1
|
Wei Q, Chen Y, Yuan D, Nie F, Li J, Yu K, Zhang C. Chest-CT-based radiomics feature of epicardial adipose tissue for screening coronary atherosclerosis. Heart Vessels 2025; 40:369-379. [PMID: 39540983 PMCID: PMC12009230 DOI: 10.1007/s00380-024-02479-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS This study aims to investigate the diagnostic value of chest-CT epicardial adipose tissue (EAT) radiomics feature in coronary atherosclerotic stenosis. METHODS Clinical data from 215 individuals who underwent coronary angiography and chest-CT scan from January to July 2022 at our institution were retrospectively analyzed. Based on the coronary angiography results, the total population, men, and women were divided into the CAD group and non-CAD group. radiomics feature of EAT at the level of the bifurcation of the left-main coronary artery on the transverse level of chest CT were measured. The features contain both first-order feature and shape-order feature.The differences between groups were analyzed using the t test or Chi-square test. The diagnostic efficacy of each parameter in diagnosing atherosclerotic stenosis of coronary arteries was assessed by plotting the receiver operating characteristic (ROC) curve. RESULTS First-order features: Mean, IntDen, Median, and RawIntDen; shape-order features: Area, Perim, Round, and BSA index; and clinical index: HbA1c showed statistical significance between the CAD group and the non-CAD group. The ROC curve analysis demonstrated high diagnostic efficacy, with the best for diagnostic efficacy being Median for the first-order feature parameter (AUC, 0.753; 95% confidence interval [CI], 0.689-0.817; t = 4.785, p < 0.001), Round for the shape-order feature (AUC, 0.775; 95% CI, 0.714-0.836; t = 7.842, p < 0.001), and HbA1c for the clinical index (AUC, 0.797; 95% CI, 0.783-0.856; t = 6.406, p < 0.001). After dividing the participants into male and female subgroups, the best diagnostic efficacy was observed with the BSA index for men (AUC, 0.743; 95% CI, 0.656-0.829; t = 5.128, p < 0.001) and Round for women (AUC, 0.871; 95% CI, 0.793-0.949; t = 7.247, p < 0.001). CONCLUSIONS Median, Round in radiomics feature of EAT on chest CT may play a role in the assessment of coronary atherosclerotic stenosis.
Collapse
Affiliation(s)
- Qin Wei
- Health Management Centre, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Yanling Chen
- Occupational Diseases Section, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Deqing Yuan
- Health Management Centre, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Fumei Nie
- Health Management Centre, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Jian Li
- Radiology department,, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - KeBing Yu
- Radiology department,, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Chengwei Zhang
- Cardiovascular Medicine, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China.
| |
Collapse
|
2
|
Hara T, Sata M. Pericoronary adipose tissue: potential for pathological diagnosis and therapeutic applications. Cardiovasc Interv Ther 2025:10.1007/s12928-025-01126-5. [PMID: 40185991 DOI: 10.1007/s12928-025-01126-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Excessive accumulation of epicardial adipose tissue (EAT) is known to be a risk factor for coronary artery disease and heart failure. In particular, it is thought that inflammation of pericoronary adipose tissue (PCAT) affects the pathology of various coronary artery diseases (CAD). EAT and PCAT are thought to be new therapeutic targets for preventing cardiovascular disease. Although there are no established drugs that specifically reduce inflammation of EAT or PCAT, the basic approach is to improve lifestyle-related diseases through exercise and diet, and to use metabolic improvement drugs and anti-inflammatory drugs as soft support. Potential candidates include statins, SGLT2 inhibitors, and GLP- 1 receptor agonists. In addition to conventional treatments that target substances within blood vessels, treatments that target EAT and PCAT by directly enveloping the coronary arteries and myocardium from outside the body are expected to further suppress cardiovascular events.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3 - 18 - 15, Kuramoto-cho, Tokushima, 770 - 8503, Japan.
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3 - 18 - 15, Kuramoto-cho, Tokushima, 770 - 8503, Japan
| |
Collapse
|
3
|
Li T, Thoen ZE, Applebaum JM, Khalil RA. Menopause-related changes in vascular signaling by sex hormones. J Pharmacol Exp Ther 2025; 392:103526. [PMID: 40184819 DOI: 10.1016/j.jpet.2025.103526] [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: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/07/2025] Open
Abstract
Cardiovascular disease (CVD), such as hypertension and coronary artery disease, involves pathological changes in vascular signaling, function, and structure. Vascular signaling is regulated by multiple intrinsic and extrinsic factors that influence endothelial cells, vascular smooth muscle, and extracellular matrix. Vascular function is also influenced by environmental factors including diet, exercise, and stress, as well as genetic background, sex differences, and age. CVD is more common in adult men and postmenopausal women than in premenopausal women. Specifically, women during menopausal transition, with declining ovarian function and production of estrogen (E2) and progesterone, show marked increase in the incidence of CVD and associated vascular dysfunction. Mechanistic research suggests that E2 and E2 receptor signaling have beneficial effects on vascular function including vasodilation, decreased blood pressure, and cardiovascular protection. Also, the tangible benefits of E2 supplementation in improving menopausal symptoms have prompted clinical trials of menopausal hormone therapy (MHT) in CVD, but the results have been inconsistent. The inadequate benefits of MHT in CVD could be attributed to the E2 type, dose, formulation, route, timing, and duration as well as menopausal changes in E2/E2 receptor vascular signaling. Other factors that could affect the responsiveness to MHT are the integrated hormonal milieu including gonadotropins, progesterone, and testosterone, vascular health status, preexisting cardiovascular conditions, and menopause-related dysfunction in the renal, gastrointestinal, endocrine, immune, and nervous systems. Further analysis of these factors should enhance our understanding of menopause-related changes in vascular signaling by sex hormones and provide better guidance for management of CVD in postmenopausal women. SIGNIFICANCE STATEMENT: Cardiovascular disease is more common in adult men and postmenopausal women than premenopausal women. Earlier observations of vascular benefits of menopausal hormone therapy did not materialize in randomized clinical trials. Further examination of the cardiovascular effects of sex hormones in different formulations and regimens, and the menopausal changes in vascular signaling would help to adjust the menopausal hormone therapy protocols in order to enhance their effectiveness in reducing the risk and the management of cardiovascular disease in postmenopausal women.
Collapse
Affiliation(s)
- Tao Li
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Zachary E Thoen
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jessica M Applebaum
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
4
|
Langenbach IL, Hadzic I, Zeleznik R, Langenbach MC, Maintz D, Mayrhofer T, Lu MT, Aerts HJWL, Foldyna B, Weintraub E. Association of Epicardial Adipose Tissue Changes on Serial Chest CT Scans with Mortality: Insights from the National Lung Screening Trial. Radiology 2025; 314:e240473. [PMID: 39964263 PMCID: PMC11868846 DOI: 10.1148/radiol.240473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/01/2024] [Accepted: 12/11/2024] [Indexed: 03/03/2025]
Abstract
Background Individuals eligible for lung cancer screening with low-dose CT face a higher cardiovascular mortality risk. Purpose To investigate the association between changes in epicardial adipose tissue (EAT) at the 2-year interval and mortality in individuals undergoing serial low-dose CT lung cancer screening. Materials and Methods This secondary analysis of the National Lung Screening Trial obtained EAT volume and density from serial low-dose CT scans using a validated automated deep learning algorithm. EAT volume and density changes over 2 years were categorized into typical (decrease of 7% to increase of 11% and decrease of 3% to increase of 2%, respectively) and atypical (increase or decrease beyond typical) changes, which were associated with all-cause, cardiovascular, and lung cancer mortality. Uni- and multivariable Cox proportional hazard regression models-adjusted for baseline EAT values, age, sex, race, ethnicity, smoking, pack-years, heart disease or myocardial infarction, stroke, hypertension, diabetes, education status, body mass index, and coronary artery calcium-were performed. Results Among 20 661 participants (mean age, 61.4 years ± 5.0 [SD]; 12 237 male [59.2%]), 3483 (16.9%) died over a median follow-up of 10.4 years (IQR, 9.9-10.8 years) (cardiovascular related: 816 [23.4%]; lung cancer related: 705 [20.2%]). Mean EAT volume increased (2.5 cm3/m2 ± 11.0) and density decreased (decrease of 0.5 HU ± 3.0) over 2 years. Atypical changes in EAT volume were independent predictors of all-cause mortality (atypical increase: hazard ratio [HR], 1.15 [95% CI: 1.06, 1.25] [P < .001]; atypical decrease: HR, 1.34 [95% CI: 1.23, 1.46] [P < .001]). An atypical decrease in EAT volume was associated with cardiovascular mortality (HR, 1.27 [95% CI: 1.06, 1.51]; P = .009). EAT density increase was associated with all-cause, cardiovascular, and lung cancer mortality (HR, 1.29 [95% CI: 1.18, 1.40] [P < .001]; HR, 1.29 [95% CI: 1.08, 1.54] [P = .005]; HR, 1.30 [95% CI: 1.07, 1.57] [P = .007], respectively). Conclusion EAT volume increase and decrease and EAT density increase beyond typical on subsequent chest CT scans were associated with all-cause mortality in participants screened for lung cancer. EAT volume decrease and EAT density increase were associated with elevated risk of cardiovascular mortality after adjustment for baseline EAT values. © RSNA, 2025 Supplemental material is available for this article. See also the editorial by Fuss in this issue.
Collapse
Affiliation(s)
- Isabel L. Langenbach
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Ibrahim Hadzic
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Roman Zeleznik
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Marcel C. Langenbach
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - David Maintz
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Thomas Mayrhofer
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Michael T. Lu
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Hugo J. W. L. Aerts
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Borek Foldyna
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| | - Elizabeth Weintraub
- From the Cardiovascular Imaging Research Center, Massachusetts
General Hospital, Harvard Medical School, 165 Cambridge St, Ste 400, Boston, MA
02114 (I.L.L., M.C.L., T.M., M.T.L., B.F.); Institute for Diagnostic and
Interventional Radiology, University Hospital Cologne, Cologne, Germany (I.L.L.,
M.C.L., D.M.); Artificial Intelligence in Medicine Program, Mass General
Brigham, Harvard Medical School, Boston, Mass (I.H., R.Z., H.J.W.L.A.);
Department of Radiation Oncology, Brigham and Women’s Hospital,
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass (I.H., R.Z.,
H.J.W.L.A.); Department of Radiology and Nuclear Medicine, CARIM & GROW,
Maastricht University, Maastricht, the Netherlands (I.H., H.J.W.L.A.); and
School of Business Studies, Stralsund University of Applied Sciences, Stralsund,
Germany (T.M.)
| |
Collapse
|
5
|
Rives J, Gil-Millan P, Viladés D, García-Osuna Á, Genua I, Miñambres I, Grau-Agramunt M, Gich I, Puig N, Benitez S, Julve J, Pérez A, Sánchez-Quesada JL. Low-Density Lipoprotein Subfraction Phenotype Is Associated with Epicardial Adipose Tissue Volume in Type 2 Diabetes. J Clin Med 2025; 14:862. [PMID: 39941533 PMCID: PMC11818426 DOI: 10.3390/jcm14030862] [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: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Increased epicardial adipose tissue (EAT) volume is a common feature in type 2 diabetes (T2DM) which is directly associated with heart failure and advanced atherosclerosis. We aimed to evaluate lipoprotein-related biomarkers of EAT volume in T2DM patients before and after glycemic control. Methods: This study included 36 T2DM patients before and after optimization of glycemic control and on 14 healthy controls (HCs). EAT volume was measured using computed tomography imaging indexed to the body surface area (iEAT). Biochemical and lipid profiles were determined using commercial methods. Lipoproteins were isolated by ultracentrifugation, and variables of lipoprotein function were assessed. Multivariable regression analysis was used to find variables independently associated with iEAT. Results: iEAT was higher in T2DM than in controls and decreased with glycemic optimization. HDLs from T2DM had less apoA-I and cholesterol and more apoC-III and triglycerides. LDLs from T2DM had more triglycerides and apoB and smaller sizes than those from HCs. Significant correlations were found between iEAT and age, BMI, HbA1c, GGT, VLDLc, triglycerides, LDL size, apoA-I in HDL, and apoC-III in HDL. In the multivariable regression analysis, age, LDL size, and GGT associations remained statistically significant, and predicted 50% of the variability in EAT volume. ROC analysis using these variables showed an AUC of 0.835. Conclusions: Qualitative characteristics of lipoproteins were altered in T2DM. Multivariable analysis showed that LDL size and GGT plasma levels were independently associated with iEAT volume, suggesting that these variables might be useful biomarkers for stratifying T2DM patients with increased EAT volume.
Collapse
Affiliation(s)
- José Rives
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Pedro Gil-Millan
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - David Viladés
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- CIBER of Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Álvaro García-Osuna
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Idoia Genua
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
| | - Inka Miñambres
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Margarida Grau-Agramunt
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Ignasi Gich
- Epidemiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Nuria Puig
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Sonia Benitez
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Josep Julve
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Antonio Pérez
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| |
Collapse
|
6
|
Hara T, Sata M. Roles of perivascular adipose tissue in the pathogenesis of atherosclerosis - an update on recent findings. Front Physiol 2025; 15:1522471. [PMID: 39835204 PMCID: PMC11744021 DOI: 10.3389/fphys.2024.1522471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Lifestyle-related diseases, such as atherosclerosis and diabetes, are now considered to be a series of diseases caused by chronic inflammation. Adipose tissue is considered to be an endocrine organ that not only plays a role in lipid storage, heat production, and buffering, but also produces physiologically active substances and is involved in chronic inflammation. Perivascular adipose tissue (PVAT) surrounding blood vessels similarly produces inflammatory and anti-inflammatory physiologically active substances that act on blood vessels either directly or via the bloodstream. Epicardial adipose tissue (EAT), which is in direct contact with the coronary arteries inside the pericardium, is thought to have a direct effect on the coronary arteries as well. The presence and inflammatory status of these adipose tissues can be evaluated by imaging tests, and has been shown to be associated with the presence of current cardiovascular disease (CVD) and to be a prognostic factor. It is also expected to become a new diagnostic and therapeutic target for CVD.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | | |
Collapse
|
7
|
Wang W, Gao Y, Wang J, Ji C, Gu H, Yuan X, Yang S, Wang X. Prognostic Value of Epicardial Adipose Tissue in Heart Failure With Mid-Range and Preserved Ejection Fraction: A Multicenter Study. J Am Heart Assoc 2024; 13:e036789. [PMID: 39673347 PMCID: PMC11935535 DOI: 10.1161/jaha.124.036789] [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: 06/26/2024] [Accepted: 09/20/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) accumulation is thought to play a role in the pathophysiology of heart failure (HF) with mid-range ejection fraction and HF with preserved ejection fraction, but its effect on outcome is unknown. METHODS AND RESULTS A total of 692 patients with HF with mid-range ejection fraction or HF with preserved ejection fraction who underwent cardiovascular magnetic resonance at 2 medical centers in China between October 2016 and October 2022 were included in this study. EAT volume and extracellular volume were calculated using cardiovascular magnetic resonance. The main outcome was the composite of all-cause mortality and first HF hospitalization. Of 692 participants, 41.3% were women. The mean age, body mass index, left ventricular ejection fraction, and EAT volume were 57.0 years, 27.2 kg/m2, 50.0%, and 67.1 mL/m2, respectively. During a median follow-up of 34 months, 169 patients (24.4%) died or were hospitalized for HF. EAT volume exhibited a strong unadjusted association with the composite outcome (hazard ratio per 1 mL/m2 [HR], 1.57 [95% CI, 1.40-1.76], P<0.001). After fully adjusting, EAT remained associated with the outcome (HR, 1.62 [95% CI, 1.42-1.86], P<0.001). We constructed a baseline multivariable model including comorbidities, New York Heart Association functional class, extracellular volume, age, body mass index, left ventricular ejection fraction, and N-terminal pro-brain natriuretic peptide. Addition of EAT volume to the baseline multivariable model significantly improved model performance (C statistic improvement: 0.711-0.760; P<0.001). CONCLUSIONS EAT accumulation is associated with an adverse prognosis in patients with HF with mid-range ejection fraction and those with HF with preserved ejection fraction. In addition, EAT provides incremental prognostic value beyond left ventricular ejection fraction and New York Heart Association class.
Collapse
Affiliation(s)
- Wenxian Wang
- School of Medical Imaging, Binzhou Medical UniversityYantaiShandongP. R. China
| | - Yan Gao
- Department of RadiologyShandong Provincial Hospital, Shandong UniversityJinanShandongChina
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Jian Wang
- Department of RadiologyCentral Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Congshan Ji
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Hui Gu
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Xianshun Yuan
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Shifeng Yang
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Ximing Wang
- Department of RadiologyShandong Provincial Hospital, Shandong UniversityJinanShandongChina
- Department of RadiologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| |
Collapse
|
8
|
Gaborit B, Julla JB, Fournel J, Ancel P, Soghomonian A, Deprade C, Lasbleiz A, Houssays M, Ghattas B, Gascon P, Righini M, Matonti F, Venteclef N, Potier L, Gautier JF, Resseguier N, Bartoli A, Mourre F, Darmon P, Jacquier A, Dutour A. Fully automated epicardial adipose tissue volume quantification with deep learning and relationship with CAC score and micro/macrovascular complications in people living with type 2 diabetes: the multicenter EPIDIAB study. Cardiovasc Diabetol 2024; 23:328. [PMID: 39227844 PMCID: PMC11373274 DOI: 10.1186/s12933-024-02411-y] [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: 08/19/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The aim of this study (EPIDIAB) was to assess the relationship between epicardial adipose tissue (EAT) and the micro and macrovascular complications (MVC) of type 2 diabetes (T2D). METHODS EPIDIAB is a post hoc analysis from the AngioSafe T2D study, which is a multicentric study aimed at determining the safety of antihyperglycemic drugs on retina and including patients with T2D screened for diabetic retinopathy (DR) (n = 7200) and deeply phenotyped for MVC. Patients included who had undergone cardiac CT for CAC (Coronary Artery Calcium) scoring after inclusion (n = 1253) were tested with a validated deep learning segmentation pipeline for EAT volume quantification. RESULTS Median age of the study population was 61 [54;67], with a majority of men (57%) a median duration of the disease 11 years [5;18] and a mean HbA1c of7.8 ± 1.4%. EAT was significantly associated with all traditional CV risk factors. EAT volume significantly increased with chronic kidney disease (CKD vs no CKD: 87.8 [63.5;118.6] vs 82.7 mL [58.8;110.8], p = 0.008), coronary artery disease (CAD vs no CAD: 112.2 [82.7;133.3] vs 83.8 mL [59.4;112.1], p = 0.0004, peripheral arterial disease (PAD vs no PAD: 107 [76.2;141] vs 84.6 mL[59.2; 114], p = 0.0005 and elevated CAC score (> 100 vs < 100 AU: 96.8 mL [69.1;130] vs 77.9 mL [53.8;107.7], p < 0.0001). By contrast, EAT volume was neither associated with DR, nor with peripheral neuropathy. We further evidenced a subgroup of patients with high EAT volume and a null CAC score. Interestingly, this group were more likely to be composed of young women with a high BMI, a lower duration of T2D, a lower prevalence of microvascular complications, and a higher inflammatory profile. CONCLUSIONS Fully-automated EAT volume quantification could provide useful information about the risk of both renal and macrovascular complications in T2D patients.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France.
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France.
| | - Jean Baptiste Julla
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Féderation de Diabétologie, Université Paris Cité, Lariboisière Hospital, APHP, 75015, Paris, France
| | | | - Patricia Ancel
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Astrid Soghomonian
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Camille Deprade
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Adèle Lasbleiz
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
| | - Marie Houssays
- Medical Evaluation Department, Assistance-Publique Hôpitaux de Marseille, CIC-CPCET, 13005, Marseille, France
| | - Badih Ghattas
- Aix Marseille School of Economics, Aix Marseille University, CNRS, Marseille, France
| | - Pierre Gascon
- Centre Monticelli Paradis, 433 Bis Rue Paradis, 13008, Marseille, France
| | - Maud Righini
- Ophtalmology Department, Assistance-Publique Hôpitaux de Marseille, Aix-Marseille Univ, 13005, Marseille, France
| | - Frédéric Matonti
- Centre Monticelli Paradis, 433 Bis Rue Paradis, 13008, Marseille, France
- National Center for Scientific Research (CNRS), Timone Neuroscience Institute (INT), Aix Marseille Univ, 13008, Marseille, France
| | - Nicolas Venteclef
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
| | - Louis Potier
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Fédération de Diabétologie, Bichat Hospital, Paris, France
| | - Jean François Gautier
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Féderation de Diabétologie, Université Paris Cité, Lariboisière Hospital, APHP, 75015, Paris, France
| | - Noémie Resseguier
- Support Unit for Clinical Research and Economic Evaluation, Assistance Publique-Hôpitaux de Marseille, 13385, Marseille, France
- Aix-Marseille Univ, EA 3279 CEReSS-Health Service Research and Quality of Life Center, Marseille, France
| | - Axel Bartoli
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- Department of Radiology, Hôpital de la TIMONE, AP-HM, Marseille, France
| | - Florian Mourre
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Patrice Darmon
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Alexis Jacquier
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- Department of Radiology, Hôpital de la TIMONE, AP-HM, Marseille, France
| | - Anne Dutour
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| |
Collapse
|
9
|
Li Q, Muhib UR, Ma X, Liu Z, Gao F, Wang Z. Potential Mechanisms of Epicardial Adipose Tissue Influencing Heart Failure with Preserved Ejection Fraction. Rev Cardiovasc Med 2024; 25:311. [PMID: 39355598 PMCID: PMC11440401 DOI: 10.31083/j.rcm2509311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/28/2024] [Accepted: 04/09/2024] [Indexed: 10/03/2024] Open
Abstract
Heart failure (HF) is the predominant terminal stage and the leading cause of mortality in cardiac disease. Heart failure with preserved ejection fraction (HFpEF) affects roughly 50% of HF patients globally. Due to the global aging population, the prevalence, morbidity, and mortality of HFpEF have gradually increased. Epicardial adipose tissue (EAT), as a key visceral adipose tissue around the heart, affects cardiac diastolic function and exercise reserve capacity. EAT closely adheres to the myocardium and can produce inflammatory factors, neurotransmitters, and other factors through autocrine or paracrine mechanisms, affecting the heart function by inflammatory response, cardiac metabolism and energy supply, cardiomyocyte structure and electrical activity, and pericardial vascular function. Currently, research on the mechanism and treatment methods of HFpEF is constantly improving. EAT may play a multi-level impact on the occurrence and development of HFpEF. This review also summarizes the potential impact of EAT on the heart in HFpEF combined with other metabolism-related diseases such as obesity or diabetes over other obesity-related measures, such as body mass index (BMI) or other adipose tissue. Above all, this review comprehensively summarizes the potential mechanisms by which EAT may affect HFpEF. The objective is to enhance our comprehension and management of HFpEF. Future research should delve into the mechanistic relationship between EAT and HFpEF, and investigate interventions aimed at EAT to improve the prognosis of patients with HFpEF.
Collapse
Affiliation(s)
- Qiuxuan Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| | - Ur Rehman Muhib
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| | - Xiaoteng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| | - Zaiqiang Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| | - Fei Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, 100029 Beijing, China
| |
Collapse
|
10
|
Chen X, Xiang H, Lu J, Yang M. Epicardial Adipose Tissue and Psoriasis: A Systematic Review and Meta-Analysis. J Clin Med 2024; 13:4761. [PMID: 39200903 PMCID: PMC11355870 DOI: 10.3390/jcm13164761] [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: 05/27/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Background: As a novel biomarker for cardiovascular diseases, epicardial adipose tissue (EAT) has been linked to psoriasis. We conducted an updated systematic review, building upon a previous report on the relationship between EAT and psoriasis. Methods: We searched Medline, Embase, and the Cochrane Central Register of Controlled Trials. The methodological quality of each study was assessed using the Newcastle-Ottawa Scale. The pooled mean difference (MD) or standardized mean difference (SMD) and the corresponding confidence interval (CIs) were calculated. Results: We included 10 studies with 1287 participants. Five of the included studies were of high methodological quality, while the other five were of moderate quality. The pooled data indicated that psoriasis patients had significantly increased EAT compared to individuals in the control group (SMD 1.53, 95% CI 0.61 to 2.45, 9 studies, 1195 participants). The subgroup analysis showed that psoriasis patients had significantly increased EAT thickness compared with the controls (SMD 2.45, 95% CI 0.73 to 4.17, 5 studies, 657 participants). Similarly, EAT area in single-slice CT images was significantly higher in the psoriasis group than in the control group (SMD 0.45, 95% CI 0.14 to 0.76, 2 studies, 195 participants). The EAT volume based on CT images appeared to be higher in the psoriasis group than in the control group, but the difference was not statistically significant (SMD 0.32, 95% CI -0.06 to 0.70, 2 studies, 343 participants). Conclusions: EAT, especially echocardiographic EAT thickness and CT-determined EAT area, was significantly associated with psoriasis, but CT-determined EAT volume was not.
Collapse
Affiliation(s)
- Xiaomei Chen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China; (X.C.); (H.X.)
| | - Hongmei Xiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China; (X.C.); (H.X.)
| | - Jing Lu
- Medical Insurance Office, West China Hospital, Sichuan University, Chengdu 610041, China
- Chinese Cochrane Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming Yang
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Zhao J, Cheng W, Dai Y, Li Y, Feng Y, Tan Y, Xue Q, Bao X, Sun X, Kang L, Mu D, Xu B. Excessive accumulation of epicardial adipose tissue promotes microvascular obstruction formation after myocardial ischemia/reperfusion through modulating macrophages polarization. Cardiovasc Diabetol 2024; 23:236. [PMID: 38970123 PMCID: PMC11227217 DOI: 10.1186/s12933-024-02342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Owing to its unique location and multifaceted metabolic functions, epicardial adipose tissue (EAT) is gradually emerging as a new metabolic target for coronary artery disease risk stratification. Microvascular obstruction (MVO) has been recognized as an independent risk factor for unfavorable prognosis in acute myocardial infarction patients. However, the concrete role of EAT in the pathogenesis of MVO formation in individuals with ST-segment elevation myocardial infarction (STEMI) remains unclear. The objective of the study is to evaluate the correlation between EAT accumulation and MVO formation measured by cardiac magnetic resonance (CMR) in STEMI patients and clarify the underlying mechanisms involved in this relationship. METHODS Firstly, we utilized CMR technique to explore the association of EAT distribution and quantity with MVO formation in patients with STEMI. Then we utilized a mouse model with EAT depletion to explore how EAT affected MVO formation under the circumstances of myocardial ischemia/reperfusion (I/R) injury. We further investigated the immunomodulatory effect of EAT on macrophages through co-culture experiments. Finally, we searched for new therapeutic strategies targeting EAT to prevent MVO formation. RESULTS The increase of left atrioventricular EAT mass index was independently associated with MVO formation. We also found that increased circulating levels of DPP4 and high DPP4 activity seemed to be associated with EAT increase. EAT accumulation acted as a pro-inflammatory mediator boosting the transition of macrophages towards inflammatory phenotype in myocardial I/R injury through secreting inflammatory EVs. Furthermore, our study declared the potential therapeutic effects of GLP-1 receptor agonist and GLP-1/GLP-2 receptor dual agonist for MVO prevention were at least partially ascribed to its impact on EAT modulation. CONCLUSIONS Our work for the first time demonstrated that excessive accumulation of EAT promoted MVO formation by promoting the polarization state of cardiac macrophages towards an inflammatory phenotype. Furthermore, this study identified a very promising therapeutic strategy, GLP-1/GLP-2 receptor dual agonist, targeting EAT for MVO prevention following myocardial I/R injury.
Collapse
Affiliation(s)
- Jinxuan Zhao
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wei Cheng
- Division of Colorectal Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yang Dai
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Yao Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuting Feng
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Ying Tan
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Qiucang Xue
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xue Bao
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xuan Sun
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Lina Kang
- Department of Cardiology, MOE Key Laboratory of Model Animal for Disease Study, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China.
| | - Dan Mu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Yin L, Yan C, Guo W, Yang C, Dong H, Zhang Y, Xu S, Zeng M. Correlation between clinical characteristics and epicardial adipose tissue features in acute myocarditis patients using coronary computed tomography (CT) vascular imaging: a case-control study with retrospective data collection. Quant Imaging Med Surg 2024; 14:5072-5083. [PMID: 39022264 PMCID: PMC11250336 DOI: 10.21037/qims-23-1407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Background Epicardial adipose tissue (EAT) is unique type of visceral adipose tissue, sharing the same microcirculation with myocardium. This study aimed to assess the imaging features of EAT in patients with acute myocarditis (AM) and explore the relationships with clinical characteristics. Methods For this retrospective case-control study, totally 38 AM patients and 52 controls were screened retrospectively from January 2019 to December 2022, and the EAT volume was measured from coronary computed tomography (CT) angiography imaging. Histogram analysis was performed to calculate parameters like the mean, standard deviation, interquartile range and percentiles of EAT attenuation. Whether EAT features change was assessed when clinical characteristics including symptoms, T wave abnormalities, pericardial effusion (PE), impairment of systolic function, and the need for intensive care presented. Results The EAT volume (75.2±22.8 mL) and mean EAT attenuation [-75.8±4.4 Hounsfield units (HU)] of the AM group was significantly larger than the control group (64.7±26.0 mL, P=0.049; -77.9±5.0 HU, P=0.044). Among the clinical characteristics, only the presence of PE was associated with changes in EAT features. Patients with PE showed significantly changes in EAT attenuation including mean attenuation [analysis of variance (ANOVA) P=0.001] and quantitative histogram parameters. The mean attenuation of patients with PE (-71.9±4.0 HU) was significantly larger than controls (-77.9±5.0 HU, Bonferroni corrected P<0.001) and patients without PE (-77.4±3.5 HU, Bonferroni corrected P=0.003). Observed in histogram, the overall increase in EAT attenuation could lead to decrease in EAT volume, which resulted in no statistically significant difference in EAT volume between the AM patients with PE and controls (64.7±26.0 vs. 72.2±28.3 mL, Bonferroni corrected P>0.99). Conclusions Compared to controls, EAT volume was significantly larger in AM, and EAT attenuation increased notably in the presence of PE. We recommend evaluating EAT volume and attenuation simultaneously when quantifying EAT using CT attenuation thresholds.
Collapse
Affiliation(s)
- Lekang Yin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yan
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weifeng Guo
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Radiology, Shanghai Geriatric Medical Center, Shanghai, China
| | - Chun Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Dong
- Department of Radiology, First People’s Hospital of Xiaoshan District, Hangzhou, China
| | - Yang Zhang
- Department of Radiology, Dongying People’s Hospital, Dongying, China
| | - Shijie Xu
- Shanghai United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Medical Imaging, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Morrissette-McAlmon J, Xu WR, Teuben R, Boheler KR, Tung L. Adipocyte-mediated electrophysiological remodeling of human stem cell - derived cardiomyocytes. J Mol Cell Cardiol 2024; 189:52-65. [PMID: 38346641 DOI: 10.1016/j.yjmcc.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Adipocytes normally accumulate in the epicardial and pericardial layers around the human heart, but their infiltration into the myocardium can be proarrhythmic. METHODS AND RESULTS: Human adipose derived stem/stromal cells and human induced pluripotent stem cells (hiPSC) were differentiated, respectively into predominantly white fat-like adipocytes (hAdip) and ventricular cardiomyocytes (CMs). Adipocytes cultured in CM maintenance medium (CM medium) maintained their morphology, continued to express adipogenic markers, and retained clusters of intracellular lipid droplets. In contrast, hiPSC-CMs cultivated in adipogenic growth medium displayed abnormal cell morphologies and more clustering across the monolayer. Pre-plated hiPSC-CMs co-cultured in direct contact with hAdips in CM medium displayed prolonged action potential durations, increased triangulation, slowed conduction velocity, increased conduction velocity heterogeneity, and prolonged calcium transients. When hAdip-conditioned medium was added to monolayer cultures of hiPSC-CMs, results similar to those recorded with direct co-cultures were observed. Both co-culture and conditioned medium experiments resulted in increases in transcript abundance of SCN10A, CACNA1C, SLC8A1, and RYR2, with a decrease in KCNJ2. Human adipokine immunoblots revealed the presence of cytokines that were elevated in adipocyte-conditioned medium, including MCP-1, IL-6, IL-8 and CFD that could induce electrophysiological changes in cultured hiPSC-CMs. CONCLUSIONS: Co-culture of hiPSC-CMs with hAdips reveals a potentially pathogenic role of infiltrating human adipocytes on myocardial tissue. In the absence of structural changes, hAdip paracrine release alone is sufficient to cause CM electrophysiological dysfunction mirroring the co-culture conditions. These effects, mediated largely by paracrine mechanisms, could promote arrhythmias in the heart.
Collapse
Affiliation(s)
| | - William R Xu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roald Teuben
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth R Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Bogdański A, Niziołek P, Kopeć S, Moszak M. Epicardial Adipose Tissue: A Precise Biomarker for Cardiovascular Risk, Metabolic Diseases, and Target for Therapeutic Interventions. Cardiol Rev 2024:00045415-990000000-00230. [PMID: 38477580 DOI: 10.1097/crd.0000000000000670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Epicardial adipose tissue (EAT) is located between the heart muscle and visceral pericardium, where it has direct contact with coronary blood vessels. Elevated thickness of this tissue can induce local inflammation affecting the myocardium and the underlying coronary arteries, contributing to various cardiovascular diseases such as coronary artery disease, atrial fibrillation, or heart failure with preserved ejection fraction. Recent studies have identified EAT thickness as a simple and reliable biomarker for certain cardiovascular outcomes. Examples include the presence of atherosclerosis, incident cardiovascular disease (CVD) in individuals with type 2 diabetes mellitus (T2DM), and the prevalence of atrial fibrillation. Furthermore, EAT measurements can help to identify patients with a higher risk of developing metabolic syndrome. Since the EAT thickness can be easily measured using echocardiography, such examinations could serve as a useful and cost-effective preventive tool for assessing cardiovascular health. This review also summarizes therapeutical interventions aimed at reducing EAT. Reducing EAT thickness has been shown to be possible through pharmacological, surgical, or lifestyle-change interventions. Pharmaceutical therapies, including thiazolidinediones, glucagon-like peptide 1-receptor agonists, sodium-glucose cotransporter 2 inhibitors, dipeptidyl peptidase-4 inhibitors, and statins, have been shown to influence EAT thickness. Additionally, EAT thickness can also be managed more invasively through bariatric surgery, or noninvasively through lifestyle changes to diet and exercise routines.
Collapse
Affiliation(s)
| | | | | | - Małgorzata Moszak
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Szamarzewskiego, Poznan, Poland
| |
Collapse
|
15
|
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: 10] [Impact Index Per Article: 5.0] [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
|
16
|
Napoli G, Pergola V, Basile P, De Feo D, Bertrandino F, Baggiano A, Mushtaq S, Fusini L, Fazzari F, Carrabba N, Rabbat MG, Motta R, Ciccone MM, Pontone G, Guaricci AI. Epicardial and Pericoronary Adipose Tissue, Coronary Inflammation, and Acute Coronary Syndromes. J Clin Med 2023; 12:7212. [PMID: 38068263 PMCID: PMC10707039 DOI: 10.3390/jcm12237212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 01/22/2025] Open
Abstract
Vascular inflammation is recognized as the primary trigger of acute coronary syndrome (ACS). However, current noninvasive methods are not capable of accurately detecting coronary inflammation. Epicardial adipose tissue (EAT) and pericoronary adipose tissue (PCAT), in addition to their role as an energy reserve system, have been found to contribute to the development and progression of coronary artery calcification, inflammation, and plaque vulnerability. They also participate in the vascular response during ischemia, sympathetic stimuli, and arrhythmia. As a result, the evaluation of EAT and PCAT using imaging techniques such as computed tomography (CT), cardiac magnetic resonance (CMR), and nuclear imaging has gained significant attention. PCAT-CT attenuation, which measures the average CT attenuation in Hounsfield units (HU) of the adipose tissue, reflects adipocyte differentiation/size and leukocyte infiltration. It is emerging as a marker of tissue inflammation and has shown prognostic value in coronary artery disease (CAD), being associated with plaque development, vulnerability, and rupture. In patients with acute myocardial infarction (AMI), an inflammatory pericoronary microenvironment promoted by dysfunctional EAT/PCAT has been demonstrated, and more recently, it has been associated with plaque rupture in non-ST-segment elevation myocardial infarction (NSTEMI). Endothelial dysfunction, known for its detrimental effects on coronary vessels and its association with plaque progression, is bidirectionally linked to PCAT. PCAT modulates the secretory profile of endothelial cells in response to inflammation and also plays a crucial role in regulating vascular tone in the coronary district. Consequently, dysregulated PCAT has been hypothesized to contribute to type 2 myocardial infarction with non-obstructive coronary arteries (MINOCA) and coronary vasculitis. Recently, quantitative measures of EAT derived from coronary CT angiography (CCTA) have been included in artificial intelligence (AI) models for cardiovascular risk stratification. These models have shown incremental utility in predicting major adverse cardiovascular events (MACEs) compared to plaque characteristics alone. Therefore, the analysis of PCAT and EAT, particularly through PCAT-CT attenuation, appears to be a safe, valuable, and sufficiently specific noninvasive method for accurately identifying coronary inflammation and subsequent high-risk plaque. These findings are supported by biopsy and in vivo evidence. Although speculative, these pieces of evidence open the door for a fascinating new strategy in cardiovascular risk stratification. The incorporation of PCAT and EAT analysis, mainly through PCAT-CT attenuation, could potentially lead to improved risk stratification and guide early targeted primary prevention and intensive secondary prevention in patients at higher risk of cardiac events.
Collapse
Affiliation(s)
- Gianluigi Napoli
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Valeria Pergola
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padua, 35122 Padua, Italy;
| | - Paolo Basile
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Daniele De Feo
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Fulvio Bertrandino
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Andrea Baggiano
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Saima Mushtaq
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Laura Fusini
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Fabio Fazzari
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Nazario Carrabba
- Department of Cardiothoracovascular Medicine, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy;
| | - Mark G. Rabbat
- Division of Cardiology, Loyola University of Chicago, Chicago, IL 60611, USA;
- Edward Hines Jr. VA Hospital, Hines, IL 60141, USA
| | - Raffaella Motta
- Radiology Unit, University Hospital of Padova, 35128 Padua, Italy;
| | - Marco Matteo Ciccone
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Gianluca Pontone
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Andrea Igoren Guaricci
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| |
Collapse
|
17
|
O'Hagan R, Hsu LY, Li H, Hong CG, Parel PM, Berg AR, Manyak GA, Bui V, Patel NH, Florida EM, Teague HL, Playford MP, Zhou W, Dey D, Chen MY, Mehta NN, Sorokin AV. Longitudinal association of epicardial and thoracic adipose tissues with coronary and cardiac characteristics in psoriasis. Heliyon 2023; 9:e20732. [PMID: 37867905 PMCID: PMC10585224 DOI: 10.1016/j.heliyon.2023.e20732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background s: Psoriasis is a disease of systemic inflammation associated with increased cardiometabolic risk. Epicardial adipose tissue (EAT) and thoracic adipose tissue (TAT) are contributing factors for atherosclerosis and cardiac dysfunction. We strove to assess the longitudinal impact of the EAT and TAT on coronary and cardiac characteristics in psoriasis. Methods The study consisted of 301 patients with baseline coronary computed tomography angiography (CTA), of which 139 had four-year follow up scans. EAT and TAT volumes from non-contrast computed tomography scans were quantified by an automated segmentation framework. Coronary plaque characteristics and left ventricular (LV) mass were quantified by CTA. Results When stratified by baseline EAT and TAT volume quartiles, a stepwise significant increase in cardiometabolic parameters was observed. EAT and TAT volumes associated with fibro-fatty burden (FFB) (TAT: ρ = 0.394, P < 0.001; EAT: ρ = 0.459, P < 0.001) in adjusted models. Only EAT had a significant four-year time-dependent association with FFB in fully adjusted models (β = 0.307 P = 0.003), whereas only TAT volume associated with myocardial injury in fully adjusted models (TAT: OR = 1.57 95 % CI = (1.00-2.60); EAT: OR = 1.46 95 % CI = (0.91-2.45). Higher quartiles of EAT and TAT had increased LV mass and developed strong correlation (TAT: ρ = 0.370, P < 0.001; EAT: ρ = 0.512, P < 0.001). Conclusions Our study is the first to explore how both EAT and TAT volumes associate with increased cardiometabolic risk profile in an inflamed psoriasis cohorts and highlight the need for further studies on its use as a potential prognostic tool for high-risk coronary plaques and cardiac dysfunction.
Collapse
Affiliation(s)
- Ross O'Hagan
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Li-Yueh Hsu
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Haiou Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christin G. Hong
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M. Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander R. Berg
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grigory A. Manyak
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vy Bui
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nidhi H. Patel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth M. Florida
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L. Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wunan Zhou
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcus Y. Chen
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V. Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Cheng YC, Ma WC, Li YH, Wu J, Liang KW, Lee WJ, Liu HC, Sheu WHH, Lee IT. Plasma aryl hydrocarbon receptor associated with epicardial adipose tissue in men: a cross-sectional study. Diabetol Metab Syndr 2023; 15:188. [PMID: 37749614 PMCID: PMC10519097 DOI: 10.1186/s13098-023-01166-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) is a type of ectopic fat with endocrine and paracrine functions. Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that responds to environmental stimuli. AhR expression is associated with obesity. In this cross-sectional study, we aimed to determine the relationship between circulating AhR concentrations and EAT. METHODS A total of 30 men with obesity and 23 age-matched men as healthy controls were enrolled. Plasma AhR concentrations were determined at fasting. The EAT thickness was measured on the free wall of the right ventricle from the basal short-axis plane by magnetic resonance imaging. RESULTS The participants with obesity had a higher plasma AhR level than the controls (81.0 ± 24.5 vs. 65.1 ± 16.4 pg/mL, P = 0.010). The plasma AhR level was positively correlated with EAT thickness (correlation coefficient = 0.380, P = 0.005). After adjusting for fasting glucose levels, plasma AhR levels were still significantly associated with EAT thickness (95% CI 0.458‒5.357, P = 0.021) but not with body mass index (P = 0.168). CONCLUSION Plasma AhR concentrations were positively correlated with EAT thickness on the free wall of the right ventricle in men. Further investigations are needed to evaluate the causal effects and underlying mechanisms between AhR and EAT.
Collapse
Affiliation(s)
- Yu-Cheng Cheng
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, No. 1650 Taiwan Boulevard, Sect. 4, Taichung, 40705, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Wei-Chun Ma
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Feng Yuan Hospital, Ministry of Health and Welfare, Taichung, 42055, Taiwan
| | - Yu-Hsuan Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, No. 1650 Taiwan Boulevard, Sect. 4, Taichung, 40705, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Department of Computer Science & Information Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Junyi Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, No. 1650 Taiwan Boulevard, Sect. 4, Taichung, 40705, Taiwan
| | - Kae-Woei Liang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, School of Medicine, National Chung Hsing University, Taichung, 402204, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | - Hsiu-Chen Liu
- Department of Nursing, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | | | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, No. 1650 Taiwan Boulevard, Sect. 4, Taichung, 40705, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan.
| |
Collapse
|
19
|
Lin T, Lee C, Huang K, Wu C, Lee J, Lan C, Su MM, Hwang J, Wang Y, Lin L. Differentiating the Prognostic Determinants of Myocardial Steatosis for Heart Failure With Preserved Ejection Fraction by Cardiac Magnetic Resonance Imaging. J Am Heart Assoc 2023; 12:e027781. [PMID: 37642018 PMCID: PMC10547328 DOI: 10.1161/jaha.122.027781] [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: 08/10/2022] [Accepted: 07/10/2023] [Indexed: 08/31/2023]
Abstract
Background Myocardial steatosis and fibrosis may play a role in the pathophysiology of heart failure with preserved ejection fraction. We therefore investigated the prognostic significance of epicardial fat (epicardial adipose tissue [EAT]) and myocardial diffuse fibrosis. Methods and Results Myocardial fibrosis, estimated as extracellular volume (ECV), and EAT were measured using cardiac magnetic resonance imaging in 163 subjects with heart failure with preserved ejection fraction. We also evaluated cardiac structure and diastolic and systolic function by echocardiography and cardiac magnetic resonance imaging. After 24 months' follow-up, 39 (24%) subjects had experienced cardiovascular events, including hospitalization for heart failure, acute coronary syndrome, and cardiovascular death. Median EAT and mean ECV were significantly higher in subjects with cardiovascular events than survivors (EAT, 35 [25-45] versus 31 [21-38], P=0.006 and ECV, 28.9±3.16% versus 27.2±3.56%, P=0.04). Subjects with high EAT (≥42 g) had increased risk of cardiovascular events (hazard ratio [HR], 2.528 [95% CI, 1.704-4.981]; P=0.032). High ECV (>29%) was also significantly associated with poorer outcomes (HR, 1.647 [95% CI, 1.263-2.548]; P=0.013). With respect to secondary end points, high EAT and high ECV were associated with increased risk of the incident acute coronary syndrome (HR, 1.982 [95% CI, 1.008-4.123]; P=0.049) and hospitalization for heart failure (HR, 1.789 [95% CI, 1.102-6.987]; P=0.033), respectively. Conclusions Our study suggested that increased epicardial fat and ECV detected by cardiac magnetic resonance imaging have an impact on cardiovascular prognosis, in particular acute coronary syndrome and hospitalization for heart failure, respectively.
Collapse
Affiliation(s)
- Ting‐Tse Lin
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Chih‐Kuo Lee
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University Hospital Hsin‐Chu BranchHsinchuTaiwan
| | - Kuan‐Chih Huang
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University Hospital Hsin‐Chu BranchHsinchuTaiwan
| | - Cho‐Kai Wu
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Jen‐Kuang Lee
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Chen‐Wei Lan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Mao‐Yuan M. Su
- Department of Medical ImagingNational Taiwan University HospitalTaipeiTaiwan
| | - Juey‐Jen Hwang
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Yi‐Chih Wang
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Lian‐Yu Lin
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| |
Collapse
|
20
|
Bernstein SR, Kelleher C, Khalil RA. Gender-based research underscores sex differences in biological processes, clinical disorders and pharmacological interventions. Biochem Pharmacol 2023; 215:115737. [PMID: 37549793 PMCID: PMC10587961 DOI: 10.1016/j.bcp.2023.115737] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Earlier research has presumed that the male and female biology is similar in most organs except the reproductive system, leading to major misconceptions in research interpretations and clinical implications, with serious disorders being overlooked or misdiagnosed. Careful research has now identified sex differences in the cardiovascular, renal, endocrine, gastrointestinal, immune, nervous, and musculoskeletal systems. Also, several cardiovascular, immunological, and neurological disorders have shown differences in prevalence and severity between males and females. Genetic variations in the sex chromosomes have been implicated in several disorders at young age and before puberty. The levels of the gonadal hormones estrogen, progesterone and testosterone and their receptors play a role in the sex differences between adult males and premenopausal women. Hormonal deficiencies and cell senescence have been implicated in differences between postmenopausal and premenopausal women. Specifically, cardiovascular disorders are more common in adult men vs premenopausal women, but the trend is reversed with age with the incidence being greater in postmenopausal women than age-matched men. Gender-specific disorders in females such as polycystic ovary syndrome, hypertension-in-pregnancy and gestational diabetes have attained further research recognition. Other gender-related research areas include menopausal hormone therapy, the "Estrogen Paradox" in pulmonary arterial hypertension being more predominant but less severe in young females, and how testosterone may cause deleterious effects in the kidney while having vasodilator effects in the coronary circulation. This has prompted the National Institutes of Health (NIH) initiative to consider sex as a biological variable in research. The NIH and other funding agencies have provided resources to establish state-of-the-art centers for women health and sex differences in biology and disease in several academic institutions. Scientific societies and journals have taken similar steps to organize specialized conferences and publish special issues on gender-based research. These combined efforts should promote research to enhance our understanding of the sex differences in biological systems beyond just the reproductive system, and provide better guidance and pharmacological tools for the management of various clinical disorders in a gender-specific manner.
Collapse
Affiliation(s)
- Sofia R Bernstein
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Caroline Kelleher
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Vershinina YS, Krasnov GS, Garbuz DG, Shaposhnikov MV, Fedorova MS, Pudova EA, Katunina IV, Kornev AB, Zemskaya NV, Kudryavtsev AA, Bulavkina EV, Matveeva AA, Ulyasheva NS, Guvatova ZG, Anurov AA, Moskalev AA, Kudryavtseva AV. Transcriptomic Analysis of the Effect of Torin-2 on the Central Nervous System of Drosophila melanogaster. Int J Mol Sci 2023; 24:ijms24109095. [PMID: 37240439 DOI: 10.3390/ijms24109095] [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: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Torin-2, a synthetic compound, is a highly selective inhibitor of both TORC1 and TORC2 (target of rapamycin) complexes as an alternative to the well-known immunosuppressor, geroprotector, and potential anti-cancer natural compound rapamycin. Torin-2 is effective at hundreds of times lower concentrations and prevents some negative side effects of rapamycin. Moreover, it inhibits the rapamycin-resistant TORC2 complex. In this work, we evaluated transcriptomic changes in D. melanogaster heads induced with lifetime diets containing Torin-2 and suggested possible neuroprotective mechanisms of Torin-2. The analysis included D. melanogaster of three ages (2, 4, and 6 weeks old), separately for males and females. Torin-2, taken at the lowest concentration being tested (0.5 μM per 1 L of nutrient paste), had a slight positive effect on the lifespan of D. melanogaster males (+4% on the average) and no positive effect on females. At the same time, RNA-Seq analysis revealed interesting and previously undiscussed effects of Torin-2, which differed between sexes as well as in flies of different ages. Among the cellular pathways mostly altered by Torin-2 at the gene expression level, we identified immune response, protein folding (heat shock proteins), histone modification, actin cytoskeleton organization, phototransduction and sexual behavior. Additionally, we revealed that Torin-2 predominantly reduced the expression of Srr gene responsible for the conversion of L-serine to D-serine and thus regulating activity of NMDA receptor. Via western blot analysis, we showed than in old males Torin-2 tends to increase the ratio of the active phosphorylated form of ERK, the lowest node of the MAPK cascade, which may play a significant role in neuroprotection. Thus, the complex effect of Torin-2 may be due to the interplay of the immune system, hormonal background, and metabolism. Our work is of interest for further research in the field of NMDA-mediated neurodegeneration.
Collapse
Affiliation(s)
- Yulia S Vershinina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - David G Garbuz
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina V Katunina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey B Kornev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Nadezhda V Zemskaya
- Institute of Biology, Komi Science Center, Ural Branch of RAS, 167000 Syktyvkar, Russia
| | - Alexander A Kudryavtsev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elizaveta V Bulavkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Natalia S Ulyasheva
- Institute of Biology, Komi Science Center, Ural Branch of RAS, 167000 Syktyvkar, Russia
| | - Zulfiya G Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Artemiy A Anurov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
22
|
Myasoedova VA, Parisi V, Moschetta D, Valerio V, Conte M, Massaiu I, Bozzi M, Celeste F, Leosco D, Iaccarino G, Genovese S, Poggio P. Efficacy of cardiometabolic drugs in reduction of epicardial adipose tissue: a systematic review and meta-analysis. Cardiovasc Diabetol 2023; 22:23. [PMID: 36721184 PMCID: PMC9890718 DOI: 10.1186/s12933-023-01738-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/06/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) plays an important role in cardiometabolic risk. EAT is a modifiable risk factor and could be a potential therapeutic target for drugs that already show cardiovascular benefits. The aim of this study is to evaluate the effect of cardiometabolic drugs on EAT reduction. METHODS A detailed search related to the effect on EAT reduction due to cardiometabolic drugs, such as glucagon-like peptide-1 receptor agonist (GLP-1 RA), sodium-glucose cotransporter-2 inhibitors (SGLT2-i), and statins was conducted according to PRISMA guidelines. Eighteen studies enrolling 1064 patients were included in the qualitative and quantitative analyses. RESULTS All three analyzed drug classes, in particular GLP-1 RA, show a significant effect on EAT reduction (GLP-1 RA standardize mean difference (SMD) = - 1.005; p < 0.001; SGLT2-i SMD = - 0.552; p < 0.001, and statin SMD = - 0.195; p < 0.001). The sensitivity analysis showed that cardiometabolic drugs strongly benefit EAT thickness reduction, measured by ultrasound (overall SMD of - 0.663; 95%CI - 0.79, - 0.52; p < 0.001). Meta-regression analysis revealed younger age and higher BMI as significant effect modifiers of the association between cardiometabolic drugs and EAT reduction for both composite effect and effect on EAT thickness, (age Z: 3.99; p < 0.001 and Z: 1.97; p = 0.001, respectively; BMI Z: - 4.40; p < 0.001 and Z: - 2.85; p = 0.004, respectively). CONCLUSIONS Cardiometabolic drugs show a significant beneficial effect on EAT reduction. GLP-1 RA was more effective than SGLT2-i, while statins had a rather mild effect. We believe that the most effective treatment with these drugs should target younger patients with high BMI.
Collapse
Affiliation(s)
- Veronika A Myasoedova
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy.
| | - Valentina Parisi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Donato Moschetta
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Vincenza Valerio
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Maddalena Conte
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Casa Di Cura San Michele, Maddaloni, Italy
| | - Ilaria Massaiu
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Michele Bozzi
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Fabrizio Celeste
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Stefano Genovese
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Paolo Poggio
- Unit for the Study of Aortic, Valvular, and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, Via Carlo Parea 4, 20138, Milan, Italy.
| |
Collapse
|
23
|
Dong X, Li N, Zhu C, Wang Y, Shi K, Pan H, Wang S, Shi Z, Geng Y, Wang W, Zhang T. Diagnosis of coronary artery disease in patients with type 2 diabetes mellitus based on computed tomography and pericoronary adipose tissue radiomics: a retrospective cross-sectional study. Cardiovasc Diabetol 2023; 22:14. [PMID: 36691047 PMCID: PMC9869509 DOI: 10.1186/s12933-023-01748-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) are highly susceptible to cardiovascular disease, and coronary artery disease (CAD) is their leading cause of death. We aimed to assess whether computed tomography (CT) based imaging parameters and radiomic features of pericoronary adipose tissue (PCAT) can improve the diagnostic efficacy of whether patients with T2DM have developed CAD. METHODS We retrospectively recruited 229 patients with T2DM but no CAD history (146 were diagnosed with CAD at this visit and 83 were not). We collected clinical information and extracted imaging manifestations from CT images and 93 radiomic features of PCAT from all patients. All patients were randomly divided into training and test groups at a ratio of 7:3. Four models were constructed, encapsulating clinical factors (Model 1), clinical factors and imaging indices (Model 2), clinical factors and Radscore (Model 3), and all together (Model 4), to identify patients with CAD. Receiver operating characteristic curves and decision curve analysis were plotted to evaluate the model performance and pairwise model comparisons were performed via the DeLong test to demonstrate the additive value of different factors. RESULTS In the test set, the areas under the curve (AUCs) of Model 2 and Model 4 were 0.930 and 0.929, respectively, with higher recognition effectiveness compared to the other two models (each p < 0.001). Of these models, Model 2 had higher diagnostic efficacy for CAD than Model 1 (p < 0.001, 95% CI [0.129-0.350]). However, Model 4 did not improve the effectiveness of the identification of CAD compared to Model 2 (p = 0.776); similarly, the AUC did not significantly differ between Model 3 (AUC = 0.693) and Model 1 (AUC = 0.691, p = 0.382). Overall, Model 2 was rated better for the diagnosis of CAD in patients with T2DM. CONCLUSIONS A comprehensive diagnostic model combining patient clinical risk factors with CT-based imaging parameters has superior efficacy in diagnosing the occurrence of CAD in patients with T2DM.
Collapse
Affiliation(s)
- Xiaolin Dong
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Na Li
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Chentao Zhu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Yujia Wang
- Department of Interventional and Vascular, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Ke Shi
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Hong Pan
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Shuting Wang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Zhenzhou Shi
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Yayuan Geng
- Shukun (Beijing) Network Technology Co., Ltd, Jinhui Building, Qiyang Road, Beijing, 100102 China
| | - Wei Wang
- The MRI Room, First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, NanGang District, Harbin, 150001 Heilongjiang China
| | - Tong Zhang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, No. 37, YiYuan Street, NanGang District, Harbin, 150001 Heilongjiang China
| |
Collapse
|
24
|
Walpot J, Van Herck P, Van de Heyning CM, Bosmans J, Massalha S, Malbrain ML, Heidbuchel H, Inácio JR. Computed tomography measured epicardial adipose tissue and psoas muscle attenuation: new biomarkers to predict major adverse cardiac events (MACE) and mortality in patients with heart disease and critically ill patients. Part I: Epicardial adipose tissue. Anaesthesiol Intensive Ther 2023; 55:141-157. [PMID: 37728441 PMCID: PMC10496106 DOI: 10.5114/ait.2023.130922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/28/2023] [Indexed: 09/21/2023] Open
Abstract
Over the last two decades, the potential role of epicardial adipocyte tissue (EAT) as a marker for major adverse cardiovascular events has been extensively studied. Unlike other visceral adipocyte tissues (VAT), EAT is not separated from the adjacent myocardium by a fascial layer and shares the same microcirculation with the myocardium. Adipocytokines, secreted by EAT, interact directly with the myocardium through paracrine and vasocrine pathways. The role of the Randle cycle, linking VAT accumulation to insulin resistance, and the relevance of blood flow and mitochondrial function of VAT, are briefly discussed. The three available imaging modalities for the assessment of EAT are discussed. The advantages of echocardiography, cardiac CT, and cardiac magnetic resonance (CMR) are compared. The last section summarises the current stage of knowledge on EAT as a clinical marker for major adverse cardiovascular events (MACE). The association between EAT volume and coronary artery disease (CAD) has robustly been validated. There is growing evidence that EAT volume is associated with computed tomography coronary angiography (CTCA) assessed high-risk plaque features. The EAT CT attenuation coefficient predicts coronary events. Many studies have established EAT volume as a predictor of atrial fibrillation after cardiac surgery. Moreover, EAT thickness has been independently associated with severe aortic stenosis and mitral annular calcification. Studies have demonstrated that EAT volume is associated with heart failure. Finally, we discuss the potential role of EAT in critically ill patients admitted to the intensive care unit. In conclusion, EAT seems to be a promising new biomarker to predict MACE.
Collapse
Affiliation(s)
| | - Paul Van Herck
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - Caroline M. Van de Heyning
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Bosmans
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Manu L.N.G. Malbrain
- International Fluid Academy, Lovenjoel, Belgium
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Lublin, Poland
| | - Hein Heidbuchel
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - João R. Inácio
- Centro Universitario Hospitalar Lisboa Norte, Faculdade de Medicina de Lisboa, UL, Portugal
| |
Collapse
|
25
|
Charpentier E, Redheuil A, Bourron O, Boussouar S, Lucidarme O, Zarai M, Kachenoura N, Bouazizi K, Salem JE, Hekimian G, Kerneis M, Amoura Z, Allenbach Y, Hatem S, Jeannin AC, Andreelli F, Phan F. Cardiac adipose tissue volume assessed by computed tomography is a specific and independent predictor of early mortality and critical illness in COVID-19 in type 2-diabetic patients. Cardiovasc Diabetol 2022; 21:294. [PMID: 36587209 PMCID: PMC9805370 DOI: 10.1186/s12933-022-01722-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/06/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Patients with type 2-diabetes mellitus (T2D), are characterized by visceral and ectopic adipose tissue expansion, leading to systemic chronic low-grade inflammation. As visceral adiposity is associated with severe COVID-19 irrespective of obesity, we aimed to evaluate and compare the predictive value for early intensive care or death of three fat depots (cardiac, visceral and subcutaneous) using computed tomography (CT) at admission for COVID-19 in consecutive patients with and without T2D. METHODS Two hundred and two patients admitted for COVID-19 were retrospectively included between February and June 2020 and distributed in two groups: T2D or non-diabetic controls. Chest CT with cardiac (CATi), visceral (VATi) and subcutaneous adipose tissue (SATi) volume measurements were performed at admission. The primary endpoint was a composite outcome criteria including death or ICU admission at day 21 after admission. Threshold values of adipose tissue components predicting adverse outcome were determined. RESULTS One hundred and eight controls [median age: 76(IQR:59-83), 61% male, median BMI: 24(22-27)] and ninety-four T2D patients [median age: 70(IQR:61-77), 70% male, median BMI: 27(24-31)], were enrolled in this study. At day 21 after admission, 42 patients (21%) had died from COVID-19, 48 (24%) required intensive care and 112 (55%) were admitted to a conventional care unit (CMU). In T2D, CATi was associated with early death or ICU independently from age, sex, BMI, dyslipidemia, CRP and coronary calcium (CAC). (p = 0.005). Concerning T2D patients, the cut-point for CATi was > 100 mL/m2 with a sensitivity of 0.83 and a specificity of 0.50 (AUC = 0.67, p = 0.004) and an OR of 4.71 for early ICU admission or mortality (p = 0.002) in the fully adjusted model. Other adipose tissues SATi or VATi were not significantly associated with early adverse outcomes. In control patients, age and male sex (OR = 1.03, p = 0.04) were the only predictors of ICU or death. CONCLUSIONS Cardiac adipose tissue volume measured in CT at admission was independently predictive of early intensive care or death in T2D patients with COVID-19 but not in non-diabetics. Such automated CT measurement could be used in routine in diabetic patients presenting with moderate to severe COVID-19 illness to optimize individual management and prevent critical evolution.
Collapse
Affiliation(s)
- Etienne Charpentier
- grid.411439.a0000 0001 2150 9058Sorbonne Université, Unité d’imagerie cardiovasculaire et thoracique, Hôpital La Pitié Salpêtrière (AP-HP), Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France
| | - Alban Redheuil
- grid.411439.a0000 0001 2150 9058Sorbonne Université, Unité d’imagerie cardiovasculaire et thoracique, Hôpital La Pitié Salpêtrière (AP-HP), Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France
| | - Olivier Bourron
- grid.462844.80000 0001 2308 1657Sorbonne Université, Département de diabétologie, Hôpital La Pitié Salpêtrière (AP-HP), Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France ,grid.417925.cCentre de Recherche Des Cordeliers, INSERM, UMR_S 1138, Paris, France
| | - Samia Boussouar
- grid.411439.a0000 0001 2150 9058Sorbonne Université, Unité d’imagerie cardiovasculaire et thoracique, Hôpital La Pitié Salpêtrière (AP-HP), Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France
| | - Olivier Lucidarme
- grid.462844.80000 0001 2308 1657Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France ,grid.462844.80000 0001 2308 1657Service d’imagerie specialisee et d’urgence SISU, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Mohamed Zarai
- grid.477396.80000 0004 3982 4357Institute of Cardiometabolism and Nutrition ICAN, Paris, France
| | - Nadjia Kachenoura
- grid.462844.80000 0001 2308 1657Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
| | - Khaoula Bouazizi
- grid.462844.80000 0001 2308 1657Laboratoire d’Imagerie Biomédicale, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
| | - Joe-Elie Salem
- grid.462844.80000 0001 2308 1657Department of Pharmacology, CIC-1901, INSERM, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France
| | - Guillaume Hekimian
- grid.462844.80000 0001 2308 1657Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Sorbonne Université, Paris, France
| | - Matthieu Kerneis
- grid.462844.80000 0001 2308 1657AP-HP, Hôpital La Pitié-Salpêtrière, ACTION Study Group, Département de Cardiologie, Sorbonne Université, Paris, France
| | - Zahir Amoura
- grid.462844.80000 0001 2308 1657Service de Médecine Interne 2, Centre National de Référence Maladies Systémiques Rares et Histiocytoses, Institut e3M, Hôpital de La Pitié-Salpêtrière, AP-HP, Sorbonne Université, 75013 Paris, France
| | - Yves Allenbach
- grid.462844.80000 0001 2308 1657AP-HP, Département de Médecine Interne Et Immunologie Clinique, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Stephane Hatem
- grid.477396.80000 0004 3982 4357Institute of Cardiometabolism and Nutrition ICAN, Paris, France
| | - Anne-Caroline Jeannin
- grid.462844.80000 0001 2308 1657Sorbonne Université, Département de diabétologie, Hôpital La Pitié Salpêtrière (AP-HP), Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France
| | - Fabrizio Andreelli
- grid.462844.80000 0001 2308 1657Sorbonne Université, Département de diabétologie, Hôpital La Pitié Salpêtrière (AP-HP), Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France ,grid.462844.80000 0001 2308 1657Nutrition and ObesitiesSystemic Approaches (NutriOmics) Research Unit, INSERM, UMRS U1269, Sorbonne Université, Paris, France
| | - Franck Phan
- grid.462844.80000 0001 2308 1657Sorbonne Université, Département de diabétologie, Hôpital La Pitié Salpêtrière (AP-HP), Institute of Cardiometabolism and Nutrition, Paris, France, Paris, France ,grid.417925.cCentre de Recherche Des Cordeliers, INSERM, UMR_S 1138, Paris, France
| | | |
Collapse
|
26
|
Waddell HMM, Moore MK, Herbert-Olsen MA, Stiles MK, Tse RD, Coffey S, Lamberts RR, Aitken-Buck HM. Identifying sex differences in predictors of epicardial fat cell morphology. Adipocyte 2022; 11:325-334. [PMID: 35531882 PMCID: PMC9122305 DOI: 10.1080/21623945.2022.2073854] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 11/12/2022] Open
Abstract
Predictors of overall epicardial adipose tissue deposition have been found to vary between males and females. Whether similar sex differences exist in epicardial fat cell morphology is currently unknown. This study aimed to determine whether epicardial fat cell size is associated with different clinical measurements in males and females. Fat cell sizes were measured from epicardial, paracardial, and appendix adipose tissues of post-mortem cases (N= 118 total, 37 females). Epicardial, extra-pericardial, and visceral fat volumes were measured by computed tomography from a subset of cases (N= 70, 22 females). Correlation analyses and stepwise linear regression were performed to identify predictors of fat cell size in males and females. Median fat cell sizes in all depots did not differ between males and females. Body mass index (BMI) and age were independently predictive of epicardial, paracardial, and appendix fat cell sizes in males, but not in females. Epicardial and appendix fat cell sizes were associated with epicardial and visceral fat volumes, respectively, in males only. In females, paracardial fat cell size was associated with extra-pericardial fat volume, while appendix fat cell size was associated with BMI only. No predictors were associated with epicardial fat cell size in females at the univariable or multivariable levels. To conclude, no clinical measurements were useful surrogates of epicardial fat cell size in females, while BMI, age, and epicardial fat volume were independent, albeit weak, predictors in males only.
Collapse
Affiliation(s)
- Helen M. M. Waddell
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Matthew K. Moore
- Department of Medicine, HeartOtago, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Morgan A. Herbert-Olsen
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Martin K. Stiles
- Department of Cardiology, Waikato District Health Board, Hamilton, New Zealand
- Waikato Clinical School, University of Auckland, Hamilton, New Zealand
| | - Rexson D. Tse
- Department of Forensic Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Sean Coffey
- Department of Medicine, HeartOtago, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Department of Cardiology, Dunedin Hospital, Southern District Health Board, Dunedin, New Zealand
| | - Regis R. Lamberts
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Hamish M. Aitken-Buck
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
27
|
Brettle H, Tran V, Drummond GR, Franks AE, Petrovski S, Vinh A, Jelinic M. Sex hormones, intestinal inflammation, and the gut microbiome: Major influencers of the sexual dimorphisms in obesity. Front Immunol 2022; 13:971048. [PMID: 36248832 PMCID: PMC9554749 DOI: 10.3389/fimmu.2022.971048] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is defined as the excessive accumulation of body fat and is associated with an increased risk of developing major health problems such as cardiovascular disease, diabetes and stroke. There are clear sexual dimorphisms in the epidemiology, pathophysiology and sequelae of obesity and its accompanying metabolic disorders, with females often better protected compared to males. This protection has predominantly been attributed to the female sex hormone estrogen and differences in fat distribution. More recently, the sexual dimorphisms of obesity have also been attributed to the differences in the composition and function of the gut microbiota, and the intestinal immune system. This review will comprehensively summarize the pre-clinical and clinical evidence for these sexual dimorphisms and discuss the interplay between sex hormones, intestinal inflammation and the gut microbiome in obesity. Major gaps and limitations of this rapidly growing area of research will also be highlighted in this review.
Collapse
Affiliation(s)
- Holly Brettle
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Vivian Tran
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Ashley E. Franks
- Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Steve Petrovski
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- *Correspondence: Maria Jelinic,
| |
Collapse
|
28
|
Relationship Between Epicardial Adipose Tissue and Biventricular Longitudinal Strain and Strain Rate in Patients with Type 2 Diabetes Mellitus. Acad Radiol 2022; 30:833-840. [PMID: 36115736 DOI: 10.1016/j.acra.2022.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) has been reported to be increased in patients with type 2 diabetes mellitus (T2DM). EAT thickness may impact left ventricular (LV) diastolic function. However, the association between EAT and right ventricular (RV) function in T2DM is unclear. We hypothesized an association between EAT volume and biventricular longitudinal strain and strain rate in patients with T2DM. MATERIALS AND METHODS A total of 20 controls and 69 T2DM patients with preserved LV ejection fraction (EF) who underwent cardiac magnetic resonance (CMR) were included. Biventricular function was evaluated by CMR Tissue-Tracking derived strain analysis, including LV global peak systolic longitudinal strain (LVGLS), peak diastolic longitudinal strain rate (LVLSR), RVGLS and RVLSR. RESULTS Compared to controls, patients with T2DM had significantly higher EAT volumes with lower LVGLS, LVLSR, RVGLS and RVLSR (all p<0.05). EAT volume was significantly correlated with LVGLS, LVLSR, RVGLS and RVLSR in T2DM patients (r=-0.45, -0.39, -0.59, -0.50, all p<0.001). Multivariate linear regression analysis revealed that EAT volume was significantly associated with LVGLS (β=0.38, p=0.001), LVLSR (β=-0.35, p=0.003), RVGLS (β=0.64, p<0.001) and RVLSR (β=-0.43, p<0.001) independently of traditional risk factors in patients with T2DM. CONCLUSION Patients with T2DM had higher EAT levels and lower biventricular function than controls. EAT volume was independently associated with biventricular longitudinal strain and strain rate in T2DM patients.
Collapse
|
29
|
Zhu J, Zhou W, Xie Z, Li W, Zhuo K. Impact of Sex and Menopausal Status on the Association Between Epicardial Adipose Tissue and Diastolic Function in Patients with Type 2 Diabetes Mellitus. Acad Radiol 2022; 30:823-832. [PMID: 36114077 DOI: 10.1016/j.acra.2022.08.017] [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: 06/20/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVE To evaluate the impact of sex and menopausal status on the association between the epicardial adipose tissue (EAT) volume and diastolic function in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS A total of 542 consecutive patients with T2DM were retrospectively included in this study. All patients underwent cardiac computed tomographic as well as echocardiography. To assess the independent association of EAT and diastolic function parameters, we performed a multivariate linear regression analysis. RESULTS The median EAT volume was 113.11 cm3 (interquartile range (IQR): 88.38, 148.03), and EAT volume was higher in men than in women (p < 0.05). We also discovered that EAT volume was significantly associated with diastolic function in both sexes after adjusting for risk factors (p < 0.05). Concerning menopausal status, EAT volume was higher in postmenopausal women than premenopausal women and was independently associated with the diastolic function only in postmenopausal women. CONCLUSION In patients with T2DM, EAT is independently associated with diastolic function in the male population and a portion of the female population. In contrast to premenopausal women, EAT volume is only significantly correlated with diastolic function in postmenopausal women.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wei Zhou
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Zhen Xie
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wenjia Li
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Kaimin Zhuo
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| |
Collapse
|
30
|
Yang MC, Liu HK, Tsai CC, Su YT, Wu JR. Epicardial Adipose Tissue Was Highly Associated with Reduction in Left Ventricular Diastolic Function as Early as in Adolescence. ACTA CARDIOLOGICA SINICA 2022; 38:601-611. [PMID: 36176364 PMCID: PMC9479045 DOI: 10.6515/acs.202209_38(5).20220331b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/31/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) is increased in adolescents with obesity and may play a role in early cardiovascular pathophysiological changes. There is a lack of evidence focusing on the association between EAT and cardiac function in adolescents. This study explored associations between EAT, left ventricle (LV) geometric, and LV functional changes in adolescents. METHODS Adolescent volunteers between 10 and 20 years of age were included. Body mass index (BMI) was presented as age- and sex-specific BMI z-scores. Blood samples for glucose metabolism, lipid profiles, and high-sensitivity C-reactive protein (hs-CRP) were obtained. EAT thickness, LV hypertrophy, and LV diastolic function were measured by echocardiography. RESULTS The mean age of the 276 adolescents was 13.51 ± 2.44 years. BMI z-score was strongly associated with EAT thickness (r = 0.77; p < 0.001). Multivariable analysis revealed that age, insulin resistance, total cholesterol to high-density lipoprotein cholesterol ratio, and hs-CRP were independent predictors of increased EAT thickness. After adjusting for sex, age, and BMI z-score by multivariable analysis, EAT thickness was a strong predictor of higher LV mass indexed to height2.7, higher relative wall thickness, lower mitral annulus e'/a', and higher E/e' of the mitral annulus. There was no association between EAT and LV ejection fraction. CONCLUSIONS EAT was highly associated with LV hypertrophy and reduction in LV diastolic function, independent of BMI z-score in the enrolled adolescents. Of note, the negative impacts of EAT on LV geometry and diastolic function occurred as early as in adolescence. This highlights the importance of preventing obesity and EAT deposition early in life.
Collapse
Affiliation(s)
- Ming-Chun Yang
- Department of Pediatrics, E-Da Hospital;
,
College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | | | - Ching-Chung Tsai
- Department of Pediatrics, E-Da Hospital;
,
College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Tsun Su
- Department of Pediatrics, E-Da Hospital;
,
College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | | |
Collapse
|
31
|
Krishnan A, Sharma H, Yuan D, Trollope AF, Chilton L. The Role of Epicardial Adipose Tissue in the Development of Atrial Fibrillation, Coronary Artery Disease and Chronic Heart Failure in the Context of Obesity and Type 2 Diabetes Mellitus: A Narrative Review. J Cardiovasc Dev Dis 2022; 9:jcdd9070217. [PMID: 35877579 PMCID: PMC9318726 DOI: 10.3390/jcdd9070217] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/07/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a significant burden globally and are especially prevalent in obese and/or diabetic populations. Epicardial adipose tissue (EAT) surrounding the heart has been implicated in the development of CVDs as EAT can shift from a protective to a maladaptive phenotype in diseased states. In diabetic and obese patients, an elevated EAT mass both secretes pro-fibrotic/pro-inflammatory adipokines and forms intramyocardial fibrofatty infiltrates. This narrative review considers the proposed pathophysiological roles of EAT in CVDs. Diabetes is associated with a disordered energy utilization in the heart, which promotes intramyocardial fat and structural remodeling. Fibrofatty infiltrates are associated with abnormal cardiomyocyte calcium handling and repolarization, increasing the probability of afterdepolarizations. The inflammatory phenotype also promotes lateralization of connexin (Cx) proteins, undermining unidirectional conduction. These changes are associated with conduction heterogeneity, together creating a substrate for atrial fibrillation (AF). EAT is also strongly implicated in coronary artery disease (CAD); inflammatory adipokines from peri-vascular fat can modulate intra-luminal homeostasis through an “outside-to-inside” mechanism. EAT is also a significant source of sympathetic neurotransmitters, which promote progressive diastolic dysfunction with eventual cardiac failure. Further investigations on the behavior of EAT in diabetic/obese patients with CVD could help elucidate the pathogenesis and uncover potential therapeutic targets.
Collapse
Affiliation(s)
- Anirudh Krishnan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Harman Sharma
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Daniel Yuan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Alexandra F. Trollope
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia;
| | - Lisa Chilton
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia
- Correspondence:
| |
Collapse
|
32
|
Measurement of epicardial adipose tissue using non-contrast routine chest-CT: a consideration of threshold adjustment for fatty attenuation. BMC Med Imaging 2022; 22:114. [PMID: 35752770 PMCID: PMC9233319 DOI: 10.1186/s12880-022-00840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/03/2022] [Indexed: 11/15/2022] Open
Abstract
Background Epicardial adipose tissue (EAT) is known as an important imaging indicator for cardiovascular risk stratification. The present study aimed to determine whether the EAT volume (EV) and mean EAT attenuation (mEA) measured by non-contrast routine chest CT (RCCT) could be more consistent with those measured by coronary CT angiography (CCTA) by adjusting the threshold of fatty attenuation. Methods In total, 83 subjects who simultaneously underwent CCTA and RCCT were enrolled. EV and mEA were quantified by CCTA using a threshold of (N30) (− 190 HU, − 30 HU) as a reference and measured by RCCT using thresholds of N30, N40 (− 190 HU, − 40 HU), and N45 (− 190 HU, − 45 HU). The correlation and agreement of EAT metrics between the two imaging modalities and differences between patients with coronary plaques (plaque ( +)) and without plaques (plaque ( −)) were analyzed. Results EV obtained from RCCT showed very strong correlation with the reference (r = 0.974, 0.976, 0.972 (N30, N40, N45), P < 0.001), whereas mEA showed a moderate correlation (r = 0.516, 0.500, 0.477 (N30, N40, N45), P < 0.001). Threshold adjustment was able to reduce the bias of EV, while increase the bias of mEA. Data obtained by CCTA and RCCT both demonstrated a significantly larger EV in the plaque ( +) group than in the plaque ( −) group (P < 0.05). A significant difference in mEA was shown only by RCCT using a threshold of N30 (plaque ( +) vs ( −): − 80.0 ± 4.4 HU vs − 78.0 ± 4.0 HU, P = 0.030). The mEA measured on RCCT using threshold of N40 and N45 showed no significant statistical difference between the two groups (P = 0.092 and 0.075), which was consistent with the result obtained on CCTA (P = 0.204). Conclusion Applying more negative threshold, the consistency of EV measurements between the two techniques improves and a consistent result can be obtained when comparing EF measurements between groups, although the bias of mEA increases. Threshold adjustment is necessary when measuring EF with non-contrast RCCT.
Collapse
|
33
|
Zhu J, Zhuo K, Zhang B, Xie Z, Li W. Sex Differences in Epicardial Adipose Tissue: Association With Atrial Fibrillation Ablation Outcomes. Front Cardiovasc Med 2022; 9:905351. [PMID: 35770221 PMCID: PMC9234200 DOI: 10.3389/fcvm.2022.905351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Background There are significant differences in the prevalence and prognosis of atrial fibrillation (AF) between sexes. Epicardial adipose tissue (EAT) has been found as a risk factor for AF. This study aimed to evaluate whether sex-based EAT differences were correlated with AF recurrence and major adverse cardiovascular events (MACE). Methods In this study, postmenopausal women and age, BMI, and type of AF matched men who had received first catheter ablation were included. EAT volume was quantified based on the pre-ablation cardiac computed tomography (CT) images. Clinical, CT, and echocardiographic variables were compared by sex groups. The predictors of AF recurrence and MACE were determined through Cox proportional hazards regression. Results Women were found with significantly lower total EAT volumes (P < 0.001) but higher periatrial/total (P/T) EAT ratios (P = 0.009). The median follow-up duration was 444.5 days. As revealed by the result of the Kaplan-Meier survival analysis, the women were found to have a significantly higher prevalence of AF recurrence (log rank, P = 0.011) but comparable MACE (log rank, P = 0.507) than men. Multivariate analysis demonstrated that female gender (HR: 1.88 [95% CI: 1.03, 4.15], P = 0.032), persistent AF (HR: 2.46 [95% CI: 1.19, 5.05], P = 0.015), left atrial (LA) dimension (HR: 1.47 [95% CI: 1.02, 2.13], P = 0.041), and P/T EAT ratio (HR: 1.73 [95% CI: 1.12, 2.67], P = 0.013) were found as the independent predictors of AF recurrence. Sex-based subgroup multivariable analysis showed that the P/T EAT ratio was an independent predictor of AF recurrence in both men (HR: 1.13 [95% CI: 1.01, 1.46], P = 0.047) and women (HR: 1.37 [95% CI: 1.11, 1.67], P = 0.028). While age (HR: 1.81 [95% CI: 1.18, 2.77], P = 0.007), BMI (HR: 1.44 [95% CI: 1.02, 2.03], P = 0.038), and periatrial EAT volume (HR: 1.31 [95% CI: 1.01, 1.91], P = 0.046) were found to be independent of MACE. Conclusion Women had a higher P/T EAT ratio and AF post-ablation recurrence but similar MACE as compared with men. Female gender and P/T EAT ratio were found to be independent predictors of AF recurrence, whereas age and periatrial EAT volume were found to be independent predictors of MACE.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- *Correspondence: Jing Zhu
| | - Kaimin Zhuo
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Bo Zhang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Zhen Xie
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wenjia Li
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
34
|
Liu J, Gao A, Liu Y, Sun Y, Zhang D, Lin X, Hu C, Zhu Y, Du Y, Han H, Li Y, Xu S, Liu T, Zhang C, Zhu J, Dong R, Zhou Y, Zhao Y. MicroRNA Expression Profiles of Epicardial Adipose Tissue-Derived Exosomes in Patients with Coronary Atherosclerosis. Rev Cardiovasc Med 2022; 23:206. [PMID: 39077165 PMCID: PMC11273655 DOI: 10.31083/j.rcm2306206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND AND AIMS Epicardial adipose tissue, exosomes, and miRNAs have important activities in atherosclerosis. The purpose of this study was to establish miRNA expression profiles of epicardial adipose tissue-derived exosomes in patients with coronary atherosclerosis. METHODS Biopsies of epicardial adipose tissue were obtained from patients with and without coronary artery disease (CAD, n = 12 and NCAD, n = 12) during elective open-heart surgeries. Tissue was incubated with DMEM-F12 for 24 hours. Exosomes were isolated, then nanoparticle tracking analysis, transmission electron microscopy, and immunoblotting were performed to confirm the existence of exosomes. Total RNA in exosomes was subjected to high-throughput sequencing to identify differentially expressed miRNAs. MicroRNA target gene prediction was performed, and target genes were analyzed by Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and mirPath to identify function. Reverse transcription quantitative PCR was performed to confirm the differentially expressed miRNAs. RESULTS Fifty-three unique miRNAs were identified (adjusted p < 0.05, fold of change > 2), among which 32 miRNAs were upregulated and 21 miRNAs were downregulated in coronary artery disease patients. Reverse transcription quantitative PCR validated the results for seven miRNAs including miR-141-3p, miR-183-5p, miR-200a-5p, miR-205-5p, miR-429, miR-382-5p and miR-485-3p, with the last two downregulated. GO and KEGG analysis by mirPath indicated that these differentially expressed miRNAs were enriched in cell survival, apoptosis, proliferation, and differentiation. CONCLUSIONS Coronary artery disease patients showed differential epicardial adipose tissue exosomal miRNA expression compared with patients without coronary artery disease. The results provide clues for further studies of mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jinxing Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Ang Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yan Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yan Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Dai Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Xuze Lin
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Chengping Hu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yong Zhu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yu Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Hongya Han
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yang Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Shijun Xu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Taoshuai Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chenhan Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Junming Zhu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Ran Dong
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| | - Yingxin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
35
|
Wang X, Tan Y, Liu D, Shen H, Deng Y, Tan Y, Wang L, Zhang Y, Ma X, Zeng X, Zhang J. Chemotherapy-associated steatohepatitis was concomitant with epicardial adipose tissue volume increasing in breast cancer patients who received neoadjuvant chemotherapy. Eur Radiol 2022; 32:4898-4908. [PMID: 35394181 DOI: 10.1007/s00330-022-08581-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To investigate the prevalence of chemotherapy-associated steatohepatitis, quantitate the epicardial adipose tissue (EAT) volume in breast cancer patients, and explore the mediating effect of liver fat content on EAT volume in breast cancer patients who received neoadjuvant chemotherapy (NAC). METHODS From October 2018 to April 2020, patients were retrospectively reviewed and divided into breast cancer non-NAC and NAC groups. The prevalence of chemotherapy-associated steatohepatitis was evaluated through quantitative MRI mDIXON-Quant examinations by using defined proton density fat fraction cutoffs of liver fat. The EAT volume was quantified on chest CT by semi-automatic volume analysis software. Bootstrap analysis was used in the breast cancer NAC group to test the significance of the mediating effect of liver fat content on EAT volume. RESULTS A total of 662 breast cancer patients (non-NAC group: 445 patients; NAC group: 217 patients) were included. The prevalence of chemotherapy-associated steatohepatitis in the NAC group was significantly higher than the prevalence of hepatic steatosis in the non-NAC group (42.8% vs. 33.3%, p < 0.001). EAT volume was measured in 561 of 662 breast cancer patients, and was significantly higher in the NAC group than in the non-NAC group (137.26 ± 53.48 mL vs. 125.14 ± 58.77 mL, p = 0.020). In the breast cancer NAC group, the indirect effect of liver fat content on EAT volume was 2.545 (p < 0.001), and the contribution rate to the effect was 69.1%. CONCLUSIONS EAT volume was significantly higher in the BC-NAC group than in the BC-non-NAC group. KEY POINTS • The prevalence of CASH was as high as 42.8% in BC patients. • NAC significantly increased the EAT volume in BC patients. • The liver fat content caused the change of EAT volume through mediating effect.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yuchuan Tan
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yongchun Deng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, People's Republic of China
| | - Yong Tan
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Lei Wang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yipeng Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Xin Ma
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, People's Republic of China.
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
36
|
Phang RJ, Ritchie RH, Hausenloy DJ, Lees JG, Lim SY. Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy. Cardiovasc Res 2022; 119:668-690. [PMID: 35388880 PMCID: PMC10153440 DOI: 10.1093/cvr/cvac049] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/16/2022] [Accepted: 03/05/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with Type 2 diabetes mellitus (T2DM) frequently exhibit a distinctive cardiac phenotype known as diabetic cardiomyopathy. Cardiac complications associated with T2DM include cardiac inflammation, hypertrophy, fibrosis and diastolic dysfunction in the early stages of the disease, which can progress to systolic dysfunction and heart failure. Effective therapeutic options for diabetic cardiomyopathy are limited and often have conflicting results. The lack of effective treatments for diabetic cardiomyopathy is due in part, to our poor understanding of the disease development and progression, as well as a lack of robust and valid preclinical human models that can accurately recapitulate the pathophysiology of the human heart. In addition to cardiomyocytes, the heart contains a heterogeneous population of non-myocytes including fibroblasts, vascular cells, autonomic neurons and immune cells. These cardiac non-myocytes play important roles in cardiac homeostasis and disease, yet the effect of hyperglycaemia and hyperlipidaemia on these cell types are often overlooked in preclinical models of diabetic cardiomyopathy. The advent of human induced pluripotent stem cells provides a new paradigm in which to model diabetic cardiomyopathy as they can be differentiated into all cell types in the human heart. This review will discuss the roles of cardiac non-myocytes and their dynamic intercellular interactions in the pathogenesis of diabetic cardiomyopathy. We will also discuss the use of sodium-glucose cotransporter 2 inhibitors as a therapy for diabetic cardiomyopathy and their known impacts on non-myocytes. These developments will no doubt facilitate the discovery of novel treatment targets for preventing the onset and progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rebecca H Ritchie
- School of Biosciences, Parkville, Victoria 3010, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung City, Taiwan
| | - Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
37
|
Iacobellis G, Basilico S, Malavazos AE. Targeting Epicardial Fat in Obesity and Diabetes Pharmacotherapy. Handb Exp Pharmacol 2022; 274:93-108. [PMID: 35156138 DOI: 10.1007/164_2021_577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Epicardial adipose tissue surrounds and infiltrates the heart. Epicardial fat displays unique anatomic, genetic, and biomolecular properties. People with obesity and in particular, those with abdominal obesity and associated type 2 diabetes mellitus, have an increased amount of epicardial adipose tissue (EAT). Epicardial fat works well as therapeutic target due to its fast-responding metabolism, organ fat specificity, and easy measurability. Epicardial fat responds to thiazolidinediones (TZD), glucagon-like peptide 1-receptor agonists (GLP1A), sodium-glucose cotransporter 2 inhibitors (SGLT2i), dipeptidyl peptidase-4 inhibitors (DPP4i), and statins. Modulating epicardial fat morphology and genetic profile with targeted pharmacological agents suggests novel strategies in the pharmacotherapy of diabetes and obesity.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Sara Basilico
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
38
|
Predictors of Epicardial Fat Volume Decrease after Dapagliflozin Treatment in Patients with Type 2 Diabetes. Medicina (B Aires) 2021; 58:medicina58010021. [PMID: 35056329 PMCID: PMC8777745 DOI: 10.3390/medicina58010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Objectives: Dapagliflozin treatment proved to reduce the epicardial fat volume (EFV) in patients with type 2 diabetes (T2D). Despite the reduction in EFV being associated with improved diastolic function in patients with T2D, EVF is not routinely evaluated in T2D because it is costly and involves radiation exposure. This study aims to identify biomarkers that predict EFV reduction after dapagliflozin treatment in patients with T2D. Materials and Methods: In a prospective, observational, consecutive-case enrollment scenario, 52 patients with T2D were initiated on dapagliflozin 10 mg q.d. as part of the standard of care. At enrollment and after six months of dapagliflozin treatment, patients were evaluated using cardiac ultrasonography, native computer tomography, transient liver elastography, and metabolic lab tests. In addition, the atherogenic index of plasma (AIP), atherogenic coefficient (AC), triglyceride glucose index (TyG), cardiac risk ratio (CRR), and visceral abdominal index (VAI) were calculated. Results: Higher AIP (r = 0.28; p = 0.04), CRR (r = 0.28; p = 0.04), and TyG (r = 0.32; p = 0.01) are associated with more important reductions in the EFV. A lower conicity index (β = −0.29; p = 0.03), visceral fat volume at the 4th vertebrae (L4VFV) (β = −0.32; p = 0.02), left atrium volume (β = −3.08; p = 0.003), and right ventricle diameter (β = −2.13; p = 0.04) are associated with higher reductions in the EFV after six months of dapagliflozin treatment. A valid performance for predicting clinically relevant decreases in EFV after dapagliflozin treatment was observed for AIP (AUROC = 0.903; Youden = 0.732; p < 0.001), CRR (AUROC = 0.772; Youden = 0.595; p = 0.004), TyG (AUROC = 0.957; Youden = 0.904; p < 0.001), and VAI (AUROC = 0.898; Youden = 0.712; p < 0.001). Conclusion: Higher initial EFV values are associated with more important reductions in EFV in patients with T2D treated for six months with dapagliflozin. TyG values have the best prediction performances for EFV reduction, having the highest sum of sensitivity and specificity at the 0.904 threshold level. AIP, CRR, VAI, conicity index, L4VF, left atrium volume, and right ventricle volume are valid biomarkers for a decrease in EFV after dapagliflozin treatment in diabetes patients.
Collapse
|
39
|
van Woerden G, van Veldhuisen DJ, Manintveld OC, van Empel VPM, Willems TP, de Boer RA, Rienstra M, Westenbrink BD, Gorter TM. Epicardial Adipose Tissue and Outcome in Heart Failure With Mid-Range and Preserved Ejection Fraction. Circ Heart Fail 2021; 15:e009238. [PMID: 34935412 PMCID: PMC8920003 DOI: 10.1161/circheartfailure.121.009238] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Supplemental Digital Content is available in the text. Epicardial adipose tissue (EAT) accumulation is thought to play a role in the pathophysiology of heart failure (HF) with mid-range and preserved ejection fraction, but its effect on outcome is unknown. We evaluated the prognostic value of EAT volume measured with cardiac magnetic resonance in patients with HF with mid-range ejection fraction and HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Gijs van Woerden
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| | - Olivier C Manintveld
- Department of Cardiology, University of Rotterdam, Erasmus Medical Center Rotterdam, the Netherlands (O.C.M.)
| | - Vanessa P M van Empel
- Department of Cardiology, University of Maastricht, Medical University Center Maastricht, the Netherlands (V.P.M.v.E.)
| | - Tineke P Willems
- Department of Radiology, University of Groningen, University Medical Center Groningen, the Netherlands. (T.P.W.)
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| | - B Daan Westenbrink
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| | - Thomas M Gorter
- Department of Cardiology, University of Groningen, University Medical Center Groningen, the Netherlands. (G.v.W., D.J.v.V., R.A.d.B., M.R., B.D.W., T.M.G.)
| |
Collapse
|
40
|
Rasmussen IKB, Zobel EH, Ripa RS, von Scholten BJ, Curovic VR, Jensen JK, Kjaer A, Hansen TW, Rossing P. Liraglutide reduces cardiac adipose tissue in type 2 diabetes: A secondary analysis of the LIRAFLAME randomized placebo-controlled trial. Diabetes Obes Metab 2021; 23:2651-2659. [PMID: 34387408 DOI: 10.1111/dom.14516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 01/13/2023]
Abstract
AIM To test the hypothesis that treatment with liraglutide can reduce cardiac adipose tissue. MATERIALS AND METHODS LIRAFLAME is a randomized placebo-controlled, double-blind, parallel clinical study. Participants with type 2 diabetes were randomized to treatment with liraglutide 1.8 mg/d or placebo for 26 weeks. Computed tomography was performed at baseline and at end of treatment to evaluate the cardiac adipose tissue volume, quantified automatically. We report the results of a secondary endpoint evaluating changes in cardiac adipose tissue. RESULTS A total of 102 participants were randomly assigned to liraglutide (n = 51) or placebo (n = 51). At baseline, the mean (SD) cardiac adipose tissue volume was comparable between the liraglutide and the placebo group (232.6 [112.8] vs. 227.0 [103.2] mL; P = 0.80). The mean change in body weight was -3.7 (-4.8, -2.6) kg in the liraglutide and -0.18 (-0.76, 0.40) kg in the placebo group. From baseline to end of treatment the mean cardiac adipose tissue change was -11.5 (95% confidence interval -17.6, -5.4) mL in the liraglutide (P < 0.001) and -0.01 (-5.3, 5.3) mL in the placebo (P = 1.00) groups. The reduction in cardiac adipose tissue was significantly greater in the liraglutide compared to the placebo group (mean difference -11.4 [-19.4, -3.3] mL; P = 0.006), but significance was lost after adjustment for changes in body mass index (P = 0.46). CONCLUSION Treatment with liraglutide for 26 weeks was associated with a reduction in cardiac adipose tissue compared to placebo. The reduction was not independent of weight loss, suggesting that this is not a drug-specific effect.
Collapse
Affiliation(s)
| | | | - Rasmus S Ripa
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Kobenhavn, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Bernt J von Scholten
- Steno Diabetes Centre Copenhagen, Gentofte, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | | | - Jacob K Jensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Kobenhavn, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Kobenhavn, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | | - Peter Rossing
- Steno Diabetes Centre Copenhagen, Gentofte, Denmark
- University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Cosson E, Nguyen MT, Rezgani I, Berkane N, Pinto S, Bihan H, Tatulashvili S, Taher M, Sal M, Soussan M, Brillet PY, Valensi P. Epicardial adipose tissue volume and myocardial ischemia in asymptomatic people living with diabetes: a cross-sectional study. Cardiovasc Diabetol 2021; 20:224. [PMID: 34819079 PMCID: PMC8613918 DOI: 10.1186/s12933-021-01420-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/14/2021] [Indexed: 12/18/2022] Open
Abstract
Background Epicardial adipose tissue (EAT) is considered a novel diagnostic marker for cardiometabolic disease. This study aimed to evaluate whether EAT volume was associated with stress-induced myocardial ischemia in asymptomatic people living with diabetes—independently of confounding factors—and whether it could predict this condition. Methods We included asymptomatic patients with diabetes and no coronary history, who had undergone both a stress a myocardial scintigraphy to diagnose myocardial ischemia, and a computed tomography to measure their coronary artery calcium (CAC) score. EAT volume was retrospectively measured from computed tomography imaging. Determinants of EAT volume and asymptomatic myocardial ischemia were evaluated. Results The study population comprised 274 individuals, including 153 men. Mean (± standard deviation) age was 62 ± 9 years, and 243, 23 and 8 had type 2, type 1, or another type of diabetes, respectively. Mean body mass index was 30 ± 6 kg/m2, and mean EAT volume 96 ± 36 cm3. Myocardial ischemia was detected in 32 patients (11.7%). EAT volume was positively correlated with age, body mass index and triglyceridemia, but negatively correlated with HbA1c, HDL- and LDL-cholesterol levels. Furthermore, EAT volume was lower in people with retinopathy, but higher in men, in current smokers, in patients with nephropathy, those with a CAC score > 100 Agatston units, and finally in individuals with myocardial ischemia (110 ± 37 cm3 vs 94 ± 37 cm3 in those without myocardial ischemia, p < 0.05). The association between EAT volume and myocardial ischemia remained significant after adjustment for gender, diabetes duration, peripheral macrovascular disease and CAC score. We also found that area under the ROC curve analysis showed that EAT volume (AROC: 0.771 [95% confidence interval 0.683–0.858]) did not provide improved discrimination of myocardial ischemia over the following classic factors: gender, diabetes duration, peripheral macrovascular disease, retinopathy, nephropathy, smoking, atherogenic dyslipidemia, and CAC score (AROC 0.773 [0.683–0.862]). Conclusions EAT may play a role in coronary atherosclerosis and coronary circulation in patients with diabetes. However, considering EAT volume is not a better marker for discriminating the risk of asymptomatic myocardial ischemia than classic clinical data.
Collapse
Affiliation(s)
- Emmanuel Cosson
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France. .,Unité de Recherche Epidémiologique Nutritionnelle, UMR U1153 INSERM/U11125 INRA/CNAM/Université Paris 13, Bobigny, France.
| | - Minh Tuan Nguyen
- Unit of Endocrinology-Diabetology-Nutrition, Jean Verdier Hospital, AP-HP, Université Paris 13, Bondy, France
| | - Imen Rezgani
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France
| | - Narimane Berkane
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France
| | - Sara Pinto
- Unit of Diabetology, Jean Verdier Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, Bondy, France
| | - Hélène Bihan
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France.,Laboratoire Educations et Pratiques de Santé UR 3412, UFR Santé, Médecine, Biologie Humaine, Université Paris Sorbonne Paris Nord, 74, Rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Sopio Tatulashvili
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France
| | - Malak Taher
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France
| | - Meriem Sal
- Department of Endocrinology-Diabetology-Nutrition, Avicenne Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, 125 Rue de Stalingrad, 93000, Bobigny Cedex, France
| | - Michael Soussan
- Department of Nuclear Medicine, Avicenne Hospital, AP-HP, Bobigny, France
| | | | - Paul Valensi
- Unit of Diabetology, Jean Verdier Hospital, CRNH-IdF, CINFO, AP-HP, Université Paris 13, Sorbonne Paris Cité, Bondy, France
| |
Collapse
|
42
|
Phan F, Boussouar S, Lucidarme O, Zarai M, Salem JE, Kachenoura N, Bouazizi K, Charpentier E, Niati Y, Bekkaoui H, Amoura Z, Mathian A, Benveniste O, Cacoub P, Allenbach Y, Saadoun D, Lacorte JM, Fourati S, Laroche S, Hartemann A, Bourron O, Andreelli F, Redheuil A. Cardiac adipose tissue volume and IL-6 level at admission are complementary predictors of severity and short-term mortality in COVID-19 diabetic patients. Cardiovasc Diabetol 2021; 20:165. [PMID: 34384426 PMCID: PMC8358546 DOI: 10.1186/s12933-021-01327-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND COVID-19 diabetic adults are at increased risk of severe forms irrespective of obesity. In patients with type-II diabetes, fat distribution is characterized by visceral and ectopic adipose tissues expansion, resulting in systemic inflammation, which may play a role in driving the COVID-19 cytokine storm. Our aim was to determine if cardiac adipose tissue, combined to interleukin-6 levels, could predict adverse short-term outcomes, death and ICU requirement, in COVID-19 diabetic patients during the 21 days after admission. METHODS Eighty one consecutive patients with type-II diabetes admitted for COVID-19 were included. Interleukin-6 measurement and chest computed tomography with total cardiac adipose tissue index (CATi) measurement were performed at admission. The primary outcome was death during the 21 days following admission while intensive care requirement with or without early death (ICU-R) defined the secondary endpoint. Associations of CATi and IL-6 and threshold values to predict the primary and secondary endpoints were determined. RESULTS Of the enrolled patients (median age 66 years [IQR: 59-74]), 73% male, median body mass index (BMI) 27 kg/m2 [IQR: 24-31]) 20 patients had died from COVID-19, 20 required intensive care and 41 were in conventional care at day 21 after admission. Increased CATi and IL-6 levels were both significantly related to increased early mortality (respectively OR = 6.15, p = 0.002; OR = 18.2, p < 0.0001) and ICU-R (respectively OR = 3.27, p = 0.01; OR = 4.86, p = 0.002). These associations remained significant independently of age, sex, BMI as well as troponin-T level and pulmonary lesion extension in CT. We combined CATi and IL-6 levels as a multiplicative interaction score (CATi*IL-6). The cut-point for this score was ≥ 6386 with a sensitivity of 0.90 and a specificity of 0.87 (AUC = 0.88) and an OR of 59.6 for early mortality (p < 0.0001). CONCLUSIONS Cardiac adipose tissue index and IL-6 determination at admission could help physicians to better identify diabetic patients with a potentially severe and lethal short term course irrespective of obesity. Diabetic patients with high CATi at admission, a fortiori associated with high IL-6 levels could be a relevant target population to promptly initiate anti-inflammatory therapies.
Collapse
Affiliation(s)
- Franck Phan
- Sorbonne Université, Paris, France.,Assistance Publique-Hôpitaux de Paris (APHP), Diabetology Department, La Pitié Salpêtrière-Charles Foix University Hospital, Paris, France.,Centre de Recherche des Cordeliers, INSERM, UMR_S 1138, Paris 06, France.,Institute of Cardiometabolism and Nutrition ICAN, Paris, France
| | - Samia Boussouar
- Cardiovascular and Thoracic Imaging Unit, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France.,Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France.,Service d'imagerie Spécialisée et d'urgence SISU, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Olivier Lucidarme
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France.,Service d'imagerie Spécialisée et d'urgence SISU, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Mohamed Zarai
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Joe-Elie Salem
- Department of Pharmacology, CIC-1901, INSERM, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Nadjia Kachenoura
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Khaoula Bouazizi
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Etienne Charpentier
- Cardiovascular and Thoracic Imaging Unit, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France.,Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France.,Service d'imagerie Spécialisée et d'urgence SISU, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Yasmine Niati
- Cardiovascular and Thoracic Imaging Unit, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France
| | - Hasnae Bekkaoui
- Cardiovascular and Thoracic Imaging Unit, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France
| | - Zahir Amoura
- Service de Médecine Interne 2, Centre National de Référence Maladies Systémiques Rares et Histiocytoses, Institut e3M, Hôpital de La Pitié-Salpêtrière, AP-HP, Sorbonne Université, 75013, Paris, France
| | - Alexis Mathian
- Service de Médecine Interne 2, Centre National de Référence Maladies Systémiques Rares et Histiocytoses, Institut e3M, Hôpital de La Pitié-Salpêtrière, AP-HP, Sorbonne Université, 75013, Paris, France
| | - Olivier Benveniste
- Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Patrice Cacoub
- Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Yves Allenbach
- Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - David Saadoun
- Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Jean-Marc Lacorte
- Department of Endocrine and Oncologic Biochemistry, Inserm, UMR_S 1166, Research Institute of Cardiovascular Disease, Metabolism and Nutrition, Paris, France
| | - Salma Fourati
- Department of Endocrine and Oncologic Biochemistry, Inserm, UMR_S 1166, Research Institute of Cardiovascular Disease, Metabolism and Nutrition, Paris, France
| | - Suzanne Laroche
- Sorbonne Université, Paris, France.,Assistance Publique-Hôpitaux de Paris (APHP), Diabetology Department, La Pitié Salpêtrière-Charles Foix University Hospital, Paris, France
| | - Agnes Hartemann
- Sorbonne Université, Paris, France.,Assistance Publique-Hôpitaux de Paris (APHP), Diabetology Department, La Pitié Salpêtrière-Charles Foix University Hospital, Paris, France.,Centre de Recherche des Cordeliers, INSERM, UMR_S 1138, Paris 06, France.,Institute of Cardiometabolism and Nutrition ICAN, Paris, France
| | - Olivier Bourron
- Sorbonne Université, Paris, France.,Assistance Publique-Hôpitaux de Paris (APHP), Diabetology Department, La Pitié Salpêtrière-Charles Foix University Hospital, Paris, France.,Centre de Recherche des Cordeliers, INSERM, UMR_S 1138, Paris 06, France.,Institute of Cardiometabolism and Nutrition ICAN, Paris, France
| | - Fabrizio Andreelli
- Sorbonne Université, Paris, France.,Assistance Publique-Hôpitaux de Paris (APHP), Diabetology Department, La Pitié Salpêtrière-Charles Foix University Hospital, Paris, France.,Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, Sorbonne Université, INSERM, UMRS U1269, Paris, France
| | - Alban Redheuil
- Cardiovascular and Thoracic Imaging Unit, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France. .,Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Institute of Cardiometabolism and Nutrition, Paris, France. .,Service d'imagerie Spécialisée et d'urgence SISU, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, INSERM, CNRS, Paris, France.
| | | |
Collapse
|
43
|
Packer M. Differential Pathophysiological Mechanisms in Heart Failure With a Reduced or Preserved Ejection Fraction in Diabetes. JACC-HEART FAILURE 2021; 9:535-549. [PMID: 34325884 DOI: 10.1016/j.jchf.2021.05.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Diabetes promotes the development of both heart failure with a reduced ejection fraction and heart failure with a preserved ejection fraction through diverse mechanisms, which are likely mediated through hyperinsulinemia rather than hyperglycemia. Diabetes promotes nutrient surplus signaling (through Akt and mammalian target of rapamycin complex 1) and inhibits nutrient deprivation signaling (through sirtuin-1 and its downstream effectors); this suppresses autophagy and promotes endoplasmic reticulum and oxidative stress and mitochondrial dysfunction, thereby undermining the health of diabetic cardiomyocytes. The hyperinsulinemia of diabetes may also activate sodium-hydrogen exchangers in cardiomyocytes (leading to injury and loss) and in the proximal renal tubules (leading to sodium retention). Diabetes may cause epicardial adipose tissue expansion, and the resulting secretion of proinflammatory adipocytokines onto the adjoining myocardium can lead to coronary microcirculatory dysfunction and myocardial inflammation and fibrosis. Interestingly, sodium-glucose cotransporter 2 (SGLT2) inhibitors-the only class of antidiabetic medication that reduces serious heart failure events-may act to mitigate each of these mechanisms. SGLT2 inhibitors up-regulate sirtuin-1 and its downstream effectors and autophagic flux, thus explaining the actions of these drugs to reduce oxidative stress, normalize mitochondrial structure and function, and mute proinflammatory pathways in the stressed myocardium. Inhibition of SGLT2 may also lead to a reduction in the activity of sodium-hydrogen exchangers in the kidney (leading to diuresis) and in the heart (attenuating the development of cardiac hypertrophy and systolic dysfunction). Finally, SGLT2 inhibitors reduce the mass and mute the adverse biology of epicardial adipose tissue (and reduce the secretion of leptin), thus explaining the capacity of these drugs to mitigate myocardial inflammation, microcirculatory dysfunction, and fibrosis, and improve ventricular filling dynamics. The pathophysiological mechanisms by which SGLT2 inhibitors may benefit heart failure likely differ depending on ejection fraction, but each represents interference with distinct pathways by which hyperinsulinemia may adversely affect cardiac structure and function.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas, USA; Imperial College, London, United Kingdom.
| |
Collapse
|
44
|
Abstract
BACKGROUND Both visceral adipose tissue and epicardial adipose tissue (EAT) have pro-inflammatory properties. The former is associated with Coronavirus Disease 19 (COVID-19) severity. We aimed to investigate whether an association also exists for EAT. MATERIAL AND METHODS We retrospectively measured EAT volume using computed tomography (CT) scans (semi-automatic software) of inpatients with COVID-19 and analyzed the correlation between EAT volume and anthropometric characteristics and comorbidities. We then analyzed the clinicobiological and radiological parameters associated with severe COVID-19 (O2 [Formula: see text] 6 l/min), intensive care unit (ICU) admission or death, and 25% or more CT lung involvement, which are three key indicators of COVID-19 severity. RESULTS We included 100 consecutive patients; 63% were men, mean age was 61.8 ± 16.2 years, 47% were obese, 54% had hypertension, 42% diabetes, and 17.2% a cardiovascular event history. Severe COVID-19 (n = 35, 35%) was associated with EAT volume (132 ± 62 vs 104 ± 40 cm3, p = 0.02), age, ferritinemia, and 25% or more CT lung involvement. ICU admission or death (n = 14, 14%) was associated with EAT volume (153 ± 67 vs 108 ± 45 cm3, p = 0.015), hypertension and 25% or more CT lung involvement. The association between EAT volume and severe COVID-19 remained after adjustment for sex, BMI, ferritinemia and lung involvement, but not after adjustment for age. Instead, the association between EAT volume and ICU admission or death remained after adjustment for all five of these parameters. CONCLUSIONS Our results suggest that measuring EAT volume on chest CT scans at hospital admission in patients diagnosed with COVID-19 might help to assess the risk of disease aggravation.
Collapse
|
45
|
Ferreira J, Martins R, Monteiro S, Teixeira R, Gonçalves L. Alternative sites of echocardiographic epicardial fat assessment and coronary artery disease. J Ultrasound 2021; 25:177-184. [PMID: 34105055 DOI: 10.1007/s40477-021-00598-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022] Open
Abstract
AIMS Increasing evidence points towards the use of epicardial fat (EF) as a reliable biomarker of coronary artery disease extent and severity. We aim to assess the different locations of echocardiographic EF thickness measurement and their relation with the presence, extent, and severity of coronary artery disease (CAD) in patients admitted with acute coronary syndromes (ACS). METHODS Prospective cohort study including patients admitted for ACS. EF was assessed by transthoracic echocardiography and compared with coronary angiography findings. Spearmen correlation analysis was used to search for EF correlations. Receiver-operating characteristic curve analysis was performed to assess the predictive value of the different sites of measurement of EF thickness for the presence of CAD. To evaluate other potential variables independently associated with CAD, we performed multivariate analysis employing logistic regression. RESULTS 196 patients were included. Significant CAD was diagnosed in 83.7% of patients. In all views, EF thickness was greater in patients with CAD (p < 0.001). We found a moderate correlation between EF thickness and CAD extent and severity. EF thickness measured at RV basal level showed a good performance in predicting significant CAD in patients with ACS (AUC = 0.885, 95% CI 0.80-0.97, p < 0.001). For a value of mean RV basal region EF thickness ≥ 12.57 mm, sensitivity was 85% and specificity was 80.8%. CONCLUSION In patients admitted with ACS, echocardiographic EF thickness predicted the presence of CAD, as well as its extent and severity. We found EF thickness measured at the RV basal region to be the best predictor of significant CAD.
Collapse
Affiliation(s)
- João Ferreira
- Serviço de Cardiologia, Centro Hospitalar e Universitário de Coimbra, Praceta, R. Prof. Mota Pinto, 3004-561, Coimbra, Portugal.
| | - Rui Martins
- Serviço de Cardiologia, Centro Hospitalar e Universitário de Coimbra, Praceta, R. Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Sílvia Monteiro
- Serviço de Cardiologia, Centro Hospitalar e Universitário de Coimbra, Praceta, R. Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Rogério Teixeira
- Serviço de Cardiologia, Centro Hospitalar e Universitário de Coimbra, Praceta, R. Prof. Mota Pinto, 3004-561, Coimbra, Portugal.,Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | - Lino Gonçalves
- Serviço de Cardiologia, Centro Hospitalar e Universitário de Coimbra, Praceta, R. Prof. Mota Pinto, 3004-561, Coimbra, Portugal.,Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
46
|
Ma W, Zhang B, Yang Y, Qi L, Zhou J, Li M, Jia J, Zhang Y, Yong H. Association of epicardial fat thickness with left ventricular diastolic function parameters in a community population. BMC Cardiovasc Disord 2021; 21:262. [PMID: 34049490 PMCID: PMC8162010 DOI: 10.1186/s12872-021-02071-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Background We examined the relationship between epicardial fat thickness (EFT) measured by echocardiography and left ventricular diastolic function parameters in a Beijing community population.
Methods We included 1004 participants in this study. Echocardiographic parameters including E and A peak velocity, the early diastolic velocities (e′) of the septal and lateral mitral annulus using tissue doppler imaging, E/e′, and EFT were measured. EFT1 was measured perpendicularly on the right ventricular free wall at end diastole in the extension line of the aortic root. EFT2 was the maximum thickness measured perpendicularly on the right ventricular free wall at end diastole. Multivariable linear regression was used to analyze the relationship between EFT and the mean e′ and E/e′. Results The mean age of the participants was 63.91 ± 9.02 years, and 51.4% were men. EFT1 and EFT2 were negatively correlated with lateral e′, septal e′, and mean e′ (p < 0.05), and the correlation coefficient for EFT1 and EFT2 and mean e′ was − 0.138 and − 0.180, respectively. EFT1 and EFT2 were positively correlated with lateral E/e′, septal E/e′, and mean E/e′ (p < 0.05), and the correlation coefficient for EFT1 and EFT2 and mean e′ was 0.100 and 0.090, respectively. Multivariable egression analysis showed that EFT2 was independently and negatively associated with e′ mean (β = − 0.078 [95% confidence interval = − 0.143, − 0.012, p = 0.020]). There were no interactions between EFT2 and any covariates, including age or heart groups, sex, BMI, or presence of hypertension, diabetes, or coronary heart disease, in relation to left ventricular diastolic dysfunction. Conclusions EFT2 was negatively and independently associated with e′ mean, which suggests that more attention to this type of adipose fat is required for cardiovascular disease therapy.
Collapse
Affiliation(s)
- Wei Ma
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China. .,Echocardiography Core Lab, Institute of Cardiovascular Disease at Peking, University First Hospital, Beijing, China. .,Division of Cardiology, Peking University First Hospital, Dahongluochang Street, Xicheng District, Beijing, 100034, China.
| | - Baowei Zhang
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China.,Echocardiography Core Lab, Institute of Cardiovascular Disease at Peking, University First Hospital, Beijing, China
| | - Ying Yang
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China.,Echocardiography Core Lab, Institute of Cardiovascular Disease at Peking, University First Hospital, Beijing, China
| | - Litong Qi
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China.,Echocardiography Core Lab, Institute of Cardiovascular Disease at Peking, University First Hospital, Beijing, China
| | - Jin Zhou
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| | - Min Li
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| | - Jia Jia
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Huo Yong
- Department of Cardiovascular Disease, Peking University First Hospital, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
47
|
Chao H, Shan H, Homayounieh F, Singh R, Khera RD, Guo H, Su T, Wang G, Kalra MK, Yan P. Deep learning predicts cardiovascular disease risks from lung cancer screening low dose computed tomography. Nat Commun 2021; 12:2963. [PMID: 34017001 PMCID: PMC8137697 DOI: 10.1038/s41467-021-23235-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer patients have a higher risk of cardiovascular disease (CVD) mortality than the general population. Low dose computed tomography (LDCT) for lung cancer screening offers an opportunity for simultaneous CVD risk estimation in at-risk patients. Our deep learning CVD risk prediction model, trained with 30,286 LDCTs from the National Lung Cancer Screening Trial, achieves an area under the curve (AUC) of 0.871 on a separate test set of 2,085 subjects and identifies patients with high CVD mortality risks (AUC of 0.768). We validate our model against ECG-gated cardiac CT based markers, including coronary artery calcification (CAC) score, CAD-RADS score, and MESA 10-year risk score from an independent dataset of 335 subjects. Our work shows that, in high-risk patients, deep learning can convert LDCT for lung cancer screening into a dual-screening quantitative tool for CVD risk estimation.
Collapse
Affiliation(s)
- Hanqing Chao
- Department of Biomedical Engineering, Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Hongming Shan
- Department of Biomedical Engineering, Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fatemeh Homayounieh
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ramandeep Singh
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruhani Doda Khera
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hengtao Guo
- Department of Biomedical Engineering, Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Timothy Su
- Niskayuna High School, Niskayuna, NY, USA
| | - Ge Wang
- Department of Biomedical Engineering, Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, NY, USA.
| | - Mannudeep K Kalra
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Pingkun Yan
- Department of Biomedical Engineering, Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, NY, USA.
| |
Collapse
|
48
|
Muzurović EM, Vujošević S, Mikhailidis DP. Can We Decrease Epicardial and Pericardial Fat in Patients With Diabetes? J Cardiovasc Pharmacol Ther 2021; 26:415-436. [PMID: 33844605 DOI: 10.1177/10742484211006997] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) is a chronic and complex metabolic disorder and also an important cause of cardiovascular (CV) disease (CVD). Patients with type 2 DM (T2DM) and obesity show a greater propensity for visceral fat deposition (and excessive fat deposits elsewhere) and the link between adiposity and CVD risk is greater for visceral than for subcutaneous (SC) adipose tissue (AT). There is growing evidence that epicardial AT (EAT) and pericardial AT (PAT) play a role in the development of DM-related atherosclerosis, atrial fibrillation (AF), myocardial dysfunction, and heart failure (HF). In this review, we will highlight the importance of PAT and EAT in patients with DM. We also consider therapeutic interventions that could have a beneficial effect in terms of reducing the amount of AT and thus CV risk. EAT is biologically active and a likely determinant of CV morbidity and mortality in patients with DM, given its anatomical characteristics and proinflammatory secretory pattern. Consequently, modification of EAT/PAT may become a therapeutic target to reduce the CV burden. In patients with DM, a low calorie diet, exercise, antidiabetics and statins may change the quantity of EAT, PAT or both, alter the secretory pattern of EAT, improve the metabolic profile, and reduce inflammation. However, well-designed studies are needed to clearly define CV benefits and a therapeutic approach to EAT/PAT in patients with DM.
Collapse
Affiliation(s)
- Emir M Muzurović
- Department of Internal Medicine, Endocrinology Section, 274294Clinical Centre of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Snežana Vujošević
- Department of Internal Medicine, Endocrinology Section, 274294Clinical Centre of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, 9687Royal Free Hospital Campus, University College London Medical School, University College London (UCL), Pond Street, London, UK.,Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
49
|
Monti CB, Capra D, Malavazos A, Florini G, Parietti C, Schiaffino S, Sardanelli F, Secchi F. Subcutaneous, Paracardiac, and Epicardial Fat CT Density Before/After Contrast Injection: Any Correlation with CAD? J Clin Med 2021; 10:jcm10040735. [PMID: 33673256 PMCID: PMC7918165 DOI: 10.3390/jcm10040735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue, in particular epicardial adipose tissue, has been identified as a potential biomarker of cardiovascular pathologies such as coronary artery disease (CAD) in the light of its metabolic activity and close anatomic and pathophysiologic relationship to the heart. Our purpose was to evaluate epicardial adipose tissue density at both unenhanced and contrast-enhanced computed tomography (CT), along with CT densities of paracardiac and subcutaneous adipose tissue, as well as the relations of such densities with CAD. We retrospectively reviewed patients who underwent cardiac CT at our institution for CAD assessment. We segmented regions of interest on epicardial, paracardiac, and subcutaneous adipose tissue on unenhanced and contrast-enhanced scans. A total of 480 patients were included, 164 of them presenting with CAD. Median epicardial adipose tissue density measured on contrast-enhanced scans (−81.5 HU; interquartile range −84.9 to −78.0) was higher than that measured on unenhanced scans (−73.4 HU; −76.9 to −69.4) (p < 0.001), whereas paracardiac and subcutaneous adipose tissue densities were not (p ≥ 0.055). Patients with or without CAD, did not show significant differences in density of epicardial, paracardiac, and subcutaneous adipose tissue either on unenhanced or contrast-enhanced scans (p ≥ 0.092). CAD patients may experience different phenomena (inflammation, fibrosis, increase in adipose depots) leading to rises or drops in epicardial adipose tissue density, resulting in variations that are difficult to detect.
Collapse
Affiliation(s)
- Caterina Beatrice Monti
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (C.B.M.); (D.C.); (F.S.)
| | - Davide Capra
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (C.B.M.); (D.C.); (F.S.)
| | - Alexis Malavazos
- Clinical Nutrition and Cardiovascular Prevention Unit and High Specialty Center for Dietetics, Nutritional Education and Cardiometabolic Prevention, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milano, Italy;
| | - Giorgia Florini
- School of Medicine and Surgery, Università degli Studi di Milano, 20122 Milano, Italy; (G.F.); (C.P.)
| | - Carlo Parietti
- School of Medicine and Surgery, Università degli Studi di Milano, 20122 Milano, Italy; (G.F.); (C.P.)
| | - Simone Schiaffino
- Department of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milano, Italy;
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (C.B.M.); (D.C.); (F.S.)
- Department of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milano, Italy;
| | - Francesco Secchi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (C.B.M.); (D.C.); (F.S.)
- Department of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milano, Italy;
- Correspondence:
| |
Collapse
|
50
|
Cosson E, Nguyen MT, Rezgani I, Tatulashvili S, Sal M, Berkane N, Allard L, Brillet PY, Bihan H. Epicardial adipose tissue volume and coronary calcification among people living with diabetes: a cross-sectional study. Cardiovasc Diabetol 2021; 20:35. [PMID: 33546697 PMCID: PMC7863354 DOI: 10.1186/s12933-021-01225-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) has anatomic and functional proximity to the heart and is considered a novel diagnostic marker and therapeutic target in cardiometabolic diseases. The aim of this study was to evaluate whether EAT volume was associated with coronary artery calcification (CAC) in people living with diabetes, independently of confounding factors. METHODS We included all consecutive patients with diabetes whose EAT volume and CAC score were measured using computed tomography between January 1, 2019 and September 30, 2020 in the Department of Diabetology-Endocrinology-Nutrition at Avicenne Hospital, France. Determinants of EAT volume and a CAC score ≥ 100 Agatston units (AU) were evaluated. RESULTS The study population comprised 409 patients (218 men). Mean (± standard deviation) age was 57 ± 12 years, and 318, 56 and 35 had type 2 (T2D), type 1 (T1D), or another type of diabetes, respectively. Mean body mass index (BMI) was 29 ± 6 kg/m2, mean AET volume 93 ± 38 cm3. EAT volume was positively correlated with age, BMI, pack-year smoking history and triglyceridaemia, but negatively correlated with HDL-cholesterol level. Furthermore, it was lower in people with retinopathy, but higher in men, in Caucasian people, in patients on antihypertensive and lipid-lowering medication, in people with nephropathy, and finally in individuals with a CAC ≥ 100 AU (CAC < 100 vs CAC ≥ 100: 89 ± 35 vs 109 ± 41 cm3, respectively, p < 0.05). In addition to EAT volume, other determinants of CAC ≥ 100 AU (n = 89, 22%) were age, T2D, ethnicity, antihypertensive and lipid-lowering medication, cumulative tobacco consumption, retinopathy, macular edema and macrovascular disease. Multivariable analysis considering all these determinants as well as gender and BMI showed that EAT volume was independently associated with CAC ≥ 100 AU (per 10 cm3 increase: OR 1.11 [1.02-1.20]). CONCLUSIONS EAT volume was independently associated with CAC. As it may play a role in coronary atherosclerosis in patients with diabetes, reducing EAT volume through physical exercise, improved diet and pharmaceutical interventions may improve future cardiovascular risk outcomes in this population.
Collapse
Affiliation(s)
- Emmanuel Cosson
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France. .,Unité de Recherche Epidémiologique Nutritionnelle, UMR U1153 INSERM/U11125 INRA/CNAM/Université Paris 13, Bobigny, France.
| | - Minh Tuan Nguyen
- Department of Functional Explorations, AP-HP, Jean Verdier Hospital, Université Paris 13, Bondy, France
| | - Imen Rezgani
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France
| | - Sopio Tatulashvili
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France
| | - Meriem Sal
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France
| | - Narimane Berkane
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France
| | - Lucie Allard
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France
| | | | - Hélène Bihan
- Department of Endocrinology-Diabetology-Nutrition, AP-HP, Avicenne Hospital, Université Paris 13, Sorbonne Paris Cité, CRNH-IdF, CINFO, Bobigny, France.,Laboratoire Educations et Pratiques de Santé UR 3412, UFR Santé, Médecine, Biologie Humaine, Université Paris Sorbonne Paris Nord, 74, rue Marcel Cachin, 93017, Bobigny Cedex, France
| |
Collapse
|