, resulting in diverse yet overlapping phenotypes. Advances in precision medicine are revolutionizing DCM management by leveraging genetic and molecular profiling for tailored diagnostic and therapeutic approaches. This review highlights comprehensive diagnostic evaluations, genetic discoveries, and multi-omics approaches integrating genomic, transcriptomic, proteomic, and metabolomic data to enhance understanding of DCM pathophysiology. Innovative risk stratification methods, including machine learning, are improving predictions of disease progression. Despite these advancements, the current one-size-fits-all management strategy contributes to persistently high morbidity and mortality. Emerging targeted therapies, such as CRISPR/Cas9 genome editing, aetiology-specific interventions, and pharmacogenomics, are reshaping treatment paradigms. Precision medicine holds promise for optimizing DCM diagnosis, treatment, and outcomes, aiming to reduce the burden of this debilitating condition.
Collapse
Affiliation(s)
- Elina Khattab
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Michael M Myrianthefs
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Stefanos Sakellaropoulos
- Department of Internal Medicine, Cardiology Clinic, Kantonsspital Baden, Baden 5404, Switzerland
| | - Kyriakos Alexandrou
- Department of Nursing, School of Health Sciences, Cyprus University of Technology; Archiepiskopou Kyprianou 30, Limassol 3036, Cyprus
| | - Andreas Mitsis
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus.
| |
Collapse
|
3
|
Jiang J, He W, Huang H. Lithium in cardiovascular health and disease: Pharmacological and clinical implications. Eur J Pharmacol 2025; 999:177657. [PMID: 40324573 DOI: 10.1016/j.ejphar.2025.177657] [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: 11/22/2024] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Lithium, a cornerstone therapy for bipolar disorder (BD), produces complex and dual effects on cardiovascular health. This review synthesizes current evidence on the direct and indirect cardiovascular effects of lithium. Direct effects include dose-dependent influences on cardiac development, hypertrophy, fibrosis, mitochondrial function, and arrhythmia. While therapeutic doses may confer cardioprotective benefits by inhibiting GSK-3β, which activates the Wnt/β-catenin signaling pathway and modulates anti-inflammatory pathways, supratherapeutic levels exacerbate oxidative stress, mitochondrial dysfunction, and arrhythmogenicity. Indirectly, lithium modulates cardiovascular outcomes caused by renal dysfunction, metabolic syndrome, and neuroendocrine-immune interactions. Clinical studies suggested that there is reduced cardiovascular mortality in lithium-treated BD patients, potentially offsetting its adverse metabolic effects. However, conflicting evidence persists, particularly regarding dose specificity and long-term outcomes. Future research should focus on high-quality clinical trials to clarify the balance between therapeutic benefits and adverse effects of lithium to ensure its safe and effective use in clinical practice.
Collapse
Affiliation(s)
- Jingya Jiang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Wanbing He
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hui Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China.
| |
Collapse
|
4
|
Zhu B, Cheng H, Li J, Hu Y, Ge X. Decoding mitochondrial stress genes in DCM: towards precision diagnosis and therapy. Hereditas 2025; 162:57. [PMID: 40217309 PMCID: PMC11987231 DOI: 10.1186/s41065-025-00399-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Mitochondrial oxidative stress (ROS) is a crucial factor in the pathogenesis of dilated cardiomyopathy (DCM). Despite its significance, robust biomarkers for assessing its role remain scarce. This study investigates ROS mechanisms in DCM and identifies associated biomarkers, offering fresh insights into diagnosis and treatment. METHODS We sourced transcriptomic data from the GEO database and mitochondrial oxidative stress-related genes from GeneCards. Using consensus clustering, we identified 64 genes associated with mitochondrial oxidative stress in DCM and further isolated five hub genes through protein-protein interaction and machine learning techniques. These genes were analyzed for functions related to immunity, drug sensitivity, and single-cell localization. Concurrently, we collected blood samples from DCM patients to validate the hub genes' expression. RESULTS The study identified five hub genes related to mitochondrial oxidative stress: VCL, ABCB1, JAK2, KDR, and NGF. Expression analysis revealed high levels of VCL, ABCB1, KDR, and NGF in the non-failing (NF) group, while JAK2 was elevated in the DCM group (p < 0.05). Diagnostic efficacy, measured by area under the curve (AUC), was significant for VCL (76.4), ABCB1 (80.1), JAK2 (68.2), KDR (78.1), and NGF (71.8). Moreover, several drugs were identified as potential regulators of these hub genes, including Topotecan, CDK9_5576, Acetalax, Afatinib, and GSK591. Notably, VCL showed increased expression in DCM patient blood samples, consistent with transcriptomic and single-cell findings. CONCLUSION This research highlights key genes associated with mitochondrial oxidative stress-VCL, ABCB1, JAK2, KDR, NGF-that show differential expression in DCM and myocardial infarction. These findings underscore their diagnostic potential and pave the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Bingbing Zhu
- Department of Cardiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu Province, 215028, China
| | - Hai Cheng
- Department of Cardiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu Province, 215028, China
| | - Jiawei Li
- Department of Cardiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu Province, 215028, China
| | - Yangyang Hu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Xiaoning Ge
- Department of Cardiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu Province, 215028, China.
| |
Collapse
|
5
|
Fan Q, Wang Y, An Q, Ling Y. Right ventricular dysfunction following surgical repair of tetralogy of Fallot: Molecular pathways and therapeutic prospects. Biomed Pharmacother 2025; 184:117924. [PMID: 39983432 DOI: 10.1016/j.biopha.2025.117924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease (CHD). Although surgical correction of TOF is possible, patients often face challenges related to right ventricle dysfunction (RVD) post-surgery, which can significantly impact their long-term survival. The causes of RVD in TOF patients are complex, involving both the unique structural characteristics of the TOF heart and damage resulting from surgical interventions. Residual anatomical issues following TOF repair are often unavoidable, placing the RV under stress and leading to the activation of multiple molecular pathways. This review comprehensively outlines the causes of RVD in patients after TOF surgery, particularly focusing the molecular pathways that contribute to RVD, including established signaling pathways as well as emerging pathways identified through transcriptomic analysis of RV myocardium in TOF patients. We also highlight the features of these molecular pathways concerning RVD, as well as the influence of gender disparities on these molecular pathways. By interpreting the causes and molecular mechanisms underlying RVD after TOF surgery, this review provides new insights for managing RVD in repaired TOF, potentially paving the way for targeted therapies aimed at improving long-term outcomes for those affected by RVD.
Collapse
Affiliation(s)
- Qiang Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yabo Wang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yunfei Ling
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
6
|
Liao Z, Tang X, Yang B, Yang J. Dopamine receptors and organ fibrosis. Biochem Biophys Rep 2025; 41:101910. [PMID: 39867679 PMCID: PMC11761258 DOI: 10.1016/j.bbrep.2024.101910] [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: 08/12/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/28/2025] Open
Abstract
Organ fibrosis, considered as a major global health concern, is a pathological condition often occurring after tissue injury in various organs. The pathogenesis of fibrosis involves multiple phases and multiple cell types. Dopamine is involved in various life activities by activating five receptors (D1, D2, D3, D4, D5). Activation or loss of function of dopamine receptors has been reported to be associated with the fibrosis of several organs, such as ocular, lung, liver, heart, and kidney. In this paper, we review dopamine receptors' potential roles in organ fibrosis and mechanisms by which organ fibrosis develops or decreases when dopamine receptors function is activated or perturbed.
Collapse
Affiliation(s)
- ZhongLi Liao
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 400030, China
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - XueFeng Tang
- Department of Pathology, Chongqing General Hospital, Chongqing University, Chongqing, 400030, China
| | - Bin Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, 361000, China
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 400030, China
| |
Collapse
|
7
|
Gergs U, Pham TH, Rayo Abella LM, Hesse C, Grundig P, Dhein S, Hofmann B, Neumann J. Contractile effects of stimulation of D 1-dopamine receptors in the isolated human atrium. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1497-1508. [PMID: 39102031 PMCID: PMC11825631 DOI: 10.1007/s00210-024-03340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
Dopamine receptors have been claimed not to directly increase contractility in the human heart. Therefore, we performed contraction experiments in isolated electrically driven human atrial preparations (HAP). For comparison, we performed contraction experiments with left atrial preparations of transgenic mice which harbor a cardiac overexpression of human D1-dopamine receptors (D1-TG). In D1-TG, first we noted that dopamine (10 nM-10 µM cumulatively applied) in the presence of propranolol exerted a concentration- and time-dependent positive inotropic effect in D1-TG. In a similar fashion, dopamine increased force of contraction in the presence of 0.4 µM propranolol in HAP and these effects were amplified by pre-treatment with inhibitor of phosphodiesterase III (1 µM) cilostamide. Moreover, contractile effects of dopamine in the presence of propranolol 0.4 µM in HAP were antagonized by odapipam, haloperidol, or raclopride. Ten micromolars of fenoldopam in the presence of cilostamide increased force of contraction in HAP and this effect was antagonized by SCH 23390. We conclude that stimulation of human D1-dopamine receptors can increase force of contraction in the HAP.
Collapse
Affiliation(s)
- U Gergs
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - T H Pham
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - L M Rayo Abella
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - C Hesse
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - P Grundig
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - S Dhein
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University Leipzig, Leipzig, Germany
| | - B Hofmann
- Department of Cardiac Surgery, Mid-German Heart Center, University Hospital Halle, Halle (Saale), Germany
| | - J Neumann
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany.
| |
Collapse
|
8
|
Zhao M, Hou Y, Yuan M, Ma S, Yue Y. Clinical efficacy and hemodynamic effects of levosimendan in cardiac surgery patients after surgery. J Cardiothorac Surg 2025; 20:43. [PMID: 39773754 PMCID: PMC11705681 DOI: 10.1186/s13019-024-03316-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To investigate the therapeutic effect of levosimendan on hemodynamics in patients undergoing major cardiac surgery and presenting with acute postoperative heart failure. METHODS The subjects of the study were 160 patients with severe cardiac conditions who underwent surgery and had acute heart failure. Eighty cases each were assigned to the research and control groups using a random number table. Document the general patient data for each of the two groups; compare the clinical outcomes of the two groups. The hemodynamic states of the two groups were compared both before and after therapy. 48 h after surgery, echocardiography was performed in both groups to determine cardiac function. 48 h after surgery, N-terminal pro-brain B-type natriuretic peptide (NT-Pro-BNP) levels were compared between the two groups. RESULTS The overall effective rate was significantly higher in the research group (92.5%) compared to the control group (76.25%, P < 0.05). Post-treatment, the research group demonstrated a significant reduction in CVP (9.25 ± 2.11 cmH2O vs. 11.36 ± 3.08 cmH2O, P < 0.001), heart rate (100.30 ± 8.69 bpm vs. 105.74 ± 7.69 bpm, P < 0.001), and lactic acid levels (1.68 ± 0.59 mmol/L vs. 2.69 ± 0.55 mmol/L, P < 0.001). The research group also showed improvements in SBP (117.23 ± 8.74 mmHg vs. 113.25 ± 7.55 mmHg, P = 0.002) and urine output (4.21 ± 1.76 mL/kg/h vs. 3.65 ± 1.23 mL/kg/h, P = 0.021). Cardiac function indicators 48 h after surgery indicated a higher LVEF (55.21 ± 8.04% vs. 47.18 ± 6.60%, P < 0.001) and lower LVEDVi and LVESVi in the research group (P < 0.001 for both). NT-Pro-BNP levels were significantly lower in the research group (6010.19 ± 1208.52 pg/mL vs. 9663.21 ± 2391.34 pg/mL, P < 0.001). The incidence of complications was lower in the research group (5% vs. 22.5%, P = 0.001). CONCLUSION Cardiac surgery patients are prone to complications with acute heart failure after surgery. Treatment with levosimendan can significantly improve clinical efficacy and reduce complications. It can also effectively improve patients' cardiac function and promote hemodynamic stability.
Collapse
Affiliation(s)
- Meiling Zhao
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Yunfeng Hou
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, JiNan, 250014, China
| | - Meng Yuan
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Shuang Ma
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Yifeng Yue
- Department of Anesthesiology, Zibo Central Hospital, No.10 Shanghai Road, Zhangdian District, Zibo City, 255000, Shandong Province, China.
| |
Collapse
|
9
|
Han S, Xu Q, Du Y, Tang C, Cui H, Xia X, Zheng R, Sun Y, Shang H. Single-cell spatial transcriptomics in cardiovascular development, disease, and medicine. Genes Dis 2024; 11:101163. [PMID: 39224111 PMCID: PMC11367031 DOI: 10.1016/j.gendis.2023.101163] [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: 03/27/2023] [Revised: 10/17/2023] [Accepted: 10/29/2023] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) impose a significant burden worldwide. Despite the elucidation of the etiology and underlying molecular mechanisms of CVDs by numerous studies and recent discovery of effective drugs, their morbidity, disability, and mortality are still high. Therefore, precise risk stratification and effective targeted therapies for CVDs are warranted. Recent improvements in single-cell RNA sequencing and spatial transcriptomics have improved our understanding of the mechanisms and cells involved in cardiovascular phylogeny and CVDs. Single-cell RNA sequencing can facilitate the study of the human heart at remarkably high resolution and cellular and molecular heterogeneity. However, this technique does not provide spatial information, which is essential for understanding homeostasis and disease. Spatial transcriptomics can elucidate intracellular interactions, transcription factor distribution, cell spatial localization, and molecular profiles of mRNA and identify cell populations causing the disease and their underlying mechanisms, including cell crosstalk. Herein, we introduce the main methods of RNA-seq and spatial transcriptomics analysis and highlight the latest advances in cardiovascular research. We conclude that single-cell RNA sequencing interprets disease progression in multiple dimensions, levels, perspectives, and dynamics by combining spatial and temporal characterization of the clinical phenome with multidisciplinary techniques such as spatial transcriptomics. This aligns with the dynamic evolution of CVDs (e.g., "angina-myocardial infarction-heart failure" in coronary artery disease). The study of pathways for disease onset and mechanisms (e.g., age, sex, comorbidities) in different patient subgroups should improve disease diagnosis and risk stratification. This can facilitate precise individualized treatment of CVDs.
Collapse
Affiliation(s)
- Songjie Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qianqian Xu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yawen Du
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chuwei Tang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Herong Cui
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaofeng Xia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
10
|
Lyu X, Fang J, Liu D, Wu Q, Li Y, Qin C, Zheng J, Hu N. Near-infrared-triggered plasmonic regulation and cardiomyocyte-based biosensing system for in vitro bradyarrhythmia treatment. Biosens Bioelectron 2024; 262:116554. [PMID: 38971038 DOI: 10.1016/j.bios.2024.116554] [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: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Bradyarrhythmia, a life-threatening cardiovascular disease, is an increasing burden for the healthcare system. Currently, surgery, implanted device, and drug are introduced to treat the bradyarrhythmia in clinical practice. However, these conventional therapeutic strategies suffer from the invasive surgery, power supply, or drug side effect, respectively, hence developing the alternative therapeutic strategy is necessarily imperative. Here, a convenient and effective strategy to treat the bradyarrhythmia is proposed using near-infrared-triggered Au nanorod (NR) based plasmonic photothermal effect (PPE). Moreover, electrophysiology of cardiomyocytes is dynamically monitored by the integrated biosensing-regulating system during and after the treatment. Cardiomyocyte-based bradyarrhythmia recover rhythmic for a long time by regulating plasmonic photothermal effect. Furthermore, the regulatory mechanism is qualitatively investigated to verify the significant thermal stimulation in the recovery process. This study establishes a reliable platform for long-term recording and evaluation of mild photothermal therapy for bradyarrhythmia in vitro, offering an efficient and non-invasive strategy for the potential clinical applications.
Collapse
Affiliation(s)
- Xuelian Lyu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Jiaru Fang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Dong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Qianni Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chunlian Qin
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Jilin Zheng
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China; General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China.
| |
Collapse
|
11
|
Xu D, Lyu X, Han H, Fang J, Xue J, Zheng J, Hu N, Gao Z. Scalable Drug-Mimicking Nanoplasmonic Therapy for Bradyarrhythmia in Cardiomyocytes. NANO LETTERS 2024; 24:11302-11310. [PMID: 39213538 DOI: 10.1021/acs.nanolett.4c03213] [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: 09/04/2024]
Abstract
Bradyarrhythmia poses a serious threat to human health, with chronic progression causing heart failure and acute onset leading to sudden death. In this study, we develop a scalable drug-mimicking nanoplasmonic therapeutic strategy by introducing gold nanorod (Au NR) mediated near-infrared (NIR) photothermal effects. An integrated sensing and regulation platform is established for in situ synchronized NIR laser regulation and electrophysiological property recording. The Au NR plasmonic regulation enables the restoration of normal cardiomyocyte rhythm from the bradyarrhythmia. By regulating the aspect ratio and concentration of Au NRs, as well as the intensity and time of NIR irradiation, we precisely optimized the plasmonic photothermal effect to explore effective therapeutic strategies. Furthermore, mRNA sequencing revealed a significant increase in the number of differentially expressed genes (DEGs) involved in the electrophysiological activities of cardiomyocytes following photothermal therapy. Au NR-mediated plasmonic photothermal therapy, as an efficient and noninvasive approach to bradyarrhythmia, holds profound implications for cardiology research.
Collapse
Affiliation(s)
- Dongxin Xu
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Xuelian Lyu
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
| | - Haote Han
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
| | - Jiaru Fang
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Jilin Zheng
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
| | - Ning Hu
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
| | - Zhigang Gao
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| |
Collapse
|
12
|
Shen J, Valentim W, Friligkou E, Overstreet C, Choi K, Koller D, O’Donnell CJ, Stein MB, Gelernter J, Posttraumatic Stress Disorder Working Group of the Psychiatric Genomics Consortium, Lv H, Sun L, Falcone GJ, Polimanti R, Pathak GA. Genetics of posttraumatic stress disorder and cardiovascular conditions using Life's Essential 8, Electronic Health Records, and Heart Imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.20.24312181. [PMID: 39228734 PMCID: PMC11370495 DOI: 10.1101/2024.08.20.24312181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Patients with post-traumatic stress disorder (PTSD) experience higher risk of adverse cardiovascular (CV) outcomes. This study explores shared loci, and genes between PTSD and CV conditions from three major domains: CV diagnoses from electronic health records (CV-EHR), cardiac and aortic imaging, and CV health behaviors defined in Life's Essential 8 (LE8). METHODS We used genome-wide association study (GWAS) of PTSD (N=1,222,882), 246 CV diagnoses based on EHR data from Million Veteran Program (MVP; N=458,061), UK Biobank (UKBB; N=420,531), 82 cardiac and aortic imaging traits (N=26,893), and GWAS of traits defined in the LE8 (N = 282,271 ~ 1,320,016). Shared loci between PTSD and CV conditions were identified using local genetic correlations (rg), and colocalization (shared causal variants). Overlapping genes between PTSD and CV conditions were identified from genetically regulated proteome expression in brain and blood tissues, and subsequently tested to identify functional pathways and gene-drug targets. Epidemiological replication of EHR-CV diagnoses was performed in AllofUS cohort (AoU; N=249,906). RESULTS Among the 76 PTSD-susceptibility risk loci, 33 loci exhibited local rg with 45 CV-EHR traits (|rg|≥0.4), four loci with eight heart imaging traits(|rg|≥0.5), and 44 loci with LE8 factors (|rg|≥0.36) in MVP. Among significantly correlated loci, we found shared causal variants (colocalization probability > 80%) between PTSD and 17 CV-EHR (in MVP) at 11 loci in MVP, that also replicated in UKBB and/or other cohorts. Of the 17 traits, the observational analysis in the AoU showed PTSD was associated with 13 CV-EHR traits after accounting for socioeconomic factors and depression diagnosis. PTSD colocalized with eight heart imaging traits on 2 loci and with LE8 factors on 31 loci. Leveraging blood and brain proteome expression, we found 33 and 122 genes, respectively, shared between PTSD and CVD. Blood proteome genes were related to neuronal and immune processes, while the brain proteome genes converged on metabolic and calcium-modulating pathways (FDR p <0.05). Drug repurposing analysis highlighted DRD2, NOS1, GFAP, and POR as common targets of psychiatric and CV drugs. CONCLUSION PTSD-CV comorbidities exhibit shared risk loci, and genes involved in tissue-specific regulatory mechanisms.
Collapse
Affiliation(s)
- Jie Shen
- Department of Cardiology, Children’s Hospital of Soochow University, Suzhou, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Wander Valentim
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, State of Minas Gerais, Brazil
| | - Eleni Friligkou
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Cassie Overstreet
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Karmel Choi
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Dora Koller
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, University of Barcelona, Catalonia, Spain
| | - Christopher J. O’Donnell
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, La Jolla, California; Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, California; Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Murray B. Stein
- Cardiology Section, Department of Medicine, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | | | - Haitao Lv
- Department of Cardiology, Children’s Hospital of Soochow University, Suzhou, China
| | - Ling Sun
- Department of Cardiology, Children’s Hospital of Soochow University, Suzhou, China
| | - Guido J. Falcone
- Center for Brain and Mind Health Yale University New Haven CT USA; Department of Neurology Yale University New Haven CT USA
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Gita A. Pathak
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
13
|
Huang Y, Xuan L, Xu Q, Wang J, Liu J. Differences in T cell-associated serum markers between ischemic cardiomyopathy and dilated cardiomyopathy. J Thorac Dis 2024; 16:4655-4665. [PMID: 39144301 PMCID: PMC11320225 DOI: 10.21037/jtd-24-901] [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: 05/31/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
Background Ischemic cardiomyopathy (ICM) and dilated cardiomyopathy (DCM) have similar clinical manifestations but differ in pathogenesis. We aimed to identify T cell-associated serum markers that can be used to distinguish between ICM and DCM. Methods We identified differentially expressed genes (DEGs) with transcriptome sequencing data in GSE116250, and then conducted enrichment analysis of DEGs in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Protein-protein interaction (PPI) networks were used to analyze the relationship between T cells-related genes and identify hub genes. Enzyme-linked immunosorbent assay (ELISA) kits were used to detect T cell-associated proteins in serum, and receiver operating characteristic (ROC) curves were used to evaluate the diagnostic efficacy of these serum markers. Results Using the limma package and Venn plots, we found that the non-failing donors (NFD) and DCM groups shared many of the same DEGs and DEGs-enriched functions compared to the ICM group, which were involved in T cell activation and differentiation, among other functions. Subsequently, the immune cell score showed no difference between NFD and DCM, but they were significantly different from ICM patients in CD8 T cells CD4 T cells memory resting and activated, T cells follicular helper, and M1 macrophage. After analyzing T cell-associated DEGs, it was found that 4 DEGs encoding secreted proteins were highly expressed in the ICM group compared with the NFD and DCM groups, namely chemokine (C-C motif) ligand 21 (CCL21), interleukin (IL)-1β, lymphocyte-activation gene 3 (LAG3), and vascular cell adhesion molecule-1 (VCAM-1). Importantly, the serum levels of CCL21, IL-1β, LAG3, and VCAM-1 in ICM patients were all significantly higher than those in DCM patients. The ROC curves showed that the area under the curve (AUC) values of serum CCL21, IL-1β, LAG3, and VCAM-1 were 0.775, 0.868, 0.934, and 0.903, respectively. Conclusions We have identified four T cell-associated serum markers, CCL21, IL-1β, LAG3, and VCAM-1, as potential diagnostic serum markers that differentiate ICM from DCM.
Collapse
Affiliation(s)
- Yuli Huang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lin Xuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Qiong Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jie Liu
- Department of Internal Medicine, Suixi County Hospital of Traditional Chinese Medicine, Huaibei, China
| |
Collapse
|
14
|
Yang Z, Li Y, Huang M, Li X, Fan X, Yan C, Meng Z, Liao B, Hamdani N, Yang X, Zhou X, El-Battrawy I, Akin I. Roles and Mechanisms of Dopamine Receptor Signaling in Catecholamine Excess Induced Endothelial Dysfunctions. Int J Med Sci 2024; 21:1964-1975. [PMID: 39113882 PMCID: PMC11302566 DOI: 10.7150/ijms.96550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial dysfunction may contribute to pathogenesis of Takotsubo cardiomyopathy, but mechanism underlying endothelial dysfunction in the setting of catecholamine excess has not been clarified. The study reports that D1/D5 dopamine receptor signaling and small conductance calcium-activated potassium channels contribute to high concentration catecholamine induced endothelial cell dysfunction. For mimicking catecholamine excess, 100 μM epinephrine (Epi) was used to treat human cardiac microvascular endothelial cells. Patch clamp, FACS, ELISA, PCR, western blot and immunostaining analyses were performed in the study. Epi enhanced small conductance calcium-activated potassium channel current (ISK1-3) without influencing the channel expression and the effect was attenuated by D1/D5 receptor blocker. D1/D5 agonists mimicked the Epi effect, suggesting involvement of D1/D5 receptors in Epi effects. The enhancement of ISK1-3 caused by D1/D5 activation involved roles of PKA, ROS and NADPH oxidases. Activation of D1/D5 and SK1-3 channels caused a hyperpolarization, reduced NO production and increased ROS production. The NO reduction was membrane potential independent, while ROS production was increased by the hyperpolarization. ROS (H2O2) suppressed NO production. The study demonstrates that high concentration catecholamine can activate D1/D5 and SK1-3 channels through NADPH-ROS and PKA signaling and reduce NO production, which may facilitate vasoconstriction in the setting of catecholamine excess.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Ophthalmology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Yingrui Li
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Mengying Huang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Xin Li
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Xuehui Fan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Sichuan, China
| | - Chen Yan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Zenghui Meng
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, 646000 Sichuan, China
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Xiaoli Yang
- Department of Ophthalmology, Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Xiaobo Zhou
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) partner site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Germany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Sichuan, China
| | - Ibrahim El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Bergmannsheil Bochum, Medical Clinic II, Department of Cardiology and Angiology, Ruhr University, Bochum, Germany
| | - Ibrahim Akin
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, 68167 Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) partner site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Germany
| |
Collapse
|
15
|
Abella LMR, Jacob H, Hesse C, Hofmann B, Schneider S, Schindler L, Keller M, Buchwalow IB, Jin C, Panula P, Dhein S, Klimas J, Hadova K, Gergs U, Neumann J. Initial characterization of a transgenic mouse with overexpression of the human D 1-dopamine receptor in the heart. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4939-4959. [PMID: 38177456 PMCID: PMC11166794 DOI: 10.1007/s00210-023-02901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/10/2023] [Indexed: 01/06/2024]
Abstract
Dopamine can exert effects in the mammalian heart via five different dopamine receptors. There is controversy whether dopamine receptors increase contractility in the human heart. Therefore, we have generated mice that overexpress the human D1-dopamine receptor in the heart (D1-TG) and hypothesized that dopamine increases force of contraction and beating rate compared to wild-type mice (WT). In D1-TG hearts, we ascertained the presence of D1-dopamine receptors by autoradiography using [3H]SKF 38393. The mRNA for human D1-dopamine receptors was present in D1-TG hearts and absent in WT. We detected by in-situ-hybridization mRNA for D1-dopamine receptors in atrial and ventricular D1-TG cardiomyocytes compared to WT but also in human atrial preparations. We noted that in the presence of 10 µM propranolol (to antagonize β-adrenoceptors), dopamine alone and the D1- and D5-dopamine receptor agonist SKF 38393 (0.1-10 µM cumulatively applied) exerted concentration- and time-dependent positive inotropic effects and positive chronotropic effects in left or right atrial preparations from D1-TG. The positive inotropic effects of SKF 38393 in left atrial preparations from D1-TG led to an increased rate of relaxation and accompanied by and probably caused by an augmented phosphorylation state of the inhibitory subunit of troponin. In the presence of 0.4 µM propranolol, 1 µM dopamine could increase left ventricular force of contraction in isolated perfused hearts from D1-TG. In this model, we have demonstrated a positive inotropic and chronotropic effect of dopamine. Thus, in principle, the human D1-dopamine receptor can couple to contractility in the mammalian heart.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- Dopamine/metabolism
- Dopamine/pharmacology
- Dopamine Agonists/pharmacology
- Heart/drug effects
- Heart/physiology
- Heart Atria/metabolism
- Heart Atria/drug effects
- Heart Rate/drug effects
- Mice, Inbred C57BL
- Mice, Transgenic
- Myocardial Contraction/drug effects
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D1/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
Collapse
Affiliation(s)
- Lina Maria Rayo Abella
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany
| | - Hannes Jacob
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany
| | - Christin Hesse
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany
| | - Britt Hofmann
- Department of Cardiac Surgery, Mid-German Heart Center, University Hospital Halle, D-06097, Halle (Saale), Germany
| | - Sarah Schneider
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany
| | - Lisa Schindler
- Institute of Pharmacy, University of Regensburg, D-93053, Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, University of Regensburg, D-93053, Regensburg, Germany
| | - Igor B Buchwalow
- Institute for Hematopathology, D-22547, Hamburg, Germany
- Scientific and Educational Resource Center for Molecular Morphology, Peoples' Friendship University of Russia, RU-117198, Moscow, Russia
| | - CongYu Jin
- Department of Anatomy, University of Helsinki, FI-00290, Helsinki, Finland
| | - Pertti Panula
- Department of Anatomy, University of Helsinki, FI-00290, Helsinki, Finland
| | - Stefan Dhein
- Rudolf-Boehm Institute for Pharmacology and Toxicology, University Leipzig, D-04107, Leipzig, Germany
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, SK-83232, Bratislava, Slovak Republic
| | - Katarína Hadova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, SK-83232, Bratislava, Slovak Republic
| | - Ulrich Gergs
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany
| | - Joachim Neumann
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, D-06097, Halle (Saale), Germany.
| |
Collapse
|
16
|
Abdelmissih S, Abdelgwad M, Ali DME, Negm MSI, Eshra MA, Youssef A. High-dose Agomelatine Combined with Haloperidol Decanoate Improves Cognition, Downregulates MT2, Upregulates D5, and Maintains Krüppel-like Factor 9 But Alters Cardiac Electrophysiology. J Pharmacol Exp Ther 2024; 390:125-145. [PMID: 38816228 DOI: 10.1124/jpet.123.002087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Haloperidol decanoate (HD) has been implicated in cognitive impairment. Agomelatine (AGO) has been claimed to improve cognition. We aimed at investigating the effects of HD + low- or high-dose AGO on cognition, verifying the melatonergic/dopaminergic to the cholinergic hypothesis of cognition and exploring relevant cardiovascular issues in adult male Wistar albino rats. HD + high-dose AGO prolonged the step-through latency by +61.47% (P < 0.0001), increased the time spent in bright light by +439.49% (P < 0.0001), reduced the time spent in dim light by -66.25% (P < 0.0001), and increased the percent of alternations by +71.25% (P < 0.0001), despite the reductions in brain acetylcholine level by -10.67% (P < 0.0001). Neurodegeneration was minimal, while the mean power frequency of the source wave was reduced by -23.39% (P < 0.05). Concurrently, the relative expression of brain melatonin type 2 receptors was reduced by -18.75% (P < 0.05), against increased expressions of dopamine type 5 receptors by +22.22% (P < 0.0001) and angiopoietin-like 4 by +119.18% (P < 0.0001). Meanwhile, electrocardiogram (ECG) demonstrated inverted P wave, reduced P wave duration by -36.15% (P < 0.0001) and PR interval by -19.91% (P < 0.0001), prolonged RR interval by +27.97% (P < 0.05), increased R wave amplitude by +523.15% (P < 0.0001), and a depressed ST segment and inverted T wave. In rats administered AGO, HD, or HD+ low-dose AGO, Alzheimer's disease (AD)-like neuropathologic features were more evident, accompanied by extensive ECG and neurochemical alterations. HD + high-dose AGO enhances cognition but alters cardiac electrophysiology. SIGNIFICANCE STATEMENT: Given the issue of cognitive impairment associated with HD and the claimed cognitive-enhancing activity of AGO, combined high-dose AGO with HD improved cognition of adult male rats, who exhibited minimal neurodegenerative changes. HD+ high-dose AGO was relatively safe regarding triggering epileptogenesis, while it altered cardiac electrophysiology. In the presence of low acetylcholine, the melatonergic/dopaminergic hypothesis, added to angiopoietin-like 4 and Krüppel-like factor 9, could offer some clue, thus offering novel targets for pharmacologic manipulation of cognition.
Collapse
Affiliation(s)
- Sherine Abdelmissih
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| | - Marwa Abdelgwad
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| | - Doaa Mohamed Elroby Ali
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| | - Mohamed Sharif Ismail Negm
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| | - Mohamed Ali Eshra
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| | - Amal Youssef
- Departments of Medical Pharmacology (S.A., A.Y.), Medical Biochemistry and Molecular Biology (M.A.), Pathology (M.S.I.N.), and Medical Physiology (M.A.E.), Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt; and Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt (D.M.E.A.)
| |
Collapse
|
17
|
Zhou T, Pan J, Yan C, Yuan J, Song H, Han Y. Unveiling shared biomarkers and therapeutic targets between systemic lupus erythematosus and heart failure through bioinformatics analysis. Front Med (Lausanne) 2024; 11:1402010. [PMID: 38912340 PMCID: PMC11190381 DOI: 10.3389/fmed.2024.1402010] [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: 03/16/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is frequently accompanied by various complications, with cardiovascular diseases being particularly concerning due to their high mortality rate. Although there is clinical evidence suggesting a potential correlation between SLE and heart failure (HF), the underlying shared mechanism is not fully understood. Therefore, it is imperative to explore the potential mechanisms and shared therapeutic targets between SLE and HF. Methods The SLE and HF datasets were downloaded from the NCBI Gene Expression Omnibus database. Differentially expressed genes (DEGs) in both SLE and HF were performed using "limma" R package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genes (KEGG) analyses were conducted to analyze the enriched functions and pathways of DEGs in both SLE and HF datasets. Protein-Protein Interaction network (PPI) and the molecular complex detection (MCODE) plugins in the Cytoscape software were performed to identify the shared hub genes between SLE and HF datasets. R package "limma" was utilized to validate the expression of hub genes based on SLE (GSE122459) and HF (GSE196656) datasets. CIBERSORT algorithm was utilized to analyze the immune cell infiltration of SLE and HF samples based on SLE (GSE112087) and HF (GSE116250) datasets. A weighted gene co-expression network analysis (WGCNA) network was established to further validate the hub genes based on HF dataset (GSE116250). Molecular biology techniques were conducted to validate the hub genes. Results 999 shared DGEs were identified between SLE and HF datasets, which were mainly enriched in pathways related to Th17 cell differentiation. 5 shared hub genes among the common DGEs between SLE and HF datasets were screened and validated, including HSP90AB1, NEDD8, RPLP0, UBB, and UBC. Additionally, 5 hub genes were identified in the central part of the MEbrown module, showing the strongest correlation with dilated cardiomyopathy. HSP90AB1 and UBC were upregulated in failing hearts compared to non-failing hearts, while UBB, NEDD8, and RPLP0 did not show significant changes. Conclusion HSP90AB1 and UBC are closely related to the co-pathogenesis of SLE and HF mediated by immune cell infiltration. They serve as promising molecular markers and potential therapeutic targets for the treatment of SLE combined with HF.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Pan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haixu Song
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
18
|
Rai R, Singh V, Ahmad Z, Jain A, Jat D, Mishra SK. Autonomic neuronal modulations in cardiac arrhythmias: Current concepts and emerging therapies. Physiol Behav 2024; 279:114527. [PMID: 38527577 DOI: 10.1016/j.physbeh.2024.114527] [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: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
The pathophysiology of atrial fibrillation and ventricular tachycardia that result in cardiac arrhythmias is related to the sustained complicated mechanisms of the autonomic nervous system. Atrial fibrillation is when the heart beats irregularly, and ventricular arrhythmias are rapid and inconsistent heart rhythms, which involves many factors including the autonomic nervous system. It's a complex topic that requires careful exploration. Cultivation of speculative knowledge on atrial fibrillation; the irregular rhythm of the heart and ventricular arrhythmias; rapid oscillating waves resulting from mistakenly inconsistent P waves, and the inclusion of an autonomic nervous system is an inconceivable approach toward clinical intricacies. Autonomic modulation, therefore, acquires new expansions and conceptions of appealing therapeutic intelligence to prevent cardiac arrhythmia. Notably, autonomic modulation uses the neural tissue's flexibility to cause remodeling and, hence, provide therapeutic effects. In addition, autonomic modulation techniques included stimulation of the vagus nerve and tragus, renal denervation, cardiac sympathetic denervation, and baroreceptor activation treatment. Strong preclinical evidence and early human studies support the annihilation of cardiac arrhythmias by sympathetic and parasympathetic systems to transmigrate the cardiac myocytes and myocardium as efficient determinants at the cellular and physiological levels. However, the goal of this study is to draw attention to these promising early pre-clinical and clinical arrhythmia treatment options that use autonomic modulation as a therapeutic modality to conquer the troublesome process of irregular heart movements. Additionally, we provide a summary of the numerous techniques for measuring autonomic tone such as heart rate oscillations and its association with cutaneous sympathetic nerve activity appear to be substitute indicators and predictors of the outcome of treatment.
Collapse
Affiliation(s)
- Ravina Rai
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India
| | - Virendra Singh
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005 UP, India
| | - Zaved Ahmad
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India
| | - Abhishek Jain
- Sanjeevani Diabetes and Heart Care Centre, Shri Chaitanya Hospital, Sagar, 470002, MP, India
| | - Deepali Jat
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India.
| | | |
Collapse
|
19
|
Li Y, Hu Y, Jiang F, Chen H, Xue Y, Yu Y. Combining WGCNA and machine learning to identify mechanisms and biomarkers of ischemic heart failure development after acute myocardial infarction. Heliyon 2024; 10:e27165. [PMID: 38455553 PMCID: PMC10918227 DOI: 10.1016/j.heliyon.2024.e27165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Background Ischemic heart failure (IHF) is a serious complication after acute myocardial infarction (AMI). Understanding the mechanism of IHF after AMI will help us conduct early diagnosis and treatment. Methods We obtained the AMI dataset GSE66360 and the IHF dataset GSE57338 from the GEO database, and screened overlapping genes common to both diseases through WGCNA analysis. Subsequently, we performed GO and KEGG enrichment analysis on overlapping genes to elucidate the common mechanism of AMI and IHF. Machine learning algorithms are also used to identify key biomarkers. Finally, we performed immune cell infiltration analysis on the dataset to further evaluate immune cell changes in AMI and IHF. Results We obtained 74 overlapping genes of AMI and IHF through WGCNA analysis, and the enrichment analysis results mainly focused on immune and inflammation-related mechanisms. Through the three machine learning algorithms of LASSO, RF and SVM-RFE, we finally obtained the four Hub genes of IL1B, TIMP2, IFIT3, and P2RY2, and verified them in the IHF dataset GSE116250, and the diagnostic model AUC = 0.907. The results of immune infiltration analysis showed that 8 types of immune cells were significantly different in AMI samples, and 6 types of immune cells were significantly different in IHF samples. Conclusion We explored the mechanism of IHF after AMI by WGCNA, enrichment analysis, and immune infiltration analysis. Four potential diagnostic candidate genes and therapeutic targets were identified by machine learning algorithms. This provides a new idea for the pathogenesis, diagnosis, and treatment of IHF after AMI.
Collapse
Affiliation(s)
- Yan Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ying Hu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Haoyu Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
20
|
Tonko JB, Lambiase PD. The proarrhythmogenic role of autonomics and emerging neuromodulation approaches to prevent sudden death in cardiac ion channelopathies. Cardiovasc Res 2024; 120:114-131. [PMID: 38195920 PMCID: PMC10936753 DOI: 10.1093/cvr/cvae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
Ventricular arrhythmias in cardiac channelopathies are linked to autonomic triggers, which are sub-optimally targeted in current management strategies. Improved molecular understanding of cardiac channelopathies and cellular autonomic signalling could refine autonomic therapies to target the specific signalling pathways relevant to the specific aetiologies as well as the central nervous system centres involved in the cardiac autonomic regulation. This review summarizes key anatomical and physiological aspects of the cardiac autonomic nervous system and its impact on ventricular arrhythmias in primary inherited arrhythmia syndromes. Proarrhythmogenic autonomic effects and potential therapeutic targets in defined conditions including the Brugada syndrome, early repolarization syndrome, long QT syndrome, and catecholaminergic polymorphic ventricular tachycardia will be examined. Pharmacological and interventional neuromodulation options for these cardiac channelopathies are discussed. Promising new targets for cardiac neuromodulation include inhibitory and excitatory G-protein coupled receptors, neuropeptides, chemorepellents/attractants as well as the vagal and sympathetic nuclei in the central nervous system. Novel therapeutic strategies utilizing invasive and non-invasive deep brain/brain stem stimulation as well as the rapidly growing field of chemo-, opto-, or sonogenetics allowing cell-specific targeting to reduce ventricular arrhythmias are presented.
Collapse
Affiliation(s)
- Johanna B Tonko
- Institute of Cardiovascular Science, University College London, 5 University Street, London WC1E 6JF, London, UK
| | - Pier D Lambiase
- Institute of Cardiovascular Science, University College London, 5 University Street, London WC1E 6JF, London, UK
- Department for Cardiology, Bart’s Heart Centre, West Smithfield EC1A 7BE, London, UK
| |
Collapse
|
21
|
Khan SU, Huang Y, Ali H, Ali I, Ahmad S, Khan SU, Hussain T, Ullah M, Lu K. Single-cell RNA Sequencing (scRNA-seq): Advances and Challenges for Cardiovascular Diseases (CVDs). Curr Probl Cardiol 2024; 49:102202. [PMID: 37967800 DOI: 10.1016/j.cpcardiol.2023.102202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
Implementing Single-cell RNA sequencing (scRNA-seq) has significantly enhanced our comprehension of cardiovascular diseases (CVDs), providing new opportunities to strengthen the prevention of CVDs progression. Cardiovascular diseases continue to be the primary cause of death worldwide. Improving treatment strategies and patient risk assessment requires a deeper understanding of the fundamental mechanisms underlying these disorders. The advanced and widespread use of Single-cell RNA sequencing enables a comprehensive investigation of the complex cellular makeup of the heart, surpassing essential descriptive aspects. This enhances our understanding of disease causes and directs functional research. The significant advancement in understanding cellular phenotypes has enhanced the study of fundamental cardiovascular science. scRNA-seq enables the identification of discrete cellular subgroups, unveiling previously unknown cell types in the heart and vascular systems that may have relevance to different disease pathologies. Moreover, scRNA-seq has revealed significant heterogeneity in phenotypes among distinct cell subtypes. Finally, we will examine current and upcoming scRNA-seq studies about various aspects of the cardiovascular system, assessing their potential impact on our understanding of the cardiovascular system and offering insight into how these technologies may revolutionise the diagnosis and treatment of cardiac conditions.
Collapse
Affiliation(s)
- Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China; Women Medical and Dental College, Khyber Medical University, Peshawar, KPK, 22020, Pakistan
| | - Yuqing Huang
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hamid Ali
- Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad-44000
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Hawally 32093, Kuwait
| | - Saleem Ahmad
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans 70112 LA, USA
| | - Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, People's Republic of China
| | - Talib Hussain
- Women Dental College Abbottabad, KPK, 22020, Pakistan
| | - Muneeb Ullah
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, KPK, Pakistan
| | - Kun Lu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China.
| |
Collapse
|
22
|
Nishiyama K. The role of P2Y 6 receptor in the pathogenesis of cardiovascular and inflammatory diseases. J Pharmacol Sci 2024; 154:108-112. [PMID: 38246724 DOI: 10.1016/j.jphs.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
The purinergic receptor P2Y6 receptor (P2Y6R) is a member of the G protein-coupled receptors (GPCR) family. P2Y6R is widely expressed in various cell types and plays a critical role in physiological processes, where it is activated by extracellular uridine diphosphate (UDP) and mobilizes Ca2+ via the Gαq/11 protein pathway. We have recently discovered the pathophysiological role of P2Y6R in cardiovascular and inflammatory diseases, including inflammatory bowel disease and non-alcoholic fatty liver disease. Furthermore, we uncovered the redox-dependent internalization of P2Y6R. In this review, we provide a comprehensive overview of the pathophysiological activity of P2Y6R in cardiovascular and inflammatory diseases. Additionally, we discuss the concept of atypical internalization control of GPCRs, which may be applied in the prevention and treatment of intestinal inflammation and cardiovascular remodeling.
Collapse
Affiliation(s)
- Kazuhiro Nishiyama
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, 1-58 Rinku-ohraikita, Izumisano, Osaka, 598-8531, Japan.
| |
Collapse
|
23
|
Lian N, Tong J, Zhu W, Meng Q, Jiang M, Bian M, Li Y. Ligustrazine and liguzinediol protect against doxorubicin-induced cardiomyocytes injury by inhibiting mitochondrial apoptosis and autophagy. Clin Exp Pharmacol Physiol 2023; 50:867-877. [PMID: 37574718 DOI: 10.1111/1440-1681.13811] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023]
Abstract
Preventing or treating heart failure (HF) by blocking cardiomyocyte apoptosis is an effective strategy that improves survival and reduces ventricular remodelling and dysfunction in the chronic stage. Autophagy is a mechanism that degrades intracellular components and compensates for energy deficiency, which is commonly observed in cardiomyocytes of failed hearts. Cardiomyocytes activated by doxorubicin (DOX) exhibit strong autophagy. This study aims to investigate the potential protective effect of ligustrazine and its derivative liguzinediol on regulating DOX-induced cardiomyocyte apoptosis and explore the use of the embryonic rat heart-derived myoblast cell line H9C2 for identifying novel treatments for HF. The results indicated that it has been demonstrated to reverse myocardial infarction remodelling in failed hearts by promoting autophagy in salvaged cardiomyocytes and anti-apoptosis of cardiomyocytes in granulation tissue. Our study suggests that ligustrazine and liguzinediol can be a promising agents and autophagy is potential pathway in the management of HF.
Collapse
Affiliation(s)
- Naqi Lian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Tong
- Xi'an International Medical Center Hospital, Xi'an, China
| | - Weijie Zhu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinghai Meng
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Miao Jiang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mianli Bian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Li X, Xu C, Li Q, Shen Q, Zeng L. Exploring key genes associated with neutrophil function and neutrophil extracellular traps in heart failure: a comprehensive analysis of single-cell and bulk sequencing data. Front Cell Dev Biol 2023; 11:1258959. [PMID: 37941896 PMCID: PMC10628466 DOI: 10.3389/fcell.2023.1258959] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Background: Heart failure (HF) is a complex and heterogeneous manifestation of multiple cardiovascular diseases that usually occurs in the advanced stages of disease progression. The role of neutrophil extracellular traps (NETs) in the pathogenesis of HF remains to be explored. Methods: Bioinformatics analysis was employed to investigate general and single-cell transcriptome sequencing data downloaded from the GEO datasets. Differentially expressed genes (DEGs) associated with NETs in HF patients and healthy controls were identified using transcriptome sequencing datasets and were subsequently subjected to functional enrichment analysis. To identify potential diagnostic biomarkers, the random forest algorithm (RF) and the least absolute shrinkage and selection operator (LASSO) were applied, followed by the construction of receiver operating characteristic (ROC) curves to assess accuracy. Additionally, single-cell transcriptome sequencing data analysis identified key immune cell subpopulations in TAC (transverse aortic constriction) mice potentially involved in NETs regulation. Cell-cell communication analysis and trajectory analysis was then performed on these key cell subpopulations. Results: We identified thirteen differentially expressed genes (DEGs) associated with NET through differential analysis of transcriptome sequencing data from HF (heart failure) samples. Utilizing the Random Forest and Lasso algorithms, along with experimental validation, we successfully pinpointed four diagnostic markers (CXCR2, FCGR3B, VNN3, and FPR2) capable of predicting HF risk. Furthermore, our analysis of intercellular communication, leveraging single-cell sequencing data, highlighted macrophages and T cells as the immune cell subpopulations with the closest interactions with neutrophils. Pseudo-trajectory analysis sheds light on the differentiation states of distinct neutrophil subpopulations. Conclusion: In this study, we conducted an in-depth investigation into the functions of neutrophil subpopulations that infiltrate cardiac tissue in TAC mice. Additionally, we identified four biomarkers (CXCR2, FCGR3B, VNN3, and FPR2) associated with NETs in HF. Our findings enhance the understanding of immunology in HF.
Collapse
Affiliation(s)
- Xudong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changhao Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiaoqiao Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingxiang Shen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of the University of South China, University of South China, Hengyang, Hunan, China
| | - Long Zeng
- Department of Cardiology, Shangrao People’s Hospital, Shangrao, Jiangxi, China
| |
Collapse
|
25
|
Yadollah B, Zahra Roudbari AB. Broiler Heart Muscle Monoaminergic Receptors Alteration in Response to Chronic Heat Stress: Based on Transcription Analysis. ARCHIVES OF RAZI INSTITUTE 2023; 78:1594-1602. [PMID: 38590690 PMCID: PMC10998952 DOI: 10.22092/ari.2023.78.5.1594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/08/2023] [Indexed: 04/10/2024]
Abstract
Chronic heat stress affects numerous physiological and behavioral mechanisms. Epigenetic changes following prolonged cyclic heat stress, creating new opportunities for molecular biology research. One of these changes involves monoamines, such as serotonin, epinephrine, norepinephrine, dopamine, and their transmission. Broiler chickens are highly susceptible to heat stress, and their hearts become insufficient during the growth phase, leading to hypertrophy of the left heart. RNA-seq data were obtained from NCBI with accession number SRP082125. The expression level of genes was determined with DESeq2 packages. Gene Ontology qualification, including biological processes, cellular components, and molecular role (MF), was performed from the Gene Ontology Resource. Cyclic heat stress in broilers significantly altered monoamine receptor expression. Twenty-nine genes of the monoamine pathway changed their expression in the left heart. Significant downregulation of expression was statistically associated with the ADRB1, HTR2A, and PNMT genes and upregulation of the MAOA gene (P<0.01). STRING database was used to construct the protein-protein interaction network; based on network analysis, the HTR2C, HTR2A, and HTR5A genes were identified as the major nodal genes in the network followed by MAOA, DRD2, DRD5, HTR1B, DRD1, DRD3, and HTR2B genes occupying the second important place in the network module. In conclusion, heat stress treatment prevented cardiac hypertrophy and altered the expression of monoamine genes. This would imply that monoamine transmission plays an important role in the development of cardiac hypertrophy, and that cyclic-chronic heat treatment modulates the cardiac monoaminergic system. These molecular biomarkers could be useful for screening, diagnosis, and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Badakhshan Yadollah
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft, Iran
| | | |
Collapse
|
26
|
Arfaras-Melainis A, Ventoulis I, Polyzogopoulou E, Boultadakis A, Parissis J. The current and future status of inotropes in heart failure management. Expert Rev Cardiovasc Ther 2023; 21:573-585. [PMID: 37458248 DOI: 10.1080/14779072.2023.2237869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/25/2023]
Abstract
INTRODUCTION Heart failure (HF) is a complex syndrome with a wide range of presentations and acuity, ranging from outpatient care to inpatient management due to acute decompensated HF, cardiogenic shock or advanced HF. Frequently, the etiology of a patient's decompensation is diminished cardiac output and peripheral hypoperfusion. Consequently, there is a need for use of inotropes, agents that increase cardiac contractility, optimize hemodynamics and ensure adequate perfusion. AREAS COVERED Inotropes are divided into 3 major classes: beta agonists, phosphodiesterase III inhibitors and calcium sensitizers. Additionally, as data from prospective studies accumulates, novel agents are emerging, including omecamtiv mecarbil and istaroxime. The aim of this review is to summarize current data on the optimal use of inotropes and to provide an expert opinion regarding their current and future use in the management of HF. EXPERT OPINION The use of inotropes has long been linked to worsening mortality, tachyarrhythmias, increased myocardial oxygen consumption and ischemia. Therefore, individualized and evidence-based treatment plans for patients who require inotropic support are necessary. Also, better quality data on the use of existing inotropes is imperative, while the development of newer and safer agents will lead to more effective management of patients with HF in the future.
Collapse
Affiliation(s)
- Angelos Arfaras-Melainis
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, Ptolemaida, Greece
| | - Effie Polyzogopoulou
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Boultadakis
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - John Parissis
- Emergency Department, Heart Failure Unit, Attikon University Hospital, Athens, Greece
| |
Collapse
|
27
|
Yamada S, Ko T, Katagiri M, Morita H, Komuro I. Recent Advances in Translational Research for Heart Failure in Japan. J Card Fail 2023; 29:931-938. [PMID: 37321698 DOI: 10.1016/j.cardfail.2022.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Despite decades of intensive research and therapeutic development, heart failure remains a leading cause of death worldwide. However, recent advances in several basic and translational research fields, such as genomic analysis and single-cell analysis, have increased the possibility of developing novel diagnostic approaches to heart failure. Most cardiovascular diseases that predispose individuals to heart failure are caused by genetic and environmental factors. It follows that genomic analysis can contribute to the diagnosis and prognostic stratification of patients with heart failure. In addition, single-cell analysis has shown great potential for unveiling the pathogenesis and/or pathophysiology and for discovering novel therapeutic targets for heart failure. Here, we summarize the recent advances in translational research on heart failure in Japan, based mainly on our studies.
Collapse
Affiliation(s)
- Shintaro Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mikako Katagiri
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, International University of Health and Welfare, Tokyo, Japan.
| |
Collapse
|
28
|
Long X, Yuan X, Du J. Single-cell and spatial transcriptomics: Advances in heart development and disease applications. Comput Struct Biotechnol J 2023; 21:2717-2731. [PMID: 37181659 PMCID: PMC10173363 DOI: 10.1016/j.csbj.2023.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Current transcriptomics technologies, including bulk RNA-seq, single-cell RNA sequencing (scRNA-seq), single-nucleus RNA-sequencing (snRNA-seq), and spatial transcriptomics (ST), provide novel insights into the spatial and temporal dynamics of gene expression during cardiac development and disease processes. Cardiac development is a highly sophisticated process involving the regulation of numerous key genes and signaling pathways at specific anatomical sites and developmental stages. Exploring the cell biological mechanisms involved in cardiogenesis also contributes to congenital heart disease research. Meanwhile, the severity of distinct heart diseases, such as coronary heart disease, valvular disease, cardiomyopathy, and heart failure, is associated with cellular transcriptional heterogeneity and phenotypic alteration. Integrating transcriptomic technologies in the clinical diagnosis and treatment of heart diseases will aid in advancing precision medicine. In this review, we summarize applications of scRNA-seq and ST in the cardiac field, including organogenesis and clinical diseases, and provide insights into the promise of single-cell and spatial transcriptomics in translational research and precision medicine.
Collapse
Affiliation(s)
- Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
29
|
Nomura S, Ono M. Precision and genomic medicine for dilated and hypertrophic cardiomyopathy. Front Cardiovasc Med 2023; 10:1137498. [PMID: 36950287 PMCID: PMC10025380 DOI: 10.3389/fcvm.2023.1137498] [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: 01/04/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Cardiomyopathy develops through an interaction of genetic and environmental factors. The clinical manifestations of both dilated cardiomyopathy and hypertrophic cardiomyopathy are diverse, but genetic testing defines the causative genes in about half of cases and can predict clinical prognosis. It has become clear that cardiomyopathy is caused not only by single rare variants but also by combinations of multiple common variants, and genome-wide genetic research is important for accurate disease risk assessment. Single-cell analysis research aimed at understanding the pathophysiology of cardiomyopathy is progressing rapidly, and it is expected that genomic analysis and single-cell molecular profiling will be combined to contribute to more detailed stratification of cardiomyopathy.
Collapse
Affiliation(s)
- Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Minoru Ono
- Department of Cardiac Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Neumann J, Hofmann B, Dhein S, Gergs U. Role of Dopamine in the Heart in Health and Disease. Int J Mol Sci 2023; 24:ijms24055042. [PMID: 36902474 PMCID: PMC10003060 DOI: 10.3390/ijms24055042] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Dopamine has effects on the mammalian heart. These effects can include an increase in the force of contraction, and an elevation of the beating rate and the constriction of coronary arteries. Depending on the species studied, positive inotropic effects were strong, very modest, or absent, or even negative inotropic effects occurred. We can discern five dopamine receptors. In addition, the signal transduction by dopamine receptors and the regulation of the expression of cardiac dopamine receptors will be of interest to us, because this might be a tempting area of drug development. Dopamine acts in a species-dependent fashion on these cardiac dopamine receptors, but also on cardiac adrenergic receptors. We will discuss the utility of drugs that are currently available as tools to understand cardiac dopamine receptors. The molecule dopamine itself is present in the mammalian heart. Therefore, cardiac dopamine might act as an autocrine or paracrine compound in the mammalian heart. Dopamine itself might cause cardiac diseases. Moreover, the cardiac function of dopamine and the expression of dopamine receptors in the heart can be altered in diseases such as sepsis. Various drugs for cardiac and non-cardiac diseases are currently in the clinic that are, at least in part, agonists or antagonists at dopamine receptors. We define the research needs in order to understand dopamine receptors in the heart better. All in all, an update on the role of dopamine receptors in the human heart appears to be clinically relevant, and is thus presented here.
Collapse
Affiliation(s)
- Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
- Correspondence: ; Tel.: +49-345-557-1686; Fax: +49-345-557-1835
| | - Britt Hofmann
- Herzchirurgie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
| | - Stefan Dhein
- Medizinische Fakultät, Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107 Leipzig, Germany
| | - Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
| |
Collapse
|
31
|
Preoperative mental illness and postoperative atrial fibrillation in cardiac surgery patients: Identifying a vulnerable population. J Clin Transl Sci 2023; 7:e15. [PMID: 36755550 PMCID: PMC9879927 DOI: 10.1017/cts.2022.493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
|
32
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
33
|
Deviatiiarov RM, Gams A, Kulakovskiy IV, Buyan A, Meshcheryakov G, Syunyaev R, Singh R, Shah P, Tatarinova TV, Gusev O, Efimov IR. An atlas of transcribed human cardiac promoters and enhancers reveals an important role of regulatory elements in heart failure. NATURE CARDIOVASCULAR RESEARCH 2023; 2:58-75. [PMID: 39196209 DOI: 10.1038/s44161-022-00182-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/02/2022] [Indexed: 08/29/2024]
Abstract
A deeper knowledge of the dynamic transcriptional activity of promoters and enhancers is needed to improve mechanistic understanding of the pathogenesis of heart failure and heart diseases. In this study, we used cap analysis of gene expression (CAGE) to identify and quantify the activity of transcribed regulatory elements (TREs) in the four cardiac chambers of 21 healthy and ten failing adult human hearts. We identified 17,668 promoters and 14,920 enhancers associated with the expression of 14,519 genes. We showed how these regulatory elements are alternatively transcribed in different heart regions, in healthy versus failing hearts and in ischemic versus non-ischemic heart failure samples. Cardiac-disease-related single-nucleotide polymorphisms (SNPs) appeared to be enriched in TREs, potentially affecting the allele-specific transcription factor binding. To conclude, our open-source heart CAGE atlas will serve the cardiovascular community in improving the understanding of the role of the cardiac gene regulatory networks in cardiovascular disease and therapy.
Collapse
Affiliation(s)
- Ruslan M Deviatiiarov
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Anna Gams
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Ivan V Kulakovskiy
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Buyan
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia
| | | | - Roman Syunyaev
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ramesh Singh
- Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Palak Shah
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
- Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Tatiana V Tatarinova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.
- Department of Biology, University of La Verne, La Verne, CA, USA.
| | - Oleg Gusev
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Graduate School of Medicine, Juntendo University, Tokyo, Japan.
- RIKEN Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan.
- Endocrinology Research Center, Moscow, Russia.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA.
- Department of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
34
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
35
|
Zou S, Khoo BL. Subtyping based on immune cell fractions reveal heterogeneity of cardiac fibrosis in end-stage heart failure. Front Immunol 2023; 14:1053793. [PMID: 36875078 PMCID: PMC9975711 DOI: 10.3389/fimmu.2023.1053793] [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: 09/26/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Background A central issue hindering the development of effective anti-fibrosis drugs for heart failure is the unclear interrelationship between fibrosis and the immune cells. This study aims at providing precise subtyping of heart failure based on immune cell fractions, elaborating their differences in fibrotic mechanisms, and proposing a biomarker panel for evaluating intrinsic features of patients' physiological statuses through subtype classification, thereby promoting the precision medicine for cardiac fibrosis. Methods We inferred immune cell type abundance of the ventricular samples by a computational method (CIBERSORTx) based on ventricular tissue samples from 103 patients with heart failure, and applied K-means clustering to divide patients into two subtypes based on their immune cell type abundance. We also designed a novel analytic strategy: Large-Scale Functional Score and Association Analysis (LAFSAA), to study fibrotic mechanisms in the two subtypes. Results Two subtypes of immune cell fractions: pro-inflammatory and pro-remodeling subtypes, were identified. LAFSAA identified 11 subtype-specific pro-fibrotic functional gene sets as the basis for personalised targeted treatments. Based on feature selection, a 30-gene biomarker panel (ImmunCard30) established for diagnosing patient subtypes achieved high classification performance, with the area under the receiver operator characteristic curve corresponding to 0.954 and 0.803 for the discovery and validation sets, respectively. Conclusion Patients with the two subtypes of cardiac immune cell fractions were likely having different fibrotic mechanisms. Patients' subtypes can be predicted based on the ImmunCard30 biomarker panel. We envision that our unique stratification strategy revealed in this study will unravel advance diagnostic techniques for personalised anti-fibrotic therapy.
Collapse
Affiliation(s)
- Shangjie Zou
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China.,Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| |
Collapse
|
36
|
Sallam M, Mysara M, Benotmane MA, Tamarat R, Santos SCR, Crijns APG, Spoor D, Van Nieuwerburgh F, Deforce D, Baatout S, Guns PJ, Aerts A, Ramadan R. DNA Methylation Alterations in Fractionally Irradiated Rats and Breast Cancer Patients Receiving Radiotherapy. Int J Mol Sci 2022; 23:16214. [PMID: 36555856 PMCID: PMC9783664 DOI: 10.3390/ijms232416214] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Radiation-Induced CardioVascular Disease (RICVD) is an important concern in thoracic radiotherapy with complex underlying pathophysiology. Recently, we proposed DNA methylation as a possible mechanism contributing to RICVD. The current study investigates DNA methylation in heart-irradiated rats and radiotherapy-treated breast cancer (BC) patients. Rats received fractionated whole heart X-irradiation (0, 0.92, 6.9 and 27.6 Gy total doses) and blood was collected after 1.5, 3, 7 and 12 months. Global and gene-specific methylation of the samples were evaluated; and gene expression of selected differentially methylated regions (DMRs) was validated in rat and BC patient blood. In rats receiving an absorbed dose of 27.6 Gy, DNA methylation alterations were detected up to 7 months with differential expression of cardiac-relevant DMRs. Of those, SLMAP showed increased expression at 1.5 months, which correlated with hypomethylation. Furthermore, E2F6 inversely correlated with a decreased global longitudinal strain. In BC patients, E2F6 and SLMAP exhibited differential expression directly and 6 months after radiotherapy, respectively. This study describes a systemic radiation fingerprint at the DNA methylation level, elucidating a possible association of DNA methylation to RICVD pathophysiology, to be validated in future mechanistic studies.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Wilrijk, Belgium;
| | - Mohamed Mysara
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Radia Tamarat
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, LR2I, 92260 Fontenay-aux-Roses, France;
| | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine of the Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Anne P. G. Crijns
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.P.G.C.); (D.S.)
| | - Daan Spoor
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.P.G.C.); (D.S.)
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium; (F.V.N.); (D.D.)
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium; (F.V.N.); (D.D.)
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
- Department of Molecular Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Wilrijk, Belgium;
| | - An Aerts
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Raghda Ramadan
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| |
Collapse
|
37
|
Zhang H, Wang L, Yin D, Zhou Q, Lv L, Dong Z, Shi Y. Integration of proteomic and metabolomic characterization in atrial fibrillation-induced heart failure. BMC Genomics 2022; 23:789. [PMID: 36456901 PMCID: PMC9714089 DOI: 10.1186/s12864-022-09044-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The exact mechanism of atrial fibrillation (AF)-induced heart failure (HF) remains unclear. Proteomics and metabolomics were integrated to in this study, as to describe AF patients' dysregulated proteins and metabolites, comparing patients without HF to patients with HF. METHODS Plasma samples of 20 AF patients without HF and another 20 with HF were analyzed by multi-omics platforms. Proteomics was performed with data independent acquisition-based liquid chromatography-tandem mass spectrometry (LC-MS/MS), as metabolomics was performed with LC-MS/MS platform. Proteomic and metabolomic results were analyzed separately and integrated using univariate statistical methods, multivariate statistical methods or machine learning model. RESULTS We found 35 up-regulated and 15 down-regulated differentially expressed proteins (DEPs) in AF patients with HF compared to AF patients without HF. Moreover, 121 up-regulated and 14 down-regulated differentially expressed metabolites (DEMs) were discovered in HF patients compared to AF patients without HF. An integrated analysis of proteomics and metabolomics revealed several significantly enriched pathways, including Glycolysis or Gluconeogenesis, Tyrosine metabolism and Pentose phosphate pathway. A total of 10 DEPs and DEMs selected as potential biomarkers provided excellent predictive performance, with an AUC of 0.94. In addition, subgroup analysis of HF classification was performed based on metabolomics, which yielded 9 DEMs that can distinguish between AF and HF for HF classification. CONCLUSIONS This study provides novel insights to understanding the mechanisms of AF-induced HF progression and identifying novel biomarkers for prognosis of AF with HF by using metabolomics and proteomics analyses.
Collapse
Affiliation(s)
- Haiyu Zhang
- grid.410736.70000 0001 2204 9268Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Lu Wang
- grid.410736.70000 0001 2204 9268Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Dechun Yin
- grid.410736.70000 0001 2204 9268Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Qi Zhou
- grid.410736.70000 0001 2204 9268Research Management Office, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Lin Lv
- grid.410736.70000 0001 2204 9268Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Zengxiang Dong
- grid.410736.70000 0001 2204 9268Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| | - Yuanqi Shi
- grid.410736.70000 0001 2204 9268Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001 China
| |
Collapse
|
38
|
Nakamura S, Numata G, Yamaguchi T, Tokiwa H, Higashikuni Y, Nomura S, Sasano T, Takimoto E, Komuro I. Endoplasmic reticulum stress-activated nuclear factor-kappa B signaling pathway induces the upregulation of cardiomyocyte dopamine D1 receptor in heart failure. Biochem Biophys Res Commun 2022; 637:247-253. [DOI: 10.1016/j.bbrc.2022.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
|
39
|
Ke Y, Jian-yuan H, Ping Z, Yue W, Na X, Jian Y, Kai-xuan L, Yi-fan S, Han-bin L, Rong L. The progressive application of single-cell RNA sequencing technology in cardiovascular diseases. Biomed Pharmacother 2022; 154:113604. [DOI: 10.1016/j.biopha.2022.113604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
|
40
|
Byrne SE, Vishwakarma N, Sriramula S, Katwa LC. Dopamine receptor 3: A mystery at the heart of cardiac fibrosis. Life Sci 2022; 308:120918. [PMID: 36041503 DOI: 10.1016/j.lfs.2022.120918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/27/2022] [Accepted: 08/24/2022] [Indexed: 10/31/2022]
Abstract
Dopamine receptors have been extensively studied in the mammalian brain and spinal cord, as dopamine is a vital determinant of bodily movement, cognition, and overall behavior. Thus, dopamine receptor antagonist antipsychotic drugs are commonly used to treat multiple psychiatric disorders. Although less discussed, these receptors are also expressed in other peripheral organ systems, such as the kidneys, eyes, gastrointestinal tract, and cardiac tissue. Consequently, therapies for certain psychiatric disorders which target dopamine receptors could have unidentified consequences on certain functions of these peripheral tissues. The existence of an intrinsic dopaminergic system in the human heart remains controversial and debated within the literature. Therefore, this review focuses on literature related to dopamine receptors within cardiac tissue, specifically dopamine receptor 3 (D3R), and summarizes the current state of knowledge while highlighting areas of research which may be lacking. Additionally, recent findings regarding crosstalk between D3R and dopamine receptor 1 (D1R) are examined. This review discusses the novel concept of understanding the role of the loss of function of D3R may play in collagen accumulation and cardiac fibrosis, eventually leading to heart failure.
Collapse
Affiliation(s)
- Shannon E Byrne
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Nandini Vishwakarma
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Laxmansa C Katwa
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
41
|
Zheng Y, Qi B, Gao W, Qi Z, Liu Y, Wang Y, Feng J, Cheng X, Luo Z, Li T. Macrophages-Related Genes Biomarkers in the Deterioration of Atherosclerosis. Front Cardiovasc Med 2022; 9:890321. [PMID: 35845072 PMCID: PMC9282674 DOI: 10.3389/fcvm.2022.890321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The macrophages are involved in all stages of cardiovascular diseases, demonstrating the correlation between inflammation, atherosclerosis, and myocardial infarction (MI). Here, we aim to investigate macrophages-related genes in the deterioration of atherosclerosis. METHODS GSE41571 was downloaded and the abundance of immune cells was estimated by utilizing the xCell. By utilizing the limma test and correlation analysis, differentially expressed macrophages-related genes (DEMRGs) were documented. The functional pathways and the protein-protein interaction (PPI) network were analyzed and the hub DEMRGs were obtained. The hub DEMRGs and their interactions were analyzed using NetworkAnalyst 3.0 and for validation, the expressions of hub DEMRGs were analyzed using the GSE135055 and GSE116250 datasets as well as atherosclerosis and MI mice model. RESULTS A total of 509 differentially expressed genes (DEGs) were correlated with the abundance of macrophages and were identified as DEMRGs (Pearson correlation coefficients (PCC) > 0.6), which were mainly enriched in extracellular structure organization, lysosomal membrane, MHC protein complex binding, and so on. After screening out, 28 hub DEMRGs were obtained with degrees ≥20, including GNAI1 (degree = 113), MRPS2 (degree = 56), HCK (degree = 45), SOCS3 (degree = 40), NET1 (degree = 28), and so on. After validating using Gene Expression Omnibus (GEO) datasets and the atherosclerosis and MI mice model, eight proteins were validated using ApoE-/- and C57 mice. The expression levels of proteins, including SYNJ2, NET1, FZD7, LCP2, HCK, GNB2, and PPP4C were positively correlated to left ventricular ejection fraction (LVEF), while that of EIF4EBP1 was negatively correlated to LVEF. CONCLUSION The screened hub DEMRGs, SYNJ2, NET1, FZD7, LCP2, HCK, GNB2, EIF4EBP1, and PPP4C, may be therapeutic targets for treatment and prediction in the patients with plaque progression and MI recurrent events. The kit of the eight hub DEMRGs may test plaque progression and MI recurrent events and help in the diagnosis and treatment of MI-induced heart failure (HF), thus decreasing mortality and morbidity.
Collapse
Affiliation(s)
- Yue Zheng
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Bingcai Qi
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Zhenchang Qi
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yanwu Liu
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yuchao Wang
- School of Medicine, Nankai University, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Jianyu Feng
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xian Cheng
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Zhiqiang Luo
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Tong Li
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Tianjin, China
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| |
Collapse
|
42
|
Zheng Y, Gao W, Zhang Q, Cheng X, Liu Y, Qi Z, Li T. Ferroptosis and Autophagy-Related Genes in the Pathogenesis of Ischemic Cardiomyopathy. Front Cardiovasc Med 2022; 9:906753. [PMID: 35845045 PMCID: PMC9279674 DOI: 10.3389/fcvm.2022.906753] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Obesity plays an important role in type 2 diabetes mellitus (T2DM) and myocardial infarction (MI). Ferroptosis and ferritinophagy are related to metabolic pathways, such as fatty acid metabolism and mitochondrial respiration. We aimed to investigate the ferroptosis- and autophagy-related differentially expressed genes (DEGs) that might be potential targets for MI progression. METHODS GSE116250 was analyzed to obtain DEGs. A Venn diagram was used to obtain the overlapping ferroptosis- and autophagy-related DEGs. The enrichment pathway analysis was performed and the hub genes were obtained. Pivotal miRNAs, transcription factors, and drugs with the hub genes interactions were also predicted. The MI mice model was constructed, and qPCR analysis and single-cell sequencing were used to validate the hub genes. RESULTS Utilizing the limma package and the Venn diagram, 26 ferroptosis-related and 29 autophagy-related DEGs were obtained. The list of ferroptosis-related DEGs was analyzed, which were involved in the cellular response to a toxic substance, cellular oxidant detoxification, and the IL-17 signaling pathway. The list of autophagy-related DEGs was involved in the regulation of autophagy, the regulation of JAK-STAT signaling pathway, and the regulation of MAPK cascade. In the protein-protein interaction network, the hub DEGs, such as IL-6, PTGS2, JUN, NQO1, NOS3, LEPR, NAMPT, CDKN2A, CDKN1A, and Snai1, were obtained. After validation using qPCR analysis in the MI mice model and single-cell sequencing, the 10 hub genes can be the potential targets for MI deterioration. CONCLUSION The screened hub genes, IL-6, PTGS2, JUN, NQO1, NOS3, LEPR, NAMPT, CDKN2A, CDKN1A, and Snai1, may be therapeutic targets for patients with MI and may prevent adverse cardiovascular events.
Collapse
Affiliation(s)
- Yue Zheng
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Qiang Zhang
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xian Cheng
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Department of Heart Center, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Yanwu Liu
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Department of Heart Center, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Zhenchang Qi
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Department of Heart Center, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Tong Li
- School of Medicine, Nankai University, Tianjin, China
- Department of Heart Center, The Third Central Hospital of Tianjin, Tianjin, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Department of Heart Center, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| |
Collapse
|
43
|
Jiang C, Jiang W. Integrated Bioinformatics Identifies FREM1 as a Diagnostic Gene Signature for Heart Failure. Appl Bionics Biomech 2022; 2022:1425032. [PMID: 35726312 PMCID: PMC9206587 DOI: 10.1155/2022/1425032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/20/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
Objective This study is aimed at integrating bioinformatics and machine learning to determine novel diagnostic gene signals in the progression of heart failure disease. Methods The heart failure microarray datasets and RNA-seq datasets have been downloaded from the public database. Differentially expressed genes (DE genes) are screened out, and then, we analyze their biological functions and pathways. Integrating three machine learning methods, the least absolute shrinkage and selection operator (LASSO) algorithm, random forest (RF) algorithm, and support vector machine recursive feature elimination (SVM-RFE) are used to determine candidate diagnostic gene signals. Then, external independent RNA-seq datasets evaluate the diagnostic value of gene signals. Finally, the convolution tool CIBERSORT estimated the composition pattern of immune cell subtypes in heart failure and carried out a correlation analysis combined with gene signals. Results Under the set threshold, we obtained 47 DE genes with the most significant differences. Enrichment analysis shows that most of them are related to hypertrophy, matrix structural constituent, protein binding, inflammatory immune pathway, cardiovascular disease, and inflammatory disease. Three machine learning methods assisted in determining the potential characteristic signals Fras1-related extracellular matrix 1 (FREM1) and meiosis-specific nuclear structural 1 (MNS1). Validation of external datasets confirms that FREM1 is a diagnostic gene signal for heart failure. Immune cell subtypes of tissue specimens found T cell CD8, mast cell resting, T cell CD4 memory resting, T cell regulation (Tregs), monocytes, macrophages M2, T cell CD4 naive, macrophages M0, and neutrophils are associated with HF. Conclusion The gene signal FREM1 may be a potential molecular target in the development of HF and is related to the difference in immune infiltration of HF tissue.
Collapse
Affiliation(s)
- Chenyang Jiang
- The First Clinical Medical College of Guangxi Medical University, Nanning 530021, China
| | - Weidong Jiang
- Department of Cardiology, Nantong Hospital of Traditional Chinese Medicine, Nantong 226000, China
| |
Collapse
|
44
|
Anto Michel N, Ljubojevic-Holzer S, Bugger H, Zirlik A. Cellular Heterogeneity of the Heart. Front Cardiovasc Med 2022; 9:868466. [PMID: 35548426 PMCID: PMC9081371 DOI: 10.3389/fcvm.2022.868466] [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: 02/02/2022] [Accepted: 03/23/2022] [Indexed: 11/18/2022] Open
Abstract
Recent advances in technology such as the introduction of high throughput multidimensional tools like single cell sequencing help to characterize the cellular composition of the human heart. The diversity of cell types that has been uncovered by such approaches is by far greater than ever expected before. Accurate identification of the cellular variety and dynamics will not only facilitate a much deeper understanding of cardiac physiology but also provide important insights into mechanisms underlying its pathological transformation. Distinct cellular patterns of cardiac cell clusters may allow differentiation between a healthy heart and a sick heart while potentially predicting future disease at much earlier stages than currently possible. These advances have already extensively improved and will ultimately revolutionize our knowledge of the mechanisms underlying cardiovascular disease as such. In this review, we will provide an overview of the cells present in the human and rodent heart as well as genes that may be used for their identification.
Collapse
|
45
|
Edinoff AN, Ellis ED, Nussdorf LM, Hill TW, Cornett EM, Kaye AM, Kaye AD. Antipsychotic Polypharmacy-Related Cardiovascular Morbidity and Mortality: A Comprehensive Review. Neurol Int 2022; 14:294-309. [PMID: 35324580 PMCID: PMC8954521 DOI: 10.3390/neurolint14010024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Schizophrenia is a psychotic disorder that exists at the more extreme end of a spectrum of diseases, and significantly affects daily functioning. Cardiovascular adverse effects of antipsychotic medications are well known, and include changes in blood pressure and arrhythmias. Sudden cardiac death is the leading cause of death worldwide, and antipsychotic medications are associated with numerous cardiac side effects. A possible link exists between antipsychotic medications and sudden cardiac death. Common prescribing patterns that may influence cardiovascular events include the use of multiple antipsychotics and/or additional drugs commonly prescribed to patients on antipsychotics. The results of this review reflect an association between antipsychotic drugs and increased risk of ventricular arrhythmias and sudden cardiac death by iatrogenic prolongation of the QTc interval. QTc prolongation and sudden cardiac death exist in patients taking antipsychotic monotherapy. The risk increases for the concomitant use of specific drugs that prolong the QTc interval, such as opioids, antibiotics, and illicit drugs. However, evidence suggests that QTc intervals may not adequately predict sudden cardiac death. In considering the findings of this narrative review, we conclude that it is unclear whether there is a precise association between antipsychotic polypharmacy and sudden cardiac death with QTc interval changes. The present narrative review warrants further research on this important potential association.
Collapse
Affiliation(s)
- Amber N. Edinoff
- Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Science Center Shreveport, 1501 Kings Hwy, Shreveport, LA 71103, USA
- Correspondence: ; Tel.: +1-(318)-675-8969
| | - Emily D. Ellis
- School of Medicine, Louisiana State University Shreveport, Shreveport, LA 71103, USA; (E.D.E.); (L.M.N.); (T.W.H.)
| | - Laura M. Nussdorf
- School of Medicine, Louisiana State University Shreveport, Shreveport, LA 71103, USA; (E.D.E.); (L.M.N.); (T.W.H.)
| | - Taylor W. Hill
- School of Medicine, Louisiana State University Shreveport, Shreveport, LA 71103, USA; (E.D.E.); (L.M.N.); (T.W.H.)
| | - Elyse M. Cornett
- Department of Anesthesiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA; (E.M.C.); (A.D.K.)
| | - Adam M. Kaye
- Department of Pharmacy Practice, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Alan D. Kaye
- Department of Anesthesiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA; (E.M.C.); (A.D.K.)
| |
Collapse
|
46
|
Elfimova AE, Tipisova EV, Molodovskaya IN, Alikina VA. Adaptive potential of cardiovascular system and its relationship with sex hormones and dopamine levels in women of Arkhangelsk Oblast and Yamalo-Nenets Autonomous Okrug. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2022-2902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To assess the adaptive potential of cardiovascular system in female population of the Russian Arctic territory and its relationship with sex hormones and dopamine levels.Material and methods. The study involved 253 women living in European North (Arkhangelsk Oblast) and Asian North (Yamalo-Nenets Autonomous Okrug), who were divided into groups of fertile (n=58 and n=70, respectively) and postmenopausal women (n=59 and n=66, respectively). Enzyme immunoassay of serum was used to determine the levels of follicle-stimulating and luteinizing hormones (LH), progesterone, prolactin, total and free testosterone, estradiol, sex hormone-binding globulin. In plasma, dopamine level was identified. Adaptive potential (AP) was estimated according to R. M. Baevsky method.Results. A significant disadaptation was revealed in residents of Asian North compared to European North as follows: the proportion of persons with poor adaptation increases from 16% in European North to 26% in Asian North in fertile women, while the proportion of persons with adaptation failure increases from 9 to 23% in postmenopausal women. The strongest relationships between hormonal parameters and AP level were found in postmenopausal women: in European North, adaptation impairment is associated with higher levels of testosterone at lower concentrations of follicle-stimulating hormone, LH and sex hormone-binding globulin; in Asian North, adaptation failure is associated with lower values of LH, prolactin, testosterone, and dopamine.Conclusion. A greater AP impairment, which characterizes cardiovascular system adaptation, was noted among Asian North residents. We revealed significant relationships between cardiovascular function and hormonal parameters. In European North residents, poor adaptation is associated with a more androgenic sex hormone profile, while in Asian North — with lower values of LH, prolactin, testosterone and dopamine.
Collapse
Affiliation(s)
- A. E. Elfimova
- N. Laverov Federal Center for Integrated Arctic Research
| | - E. V. Tipisova
- N. Laverov Federal Center for Integrated Arctic Research
| | | | - V. A. Alikina
- N. Laverov Federal Center for Integrated Arctic Research
| |
Collapse
|
47
|
Shi X, Zhang L, Li Y, Xue J, Liang F, Ni HW, Wang X, Cai Z, Shen LH, Huang T, He B. Integrative Analysis of Bulk and Single-Cell RNA Sequencing Data Reveals Cell Types Involved in Heart Failure. Front Bioeng Biotechnol 2022; 9:779225. [PMID: 35071201 PMCID: PMC8766768 DOI: 10.3389/fbioe.2021.779225] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Owing to the high mortality rates of heart failure (HF), a more detailed description of the HF becomes extremely urgent. Since the pathogenesis of HF remain elusive, a thorough identification of the genetic factors will provide novel insights into the molecular basis of this cardiac dysfunction. In our research, we performed publicly available transcriptome profiling datasets, including non-failure (NF), dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) hearts tissues. Through principal component analysis (PCA), gene differential expression analysis, gene set enrichment analysis (GSEA), and gene Set Variation Analysis (GSVA), we figured out the candidate genes noticeably altered in HF, the specific biomarkers of endothelial cell (EC) and cardiac fibrosis, then validated the differences of the inflammation-related cell adhesion molecules (CAMs), extracellular matrix (ECM) genes, and immune responses. Taken together, our results suggested the EC and fibroblast could be activated in response to HF. DCM and ICM had both commonality and specificity in the pathogenesis of HF. Higher inflammation in ICM might related to autocrine CCL3/CCL4-CCR5 interaction induced chemokine signaling activation. Furthermore, the activities of neutrophil and macrophage were higher in ICM than DCM. These findings identified features of the landscape of previously underestimated cellular, transcriptomic heterogeneity between ICM and DCM.
Collapse
Affiliation(s)
- Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Li Zhang
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science, East China Normal University, Ministry of Education, Shanghai, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jieyuan Xue
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Han-Wen Ni
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ling-Hong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Huang
- Bio-Med Big Data Center, Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Bryson TD, Harding P. Prostaglandin E2 EP receptors in cardiovascular disease: An update. Biochem Pharmacol 2021; 195:114858. [PMID: 34822808 DOI: 10.1016/j.bcp.2021.114858] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022]
Abstract
This review article provides an update for the role of prostaglandin E2 receptors (EP1, EP2, EP3 and EP4) in cardiovascular disease. Where possible we have reported citations from the last decade although this was not possible for all of the topics covered due to the paucity of publications. The authors have attempted to cover the subjects of ischemia-reperfusion injury, arrhythmias, hypertension, novel protein binding partners of the EP receptors and their pathophysiological significance, and cardiac regeneration. These latter two topics bring studies of the EP receptors into new and exciting areas of research that are just beginning to be explored. Where there is peer-reviewed literature, the authors have placed particular emphasis on clinical studies although these are limited in number.
Collapse
Affiliation(s)
- Timothy D Bryson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, United States; Frankel Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pamela Harding
- Hypertension & Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
49
|
Kisling A, Byrne S, Parekh RU, Melit-Thomas D, de Castro Brás LE, Lust RM, Clemens S, Sriramula S, Katwa LC. Loss of Function in Dopamine D3 Receptor Attenuates Left Ventricular Cardiac Fibroblast Migration and Proliferation in vitro. Front Cardiovasc Med 2021; 8:732282. [PMID: 34708087 PMCID: PMC8542768 DOI: 10.3389/fcvm.2021.732282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Evidence suggests the existence of an intracardiac dopaminergic system that plays a pivotal role in regulating cardiac function and fibrosis through G-protein coupled receptors, particularly mediated by dopamine receptor 3 (D3R). However, the expression of dopamine receptors in cardiac tissue and their role in cardiac fibroblast function is unclear. In this brief report, first we determined expression of D1R and D3R both in left ventricle (LV) tissue and fibroblasts. Then, we explored the role of D3R in the proliferation and migration of fibroblast cell cultures using both genetic and pharmaceutical approaches; specifically, we compared cardiac fibroblasts isolated from LV of wild type (WT) and D3R knockout (D3KO) mice in response to D3R-specific pharmacological agents. Finally, we determined if loss of D3R function could significantly alter LV fibroblast expression of collagen types I (Col1a1) and III (Col3a1). Cardiac fibroblast proliferation was attenuated in D3KO cells, mimicking the behavior of WT cardiac fibroblasts treated with D3R antagonist. In response to scratch injury, WT cardiac fibroblasts treated with the D3R agonist, pramipexole, displayed enhanced migration compared to control WT and D3KO cells. Loss of function in D3R resulted in attenuation of both proliferation and migration in response to scratch injury, and significantly increased the expression of Col3a1 in LV fibroblasts. These findings suggest that D3R may mediate cardiac fibroblast function during the wound healing response. To our knowledge this is the first report of D3R's expression and functional significance directly in mouse cardiac fibroblasts.
Collapse
Affiliation(s)
- Andrew Kisling
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Shannon Byrne
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Rohan U Parekh
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Deepthy Melit-Thomas
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Lisandra E de Castro Brás
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States.,Department of Cardiovascular Sciences, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Robert M Lust
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Laxmansa C Katwa
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| |
Collapse
|
50
|
Liu J, Jin Y, Wang B, Wang Y, Zuo S, Zhang J. Dopamine D1 receptor alleviates doxorubicin-induced cardiac injury by inhibiting NLRP3 inflammasome. Biochem Biophys Res Commun 2021; 561:7-13. [PMID: 33992835 DOI: 10.1016/j.bbrc.2021.04.098] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Doxorubicin (DOX) is a broad-spectrum antineoplastic drug; however, its serious cardiotoxic side effects in inflammatory responses limit its use in clinical applications. Dopamine D1 receptor (DRD1), a G protein-coupled receptor, is crucial for the development and function of the nervous system; additionally, it also play a role in immune regulation. However, the specific role of DRD1 in DOX-induced cardiac inflammation has not yet been clarified. Here, we discovered that DRD1 expression was induced by DOX treatment in H9C2 cardiomyocytes. DRD1 activation by A-68930, a DRD1-specific agonist, decreased DOX-induced nucleotide-binding domain-like receptor protein 3 (NLRP3) expression, caspase-1 activation, and IL-1β maturation in H9C2 cells. Expression of the cytokines IL-1β and IL-18 in the supernatants was also inhibited by A-68930 treatment. DRD1 knockdown, using siRNA, abolished the effects of A-68930 on the DOX-induced NLRP3 inflammasome. Furthermore, we found that DRD1 signaling downregulated the NLRP3 inflammasome in H9C2 cells through cyclic adenosine monophosphate (cAMP). Moreover, application of A-68930 to activate DRD1 reduced cardiac injury and fibrosis in a DOX-treated mouse model by suppressing the NLRP3 inflammasome in the heart. These findings indicate that DRD1 signaling may protect against DOX-induced cardiac injury by inhibiting the NLRP3 inflammasome-mediated inflammation.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
| | - Yuxuan Jin
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bei Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yiran Wang
- Division of Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Shengkai Zuo
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jinying Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|