1
|
Mikami T, Yokomachi K, Mizuno K, Kobayashi M. Feasibility of Epicardial Adipose Tissue Quantification Using Non-electrocardiogram-Gated Chest Computed Tomography Images. J Comput Assist Tomogr 2025; 49:80-84. [PMID: 39146220 DOI: 10.1097/rct.0000000000001662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
OBJECTIVE Epicardial adipose tissue (EAT) is an important imaging indicator of cardiovascular risk. EAT volume is usually measured using electrocardiogram (ECG) gating. However, there are concerns regarding the influence of motion artifacts when measuring EAT volume on non-ECG-gated plain chest computed tomography (CT) images. Few studies have evaluated the EAT volume using non-ECG gating. This study aimed to validate the accuracy of EAT quantification using non-ECG-gated chest CT imaging. METHODS We included 100 patients (64 males, 36 females) who underwent simultaneous coronary artery calcification score imaging (ECG gated) and plain chest CT imaging (non-ECG gated). Images taken using non-ECG gating were reconstructed using the same field of view and slice thickness as those obtained with ECG gating. The EAT capacity of each image was measured and compared. An AZE Virtual Place (Canon) was used for the measurements. The Mann-Whitney U test and intraclass correlation coefficient were used for statistical analyses. P values <0.05 were considered statistically significant. Concordance was evaluated using Bland-Altman analysis. RESULTS The mean EAT volume measured by ECG-gated imaging was 156.5 ± 66.9 mL and 155.4 ± 67.9 mL by non-ECG-gated imaging, with no significant difference between the two groups ( P = 0.86). Furthermore, the EAT volumes measured using ECG-gated and non-ECG-gated imaging showed a strong correlation ( r = 0.95, P < 0.05). Bland-Altman analysis revealed that the mean error of the EAT volume (non-ECG-gated imaging - ECG-gated imaging) was -1.02 ± 2.95 mL (95% confidence interval, -6.49 to 4.76). CONCLUSIONS The EAT volume obtained using non-ECG-gated imaging was equivalent to that obtained using ECG-gated imaging.
Collapse
Affiliation(s)
- Tomio Mikami
- From the Department of Radiology, Ichiyokai Harada Hospital
| | | | - Kenji Mizuno
- From the Department of Radiology, Ichiyokai Harada Hospital
| | | |
Collapse
|
2
|
Zhang SJ, Wang SW, Liu SY, Li P, Huang DL, Zeng XX, Lan T, Ruan YP, Shi HJ, Zhang X. Epicardial adipose tissue: a new link between type 2 diabetes and heart failure-a comprehensive review. Heart Fail Rev 2024:10.1007/s10741-024-10478-8. [PMID: 39730926 DOI: 10.1007/s10741-024-10478-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Diabetic cardiomyopathy is a unique cardiomyopathy that is common in diabetic patients, and it is also a diabetic complication for which no effective treatment is currently available. Moreover, relevant studies have revealed that a link exists between type 2 diabetes and heart failure and that abnormal thickening of EAT is inextricably linked to the development of diabetic heart failure. Numerous clinical studies have demonstrated that EAT is implicated in the pathophysiologic process of diabetic myocardial disease. In this overview, we will introduce the physiology, pathophysiology of the disease and potential therapeutic strategies, knowledge gaps, and future directions of the role of epicardial adipose tissue in type 2 diabetes mellitus and heart failure to promote the development of novel therapeutic approaches to improve the prognosis of patients with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Si-Jia Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Si-Wei Wang
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Laboratory Animal Resources Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Shi-Yu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Ping Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - De-Lian Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Tian Lan
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Laboratory Animal Resources Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hai-Jiao Shi
- The Third Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Liaoning, 116600, China.
| | - Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China.
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
3
|
Museedi AS, Samson R, Le Jemtel TH. Menopause, epicardial adiposity and preserved ejection fraction heart failure. Int J Cardiol 2024; 415:132478. [PMID: 39179034 DOI: 10.1016/j.ijcard.2024.132478] [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: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Postmenopausal women are overrepresented in the preserved ejection heart failure population. Expansion of visceral and epicardial adipose tissue during the menopause transition leads to local and low-grade systemic inflammation that in turn contributes to left ventricular concentric remodeling, diastolic dysfunction and the development and progression of preserved ejection fraction. In contrast to visceral adipose tissue imaging, epicardial adipose tissue can be inexpensively imaged on low radiation coronary calcium score computerized tomography examination. The menopause transition provides a unique time frame to evaluate the contribution of epicardial adipose tissue expansion to the pathogenesis of preserved ejection heart failure.
Collapse
Affiliation(s)
- Abdulrahman S Museedi
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States of America
| | - Rohan Samson
- Advanced Heart Failure Therapies Program, University of Louisville Health-Jewish Hospital, 201 Abraham Flexner Way, Suite 1001, Louisville, KY 40202, United States of America
| | - Thierry H Le Jemtel
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States of America.
| |
Collapse
|
4
|
Xie L, Xiao H, Zhao M, Xu L, Tang S, Qiu Y. Screening of CAD-related secretory genes associated with type II diabetes based on comprehensive bioinformatics analysis and machine learning. BMC Cardiovasc Disord 2024; 24:620. [PMID: 39501130 PMCID: PMC11536945 DOI: 10.1186/s12872-024-04266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/15/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Type II diabetes mellitus (T2DM) is strongly linked with a heightened risk of coronary artery disease (CAD). Exploring biological targets common to T2DM and CAD is essential for CAD intervention strategies. METHODS RNA transcriptome data from CAD and T2DM patients and single-cell transcriptional data from myocardial tissue of CAD patients were used for bioinformatics analysis. Differential analysis and Weighted Gene Co-expression Network Analysis (WGCNA) were conducted to identify hub genes associated with the CAD Index (CADi) in these cells. We then intersected these genes with differentially expressed genes in the T2DM dataset to validate the key gene FGF7. Additional analyses included immune analysis, drug sensitivity, competing endogenous RNA (ceRNA) networks, and smooth muscle cell -related functional analysis. RESULTS An abnormally high proportion of smooth muscle cells was observed in CAD tissues compared to normal cardiomyocytes. The gene FGF7, which encodes the keratinocyte growth factor 7 protein, showed increased expression in both CAD and T2DM and was significantly positively correlated with the CADi (correlation = 0.24, p < 0.05). FGF7 expression was inversely correlated with CD4+ and CD8+ T-cell immune infiltration and correlated with the cardiovascular drugs. Overexpression of FGF7 in CAD samples enhanced interactions with mononuclear macrophages and influenced the metabolism of alanine, glutamate, nicotinamide, and retinol. We also identified that hsa-miR-15a-5p, hsa-miR-373-3p, hsa-miR-20a-5p, and hsa-miR-372-3p could regulate FGF7 expression. CONCLUSION FGF7 serves as a critical shared biological target for T2DM and CAD, playing a significant role in CAD progression with potential therapeutic implications.
Collapse
Affiliation(s)
- Li Xie
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Han Xiao
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Maoyu Zhao
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Li Xu
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Si Tang
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Youzhu Qiu
- Department of Cardiology, The Second Affiliated Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
5
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [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: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
6
|
Pyman E, Ernault AC, Kumar Patel KH, Ng FS, Coronel R. Subepicardial adipose tissue as a modulator of arrhythmias. Heart Rhythm 2024:S1547-5271(24)03449-0. [PMID: 39427690 DOI: 10.1016/j.hrthm.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Affiliation(s)
| | - Auriane C Ernault
- Amsterdam University Medical Centers, location AMC, Amsterdam, The Netherlands
| | | | - Fu Siong Ng
- Imperial College London, London, United Kingdom
| | - Ruben Coronel
- Amsterdam University Medical Centers, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Vyas V, Sandhar B, Keane JM, Wood EG, Blythe H, Jones A, Shahaj E, Fanti S, Williams J, Metic N, Efremova M, Ng HL, Nageswaran G, Byrne S, Feldhahn N, Marelli-Berg F, Chain B, Tinker A, Finlay MC, Longhi MP. Tissue-resident memory T cells in epicardial adipose tissue comprise transcriptionally distinct subsets that are modulated in atrial fibrillation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1067-1082. [PMID: 39271815 PMCID: PMC11399095 DOI: 10.1038/s44161-024-00532-x] [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: 09/20/2023] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia and carries an increased risk of stroke and heart failure. Here we investigated how the immune infiltrate of human epicardial adipose tissue (EAT), which directly overlies the myocardium, contributes to AF. Flow cytometry analysis revealed an enrichment of tissue-resident memory T (TRM) cells in patients with AF. Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell T cell receptor (TCR) sequencing identified two transcriptionally distinct CD8+ TRM cells that are modulated in AF. Spatial transcriptomic analysis of EAT and atrial tissue identified the border region between the tissues to be a region of intense inflammatory and fibrotic activity, and the addition of TRM populations to atrial cardiomyocytes demonstrated their ability to differentially alter calcium flux as well as activate inflammatory and apoptotic signaling pathways. This study identified EAT as a reservoir of TRM cells that can directly modulate vulnerability to cardiac arrhythmia.
Collapse
Affiliation(s)
- Vishal Vyas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Balraj Sandhar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack M Keane
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hazel Blythe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aled Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eriomina Shahaj
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack Williams
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nasrine Metic
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Mirjana Efremova
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Han Leng Ng
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Gayathri Nageswaran
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Suzanne Byrne
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Niklas Feldhahn
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Benny Chain
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Andrew Tinker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Malcolm C Finlay
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
8
|
Ruggeri F, Papadopoulou V, Kallikourdis M. Epicardial adipose tissue resident memory T cells in atrial fibrillation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1026-1027. [PMID: 39271814 DOI: 10.1038/s44161-024-00528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Affiliation(s)
- Federica Ruggeri
- Humanitas University, Milan, Italy
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Vasiliki Papadopoulou
- Humanitas University, Milan, Italy
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marinos Kallikourdis
- Humanitas University, Milan, Italy.
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
9
|
Shaaban A, Scott SS, Greenlee AN, Binda N, Noor A, Webb A, Guo S, Purdy N, Pennza N, Habib A, Mohammad SJ, Smith SA. Atrial fibrillation in cancer, anticancer therapies, and underlying mechanisms. J Mol Cell Cardiol 2024; 194:118-132. [PMID: 38897563 PMCID: PMC11500699 DOI: 10.1016/j.yjmcc.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Atrial fibrillation (AF) is a common arrhythmic complication in cancer patients and can be exacerbated by traditional cytotoxic and targeted anticancer therapies. Increased incidence of AF in cancer patients is independent of confounding factors, including preexisting myocardial arrhythmogenic substrates, type of cancer, or cancer stage. Mechanistically, AF is characterized by fast unsynchronized atrial contractions with rapid ventricular response, which impairs ventricular filling and results in various symptoms such as fatigue, chest pain, and shortness of breath. Due to increased blood stasis, a consequence of both cancer and AF, concern for stroke increases in this patient population. To compound matters, cardiotoxic anticancer therapies themselves promote AF; thereby exacerbating AF morbidity and mortality in cancer patients. In this review, we examine the relationship between AF, cancer, and cardiotoxic anticancer therapies with a focus on the shared molecular and electrophysiological mechanisms linking these disease processes. We also explore the potential role of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in the management of anticancer-therapy-induced AF.
Collapse
Affiliation(s)
- Adnan Shaaban
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Shane S Scott
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashley N Greenlee
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nkongho Binda
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Ali Noor
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Averie Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shuliang Guo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Najhee Purdy
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nicholas Pennza
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Alma Habib
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA
| | - Somayya J Mohammad
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sakima A Smith
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
10
|
Goette A, Corradi D, Dobrev D, Aguinaga L, Cabrera JA, Chugh SS, de Groot JR, Soulat-Dufour L, Fenelon G, Hatem SN, Jalife J, Lin YJ, Lip GYH, Marcus GM, Murray KT, Pak HN, Schotten U, Takahashi N, Yamaguchi T, Zoghbi WA, Nattel S. Atrial cardiomyopathy revisited-evolution of a concept: a clinical consensus statement of the European Heart Rhythm Association (EHRA) of the ESC, the Heart Rhythm Society (HRS), the Asian Pacific Heart Rhythm Society (APHRS), and the Latin American Heart Rhythm Society (LAHRS). Europace 2024; 26:euae204. [PMID: 39077825 PMCID: PMC11431804 DOI: 10.1093/europace/euae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024] Open
Abstract
AIMS The concept of "atrial cardiomyopathy" (AtCM) had been percolating through the literature since its first mention in 1972. Since then, publications using the term were sporadic until the decision was made to convene an expert working group with representation from four multinational arrhythmia organizations to prepare a consensus document on atrial cardiomyopathy in 2016 (EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication). Subsequently, publications on AtCM have increased progressively. METHODS AND RESULTS The present consensus document elaborates the 2016 AtCM document further to implement a simple AtCM staging system (AtCM stages 1-3) by integrating biomarkers, atrial geometry, and electrophysiological changes. However, the proposed AtCM staging needs clinical validation. Importantly, it is clearly stated that the presence of AtCM might serve as a substrate for the development of atrial fibrillation (AF) and AF may accelerates AtCM substantially, but AtCM per se needs to be viewed as a separate entity. CONCLUSION Thus, the present document serves as a clinical consensus statement of the European Heart Rhythm Association (EHRA) of the ESC, the Heart Rhythm Society (HRS), the Asian Pacific Heart Rhythm Society (APHRS), and the Latin American Heart Rhythm Society (LAHRS) to contribute to the evolution of the AtCM concept.
Collapse
Affiliation(s)
- Andreas Goette
- Department of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Am Busdorf 2, 33098 Paderborn, Germany
- MAESTRIA Consortium at AFNET, Münster, Germany
- Otto-von-Guericke University, Medical Faculty, Magdeburg, Germany
| | - Domenico Corradi
- Department of Medicine and Surgery, Unit of Pathology; Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy
| | - Dobromir Dobrev
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
- Montréal Heart Institute, Université de Montréal, 5000 Belanger St. E., Montréal, Québec H1T1C8, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Luis Aguinaga
- Director Centro Integral de Arritmias Tucumán, Presidente Sociedad de Cardiología de Tucumàn, Ex-PRESIDENTE DE SOLAECE (LAHRS), Sociedad Latinoamericana de EstimulaciónCardíaca y Electrofisiología, Argentina
| | - Jose-Angel Cabrera
- Hospital Universitario QuirónSalud, Madrid, Spain
- European University of Madrid, Madrid, Spain
| | - Sumeet S Chugh
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - Joris R de Groot
- Department of Cardiology; Cardiovascular Sciences, Heart Failure and Arrhythmias, University of Amsterdam, Amsterdam, The Netherlands
| | - Laurie Soulat-Dufour
- Department of Cardiology, Saint Antoine and Tenon Hospital, AP-HP, Unité INSERM UMRS 1166 Unité de recherche sur les maladies cardiovasculaires et métaboliques, Institut Hospitalo-Universitaire, Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | | | - Stephane N Hatem
- Department of Cardiology, Assistance Publique—Hôpitaux de Paris, Pitié-Salpêtrière Hospital; Sorbonne University; INSERM UMR_S1166; Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Jose Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Yenn-Jiang Lin
- Cardiovascular Center, Taipei Veterans General Hospital, and Faculty of Medicine National Yang-Ming University Taipei, Taiwan
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Gregory M Marcus
- Electrophysiology Section, Division of Cardiology, University of California, San Francisco, USA
| | - Katherine T Murray
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Ulrich Schotten
- MAESTRIA Consortium at AFNET, Münster, Germany
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University and Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University and Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Japan
| | - Takanori Yamaguchi
- Department of Cardiovascular Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - William A Zoghbi
- Department of Cardiology, Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, USA
| | - Stanley Nattel
- McGill University, 3655 Promenade Sir-William-Osler, Montréal, Québec H3G1Y6, Canada
- West German Heart and Vascular Center, Institute of Pharmacology, University Duisburg, Essen, Germany
| |
Collapse
|
11
|
Aburel OM, Brăescu L, Buriman DG, Merce AP, Bînă AM, Borza C, Mornoș C, Sturza A, Muntean DM. Methylene blue reduces monoamine oxidase expression and oxidative stress in human cardiovascular adipose tissue. Mol Cell Biochem 2024:10.1007/s11010-024-05092-z. [PMID: 39167271 DOI: 10.1007/s11010-024-05092-z] [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: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Cardiovascular diseases represent the major cause of morbidity mainly due to chronic heart failure. Epicardial (EAT) and perivascular adipose tissues (PVAT) are considered major contributors to the pathogenesis of cardiometabolic pathologies. Monoamine oxidases (MAOs) are mitochondrial enzymes recognized as sources of reactive oxygen species (ROS) in cardiometabolic pathologies. Methylene blue (MB) is one of the oldest protective agents, yet no data are available about its effects on adipose tissue. The present pilot study was aimed at assessing the effects of MB: (i) on MAO expression and (ii) oxidative stress in EAT and PVAT harvested from patients with heart failure subjected to cardiac surgery (n = 25). Adipose tissue samples were incubated with MB (0.1 µM/24 h) and used for the assessment of MAO gene and protein expression (qPCS and immune fluorescence) and ROS production (confocal microscopy and spectrophotometry). The human cardiovascular adipose tissues contain both MAO isoforms, predominantly MAO-A. Incubation with MB reduced MAOs expression and oxidative stress; co-incubation with serotonin, the MAO-A substrate, further augmented ROS generation, an effect partially reversed by MB. In conclusion, MAO-A is the major isoform expressed in EAT and PVAT and contribute to local oxidative stress; both effects can be mitigated by methylene blue.
Collapse
Affiliation(s)
- Oana-Maria Aburel
- Chair of Pathophysiology, Department III, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| | - Laurențiu Brăescu
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Doctoral School Medicine-Pharmacy, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Department VI, Clinic of Cardiovascular Surgery, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Institute for Cardiovascular Diseases, G. Adam Str. No.13A, 300310, Timișoara, Romania
| | - Darius G Buriman
- Chair of Pathophysiology, Department III, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Doctoral School Medicine-Pharmacy, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| | - Adrian P Merce
- Institute for Cardiovascular Diseases, G. Adam Str. No.13A, 300310, Timișoara, Romania
| | - Anca M Bînă
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| | - Claudia Borza
- Chair of Pathophysiology, Department III, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| | - Cristian Mornoș
- Institute for Cardiovascular Diseases, G. Adam Str. No.13A, 300310, Timișoara, Romania
- Department VI, 2nd Clinic of Cardiology, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| | - Adrian Sturza
- Chair of Pathophysiology, Department III, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania.
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania.
- Timișoara County Hospital, 156 L. Rebreanu Str, 300723, Timişoara, Romania.
| | - Danina M Muntean
- Chair of Pathophysiology, Department III, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
- Centre for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041, Timişoara, Romania
| |
Collapse
|
12
|
Li TL, Zhu NN, Yin Z, Sun J, Guo JP, Yuan HT, Shi XM, Guo HY, Li SX, Shan ZL. Transcriptomic analysis of epicardial adipose tissue reveals the potential crosstalk genes and immune relationship between type 2 diabetes mellitus and atrial fibrillation. Gene 2024; 920:148528. [PMID: 38703871 DOI: 10.1016/j.gene.2024.148528] [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: 12/02/2023] [Revised: 03/27/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The complex relationship between atrial fibrillation (AF) and type 2 diabetes mellitus (T2DM) suggests a potential role for epicardial adipose tissue (EAT) that requires further investigation. This study employs bioinformatics and experimental approaches to clarify EAT's role in linking T2DM and AF, aiming to unravel the biological mechanisms involved. METHOD Bioinformatics analysis initially identified common differentially expressed genes (DEGs) in EAT from T2DM and AF datasets. Pathway enrichment and network analyses were then performed to determine the biological significance and network connections of these DEGs. Hub genes were identified through six CytoHubba algorithms and subsequently validated biologically, with further in-depth analyses confirming their roles and interactions. Experimentally, db/db mice were utilized to establish a T2DM model. AF induction was executed via programmed transesophageal electrical stimulation and burst pacing, focusing on comparing the incidence and duration of AF. Frozen sections and Hematoxylin and Eosin (H&E) staining illuminated the structures of the heart and EAT. Moreover, quantitative PCR (qPCR) measured the expression of hub genes. RESULTS The study identified 106 DEGs in EAT from T2DM and AF datasets, underscoring significant pathways in energy metabolism and immune regulation. Three hub genes, CEBPZ, PAK1IP1, and BCCIP, emerged as pivotal in this context. In db/db mice, a marked predisposition towards AF induction and extended duration was observed, with HE staining verifying the presence of EAT. Additionally, qPCR validated significant changes in hub genes expression in db/db mice EAT. In-depth analysis identified 299 miRNAs and 33 TFs as potential regulators, notably GRHL1 and MYC. GeneMANIA analysis highlighted the hub genes' critical roles in stress responses and leukocyte differentiation, while immune profile correlations highlighted their impact on mast cells and neutrophils, emphasizing the genes' significant influence on immune regulation within the context of T2DM and AF. CONCLUSION This investigation reveals the molecular links between T2DM and AF with a focus on EAT. Targeting these pathways, especially EAT-related ones, may enable personalized treatments and improved outcomes.
Collapse
Affiliation(s)
- Tian-Lun Li
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Na-Na Zhu
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao Yin
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiao Sun
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian-Pin Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Tao Yuan
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiang-Min Shi
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Yang Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shi-Xing Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao-Liang Shan
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
13
|
Aromolaran KA, Corbin A, Aromolaran AS. Obesity Arrhythmias: Role of IL-6 Trans-Signaling. Int J Mol Sci 2024; 25:8407. [PMID: 39125976 PMCID: PMC11313575 DOI: 10.3390/ijms25158407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease that is rapidly increasing in prevalence and affects more than 600 million adults worldwide, and this figure is estimated to increase by at least double by 2030. In the United States, more than one-third of the adult population is either overweight or obese. The global obesity epidemic is a major risk factor for the development of life-threatening arrhythmias occurring in patients with long QT, particularly in conditions where multiple heart-rate-corrected QT-interval-prolonging mechanisms are simultaneously present. In obesity, excess dietary fat in adipose tissue stimulates the release of immunomodulatory cytokines such as interleukin (IL)-6, leading to a state of chronic inflammation in patients. Over the last decade, increasing evidence has been found to support IL-6 signaling as a powerful predictor of the severity of heart diseases and increased risk for ventricular arrhythmias. IL-6's pro-inflammatory effects are mediated via trans-signaling and may represent a novel arrhythmogenic risk factor in obese hearts. The first selective inhibitor of IL-6 trans-signaling, olamkicept, has shown encouraging results in phase II clinical studies for inflammatory bowel disease. Nevertheless, the connection between IL-6 trans-signaling and obesity-linked ventricular arrhythmias remains unexplored. Therefore, understanding how IL-6 trans-signaling elicits a cellular pro-arrhythmic phenotype and its use as an anti-arrhythmic target in a model of obesity remain unmet clinical needs.
Collapse
Affiliation(s)
- Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
| | - Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
14
|
Corbin A, Aromolaran KA, Aromolaran AS. STAT4 Mediates IL-6 Trans-Signaling Arrhythmias in High Fat Diet Guinea Pig Heart. Int J Mol Sci 2024; 25:7813. [PMID: 39063055 PMCID: PMC11277091 DOI: 10.3390/ijms25147813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is a major risk factor for the development of life-threatening malignant ventricular tachyarrhythmias (VT) and sudden cardiac death (SCD). Risks may be highest for patients with high levels of the proinflammatory cytokine interleukin (IL)-6. We used our guinea pig model of high-fat diet (HFD)-induced arrhythmias that exhibit a heightened proinflammatory-like pathology, which is also observed in human obesity arrhythmias, as well as immunofluorescence and confocal microscopy approaches to evaluate the pathological IL-6 trans-signaling function and explore the underlying mechanisms. Using blind-stick and electrocardiogram (ECG) techniques, we tested the hypothesis that heightened IL-6 trans-signaling would exhibit increased ventricular arrhythmia/SCD incidence and underlying arrhythmia substrates. Remarkably, compared to low-fat diet (LFD)-fed controls, HFD promoted phosphorylation of the IL-6 signal transducer and activator of transcription 4 (STAT4), leading to its activation and enhanced nuclear translocation of pSTAT4/STAT4 compared to LFD controls and pSTAT3/STAT3 nuclear expression. Overactivation of IL-6 trans-signaling in guinea pigs prolonged the QT interval, which resulted in greater susceptibility to arrhythmias/SCD with isoproterenol challenge, as also observed with the downstream Janus kinase (JAK) 2 activator. These findings may have potentially profound implications for more effective arrhythmia therapy in the vulnerable obese patient population.
Collapse
Affiliation(s)
- Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
15
|
Doukbi E, Ancel P, Dutour A, Soghomonian A, Ahmed S, Castejon V, Piperoglou C, Gariboldi V, Lenoir M, Lechevallier E, Gondran-Tellier B, Boissier R, Ebbo M, Vély F, Gaborit B. Human epicardial fat has a beige profile and contains higher type 2 innate lymphoid cells than subcutaneous fat. Obesity (Silver Spring) 2024; 32:1302-1314. [PMID: 38747118 DOI: 10.1002/oby.24023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Epicardial adipose tissue (EAT) is a visceral fat that has been associated with coronary artery disease and atrial fibrillation. Previous work has revealed that EAT exhibits beige features. METHODS First, a new pan-genomic microarray analysis was performed on previously collected paired human EAT and thoracic subcutaneous AT (thSAT) from the EPICAR study (n = 31) to decipher a specific immune signature and its link with browning genes. Then, adaptive (T and B cells) and innate lymphoid cell (ILC1, ILC2, and ILC3) immunophenotyping assay panels, including CD127, CD117, and prostaglandin D2 receptor 2, were performed on prospectively collected paired human multiorgan donors (n = 18; INTERFACE study). RESULTS In the EPICAR study, a positive correlation between the T helper cell subtype Th2 immune pathway and browning genes was found in EAT versus thSAT (r = 0.82; p < 0.0001). In the INTERFACE study, this correlation was also observed (r = 0.31; p = 0.017), and a preponderance of CD4+T cells, CD8+T cells, and a few B cells was observed in all ATs (p < 0.0001). An increase in ILCs was observed in visceral AT (VAT) (i.e., EAT + VAT; 30 ± 5 ILCs per gram of AT) compared with subcutaneous counterparts (i.e., thSAT + abdominal SAT; 8 ± 2 ILCs per gram of AT; p = 0.001), with ILC1 being the most frequent (ILC1 > ILC3 > ILC2). Numbers of ILCs per gram of AT correlated with several Th2 or browning genes (IL-13, TNF receptor superfamily member 9 [TNFRSF9], and alkaline phosphatase, biomineralization associated [ALPL]). Interestingly, a specific increase in EAT-ILC2 compared with other ATs was observed, including a significant proportion expressing CD69 and/or CD25 activation markers (97.9% ± 1.2%; p < 0.0001). Finally, more natural killer cells were observed in EAT + VAT than in thSAT + abdominal SAT (p = 0.01). Exclusion of patients with coronary artery disease in the EPICAR and INTERFACE studies did not modify the main findings. Gene expression phenotyping confirmed specific upregulation of Th2 pathway and browning genes (IL-33 and uncoupling protein 1 [UCP-1]) in EAT. CONCLUSIONS This is the first study, to our knowledge, to provide a comparison between innate and adaptive lymphoid cells in human EAT. Further studies are ongoing to decipher whether these cells could be involved in EAT beiging.
Collapse
Affiliation(s)
- Elisa Doukbi
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
| | - Patricia Ancel
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
| | - Anne Dutour
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endocrinology-Nutrition-Diabetes-Obesity, Public Assistance Marseille Hospitals, Marseille, France
| | - Astrid Soghomonian
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endocrinology-Nutrition-Diabetes-Obesity, Public Assistance Marseille Hospitals, Marseille, France
| | - Shaista Ahmed
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
- Heart Repair and Regeneration Laboratory, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Victoria Castejon
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
| | - Christelle Piperoglou
- Aix-Marseille University, CNRS, National Institute for Health and Medical Research (INSERM), Marseille-Luminy Immunology Center (CIML), Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille Immunopole, Marseille, France
| | - Vlad Gariboldi
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
- Department of Cardiac Surgery, Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille, France
| | - Marien Lenoir
- Division of Paediatric Cardiac Surgery, Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille, France
| | - Eric Lechevallier
- Department of Urology Surgery, Conception Hospital, Public Assistance Marseille Hospitals, Marseille, France
| | - Bastien Gondran-Tellier
- Department of Urology Surgery, Conception Hospital, Public Assistance Marseille Hospitals, Marseille, France
| | - Romain Boissier
- Department of Urology Surgery, Conception Hospital, Public Assistance Marseille Hospitals, Marseille, France
| | - Mikael Ebbo
- Aix-Marseille University, CNRS, National Institute for Health and Medical Research (INSERM), Marseille-Luminy Immunology Center (CIML), Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille Immunopole, Marseille, France
- Internal Medicine Department, Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille, France
| | - Frédéric Vély
- Aix-Marseille University, CNRS, National Institute for Health and Medical Research (INSERM), Marseille-Luminy Immunology Center (CIML), Marseille University Hospital Timone, Public Assistance Marseille Hospitals, Marseille Immunopole, Marseille, France
| | - Bénédicte Gaborit
- Aix-Marseille University, National Institute for Health and Medical Research (INSERM), National Research Institute for Agriculture, Food and the Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endocrinology-Nutrition-Diabetes-Obesity, Public Assistance Marseille Hospitals, Marseille, France
| |
Collapse
|
16
|
Doukbi E, Ancel P, Dutour A, Soghomonian A, Ahmed S, Castejon V, Piperoglou C, Gariboldi V, Lenoir M, Lechevallier E, Gondran-Tellier B, Boissier R, Ebbo M, Vély F, Gaborit B. Human epicardial adipose tissue contains innate and adaptive lymphoid cells and a higher proportion of innate type 2 lymphoid cells compared to other adipose tissues. ANNALES D'ENDOCRINOLOGIE 2024; 85:226-230. [PMID: 38871498 DOI: 10.1016/j.ando.2024.05.009] [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: 06/15/2024]
Abstract
IMPORTANCE Epicardial adipose tissue (EAT) is a biologically active organ surrounding myocardium and coronary arteries that has been associated with coronary artery disease (CAD) and atrial fibrillation. Previous work has shown that EAT exhibits beige features. OBJECTIVE Our objective was to determine whether the stromal vascular fraction of the human EAT contains innate or adaptive lymphoid cells compared to thoracic subcutaneous (thSAT), visceral abdominal (VAT) and subcutaneous abdominal (abSAT). PARTICIPANTS New pangenomic microarray analysis was performed on previous transcriptomic dataset using significance analysis of microarray and ingenuity pathway analysis (n=41) to identify specific immune signature and its link with browning genes. EAT, thSAT, VAT and abSAT samples from explanted patients with severe cardiomyopathies and multi-organ donor patients (n=17) were used for flow cytometry (FC) immunophenotyping assay. Patients were on average 55±16 years-old; 47% had hypertension and 6% CAD. Phenotypic adaptive and innate immune profiles were performed using a TBNK panel and a specific ILC1-2-3 panel including CD127, CD117, CRTH2 (CD294) and activation markers such as CD25 and CD69. RESULTS Transcriptomic analysis showed a significant positive correlation between the TH2 immune pathway (IL-4, IL-5, IL-13, IL-25, IL-33) and browning genes (UCP-1, PRDM16, TMEM26, CITED1, TBX1) in EAT versus thSAT (R=0.82, P<0.0001). Regarding adaptive immune cells, a preponderance of CD8T cells, a contingent of CD4T cells, and a few B cells were observed in all ATs (P<0.0001). In innate lymphoid cells (ILCs), an increase was observed in visceral ATs (i.e. EAT; VAT 35±8ILCs/g of tissue) compared to their subcutaneous counterpart (i.e. thSAT+abSAT: 8±3 ILCs/g of AT, P=0.002), with a difference in the proportion of the 3 subtypes of ILCs (ILC1>ILC3>ILC2). In addition, we observed an increase in EAT-ILC2 compared to other ATs and almost all these EAT-ILC2 expressed CD69 and/or CD25 activation markers (99.75±0.16%; P<0.0001). We also observed more NKs in EAT and VAT (1520±71 cells/g of AT) than in SATs (562±17 cells/g of AT); P=0.01. CONCLUSION This is the first study to provide a comparison between innate and adaptive lymphoid cells in human epicardial versus abdominal or thoracic adipose tissues. Further studies are ongoing to decipher whether these cells could be involved in EAT beiging. TRIAL REGISTRATION CODECOH No. DC-2021-4518 The French agency of biomedicine PFS21-005.
Collapse
Affiliation(s)
- Elisa Doukbi
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France
| | - Patricia Ancel
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France
| | - Anne Dutour
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France; Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, AP-HM, 13005 Marseille, France
| | - Astrid Soghomonian
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France; Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, AP-HM, 13005 Marseille, France
| | - Shaista Ahmed
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France; Heart repair and regeneration laboratory, Department EMC, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| | | | - Christelle Piperoglou
- Aix Marseille University, CNRS, Inserm, CIML, Timone Hospital, AP-HM, Marseille Immunopôle, Marseille, France
| | - Vlad Gariboldi
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France; Department of Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | - Marien Lenoir
- Division of Paediatric Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | - Eric Lechevallier
- Department of Urology surgery, La Conception Hospital, AP-HM, Marseille, France
| | | | - Romain Boissier
- Department of Urology surgery, La Conception Hospital, AP-HM, Marseille, France
| | - Mikael Ebbo
- Aix Marseille University, CNRS, Inserm, CIML, Timone Hospital, AP-HM, Marseille Immunopôle, Marseille, France; Internal Medicine Department, Timone Hospital, AP-HM, Marseille, France
| | - Frédéric Vély
- Aix Marseille University, CNRS, Inserm, CIML, Timone Hospital, AP-HM, Marseille Immunopôle, Marseille, France
| | - Bénédicte Gaborit
- Aix Marseille University, Inserm, INRAE, C2VN, Marseille, France; Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, AP-HM, 13005 Marseille, France.
| |
Collapse
|
17
|
Milyukov VE, Bryukhanov VA, Nguyen CC. [Morphofunctional Analysis of the Role of Epicardial Adipose Tissue in the Formation of the Obesity Paradox in Chronic Heart Failure]. KARDIOLOGIIA 2024; 64:72-80. [PMID: 38597765 DOI: 10.18087/cardio.2024.3.n2469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 04/11/2024]
Abstract
Based on the available modern medical literature, the article summarizes data on the morpho-functional significance of epicardial adipose tissue (EAT) in health and heart failure, analyzes the likelihood and reliability of the formation of the obesity paradox, and also discusses its possible morpho-functional mechanisms. The authors reviewed and analyzed the consequences of the obesity paradox in the aspect of the normal EAT phenotype protectivity. The review proposed ways of further research in this direction aimed at a deep anatomical and physiological analysis and at determining the morpho-functional role of EAT in the adaptive mechanisms of myocardial trophic provision, which may be an important part of the pathogenetic connection between obesity and CHF and, therefore, can improve outcomes in such patients.
Collapse
Affiliation(s)
- V E Milyukov
- Pirogov Russian National Research Medical University
| | | | | |
Collapse
|
18
|
Morocho-Jaramillo PA, Kotlar-Goldaper I, Zakarauskas-Seth BI, Purfürst B, Filosa A, Sawamiphak S. The zebrafish heart harbors a thermogenic beige fat depot analog of human epicardial adipose tissue. Cell Rep 2024; 43:113955. [PMID: 38507414 DOI: 10.1016/j.celrep.2024.113955] [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: 04/14/2023] [Revised: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.
Collapse
Affiliation(s)
- Paul-Andres Morocho-Jaramillo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Ilan Kotlar-Goldaper
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bhakti I Zakarauskas-Seth
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bettina Purfürst
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| |
Collapse
|
19
|
Bialobroda J, Bouazizi K, Ponnaiah M, Kachenoura N, Charpentier E, Zarai M, Clement K, Andreelli F, Aron-Wisnewsky J, Hatem SN, Redheuil A. The epicardial adipose tissue confined in the atrioventricular groove can be used to assess atrial adipose tissue and atrial dysfunction in cardiac magnetic resonance imaging. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae057. [PMID: 39224099 PMCID: PMC11367945 DOI: 10.1093/ehjimp/qyae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/17/2024] [Indexed: 09/04/2024]
Abstract
Aims The growing interest in epicardial adipose tissue (EAT) as a biomarker of atrial fibrillation is limited by the difficulties in isolating EAT from other paracardial adipose tissues. We tested the feasibility and value of measuring the pure EAT contained in the atrioventricular groove (GEAT) using cardiovascular magnetic resonance (CMR) imaging in patients with distinct metabolic disorders. Methods and results CMR was performed on 100 patients from the MetaCardis cohort: obese (n = 18), metabolic syndrome (MSD) (n = 25), type-2 diabetes (T2D) (n = 42), and age- and gender-matched healthy controls (n = 15). GEAT volume measured from long-axis views was obtained in all patients with a strong correlation between GEAT and atrial EAT (r = 0.95; P < 0.0001). GEAT volume was higher in the three groups of patients with metabolic disorders and highest in the MSD group compared with controls. GEAT volume, as well as body mass and body fat, allowed obese, T2D, and MSD patients to be distinguished from controls. GEAT T1 relaxation and peak longitudinal left atrial (LA) strain in CMR were decreased in T2D patients. Logistic regression and random forest machine learning methods were used to create an algorithm combining GEAT volume, GEAT T1, and peak LA strain to identify T2D patients from other groups with an area under curve (AUC) of 0.81 (Se: 77%, Spe: 80%; 95% confidence interval 0.72-0.91, P < 0.0001). Conclusion Atrioventricular groove adipose tissue characteristics measured during routine CMR can be used as a proxy of atrial EAT and integrated in a multi-parametric CMR biomarker for early identification of atrial cardiomyopathy.
Collapse
Affiliation(s)
- Jonathan Bialobroda
- Institute of Cardiology, Foundation for Innovation in Cardiometabolism and Nutrition—ICAN, INSERM UMRS 1166, Sorbonne Université, AP-HP Pitié-Salpêtrière University Hospital, 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Khaoula Bouazizi
- Laboratoire d’Imagerie Biomédicale, CNRS, INSERM UMR 1146, Sorbonne Université, Paris, France
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Nadjia Kachenoura
- Laboratoire d’Imagerie Biomédicale, CNRS, INSERM UMR 1146, Sorbonne Université, Paris, France
| | - Etienne Charpentier
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Mohamed Zarai
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Karine Clement
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Nutrition, Sorbonne Université, Assistance Publique- Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Fabrizio Andreelli
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Diabetology, AP-HP Pitié-Salpêtrière University Hospital, Sorbonne Université, Paris, France
| | - Judith Aron-Wisnewsky
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Nutrition, Sorbonne Université, Assistance Publique- Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Stéphane N Hatem
- Institute of Cardiology, Foundation for Innovation in Cardiometabolism and Nutrition—ICAN, INSERM UMRS 1166, Sorbonne Université, AP-HP Pitié-Salpêtrière University Hospital, 47-83, Boulevard de l’Hôpital, 75013 Paris, France
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
20
|
Cinti F, Leccisotti L, Sorice GP, Capece U, D'Amario D, Lorusso M, Gugliandolo S, Morciano C, Guarneri A, Guzzardi MA, Mezza T, Capotosti A, Indovina L, Ferraro PM, Iozzo P, Crea F, Giordano A, Giaccari A. Dapagliflozin treatment is associated with a reduction of epicardial adipose tissue thickness and epicardial glucose uptake in human type 2 diabetes. Cardiovasc Diabetol 2023; 22:349. [PMID: 38115004 PMCID: PMC10731727 DOI: 10.1186/s12933-023-02091-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
OBJECTIVE We recently demonstrated that treatment with sodium-glucose cotransporter-2 inhibitors (SGLT-2i) leads to an increase in myocardial flow reserve in patients with type 2 diabetes (T2D) with stable coronary artery disease (CAD). The mechanism by which this occurs is, however, unclear. One of the risk factors for cardiovascular disease is inflammation of epicardial adipose tissue (EAT). Since the latter is often increased in type 2 diabetes patients, it could play a role in coronary microvascular dysfunction. It is also well known that SGLT-2i modify adipose tissue metabolism. We aimed to investigate the effects of the SGLT-2i dapagliflozin on metabolism and visceral and subcutaneous adipose tissue thickness in T2D patients with stable coronary artery disease and to verify whether these changes could explain observed changes in myocardial flow. METHODS We performed a single-center, prospective, randomized, double-blind, controlled clinical trial with 14 T2D patients randomized 1:1 to SGLT-2i dapagliflozin (10 mg daily) or placebo. The thickness of visceral (epicardial, mediastinal, perirenal) and subcutaneous adipose tissue and glucose uptake were assessed at baseline and 4 weeks after treatment initiation by 2-deoxy-2-[18F]fluoro-D-glucose Positron Emission Tomography/Computed Tomography during hyperinsulinemic euglycemic clamp. RESULTS The two groups were well-matched for baseline characteristics (age, diabetes duration, HbA1c, BMI, renal and heart function). Dapagliflozin treatment significantly reduced EAT thickness by 19% (p = 0.03). There was a significant 21.6% reduction in EAT glucose uptake during euglycemic hyperinsulinemic clamp in the dapagliflozin group compared with the placebo group (p = 0.014). There were no significant effects on adipose tissue thickness/metabolism in the other depots explored. CONCLUSIONS SGLT-2 inhibition selectively reduces EAT thickness and EAT glucose uptake in T2D patients, suggesting a reduction of EAT inflammation. This could explain the observed increase in myocardial flow reserve, providing new insights into SGLT-2i cardiovascular benefits.
Collapse
Affiliation(s)
- Francesca Cinti
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucia Leccisotti
- UOC di Medicina Nucleare, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Pio Sorice
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
- Sezione di Medicina Interna, Endocrinologia, Andrologia e Malattie Metaboliche, Dipartimento di Medicina di Precisione e Rigenerativa e Area Jonica - (DiMePRe-J), Università Degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Umberto Capece
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico D'Amario
- Dipartimento di Scienze Cardiovascolari, UOC Di Cardiologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
- Università del Piemonte Orientale , Dipartimento Medicina Translazionale, Novara, Italy
| | - Margherita Lorusso
- UOC di Medicina Nucleare, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Shawn Gugliandolo
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cassandra Morciano
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze Cliniche e Sperimentali, Medicina Interna - Università degli Studi di Brescia, Brescia, BS, Italy
| | - Andrea Guarneri
- UOC di Medicina Nucleare, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Angela Guzzardi
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Teresa Mezza
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
- Pancreas Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Amedeo Capotosti
- UOSD Fisica Medica e Radioprotezione, Dipartimento di Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Radioterapia Oncologica ed Ematologia, Rome, Italy
| | - Luca Indovina
- UOSD Fisica Medica e Radioprotezione, Dipartimento di Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Radioterapia Oncologica ed Ematologia, Rome, Italy
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Patricia Iozzo
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Filippo Crea
- Dipartimento di Scienze Cardiovascolari, UOC Di Cardiologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Giordano
- UOC di Medicina Nucleare, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Andrea Giaccari
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
21
|
Song Y, Tan Y, Deng M, Shan W, Zheng W, Zhang B, Cui J, Feng L, Shi L, Zhang M, Liu Y, Sun Y, Yi W. Epicardial adipose tissue, metabolic disorders, and cardiovascular diseases: recent advances classified by research methodologies. MedComm (Beijing) 2023; 4:e413. [PMID: 37881786 PMCID: PMC10594046 DOI: 10.1002/mco2.413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023] Open
Abstract
Epicardial adipose tissue (EAT) is located between the myocardium and visceral pericardium. The unique anatomy and physiology of the EAT determines its great potential in locally influencing adjacent tissues such as the myocardium and coronary arteries. Classified by research methodologies, this study reviews the latest research progress on the role of EAT in cardiovascular diseases (CVDs), particularly in patients with metabolic disorders. Studies based on imaging techniques demonstrated that increased EAT amount in patients with metabolic disorders is associated with higher risk of CVDs and increased mortality. Then, in-depth profiling studies indicate that remodeled EAT may serve as a local mediator of the deleterious effects of cardiometabolic conditions and plays a crucial role in CVDs. Further, in vitro coculture studies provided preliminary evidence that the paracrine effect of remodeled EAT on adjacent cardiomyocytes can promote the occurrence and progression of CVDs. Considering the important role of EAT in CVDs, targeting EAT might be a potential strategy to reduce cardiovascular risks. Several interventions have been proved effective in reducing EAT amount. Our review provides valuable insights of the relationship between EAT, metabolic disorders, and CVDs, as well as an overview of the methodological constructs of EAT-related studies.
Collapse
Affiliation(s)
- Yujie Song
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yanzhen Tan
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Meng Deng
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wenju Shan
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wenying Zheng
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Bing Zhang
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Jun Cui
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Lele Feng
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Lei Shi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Miao Zhang
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yingying Liu
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yang Sun
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wei Yi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
22
|
Isidoro CA, Deniset JF. Pericardial Immune Cells and Their Evolving Role in Cardiovascular Pathophysiology. Can J Cardiol 2023; 39:1078-1089. [PMID: 37270165 DOI: 10.1016/j.cjca.2023.05.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/05/2023] Open
Abstract
The pericardium plays several homeostatic roles to support and maintain everyday cardiac function. Recent advances in techniques and experimental models have allowed for further exploration into the cellular contents of the pericardium itself. Of particular interest are the various immune cell populations present in the space within the pericardial fluid and fat. In contrast to immune cells of the comparable pleura, peritoneum and heart, pericardial immune cells appear to be distinct in their function and phenotype. Specifically, recent work has suggested these cells play critical roles in an array of pathophysiological conditions including myocardial infarction, pericarditis, and post-cardiac surgery complications. In this review, we spotlight the pericardial immune cells currently identified in mice and humans, the pathophysiological role of these cells, and the clinical significance of the immunocardiology axis in cardiovascular health.
Collapse
Affiliation(s)
- Carmina Albertine Isidoro
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Justin F Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada; Department of Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
23
|
Nesti L, Pugliese NR, Chiriacò M, Trico D, Baldi S, Natali A. Epicardial adipose tissue thickness is associated with reduced peak oxygen consumption and systolic reserve in patients with type 2 diabetes and normal heart function. Diabetes Obes Metab 2023; 25:177-188. [PMID: 36066008 PMCID: PMC10087544 DOI: 10.1111/dom.14861] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 12/14/2022]
Abstract
AIM To investigate the impact of epicardial adipose tissue (EAT) thickness on cardiopulmonary performance in patients with type 2 diabetes (T2D) and normal heart function. MATERIALS AND METHODS We analysed EAT thickness in subjects with T2D and normal biventricular systo-diastolic functions undergoing a maximal cardiopulmonary exercise test combined with stress echocardiography, speckle tracking and pulmonary function assessment, as well as serum N-terminal pro B-type natriuretic peptide (NT-proBNP). RESULTS In the 72 subjects enrolled, those with EAT thickness above the median (> 5 mm) showed higher body fat mass, smaller indexed left ventricular dimensions and marginally reduced diastolic function variables at rest. Higher EAT thickness was associated with lower peak oxygen uptake (VO2peak 17.1 ± 3.6 vs. 21.0 ± 5.7 ml/min/kg, P = .001), reduced systolic reserve (ΔS' 4.6 ± 1.6 vs. 5.8 ± 2.5 m/s, P = .02) and higher natriuretic peptides (NT-proBNP 64 [29-165] vs. 31 [26-139] pg/ml, P = .04), as well as chronotropic insufficiency and impaired heart rate recovery. Ventilatory variables and peripheral oxygen extraction were not different between groups. EAT was independently associated with VO2peak and linearly and negatively correlated with peak heart rate, heart rate recovery, workload, VO2 at the anaerobic threshold and at peak, and cardiac power output, and was directly correlated with natriuretic peptides. CONCLUSION Higher EAT thickness in T2D is associated with worse cardiopulmonary performance and multiple traits of subclinical cardiac systolic dysfunction.
Collapse
Affiliation(s)
- Lorenzo Nesti
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Riccardo Pugliese
- Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Martina Chiriacò
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Domenico Trico
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Baldi
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Natali
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
24
|
Abstract
Diabetes is a long-term chronic disease, and cardiovascular disease is the leading cause of death. Diabetic cardiomyopathy (DCM), one of the cardiovascular complications of diabetes, has many uncertain factors. Epicardial fat, as the heart fat bank, functions as fatty tissue and is the heart's endocrine organ. The existence of diabetes affects the distribution of heart fat and promotes the secretion of adipokine. In different pathological conditions, it can promote the secretion of pro-inflammatory adipokine, reactive oxygen species, oxidative stress, and even autophagy, thus affecting cardiac function. In this paper, we will elaborate on the mechanism of epicardial fat in the pathogenesis of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Xueyuan Yang
- Chest Clinical College, Tianjin Medical University, Tianjin, China
| | - Chao Feng
- Tianjin Chest Hospital, Tianjin, China
| | | |
Collapse
|
25
|
Zhang XZ, Chen XL, Tang TT, Zhang S, Li QL, Xia N, Nie SF, Zhang M, Zhu ZF, Zhou ZH, Dong NG, Cheng X. T lymphocyte characteristics and immune repertoires in the epicardial adipose tissue of heart failure patients. Front Immunol 2023; 14:1126997. [PMID: 36960061 PMCID: PMC10027920 DOI: 10.3389/fimmu.2023.1126997] [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: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Background Epicardial adipose tissue (EAT) acts as an active immune organ and plays a critical role in the pathogenesis of heart failure (HF). However, the characteristics of immune cells in EAT of HF patients have rarely been elucidated. Methods To identify key immune cells in EAT, an integrated bioinformatics analysis was performed on public datasets. EAT samples with paired subcutaneous adipose tissue (SAT), heart, and peripheral blood samples from HF patients were collected in validation experiments. T cell receptor (TCR) repertoire was assessed by high-throughput sequencing. The phenotypic characteristics and key effector molecules of T lymphocytes in EAT were assessed by flow cytometry and histological staining. Results Compared with SAT, EAT was enriched for immune activation-related genes and T lymphocytes. Compared with EAT from the controls, activation of T lymphocytes was more pronounced in EAT from HF patients. T lymphocytes in EAT of HF patients were enriched by highly expanded clonotypes and had greater TCR clonotype sharing with cardiac tissue relative to SAT. Experiments confirmed the abundance of IFN-γ+ effector memory T lymphocytes (TEM) in EAT of HF patients. CCL5 and GZMK were confirmed to be associated with T lymphocytes in EAT of HF patients. Conclusion EAT of HF patients was characterized by pronounced immune activation of clonally expanded IFN-γ+ TEM and a generally higher degree of TCR clonotypes sharing with paired cardiac tissue.
Collapse
Affiliation(s)
- Xu-Zhe Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Li Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting-Ting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin-Lin Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Fang Nie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng-Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Hua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiang Cheng, ; Nian-Guo Dong, ; Zi-Hua Zhou,
| | - Nian-Guo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiang Cheng, ; Nian-Guo Dong, ; Zi-Hua Zhou,
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiang Cheng, ; Nian-Guo Dong, ; Zi-Hua Zhou,
| |
Collapse
|
26
|
AlZaim I, Eid AH, Abd-Elrahman KS, El-Yazbi AF. Adipose Tissue Mitochondrial Dysfunction and Cardiometabolic Diseases: On the Search for Novel Molecular Targets. Biochem Pharmacol 2022; 206:115337. [DOI: 10.1016/j.bcp.2022.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
27
|
Shuaishuai D, Jingyi L, Zhiqiang Z, Guanwei F. Sex differences and related estrogenic effects in heart failure with preserved ejection fraction. Heart Fail Rev 2022:10.1007/s10741-022-10274-2. [PMID: 36190606 DOI: 10.1007/s10741-022-10274-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2022] [Indexed: 11/04/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an essential subtype of heart failure accounting for 40% of the total. However, the related pathological mechanism and drug therapy research have been stagnant for a long time. The direct cause of this dilemma is the heterogeneity of HFpEF. And some researchers believe that there is no common pathway to reach the origin of HFpEF; others argue that there is an unidentified unified pathophysiological process hidden beneath the ice surface. Aside from the debate, a series of clinical studies have shown that hypertension and obesity play a fundamental role in the pathogenesis of HFpEF. These results imply that there may be two parallel pathological processes interweaved in one disease, manifested as multiple coexistent pathological phenomena, like a shadow. Meanwhile, the prevalence of HFpEF in women is higher than in men in any given age group, especially prominent in elderly patients. These pathological processes and epidemiological data reflect gender differences, reminding us to shift our attention to estrogen. This article will review the parallel pathogenesis of HFpEF, and also introduce sex differences and the potential effect of estrogen in this condition below.
Collapse
Affiliation(s)
- Deng Shuaishuai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Lin Jingyi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Zhao Zhiqiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China
| | - Fan Guanwei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China. .,National Clinical Research Center for Chinese Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
28
|
Yang H, Xiong B, Xiong T, Wang D, Yu W, Liu B, She Q. Identification of key genes and mechanisms of epicardial adipose tissue in patients with diabetes through bioinformatic analysis. Front Cardiovasc Med 2022; 9:927397. [PMID: 36158806 PMCID: PMC9500152 DOI: 10.3389/fcvm.2022.927397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIn recent years, peri-organ fat has emerged as a diagnostic and therapeutic target in metabolic diseases, including diabetes mellitus. Here, we performed a comprehensive analysis of epicardial adipose tissue (EAT) transcriptome expression differences between diabetic and non-diabetic participants and explored the possible mechanisms using various bioinformatic tools.MethodsRNA-seq datasets GSE108971 and GSE179455 for EAT between diabetic and non-diabetic patients were obtained from the public functional genomics database Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) were identified using the R package DESeq2, then Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were analyzed. Next, a PPI (protein–protein interaction) network was constructed, and hub genes were mined using STRING and Cytoscape. Additionally, CIBERSORT was used to analyze the immune cell infiltration, and key transcription factors were predicted based on ChEA3.ResultsBy comparing EAT samples between diabetic and non-diabetic patients, a total of 238 DEGs were identified, including 161 upregulated genes and 77 downregulated genes. A total of 10 genes (IL-1β, CD274, PDCD1, ITGAX, PRDM1, LAG3, TNFRSF18, CCL20, IL1RN, and SPP1) were selected as hub genes. GO and KEGG analysis showed that DEGs were mainly enriched in the inflammatory response and cytokine activity. Immune cell infiltration analysis indicated that macrophage M2 and T cells CD4 memory resting accounted for the largest proportion of these immune cells. CSRNP1, RELB, NFKB2, SNAI1, and FOSB were detected as potential transcription factors.ConclusionComprehensive bioinformatic analysis was used to compare the difference in EAT between diabetic and non-diabetic patients. Several hub genes, transcription factors, and immune cell infiltration were identified. Diabetic EAT is significantly different in the inflammatory response and cytokine activity. These findings may provide new targets for the diagnosis and treatment of diabetes, as well as reduce potential cardiovascular complications in diabetic patients through EAT modification.
Collapse
|
29
|
Decreased Epicardial CTRP3 mRNA Levels in Patients with Type 2 Diabetes Mellitus and Coronary Artery Disease Undergoing Elective Cardiac Surgery: A Possible Association with Coronary Atherosclerosis. Int J Mol Sci 2022; 23:ijms23179988. [PMID: 36077376 PMCID: PMC9456433 DOI: 10.3390/ijms23179988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: C1q TNF-related protein 3 (CTRP3) is an adipokine with anti-inflammatory and cardioprotective properties. In our study, we explored changes in serum CTRP3 and its gene expression in epicardial (EAT) and subcutaneous (SAT) adipose tissue in patients with and without coronary artery disease (CAD) and type 2 diabetes mellitus (T2DM) undergoing elective cardiac surgery. (2) Methods: SAT, EAT, and blood samples were collected at the start and end of surgery from 34 patients: (i) 11 without CAD or T2DM, (ii) 14 with CAD and without T2DM, and (iii) 9 with both CAD and T2DM. mRNA levels of CTRP3 were assessed by quantitative reverse transcription PCR. Circulating levels of CTRP3 and other factors were measured using ELISA and Luminex Multiplex commercial kits. (3) Results: Baseline plasma levels of TNF-α and IL6 did not differ among the groups and increased at the end of surgery. Baseline circulating levels of CTRP3 did not differ among the groups and decreased after surgery. In contrast, baseline CTRP3 mRNA levels in EAT were significantly decreased in CAD/T2DM group, while no differences were found for TNF-α and IL6 gene expression. (4) Conclusions: Our data suggest that decreased EAT mRNA levels of CTRP3 could contribute to higher risk of atherosclerosis in patients with CAD and T2DM.
Collapse
|
30
|
Li Y, Song S, Sun Y, Bao N, Yang B, Xu L. Segmentation and volume quantification of epicardial adipose tissue in computed tomography images. Med Phys 2022; 49:6477-6490. [PMID: 36047382 DOI: 10.1002/mp.15965] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Many cardiovascular diseases are closely related to the composition of epicardial adipose tissue (EAT). Accurate segmentation of EAT can provide a reliable reference for doctors to diagnose the disease. The distribution and composition of EAT often have significant individual differences, and the traditional segmentation methods are not effective. In recent years, deep learning method has been gradually introduced into EAT segmentation task. PURPOSE The existing EAT segmentation methods based on deep learning have a large amount of computation and the segmentation accuracy needs to be improved. Therefore, the purpose of this paper is to develop a lightweight EAT segmentation network, which can obtain higher segmentation accuracy with less computation and further alleviate the problem of false positive segmentation. METHODS Firstly, the obtained Computed Tomography (CT) was preprocessed. That is, the threshold range of EAT was determined to be (-190, -30) HU according to prior knowledge, and the non-adipose pixels were excluded by threshold segmentation to reduce the difficulty of training. Secondly, the image obtained after thresholding was input into the lightweight RDU-Net network to perform the training, validating, and testing process. RDU-Net uses a residual multi-scale dilated convolution block in order to extract a wider range of information without changing the current resolution. At the same time, the form of residual connection is adopted to avoid the problem of gradient expansion or gradient explosion caused by too deep network, which also makes the learning easier. In order to optimize the training process, this paper proposes PNDiceLoss, which takes both positive and negative pixels as learning targets, fully considers the class imbalance problem and appropriately highlights the status of positive pixels. RESULTS In this paper, 50 CCTA images were randomly selected from the hospital, and the commonly used Dice similarity coefficient (DSC), Jaccard similarity (JS), Accuracy (ACC), Specificity (SP), Precision (PC), and Pearson correlation coefficient are used as evaluation metrics. Bland-Altman analysis results show that the extracted EAT volume is consistent with the actual volume. Compared with the existing methods, the segmentation results show that the proposed method achieves better performance on these metrics, achieving the DSC of 0.9262. The number of false positive pixels has been reduced by more than half. Pearson correlation coefficient reached 0.992, and linear regression coefficient reached 0.977 when measuring the volume of EAT obtained. In order to verify the effectiveness of the proposed method, experiments are carried out in the cardiac fat database of VisualLab. On this database, the proposed method also achieved good results, and the DSC value reached 0.927 in the case of only 878 slices. CONCLUSIONS A new method to segment and quantify EAT is proposed. Comprehensive experiments show that compared with some classical segmentation algorithms, the proposed method has the advantages of shorter time-consuming, less memory required for operations, and higher segmentation accuracy. The code is available at https://github.com/lvanlee/EAT_Seg/tree/main/EAT_seg. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yifan Li
- School of Science, Northeastern University, Shenyang, 110819, China
| | - Shuni Song
- Guangdong Peizheng College, Guangzhou, 510830, China
| | - Yu Sun
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, 110169, China.,Department of Radiology, General Hospital of Northern Theater Command, Shenyang, 110016, China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, 110169, China
| | - Nan Bao
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, 110169, China.,Key Laboratory of Medical Image Computing, Ministry of Education, Shenyang, Liaoning, 110169, China
| | - Benqiang Yang
- Department of Radiology, General Hospital of Northern Theater Command, Shenyang, 110016, China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, 110169, China
| | - Lisheng Xu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, 110169, China.,Key Laboratory of Medical Image Computing, Ministry of Education, Shenyang, Liaoning, 110169, China.,Neusoft Research of Intelligent Healthcare Technology, Co. Ltd., Shenyang, Liaoning, 110169, China
| |
Collapse
|
31
|
Akhter N, Kochumon S, Hasan A, Wilson A, Nizam R, Al Madhoun A, Al-Rashed F, Arefanian H, Alzaid F, Sindhu S, Al-Mulla F, Ahmad R. IFN-γ and LPS Induce Synergistic Expression of CCL2 in Monocytic Cells via H3K27 Acetylation. J Inflamm Res 2022; 15:4291-4302. [PMID: 35923906 PMCID: PMC9343018 DOI: 10.2147/jir.s368352] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/02/2022] [Indexed: 11/23/2022] Open
Abstract
Background Overexpression of CCL2 (MCP-1) has been implicated in pathogenesis of metabolic conditions, such as obesity and T2D. However, the mechanisms leading to increased CCL2 expression in obesity are not fully understood. Since both IFN-γ and LPS levels are found to be elevated in obesity and shown to be involved in the regulation of metabolic inflammation and insulin resistance, we investigated whether these two agents could synergistically trigger the expression of CCL2 in obesity. Methods Monocytes (Human monocytic THP-1 cells) were stimulated with IFN-γ and LPS. CCL2 gene expression was determined by real-time RT-PCR. CCL2 protein was determined by ELISA. Signaling pathways were identified by using epigenetic inhibitors and STAT1 siRNA. Acetylation of H3K27 was analyzed by Western blotting. The acetylation level of histone H3K27 in the transcriptional initiation region of CCL2 gene was determined by ChIP-qPCR. Results Our results show that the co-incubation of THP-1 monocytes with IFN-γ and LPS significantly enhanced the expression of CCL2, compared to treatment with IFN-γ or LPS alone. Similar results were obtained using primary monocytes and macrophages. Interestingly, IFN-γ priming was found to be more effective than LPS priming in inducing synergistic expression of CCL2. Moreover, STAT1 deficiency significantly suppressed this synergy for CCL2 expression. Mechanistically, we showed that IFN-γ priming induced acetylation of lysine 27 on histone 3 (H3K27ac) in THP-1 cells. Chromatin immunoprecipitation (ChIP) assay followed by qRT-PCR revealed increased H3K27ac at the CCL2 promoter proximal region, resulting in stabilized gene expression. Furthermore, inhibition of histone acetylation with anacardic acid suppressed this synergistic response, whereas trichostatin A (TSA) could substitute IFN-γ in this synergy. Conclusion Our findings suggest that IFN-γ, in combination with LPS, has the potential to augment inflammation via the H3K27ac-mediated induction of CCL2 in monocytic cells in the setting of obesity.
Collapse
Affiliation(s)
- Nadeem Akhter
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Amal Hasan
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ajit Wilson
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics & Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Genetics & Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Hossein Arefanian
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fawaz Alzaid
- Genetics & Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
- Institut Necker Enfants Malades (INEM), French Institute of Health and Medical Research (INSERM), Immunity & Metabolism of Diabetes (IMMEDIAB), Université de Paris Cité, Paris, France
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- Correspondence: Rasheed Ahmad, Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait, Tel +965 2224 2999 Ext. 4311, Email
| |
Collapse
|
32
|
Karampetsou N, Alexopoulos L, Minia A, Pliaka V, Tsolakos N, Kontzoglou K, Perrea DN, Patapis P. Epicardial Adipose Tissue as an Independent Cardiometabolic Risk Factor for Coronary Artery Disease. Cureus 2022; 14:e25578. [PMID: 35784958 PMCID: PMC9248997 DOI: 10.7759/cureus.25578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 02/07/2023] Open
Abstract
During the last decades, visceral adiposity has been at the forefront of scientific research because of its complex role in the pathogenesis of cardiovascular diseases. Epicardial adipose tissue (EAT) is the visceral lipid compartment between the myocardium and the visceral pericardium. Due to their unobstructed anatomic vicinity, epicardial fat and myocardium are nourished by the same microcirculation. It is widely known that EAT serves as an energy lipid source and thermoregulator for the human heart. In addition to this, epicardial fat exerts highly protective effects since it releases a great variety of anti-inflammatory molecules to the adjacent cardiac muscle. Taking into account the unique properties of human EAT, it is undoubtedly a key factor in cardiac physiology since it facilitates complex heart functions. Under pathological circumstances, however, epicardial fat promotes coronary atherosclerosis in a variety of ways. Therefore, the accurate estimation of epicardial fat thickness and volume could be utilized as an early detecting method and future medication target for coronary artery disease (CAD) elimination. Throughout the years, several therapeutic approaches for dysfunctional human EAT have been proposed. A balanced healthy diet, aerobic and anaerobic physical activity, bariatric surgery, and pharmacological treatment with either traditional or novel antidiabetic and antilipidemic drugs are some of the established medical approaches. In the present article, we review the current knowledge regarding the anatomic and physiological characteristics of epicardial fat. In addition to this, we describe the pathogenic mechanisms which refer to the crosstalk between epicardial fat alteration and coronary arterial atherosclerosis development. Lastly, we present both lifestyle and pharmacological methods as possible treatment options for EAT dysfunction.
Collapse
Affiliation(s)
- Nikoleta Karampetsou
- Experimental Surgery and Surgical Research, National and Kapodistrian University of Athens, Athens, GRC
| | | | | | | | | | | | - Despoina N Perrea
- Experimental Surgery and Surgical Research, National and Kapodistrian University of Athens, Athens, GRC
| | - Paulos Patapis
- Surgery, National and Kapodistrian University of Athens, Athens, GRC
| |
Collapse
|
33
|
Inflammasome Signaling in Atrial Fibrillation. J Am Coll Cardiol 2022; 79:2349-2366. [DOI: 10.1016/j.jacc.2022.03.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/26/2022]
|
34
|
Wolosowicz M, Prokopiuk S, Kaminski TW. Recent Advances in the Treatment of Insulin Resistance Targeting Molecular and Metabolic Pathways: Fighting a Losing Battle? MEDICINA (KAUNAS, LITHUANIA) 2022; 58:472. [PMID: 35454311 PMCID: PMC9029454 DOI: 10.3390/medicina58040472] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022]
Abstract
Diabetes Mellitus (DM) is amongst the most notable causes of years of life lost worldwide and its prevalence increases perpetually. The disease is characterized as multisystemic dysfunctions attributed to hyperglycemia resulting directly from insulin resistance (IR), inadequate insulin secretion, or enormous glucagon secretion. Insulin is a highly anabolic peptide hormone that regulates blood glucose levels by hastening cellular glucose uptake as well as controlling carbohydrate, protein, and lipid metabolism. In the course of Type 2 Diabetes Mellitus (T2DM), which accounts for nearly 90% of all cases of diabetes, the insulin response is inadequate, and this condition is defined as Insulin Resistance. IR sequela include, but are not limited to, hyperglycemia, cardiovascular system impairment, chronic inflammation, disbalance in oxidative stress status, and metabolic syndrome occurrence. Despite the substantial progress in understanding the molecular and metabolic pathways accounting for injurious effects of IR towards multiple body organs, IR still is recognized as a ferocious enigma. The number of widely available therapeutic approaches is growing, however, the demand for precise, safe, and effective therapy is also increasing. A literature search was carried out using the MEDLINE/PubMed, Google Scholar, SCOPUS and Clinical Trials Registry databases with a combination of keywords and MeSH terms, and papers published from February 2021 to March 2022 were selected as recently published papers. This review paper aims to provide critical, concise, but comprehensive insights into the advances in the treatment of IR that were achieved in the last months.
Collapse
Affiliation(s)
- Marta Wolosowicz
- Department of Physiology, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Slawomir Prokopiuk
- Faculty of Health Sciences, Lomza State University of Applied Sciences, 18-400 Lomza, Poland;
| | - Tomasz W. Kaminski
- Department of Medicine, Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
35
|
Abstract
Interest in epicardial adipose tissue (EAT) is growing rapidly, and research in this area appeals to a broad, multidisciplinary audience. EAT is unique in its anatomy and unobstructed proximity to the heart and has a transcriptome and secretome very different from that of other fat depots. EAT has physiological and pathological properties that vary depending on its location. It can be highly protective for the adjacent myocardium through dynamic brown fat-like thermogenic function and harmful via paracrine or vasocrine secretion of pro-inflammatory and profibrotic cytokines. EAT is a modifiable risk factor that can be assessed with traditional and novel imaging techniques. Coronary and left atrial EAT are involved in the pathogenesis of coronary artery disease and atrial fibrillation, respectively, and it also contributes to the development and progression of heart failure. In addition, EAT might have a role in coronavirus disease 2019 (COVID-19)-related cardiac syndrome. EAT is a reliable potential therapeutic target for drugs with cardiovascular benefits such as glucagon-like peptide 1 receptor agonists and sodium–glucose co-transporter 2 inhibitors. This Review provides a comprehensive and up-to-date overview of the role of EAT in cardiovascular disease and highlights the translational nature of EAT research and its applications in contemporary cardiology. In this Review, Iacobellis provides a comprehensive overview of the role of epicardial adipose tissue (EAT) in cardiovascular disease, including coronary artery disease, heart failure and atrial fibrillation, discusses imaging techniques for EAT assessment and highlights the therapeutic potential of targeting EAT in cardiovascular disease. Epicardial adipose tissue (EAT) has anatomical and functional interactions with the heart owing to the shared circulation and the absence of muscle fascia separating the two organs. EAT can be clinically measured with cardiac imaging techniques that can help to predict and stratify cardiovascular risk. Regional distribution of EAT is important because pericoronary EAT and left atrial EAT differently affect the risk of coronary artery diseases and atrial fibrillation, respectively. EAT has a role in the development of several cardiovascular diseases through complex mechanisms, including gene expression profile, pro-inflammatory and profibrotic proteome, neuromodulation, and glucose and lipid metabolism. EAT could be a potential therapeutic target for novel cardiometabolic medications that modulate adipose tissue such as glucagon-like peptide 1 receptor agonists and sodium–glucose co-transporter 2 inhibitors. EAT might be a reservoir of severe acute respiratory syndrome coronavirus 2 and an amplifier of coronavirus disease 2019 (COVID-19)-related cardiac syndrome.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
36
|
Schiattarella GG, Alcaide P, Condorelli G, Gillette TG, Heymans S, Jones EAV, Kallikourdis M, Lichtman A, Marelli-Berg F, Shah S, Thorp EB, Hill JA. Immunometabolic Mechanisms of Heart Failure with Preserved Ejection Fraction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:211-222. [PMID: 35755006 PMCID: PMC9229992 DOI: 10.1038/s44161-022-00032-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing in prevalence worldwide, already accounting for at least half of all heart failure (HF). As most patients with HFpEF are obese with metabolic syndrome, metabolic stress has been implicated in syndrome pathogenesis. Recently, compelling evidence for bidirectional crosstalk between metabolic stress and chronic inflammation has emerged, and alterations in systemic and cardiac immune responses are held to participate in HFpEF pathophysiology. Indeed, based on both preclinical and clinical evidence, comorbidity-driven systemic inflammation, coupled with metabolic stress, have been implicated together in HFpEF pathogenesis. As metabolic alterations impact immune function(s) in HFpEF, major changes in immune cell metabolism are also recognized in HFpEF and in HFpEF-predisposing conditions. Both arms of immunity - innate and adaptive - are implicated in the cardiomyocyte response in HFpEF. Indeed, we submit that crosstalk among adipose tissue, the immune system, and the heart represents a critical component of HFpEF pathobiology. Here, we review recent evidence in support of immunometabolic mechanisms as drivers of HFpEF pathogenesis, discuss pivotal biological mechanisms underlying the syndrome, and highlight questions requiring additional inquiry.
Collapse
Affiliation(s)
- Gabriele G. Schiattarella
- Center for Cardiovascular Research (CCR), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gianluigi Condorelli
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Cardio Center, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Elizabeth A. V. Jones
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marinos Kallikourdis
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Andrew Lichtman
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sanjiv Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Edward B. Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
37
|
Patel KHK, Hwang T, Se Liebers C, Ng FS. Epicardial adipose tissue as a mediator of cardiac arrhythmias. Am J Physiol Heart Circ Physiol 2022; 322:H129-H144. [PMID: 34890279 PMCID: PMC8742735 DOI: 10.1152/ajpheart.00565.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Obesity is associated with higher risks of cardiac arrhythmias. Although this may be partly explained by concurrent cardiometabolic ill-health, growing evidence suggests that increasing adiposity independently confers risk for arrhythmias. Among fat depots, epicardial adipose tissue (EAT) exhibits a proinflammatory secretome and, given the lack of fascial separation, has been implicated as a transducer of inflammation to the underlying myocardium. The present review explores the mechanisms underpinning adverse electrophysiological remodeling as a consequence of EAT accumulation and the consequent inflammation. We first describe the physiological and pathophysiological function of EAT and its unique secretome and subsequently discuss the evidence for ionic channel and connexin expression modulation as well as fibrotic remodeling induced by cytokines and free fatty acids that are secreted by EAT. Finally, we highlight how weight reduction and regression of EAT volume may cause reverse remodeling to ameliorate arrhythmic risk.
Collapse
Affiliation(s)
| | - Taesoon Hwang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Curtis Se Liebers
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
38
|
Song M. Response to Letter to the Editor by Scarano-Pereira et al.: Response to: Recent advances in the mechanisms underlying the beneficial effects of bariatric and metabolic surgery. Surg Obes Relat Dis 2021; 18:298. [PMID: 34866015 DOI: 10.1016/j.soard.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/07/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky; Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|