1
|
Mondal S, Rathor R, Singh SN, Suryakumar G. miRNA and leptin signaling in metabolic diseases and at extreme environments. Pharmacol Res Perspect 2024; 12:e1248. [PMID: 39017237 PMCID: PMC11253706 DOI: 10.1002/prp2.1248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
The burden of growing concern about the dysregulation of metabolic processes arises due to complex interplay between environment and nutrition that has great impact on genetics and epigenetics of an individual. Thereby, any abnormality at the level of food intake regulating hormones may contribute to the development of metabolic diseases in any age group due to malnutrition, overweight, changing lifestyle, and exposure to extreme environments such as heat stress (HS), cold stress, or high altitude (HA). Hormones such as leptin, adiponectin, ghrelin, and cholecystokinin regulate appetite and satiety to maintain energy homeostasis. Leptin, an adipokine and a pleiotropic hormone, play major role in regulating the food intake, energy gain and energy expenditure. Using in silico approach, we have identified the major genes (LEP, LEPR, JAK2, STAT3, NPY, POMC, IRS1, SOCS3) that play crucial role in leptin signaling pathway. Further, eight miRNAs (hsa-miR-204-5p, hsa-miR-211-5p, hsa-miR-30, hsa-miR-3163, hsa-miR-33a-3p, hsa-miR-548, hsa-miR-561-3p, hsa-miR-7856-5p) from TargetScan 8.0 database were screened out that commonly target these genes. The role of these miRNAs should be explored as they might play vital role in regulating the appetite, energy metabolism, metabolic diseases (obesity, type 2 diabetes, cardiovascular diseases, inflammation), and to combat extreme environments. The miRNAs regulating leptin signaling and appetite may be useful for developing novel therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Samrita Mondal
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Richa Rathor
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Som Nath Singh
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | | |
Collapse
|
2
|
Shibasaki I, Nakajima T, Fukuda T, Hasegawa T, Ogawa H, Tsuchiya G, Takei Y, Tezuka M, Kato T, Kanazawa Y, Kano Y, Kuwata T, Ouchi M, Toyoda S, Aso Y, Fukuda H. Serum and Adipose Dipeptidyl Peptidase 4 in Cardiovascular Surgery Patients: Influence of Dipeptidyl Peptidase 4 Inhibitors. J Clin Med 2022; 11:jcm11154333. [PMID: 35893426 PMCID: PMC9331841 DOI: 10.3390/jcm11154333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 12/25/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP-4) is a novel adipokine and may be involved in the association between adipose tissue and metabolic syndrome. We investigated DPP-4 and adiponectin levels in the serum, subcutaneous adipose tissue (SAT), and epicardial adipose tissue (EAT), and their relationship with preoperative factors, as well as comparing the DPP-4 levels in SAT and EAT with and without DPP-4 inhibitors. This study included 40 patients (25 men, age 67.5 ± 13.8 years). The serum adipokine, DPP-4, and adiponectin levels in SAT and EAT were measured using ELISA and Western blotting. The DPP-4 and adiponectin levels were significantly higher in the SAT than in the EAT. The serum DPP-4 and DPP-4 activity levels had no correlation with the DPP-4 levels in the SAT and EAT, but the DPP-4 levels in the SAT and EAT had a positive correlation. The DPP-4 levels in the SAT were positively correlated with atherosclerosis, diabetes mellitus, DPP-4-inhibitor use, and fasting blood glucose. The DPP-4 levels in the EAT showed a negative correlation with eGFR and a positive correlation with atrial fibrillation. The DPP-4 activity in the serum had a lower tendency in the group taking DPP-4 inhibitors than in the group not taking them. DPP-4 inhibitors may suppress angiogenesis and adipose-tissue hypertrophy.
Collapse
Affiliation(s)
- Ikuko Shibasaki
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
- Correspondence:
| | - Toshiaki Nakajima
- Department of Medical KAATSU Training, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (T.N.); (T.H.)
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan;
| | - Taira Fukuda
- Department of Liberal Arts and Human Development, Kanagawa University of Human Services, Yokosuka 238-8522, Kanagawa, Japan;
| | - Takaaki Hasegawa
- Department of Medical KAATSU Training, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (T.N.); (T.H.)
| | - Hironaga Ogawa
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Go Tsuchiya
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Yusuke Takei
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Masahiro Tezuka
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Takashi Kato
- Department of Cardiovascular Surgery, Maebashi Red Cross Hospital, Maebashi 371-0811, Gunma, Japan; (T.K.); (T.K.)
| | - Yuta Kanazawa
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Yasuyuki Kano
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| | - Toshiyuki Kuwata
- Department of Cardiovascular Surgery, Maebashi Red Cross Hospital, Maebashi 371-0811, Gunma, Japan; (T.K.); (T.K.)
| | - Motoshi Ouchi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan;
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan;
| | - Yoshimasa Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan;
| | - Hirotsugu Fukuda
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan; (H.O.); (G.T.); (Y.T.); (M.T.); (Y.K.); (Y.K.); (H.F.)
| |
Collapse
|
3
|
Sun Y, Li XG, Xu K, Hou J, You HR, Zhang RR, Qi M, Zhang LB, Xu LS, Greenwald SE, Yang BQ. Relationship between epicardial fat volume on cardiac CT and atherosclerosis severity in three-vessel coronary artery disease: a single-center cross-sectional study. BMC Cardiovasc Disord 2022; 22:76. [PMID: 35246047 PMCID: PMC8895769 DOI: 10.1186/s12872-022-02527-7] [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: 12/08/2021] [Accepted: 02/24/2022] [Indexed: 12/03/2022] Open
Abstract
Background The ideal treatment strategy for stable three-vessel coronary artery disease (CAD) patients are difficult to determine and for patients undergoing conservative treatment, imaging evidence of coronary atherosclerotic severity progression remains limited. Epicardial fat volume (EFV) on coronary CT angiography (CCTA) has been considered to be associated with coronary atherosclerosis. Therefore, this study aims to evaluate the relationship between EFV level and coronary atherosclerosis severity in three-vessel CAD. Methods This retrospective study enrolled 252 consecutive patients with three-vessel CAD and 252 normal control group participants who underwent CCTA between January 2018 and December 2019. A semi-automatic method was developed for EFV quantification on CCTA images, standardized by body surface area. Coronary atherosclerosis severity was evaluated and scored by the number of coronary arteries with ≥ 50% stenosis on coronary angiography. Patients were subdivided into groups on the basis of lesion severity: mild (score = 3 vessels, n = 85), moderate (3.5 vessels ≤ score < 4 vessels, n = 82), and severe (4 vessels ≤ score ≤ 7 vessels, n = 85). The independent sample t-test, analysis of variance, and logistic regression analysis were used to evaluate the associations between EFV level and severity of coronary atherosclerosis. Results Compared with normal controls, three-vessel CAD patients had significantly higher EFV level (65 ± 22 mL/m2 vs. 48 ± 19 mL/m2; P < 0.001). In patients with three-vessel CAD, there was a progressive decline in EFV level as the score of coronary atherosclerosis severity increased, especially in those patients with a body mass index (BMI) ≥ 25 kg/m2 (75 ± 21 mL/m2 vs. 72 ± 22 mL/m2 vs. 62 ± 17 mL/m2; P < 0.05). Multivariable regression analysis showed that both BMI (OR 3.40, 95% CI 2.00–5.78, P < 0.001) and the score of coronary atherosclerosis severity (OR 0.49, 95% CI 0.26–0.93, P < 0.05) were independently related to the change of EFV level. Conclusion Three-vessel CAD patients do have higher EFV level than the normal controls. While, there may be an inverse relationship between EFV level and the severity of coronary atherosclerosis in patients with three-vessel CAD.
Collapse
Affiliation(s)
- Yu Sun
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, People's Republic of China.,Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China
| | - Xiao-Gang Li
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China
| | - Kai Xu
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Jie Hou
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, People's Republic of China.,Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China
| | - Hong-Rui You
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China
| | - Rong-Rong Zhang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China
| | - Miao Qi
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, People's Republic of China.,Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China
| | - Li-Bo Zhang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China.,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China
| | - Li-Sheng Xu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, People's Republic of China
| | - Stephen E Greenwald
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ben-Qiang Yang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua RoadLiaoning Province, Shenyang, 110016, People's Republic of China. .,Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, People's Republic of China.
| |
Collapse
|
4
|
Konwerski M, Gąsecka A, Opolski G, Grabowski M, Mazurek T. Role of Epicardial Adipose Tissue in Cardiovascular Diseases: A Review. BIOLOGY 2022; 11:355. [PMID: 35336728 PMCID: PMC8945130 DOI: 10.3390/biology11030355] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide. Epicardial adipose tissue (EAT) is defined as a fat depot localized between the myocardial surface and the visceral layer of the pericardium and is a type of visceral fat. EAT is one of the most important risk factors for atherosclerosis and cardiovascular events and a promising new therapeutic target in CVDs. In health conditions, EAT has a protective function, including protection against hypothermia or mechanical stress, providing myocardial energy supply from free fatty acid and release of adiponectin. In patients with obesity, metabolic syndrome, or diabetes mellitus, EAT becomes a deleterious tissue promoting the development of CVDs. Previously, we showed an adverse modulation of gene expression in pericoronary adipose tissue in patients with coronary artery disease (CAD). Here, we summarize the currently available evidence regarding the role of EAT in the development of CVDs, including CAD, heart failure, and atrial fibrillation. Due to the rapid development of the COVID-19 pandemic, we also discuss data regarding the association between EAT and the course of COVID-19. Finally, we present the potential therapeutic possibilities aiming at modifying EAT's function. The development of novel therapies specifically targeting EAT could revolutionize the prognosis in CVDs.
Collapse
Affiliation(s)
| | | | | | | | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warszawa, Poland; (M.K.); (A.G.); (G.O.); (M.G.)
| |
Collapse
|
5
|
Konwerski M, Gromadka A, Arendarczyk A, Koblowska M, Iwanicka-Nowicka R, Wilimski R, Czub P, Filipiak KJ, Hendzel P, Zielenkiewicz P, Opolski G, Gąsecka A, Mazurek T. Atherosclerosis Pathways are Activated in Pericoronary Adipose Tissue of Patients with Coronary Artery Disease. J Inflamm Res 2021; 14:5419-5431. [PMID: 34707383 PMCID: PMC8542577 DOI: 10.2147/jir.s326769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Perivascular release of inflammatory mediators may accelerate coronary lesion formation and contribute to plaque instability. Accordingly, we compared gene expression in pericoronary adipose tissue (PCAT) in patients with advanced coronary artery disease (CAD) and non-CAD controls. PATIENTS AND METHODS PCAT samples were collected during coronary bypass grafting from CAD patients (n = 21) and controls undergoing valve replacement surgery, with CAD excluded by coronary angiography (n = 19). Gene expression was measured by GeneChip™ Human Transcriptome Array 2.0. Obtained list of 1348 transcripts (2.0%) that passed the filter criteria was further analyzed by Ingenuity Pathway Analysis software, identifying 735 unique differentially expressed genes (DEGs). RESULTS Among the CAD patients, 416 (30.9%) transcripts were upregulated, and 932 (69.1%) were downregulated, compared to controls. The top upregulated genes were involved in inflammation and atherosclerosis (chemokines, interleukin-6, selectin E and low-density lipoprotein cholesterol (LDL-C) receptor), whereas the downregulated genes were involved in cardiac ischaemia and remodelling, platelet function and mitochondrial function (miR-3671, miR-4524a, multimerin, biglycan, tissue factor pathway inhibitor (TFPI), glucuronidases, miR-548, collagen type I, III, IV). Among the top upstream regulators, we identified molecules that have proinflammatory and atherosclerotic features (High Mobility Group Box 2 (HMGB2), platelet-derived growth platelet (PDGF) and evolutionarily conserved signaling intermediate in Toll pathways (ESCIT)). The activated pathway related to DEGs consisted of molecules with well-established role in the pathogenesis of atherosclerosis (TFPI, plasminogen activator, plasminogen activator, urokinase receptor (PLAUR), thrombomodulin). Moreover, we showed that 22 of the altered genes form a pro-atherogenic network. CONCLUSION Altered gene expression in PCAT of CAD patients, with genes upregulation and activation of pathway involved in inflammation and atherosclerosis, may be involved in CAD development and progression.
Collapse
Affiliation(s)
- Michał Konwerski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Gromadka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Adam Arendarczyk
- Department of Cardiac Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Marta Koblowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Radosław Wilimski
- Department of Cardiac Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Czub
- Department of Cardiac Surgery, Medical University of Warsaw, Warsaw, Poland
| | | | - Piotr Hendzel
- Department of Cardiac Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Zielenkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
Bermúdez V, Durán P, Rojas E, Díaz MP, Rivas J, Nava M, Chacín M, Cabrera de Bravo M, Carrasquero R, Ponce CC, Górriz JL, D´Marco L. The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front Endocrinol (Lausanne) 2021; 12:735070. [PMID: 34603210 PMCID: PMC8479191 DOI: 10.3389/fendo.2021.735070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue (AT) biology is linked to cardiovascular health since obesity is associated with cardiovascular disease (CVD) and positively correlated with excessive visceral fat accumulation. AT signaling to myocardial cells through soluble factors known as adipokines, cardiokines, branched-chain amino acids and small molecules like microRNAs, undoubtedly influence myocardial cells and AT function via the endocrine-paracrine mechanisms of action. Unfortunately, abnormal total and visceral adiposity can alter this harmonious signaling network, resulting in tissue hypoxia and monocyte/macrophage adipose infiltration occurring alongside expanded intra-abdominal and epicardial fat depots seen in the human obese phenotype. These processes promote an abnormal adipocyte proteomic reprogramming, whereby these cells become a source of abnormal signals, affecting vascular and myocardial tissues, leading to meta-inflammation, atrial fibrillation, coronary artery disease, heart hypertrophy, heart failure and myocardial infarction. This review first discusses the pathophysiology and consequences of adipose tissue expansion, particularly their association with meta-inflammation and microbiota dysbiosis. We also explore the precise mechanisms involved in metabolic reprogramming in AT that represent plausible causative factors for CVD. Finally, we clarify how lifestyle changes could promote improvement in myocardiocyte function in the context of changes in AT proteomics and a better gut microbiome profile to develop effective, non-pharmacologic approaches to CVD.
Collapse
Affiliation(s)
- Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Pablo Durán
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Edward Rojas
- Cardiovascular Division, University Hospital, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - María P. Díaz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - José Rivas
- Department of Medicine, Cardiology Division, University of Florida-College of Medicine, Jacksonville, FL, United States
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | - Rubén Carrasquero
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Clímaco Cano Ponce
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - José Luis Górriz
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Luis D´Marco
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
7
|
Sardu C, D’Onofrio N, Torella M, Portoghese M, Mureddu S, Loreni F, Ferraraccio F, Panarese I, Trotta MC, Gatta G, Galdiero M, Sasso FC, D’Amico M, De Feo M, Balestrieri ML, Paolisso G, Marfella R. Metformin Therapy Effects on the Expression of Sodium-Glucose Cotransporter 2, Leptin, and SIRT6 Levels in Pericoronary Fat Excised from Pre-Diabetic Patients with Acute Myocardial Infarction. Biomedicines 2021; 9:biomedicines9080904. [PMID: 34440108 PMCID: PMC8389537 DOI: 10.3390/biomedicines9080904] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND PURPOSE pericoronary fat over-inflammation might lead to the development and destabilization of coronary plaque in patients with pre-diabetes (PDM). Notably, pericoronary fat could over-express the sodium-glucose cotransporter 2 (SGLT2) and leptin, along with decreased sirtuin 6 (SIRT6) expression in PDM vs. normoglycemic (NG) patients undergoing coronary artery bypass grafting (CABG) for acute myocardial infarction (AMI). However, in the current study, we evaluated inflammatory markers, SGLT2, SIRT6, and leptin levels in pericoronary fat and, subsequently, 12-month prognosis comparing PDM to NG subjected to CABG for AMI. In addition, we evaluated in PDM patients the effects of metformin therapy on SIRT6 expression, leptin, and SGLT2 levels, and assessed its beneficial effect on nitrotyrosine and inflammatory cytokine levels. METHODS we studied AMI patients referred for CABG, divided into PDM and NG-patients. PDM patients were divided into never-metformin users and metformin users. Finally, we evaluated major adverse cardiac events (MACE) at a 12-month follow-up. RESULTS the MACE was 9.1% in all PDM and 3% in NG patients (p < 0.05). Metformin users presented a significantly lower MACE rate in PDM than never-metformin users (p < 0.05). PDM showed higher inflammatory cytokines, 3-nitrotyrosine levels, SGLT2, and leptin content, and decreased SIRT6 protein levels in pericoronary fat compared to NG-patients (p < 0.05). PDM never-metformin-users showed higher SGLT2 and leptin levels in pericoronary fat than current-metformin-users (p < 0.05). CONCLUSIONS metformin therapy might ameliorate cardiovascular outcomes by reducing inflammatory parameters, SGLT2, and leptin levels, and finally improving SIRT6 levels in AMI-PDM patients treated with CABG.
Collapse
Affiliation(s)
- Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.C.S.); (G.P.); (R.M.)
- Correspondence: ; Tel.: +39-0815665110; Fax: +39-0815665303
| | - Nunzia D’Onofrio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.D.); (M.L.B.)
| | - Michele Torella
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.T.); (F.L.); (F.F.); (I.P.); (M.D.F.)
| | - Michele Portoghese
- Department of Cardiac Surgery, Santissima Annunziata Hospital, 07100 Sassari, Italy; (M.P.); (S.M.)
| | - Simone Mureddu
- Department of Cardiac Surgery, Santissima Annunziata Hospital, 07100 Sassari, Italy; (M.P.); (S.M.)
| | - Francesco Loreni
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.T.); (F.L.); (F.F.); (I.P.); (M.D.F.)
| | - Franca Ferraraccio
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.T.); (F.L.); (F.F.); (I.P.); (M.D.F.)
| | - Iacopo Panarese
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.T.); (F.L.); (F.F.); (I.P.); (M.D.F.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (M.G.); (M.D.)
| | - Gianluca Gatta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (M.G.); (M.D.)
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (M.G.); (M.D.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.C.S.); (G.P.); (R.M.)
| | - Michele D’Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (G.G.); (M.G.); (M.D.)
| | - Marisa De Feo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.T.); (F.L.); (F.F.); (I.P.); (M.D.F.)
| | - Maria Luisa Balestrieri
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.D.); (M.L.B.)
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.C.S.); (G.P.); (R.M.)
- Mediterranea Cardiocentro, 80122 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.C.S.); (G.P.); (R.M.)
- Mediterranea Cardiocentro, 80122 Naples, Italy
| |
Collapse
|
8
|
Konwerski M, Postuła M, Barczuk-Falęcka M, Czajkowska A, Mróz A, Witek K, Bakalarski W, Gąsecka A, Małek ŁA, Mazurek T. Epicardial Adipose Tissue and Cardiovascular Risk Assessment in Ultra-Marathon Runners: A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18063136. [PMID: 33803664 PMCID: PMC8002849 DOI: 10.3390/ijerph18063136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 01/04/2023]
Abstract
Epicardial adipose tissue (EAT) volume is associated with cardiovascular disease (CVD). Data regarding the influence of extremely intensive training on CVD are scarce. We compared EAT volume among ultra-marathon runners and in the sedentary control group, and assessed the correlations between EAT and risk factors of coronary artery disease (CAD). EAT volume around three main coronary vessels and right ventricle (RV) was measured in 30 healthy amateur ultrarunners and 9 sex- and age-matched sedentary controls using cardiac magnetic resonance. In addition, body composition, lipid profile, interleukin-6 (IL-6) plasma concentration, and intima-media thickness (IMT) were measured as well. The EAT volume was lower in all measured locations in the ultrarunners' group compared to control group (p < 0.001 for all). Ultrarunners had lower BMI and fat percentage (FAT%) and more favorable lipid profile compared to the control group (p < 0.05 for all). Ultrarunners had lower rate of pathologically high levels of plasma IL-6 (>1 pg/mL) compared to the control group (17% vs. 56%, p < 0.05). IMT was similar in both groups. In the ultrarunners' group, there was a positive correlation between EAT surrounding left anterior descending artery, circumflex artery, and RV and FAT%, and between EAT around circumflex artery and LDL and non-HDL cholesterol (p < 0.05 for all). In summary, extremely intensive training may decrease the risk of cardiovascular events in adult population of amateur athletes by reducing the amount and pro-inflammatory activity of EAT. However, more research is needed to draw firm conclusions regarding the anti- and pro-inflammatory effects of intensive training.
Collapse
Affiliation(s)
- Michał Konwerski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.K.); (A.G.)
| | - Marek Postuła
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | | | - Anna Czajkowska
- Faculty of Tourism and Recreation, Józef Piłsudski University of Physical Education in Warsaw, 00-968 Warsaw, Poland;
| | - Anna Mróz
- Faculty of Physical Education, Józef Piłsudski University of Physical Education in Warsaw, 00-968 Warsaw, Poland; (A.M.); (K.W.); (W.B.)
| | - Katarzyna Witek
- Faculty of Physical Education, Józef Piłsudski University of Physical Education in Warsaw, 00-968 Warsaw, Poland; (A.M.); (K.W.); (W.B.)
| | - Wawrzyniec Bakalarski
- Faculty of Physical Education, Józef Piłsudski University of Physical Education in Warsaw, 00-968 Warsaw, Poland; (A.M.); (K.W.); (W.B.)
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.K.); (A.G.)
| | - Łukasz A. Małek
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.K.); (A.G.)
- Correspondence: ; Tel.: +48-22-599-1958
| |
Collapse
|
9
|
Tian W, Zhang W, Zhang Y, Zhu T, Hua Y, Li H, Zhang Q, Xia M. FABP4 promotes invasion and metastasis of colon cancer by regulating fatty acid transport. Cancer Cell Int 2020; 20:512. [PMID: 33088219 PMCID: PMC7574203 DOI: 10.1186/s12935-020-01582-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/28/2020] [Indexed: 01/29/2023] Open
Abstract
Background The prognosis of colon cancer is poor for metastasis, while the mechanism, especially adipocytes related, is not yet clear. The purpose of this study is to determine the effects of fatty acid binding protein 4 (FABP4), a transporter for lipids, on colon cancer progression. Methods The distribution of lipids and FABP4 was tested in the colon cancer tissues and adjacent normal tissues, and their relationship was also verified in vitro. Experiments about cellular invasion, migration and proliferation were performed to detect the impacts of FABP4 on the biological behaviors of colon cancer, and the positive results were checked in vivo. Meanwhile, the regulatory role of FABP4 in the energy and lipid metabolism was evaluated by the levels of triglyceride, ATP, LDH, glycerol and NEFA. At last, GO and KEGG analysis based on FABP4 overexpressed cells was performed, and the AKT pathway and epithelial-mesenchymal transition (EMT)-related proteins were determined by Western blot. Results Higher accumulation of lipids and stronger FABP4 transcription were observed in colon cancer tissues. Having been incubated with adipose tissue extract and overexpressed FABP4, colon cancer cells demonstrated enhanced lipid accumulation. In functional experiments, co-culture with adipose tissue extract significantly enhanced the invasion and migration of colon cancer cells, as well as the energy and lipid metabolism, and all these processes were reversed by FABP4 inhibitor. In addition, the metastasis of FABP4-overexpressed colon cancer cells was also significantly enhanced in vitro and in vivo. In terms of mechanism, the bioinformatics analysis showed that FABP4 was enriched in 11 pathways related to metabolic processes in FABP4 overexpressed cells. Finally, FABP4 overexpression improved EMT progression of colon cancer, as evidenced by the upregulation of Snail, MMP-2 and MMP-9, the downregulation of E-cadherin. The expression of p-Akt was also elevated. Conclusion FABP4 overexpression could increase FAs transport to enhance energy and lipid metabolism, and activate AKT pathway and EMT to promote the migration and invasion of colon cancer cells.
Collapse
Affiliation(s)
- Wenying Tian
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Wenjia Zhang
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Yan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu China
| | - Tianyue Zhu
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Yuting Hua
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Hui Li
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Qinglin Zhang
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Min Xia
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qing Yang Road, Wuxi, 214023 Jiangsu People's Republic of China
| |
Collapse
|
10
|
Iacobellis G, Gra-Menendez S. Effects of Dapagliflozin on Epicardial Fat Thickness in Patients with Type 2 Diabetes and Obesity. Obesity (Silver Spring) 2020; 28:1068-1074. [PMID: 32352644 DOI: 10.1002/oby.22798] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Epicardial adipose tissue (EAT) thickness is a marker of visceral fat and an emerging therapeutic target. Dapagliflozin, a selective sodium-glucose cotransporter 2 inhibitor, improves glucose control and induces moderate weight loss in patients with type 2 diabetes mellitus. Dapagliflozin has recently been shown to reduce cardiovascular risk. Nevertheless, whether dapagliflozin could reduce EAT thickness is unknown. METHODS This hypothesis was tested in a 24-week, randomized, double-blind, placebo-controlled clinical trial in 100 patients with type 2 diabetes mellitus with BMI ≥ 27 kg/m2 and a hemoglobin A1c level ≤ 8% on metformin monotherapy. Individuals were randomly assigned to 2 groups to receive additional dapagliflozin up to 10 mg once daily or to remain on metformin up to 1,000 mg twice daily. Ultrasound-measured EAT thickness was measured at baseline, 12 weeks, and 24 weeks. RESULTS In the dapagliflozin group, EAT decreased by 20% from baseline to 24 weeks, by 15% after 12 weeks, and by 7% between 12 and 24 weeks, respectively (P < 0.01 for all), whereas in the metformin group, there was a significant but smaller EAT reduction. There was no statistically significant correlation between EAT and body weight changes. CONCLUSIONS Dapagliflozin causes a rapid and significant EAT reduction that could be independent of weight loss.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Silvia Gra-Menendez
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
11
|
Niedziela M, Wojciechowska M, Zarębiński M, Cudnoch-Jędrzejewska A, Mazurek T. Adiponectin promotes ischemic heart preconditioning- PRO and CON. Cytokine 2020; 127:154981. [PMID: 31911263 DOI: 10.1016/j.cyto.2019.154981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/07/2019] [Accepted: 12/27/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Magdalena Niedziela
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Wojciechowska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland.
| | - Maciej Zarębiński
- Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland
| |
Collapse
|
12
|
Christensen RH, von Scholten BJ, Lehrskov LL, Rossing P, Jørgensen PG. Epicardial adipose tissue: an emerging biomarker of cardiovascular complications in type 2 diabetes? Ther Adv Endocrinol Metab 2020; 11:2042018820928824. [PMID: 32518616 PMCID: PMC7252363 DOI: 10.1177/2042018820928824] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/03/2020] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease and heart failure, which highlights the need for improved understanding of factors contributing to the pathophysiology of these complications as they are the leading cause of mortality in T2D. Patients with T2D have high levels of epicardial adipose tissue (EAT). EAT is known to secrete inflammatory factors, lipid metabolites, and has been proposed to apply mechanical stress on the cardiac muscle that may accelerate atherosclerosis, cardiac remodeling, and heart failure. High levels of EAT in patients with T2D have been associated with atherosclerosis, diastolic dysfunction, and incident cardiovascular events, and this fat depot has been suggested as an important link coupling diabetes, obesity, and cardiovascular disease. Despite this, the predictive potential of EAT in general, and in patients with diabetes, is yet to be established, and, up until now, the clinical relevance of EAT is therefore limited. Should this link be established, importantly, studies show that this fat depot can be modified both by pharmacological and lifestyle interventions. In this review, we first introduce the role of adipose tissue in T2D and present mechanisms involved in the pathophysiology of EAT and pericardial adipose tissue (PAT) in general, and in patients with T2D. Next, we summarize the evidence that these fat depots are elevated in patients with T2D, and discuss whether they might drive the high cardiometabolic risk in patients with T2D. Finally, we discuss the clinical potential of cardiac adipose tissues, address means to target this depot, and briefly touch upon underlying mechanisms and future research questions.
Collapse
Affiliation(s)
| | | | - Louise Lang Lehrskov
- Center for Inflammation and Metabolism/Center for Physical Activity Research, Rigshospitalet, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
13
|
|
14
|
Sardu C, D'Onofrio N, Torella M, Portoghese M, Loreni F, Mureddu S, Signoriello G, Scisciola L, Barbieri M, Rizzo MR, Galdiero M, De Feo M, Balestrieri ML, Paolisso G, Marfella R. Pericoronary fat inflammation and Major Adverse Cardiac Events (MACE) in prediabetic patients with acute myocardial infarction: effects of metformin. Cardiovasc Diabetol 2019; 18:126. [PMID: 31570103 PMCID: PMC6767640 DOI: 10.1186/s12933-019-0931-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/18/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/OBJECTIVES Pericoronary adipose tissue inflammation might lead to the development and destabilization of coronary plaques in prediabetic patients. Here, we evaluated inflammation and leptin to adiponectin ratio in pericoronary fat from patients subjected to coronary artery bypass grafting (CABG) for acute myocardial infarction (AMI). Furthermore, we compared the 12-month prognosis of prediabetic patients compared to normoglycemic patients (NG). Finally, the effect of metformin therapy on pericoronary fat inflammation and 12-months prognosis in AMI-prediabetic patients was also evaluated. METHODS An observational prospective study was conducted on patients with first AMI referred for CABG. Participants were divided in prediabetic and NG-patients. Prediabetic patients were divided in two groups; never-metformin-users and current-metformin-users receiving metformin therapy for almost 6 months before CABG. During the by-pass procedure on epicardial coronary portion, the pericoronary fat was removed from the surrounding stenosis area. The primary endpoints were the assessments of Major-Adverse-Cardiac-Events (MACE) at 12-month follow-up. Moreover, inflammatory tone was evaluated by measuring pericoronary fat levels of tumor necrosis factor-α (TNF-α), sirtuin 6 (SIRT6), and leptin to adiponectin ratio. Finally, inflammatory tone was correlated to the MACE during the 12-months follow-up. RESULTS The MACE was 9.1% in all prediabetic patients and 3% in NG-patients. In prediabetic patients, current-metformin-users presented a significantly lower rate of MACE compared to prediabetic patients never-metformin-users. In addition, prediabetic patients showed higher inflammatory tone and leptin to adiponectin ratio in pericoronary fat compared to NG-patients (P < 0.001). Prediabetic never-metformin-users showed higher inflammatory tone and leptin to adiponectin ratio in pericoronary fat compared to current-metformin-users (P < 0.001). Remarkably, inflammatory tone and leptin to adiponectin ratio was significantly related to the MACE during the 12-months follow-up. CONCLUSION Prediabetes increase inflammatory burden in pericoronary adipose tissue. Metformin by reducing inflammatory tone and leptin to adiponectin ratio in pericoronary fat may improve prognosis in prediabetic patients with AMI. Trial registration Clinical Trial NCT03360981, Retrospectively Registered 7 January 2018.
Collapse
Affiliation(s)
- Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Torella
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Portoghese
- Department of Cardiac Surgery, Santissima Annunziata Hospital, Sassari, Italy
| | - Francesco Loreni
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Mureddu
- Department of Cardiac Surgery, Santissima Annunziata Hospital, Sassari, Italy
| | - Giuseppe Signoriello
- Department of Mental Health and Public Medicine, Section of Statistic, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marisa De Feo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia, 2, 80138, Naples, Italy.
| |
Collapse
|
15
|
Reynés B, van Schothorst EM, Keijer J, Ceresi E, Oliver P, Palou A. Cold Induced Depot-Specific Browning in Ferret Aortic Perivascular Adipose Tissue. Front Physiol 2019; 10:1171. [PMID: 31620014 PMCID: PMC6759601 DOI: 10.3389/fphys.2019.01171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 08/29/2019] [Indexed: 12/23/2022] Open
Abstract
Brown adipose tissue is responsible for facultative thermogenesis to produce heat and increase energy expenditure in response to proper stimuli, e.g., cold. Acquisition of brown-like features (browning) in perivascular white adipose tissue (PVAT) may protect against obesity/cardiovascular disease. Most browning studies are performed in rodents, but translation to humans would benefit from a closer animal model. Therefore, we studied the browning response of ferret thoracic aortic PVAT (tPVAT) to cold. We performed global transcriptome analysis of tPVAT of 3-month-old ferrets acclimatized 1 week to 22 or 4°C, and compared the results with those of inguinal subcutaneous adipose tissue. Immunohistochemistry was used to visualize browning. Transcriptome data revealed a stronger cold exposure response of tPVAT, including increased expression of key brown/brite markers, compared to subcutaneous fat. This translated into a clear white-to-brown remodeling of tPVAT, with the appearance of multilocular highly UCP1-stained adipocytes. The pathway most affected by cold exposure in tPVAT was immune response, characterized by down-regulation of immune-related genes, with cardio protective implications. On the other hand, subcutaneous fat responded to cold by increasing energy metabolism based on increased expression of fatty acid oxidation and tricarboxylic acid cycle genes, concordant with lower inguinal adipose tissue weight in cold-exposed animals. Thus, ferret tPVAT responds to cold acclimation with a strong induction of browning and immunosuppression compared to subcutaneous fat. Our results present ferrets as an accessible translational animal model displaying functional responses relevant for obesity and cardiovascular disease prevention.
Collapse
Affiliation(s)
- Bàrbara Reynés
- Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Palma, Spain.,CIBER de Fisiopatología de la Obesidad y Nutrición, Madrid, Spain.,Institut d'Investigació Sanitària Illes Balears, Palma, Spain
| | - Evert M van Schothorst
- Human and Animal Physiology, Wageningen University and Research, Wageningen, Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University and Research, Wageningen, Netherlands
| | - Enzo Ceresi
- Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Palma, Spain.,CIBER de Fisiopatología de la Obesidad y Nutrición, Madrid, Spain.,Institut d'Investigació Sanitària Illes Balears, Palma, Spain
| | - Paula Oliver
- Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Palma, Spain.,CIBER de Fisiopatología de la Obesidad y Nutrición, Madrid, Spain.,Institut d'Investigació Sanitària Illes Balears, Palma, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Palma, Spain.,CIBER de Fisiopatología de la Obesidad y Nutrición, Madrid, Spain.,Institut d'Investigació Sanitària Illes Balears, Palma, Spain
| |
Collapse
|
16
|
Boucher JM, Robich M, Scott SS, Yang X, Ryzhova L, Turner JE, Pinz I, Liaw L. Rab27a Regulates Human Perivascular Adipose Progenitor Cell Differentiation. Cardiovasc Drugs Ther 2019; 32:519-530. [PMID: 30105417 DOI: 10.1007/s10557-018-6813-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Perivascular adipose tissue (PVAT) surrounds blood vessels and regulates vascular tone through paracrine secretion of cytokines. During conditions promoting cardiometabolic dysfunction, such as obesity, cytokine secretion is altered towards a proinflammatory and proatherogenic profile. Despite the clinical implications for cardiovascular disease, studies addressing the biology of human PVAT remain limited. We are interested in characterizing the resident adipose progenitor cells (APCs) because of their potential role in PVAT expansion during obesity. We also focused on proteins regulating paracrine interactions, including the small GTPase Rab27a, which regulates protein trafficking and secretion. METHODS PVAT from the ascending aorta was collected from patients with severe cardiovascular disease undergoing coronary artery bypass grafting (CABG). Freshly-isolated PVAT was digested and APC expanded in culture for characterizing progenitor markers, evaluating adipogenic potential and assessing the function(s) of Rab27a. RESULTS Using flow cytometry, RT-PCR, and immunoblot, we characterized APC from human PVAT as negative for CD45 and CD31 and expressing CD73, CD105, and CD140A. These APCs differentiate into multilocular, UCP1-producing adipocytes in vitro. Rab27a was detected in interstitial cells of human PVAT in vivo and along F-actin tracks of PVAT-APC in vitro. Knockdown of Rab27a using siRNA in PVAT-APC prior to induction resulted in a marked reduction in lipid accumulation and reduced expression of adipogenic differentiation markers. CONCLUSIONS PVAT-APC from CABG donors express common adipocyte progenitor markers and differentiate into UCP1-containing adipocytes. Rab27a has an endogenous role in promoting the maturation of adipocytes from human PVAT-derived APC.
Collapse
Affiliation(s)
- Joshua M Boucher
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Michael Robich
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
- Division of Thoracic and Cardiac Surgery, Maine Medical Center, Portland, ME, 04102, USA
| | - S Spencer Scott
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Larisa Ryzhova
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Jacqueline E Turner
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Ilka Pinz
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04072, USA.
| |
Collapse
|
17
|
Zuo H, Zhang Y, Ma Q. Correlation between coronary atherosclerosis calcification and epicardial adipose tissue volume in patients with nephropathy. Exp Ther Med 2018; 16:4669-4673. [PMID: 30546396 PMCID: PMC6256921 DOI: 10.3892/etm.2018.6801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022] Open
Abstract
Correlation between coronary atherosclerosis calcification and epicardial adipose tissue (EAT) volume in patients with nephropathy was investigated. A total of 529 patients with high risk of coronary atherosclerotic nephropathy were selected from August 2013 to September 2016 in Xianyang Central Hospital to serve as research subjects, and their clinical data were retrospectively analyzed. All patients underwent coronary artery and EAT examination using dual-source CT. Correlation between EAT and severity of coronary atherosclerosis, calcification, lesions, and BMI were analyzed. Volume of EAT in patients with atherosclerosis was significantly higher than that in patients without atherosclerosis (p<0.05). EAT volume was positively correlated with BMI (r=0.61, p<0.05), calcification scores (r=0.72, p<0.05) and the number of coronary artery lesions (r=0.64, p<0.05) in patients with nephropathy. Coronary atherosclerosis calcification score, number of lesions and BMI are positively correlated with the EAT volume in patients with nephropathy. Detection of EAT volume may serve as a predictor of the occurrence and develop-ment of coronary atherosclerosis in the future.
Collapse
Affiliation(s)
- Hong Zuo
- Department of Cardiovascular Medicine, Xianyang Central Hospital, Xianyang, Shaanxi 712000, P.R. China
| | - Ying Zhang
- Department of Nephrology, Hanzhong Central Hospital, Hanzhong, Shaanxi 723000, P.R. China
| | - Qiaojuan Ma
- Department of Cardiovascular Medicine, The Central Hospital of Tongchuan Mining Bureau, Tongchuan, Shaanxi 727000, P.R. China
| |
Collapse
|
18
|
RPS3A positively regulates the mitochondrial function of human periaortic adipose tissue and is associated with coronary artery diseases. Cell Discov 2018; 4:52. [PMID: 30131868 PMCID: PMC6102269 DOI: 10.1038/s41421-018-0041-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 05/27/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
Pericardial adipose tissue, which comprises both epicardial adipose tissue (EAT) and paracardial adipose tissue (PAT), has recently been recognized as a novel factor in the pathophysiology of cardiovascular diseases, especially coronary artery disease (CAD). The goal of this study was to evaluate differences in the brown-like characteristic and proteome among human EAT, PAT, and subcutaneous adipose tissue (SAT) to identify candidate molecules causing CAD. Uncoupling protein 1 (UCP-1) and other brown-related proteins were highly expressed in pericardial adipose tissue but was weakly expressed in SAT from the same non-CAD patient. Moreover, pericardial adipose tissues displayed a higher thermogenesis than SAT. However, brown-related genes were lower in CAD pericardial fat. Remarkably, there were lower levels of metabolic enzymes involved in glycolysis, tricarboxylic acid cycle, and fatty acid metabolism in pericardial adipose tissues of CAD. EAT is an organ adjacent to aortic root without anatomy barriers, which differs from PAT. We found that the expression of ribosomal protein S3A (RPS3A) was decreased in human EAT as well as in mouse perivascular adipose tissue (PVAT). Knockdown of RPS3A significantly inhibited adipocyte differentiation in preadipocytes and impaired the function of mitochondria in mature adipocytes. Moreover, RPS3A knockdown in mouse periaortic adipose tissue impaired browning of PVAT, accelerated vascular inflammation, and atherosclerosis progression. Mechanistically, RPS3A can migrate to the mitochondria to maintain the function of brown adipocytes. These findings provide compelling evidence that RPS3A was a key factor for modulating the brown fat-specific gene UCP-1 and carbon metabolic enzymes in EAT for preventing CAD.
Collapse
|
19
|
Bornachea O, Vea A, Llorente-Cortes V. Interplay between epicardial adipose tissue, metabolic and cardiovascular diseases. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 30:230-239. [PMID: 29903689 DOI: 10.1016/j.arteri.2018.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/24/2018] [Accepted: 03/19/2018] [Indexed: 01/26/2023]
Abstract
Cardiovascular disease is the primary cause of death in obese and diabetic patients. In these groups of patients, the alterations of epicardial adipose tissue (EAT) contribute to both vascular and myocardial dysfunction. Therefore, it is of clinical interest to determine the mechanisms by which EAT influences cardiovascular disease. Two key factors contribute to the tight intercommunication among EAT, coronary arteries and myocardium. One is the close anatomical proximity between these tissues. The other is the capacity of EAT to secrete cytokines and other molecules with paracrine and vasocrine effects on the cardiovascular system. Epidemiological studies have demonstrated that EAT thickness is associated with not only metabolic syndrome but also atherosclerosis and heart failure. The evaluation of EAT using imaging modalities, although effective, presents several disadvantages including radiation exposure, limited availability and elevated costs. Therefore, there is a clinical interest in EAT as a source of new biomarkers of cardiovascular and endocrine alterations. In this review, we revise the mechanisms involved in the protective and pathological role of EAT and present the molecules released by EAT with greater potential to become biomarkers of cardiometabolic alterations.
Collapse
Affiliation(s)
- Olga Bornachea
- Institute of Biomedical Research IIB-Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IibB)-CSIC, Barcelona, Spain
| | - Angela Vea
- Institute of Biomedical Research IIB-Sant Pau, Barcelona, Spain
| | - Vicenta Llorente-Cortes
- Institute of Biomedical Research IIB-Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IibB)-CSIC, Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
20
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
21
|
Akoumianakis I, Akawi N, Antoniades C. Exploring the Crosstalk between Adipose Tissue and the Cardiovascular System. Korean Circ J 2017; 47:670-685. [PMID: 28955384 PMCID: PMC5614942 DOI: 10.4070/kcj.2017.0041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/04/2017] [Indexed: 12/28/2022] Open
Abstract
Obesity is a clinical entity critically involved in the development and progression of cardiovascular disease (CVD), which is characterised by variable expansion of adipose tissue (AT) mass across the body as well as by phenotypic alterations in AT. AT is able to secrete a diverse spectrum of biologically active substances called adipocytokines, which reach the cardiovascular system via both endocrine and paracrine routes, potentially regulating a variety of physiological and pathophysiological responses in the vasculature and heart. Such responses include regulation of inflammation and oxidative stress as well as cell proliferation, migration and hypertrophy. Furthermore, clinical observations such as the “obesity paradox,” namely the fact that moderately obese patients with CVD have favourable clinical outcome, strongly indicate that the biological “quality” of AT may be far more crucial than its overall mass in the regulation of CVD pathogenesis. In this work, we describe the anatomical and biological diversity of AT in health and metabolic disease; we next explore its association with CVD and, importantly, novel evidence for its dynamic crosstalk with the cardiovascular system, which could regulate CVD pathogenesis.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Nadia Akawi
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
22
|
Camarena V, Sant D, Mohseni M, Salerno T, Zaleski ML, Wang G, Iacobellis G. Novel atherogenic pathways from the differential transcriptome analysis of diabetic epicardial adipose tissue. Nutr Metab Cardiovasc Dis 2017; 27:739-750. [PMID: 28739185 PMCID: PMC7540222 DOI: 10.1016/j.numecd.2017.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/25/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM To evaluate the epicardial adipose tissue (EAT) transcriptome in comparison to subcutaneous fat (SAT) in coronary artery disease (CAD) and type 2 diabetes (T2DM). METHODS AND RESULTS SAT and EAT samples were obtained from subjects with T2DM and CAD (n = 5) and those without CAD with or without T2DM (=3) undergoing elective cardiac surgery. RNA-sequencing analysis was performed in both EAT and SAT. Gene enrichment analysis was conducted to identify pathways affected by the differentially expressed genes. Changes of top genes were verified by quantitative RT-PCR (qRT-PCR), western blot, and immunofluorescence. A total of 592 genes were differentially expressed in diabetic EAT, whereas there was no obvious changes in SAT transcriptome between diabetics and non-diabetics. Diabetic EAT was mainly enriched in inflammatory genes, such as Colony Stimulating Factor 3 (CSF3), Interleukin-1b (IL-1b), IL-6. KEGG pathway analysis confirmed that upregulated genes were involved in inflammatory pathways, such as Tumor Necrosis Factor (TNF), Nuclear Factor-κB (NF-κB) and advanced glycation end-products-receptor advanced glycation end products (AGE-RAGE). The overexpression of inflammatory genes in diabetic EAT was largely correlated with upregulated transcription factors such as NF-κB and FOS. CONCLUSIONS Diabetic EAT transcriptome is significantly different when compared to diabetic SAT and highly enriched with genes involved in innate immune response and endothelium, like Pentraxin3 (PTX3) and Endothelial lipase G (LIPG). EAT inflammatory genes expression could be induced by upregulated transcription factors, mainly NF-kB and FOSL, primarily activated by the overexpressed AGE-RAGE signaling. This suggests a unique and novel atherogenic pathway in diabetes.
Collapse
Affiliation(s)
- V Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miami, FL, USA
| | - D Sant
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miami, FL, USA
| | - M Mohseni
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miami, FL, USA
| | - T Salerno
- Department of Surgery, Division of Thoracic and Cardiac Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M L Zaleski
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miami, FL, USA
| | - G Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miami, FL, USA.
| | - G Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miami, FL, USA.
| |
Collapse
|
23
|
Nagy E, Jermendy AL, Merkely B, Maurovich-Horvat P. Clinical importance of epicardial adipose tissue. Arch Med Sci 2017; 13:864-874. [PMID: 28721155 PMCID: PMC5507110 DOI: 10.5114/aoms.2016.63259] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Different visceral fat compartments have several systemic effects and may play a role in the development of both insulin resistance and cardiovascular diseases. In the last couple of years special attention has been paid to the epicardial adipose tissue (EAT), which can be quantified by non-invasive cardiac imaging techniques. The epicardial fat is a unique fat compartment between the myocardium and the visceral pericardium sharing a common embryologic origin with the visceral fat depot. Epicardial adipose tissue has several specific roles, and its local effects on cardiac function are incorporated in the complex pathomechanism of coronary artery disease. Importantly, EAT may produce several adipocytokines and chemokines that may influence - through paracrine and vasocrine effects - the development and progression of coronary atherosclerosis. Epicardial adipose tissue volume has a relatively strong genetic dependence, similarly to other visceral fat depots. In this article, the anatomical and physiological as well as pathophysiological characteristics of the epicardial fat compartment are reviewed.
Collapse
Affiliation(s)
- Eszter Nagy
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | - Adam L Jermendy
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | - Bela Merkely
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | | |
Collapse
|
24
|
Mazurek T, Kobylecka M, Zielenkiewicz M, Kurek A, Kochman J, Filipiak KJ, Mazurek K, Huczek Z, Królicki L, Opolski G. PET/CT evaluation of 18F-FDG uptake in pericoronary adipose tissue in patients with stable coronary artery disease: Independent predictor of atherosclerotic lesions' formation? J Nucl Cardiol 2017; 24:1075-1084. [PMID: 26951555 DOI: 10.1007/s12350-015-0370-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/01/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND Inflammatory infiltrations in EAT which releases inflammatory cytokines correspond anatomically to the atheromatous plaques in underlying coronary vessels. However, it is unknown whether inflammatory activity of pericoronary adipose tissue (PCAT) promotes coronary atherosclerosis. METHODS AND RESULTS 35 non-diabetic patients with confirmed CAD and 35 non-CAD controls matched for age and BMI underwent 18F-FDG-PET/CT. Maximal SUV normalized by LA blood activity was measured on the sections corresponding to the respective coronaries (RCA, LCX, LAD), as well, as in subcutaneous fat, visceral fat, and epicardial fat. Extent of CAD was determined by % stenosis in segments corresponding to 18F-FDG-PET/CT sections in coronarography using quantitative coronary analysis. PCAT SUV was significantly greater than SUV in other fat locations, as well as PCAT SUV in the controls. In CAD patients with BMI >25, PCAT SUV was positively related to % stenosis of a respective coronary artery (RCA: 0.43; P < .05; LCX 0.58; P < .05; LAD 0.65; P < .05). PCAT SUV was the only independent predictor of coronary stenosis of LAD and RCA. CONCLUSIONS Inflammatory activity of PCAT is greater than in other fat locations, in CAD is greater than in non-CAD controls, and is independently associated with coronary stenosis. In overweight patients, PCAT SUV correlates with the extent of CAD.
Collapse
Affiliation(s)
- Tomasz Mazurek
- First Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland.
| | | | | | - Aleksandra Kurek
- The International Institute of Molecular and Cell Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Janusz Kochman
- First Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Krzysztof J Filipiak
- First Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Krzysztof Mazurek
- Józef Piłsudski University of Physical Education in Warsaw, Warsaw, Poland
| | - Zenon Huczek
- First Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Leszek Królicki
- Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Opolski
- First Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| |
Collapse
|
25
|
Comparison of Antidiabetic Medications during the Treatment of Atherosclerosis in T2DM Patients. Mediators Inflamm 2017; 2017:5032708. [PMID: 28539704 PMCID: PMC5429949 DOI: 10.1155/2017/5032708] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/06/2017] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes is often associated with arterial atherosclerosis in large blood vessels. We set out to elucidate whether commonly used antidiabetic drugs metformin, sitagliptin, and pioglitazone will reduce atherosclerosis in T2DM patients. We enrolled 176 individuals with type 2 diabetes, which were divided into four treatment groups according to different oral drugs: metformin alone, sitagliptin alone, pioglitazone alone, or combination of metformin and sitagliptin. We assessed changes in glycometabolism, lipid metabolism, cytokine released, and carotid artery intima-media thickness as the readout for improvement in atherosclerosis. HbA1c levels were significantly decreased in all treatment groups (p < 0.05), and FBG levels were also decreased in metformin and combined groups (p < 0.05). In addition, we found IL-6 levels significantly decreased in all treatment groups (p < 0.05). Treatment with pioglitazone showed a significant increase in BMI, HDL, and ADPN levels (p < 0.05). We also observed a significant decrease in NHDL levels in the combined treatment group (p < 0.05). Our data revealed that in addition to hypoglycemic properties of metformin, sitagliptin, and pioglitazone, these drugs also have the potential to promote an anti-inflammatory response. Therefore, combination therapy may be more beneficial for reducing atherosclerosis in patients with type 2 diabetes. The clinical trial is registered with ChiCTR-ORC-17010835.
Collapse
|
26
|
Fernández-Alfonso MS, Gil-Ortega M, Aranguez I, Souza D, Dreifaldt M, Somoza B, Dashwood MR. Role of PVAT in coronary atherosclerosis and vein graft patency: friend or foe? Br J Pharmacol 2017; 174:3561-3572. [PMID: 28150299 DOI: 10.1111/bph.13734] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/02/2017] [Accepted: 01/19/2017] [Indexed: 01/18/2023] Open
Abstract
Perivascular adipose tissue (PVAT) releases numerous factors and adipokines with paracrine effects on both vascular structure and function. These effects are variable as they depend on regional differences in PVAT among blood vessels and vary with changes in adiposity. There is considerable evidence demonstrating an association between coronary PVAT and the development and progression of coronary artery disease, which is associated with inflammation, oxidative stress, angiogenesis, vascular remodelling and blood clotting. However, PVAT also has a protective role in vascular grafts, especially the no-touch saphenous vein, in patients undergoing coronary artery bypass. This beneficial influence of PVAT involves factors such as adipocyte-derived relaxing factor, nitric oxide (NO), leptin, adiponectin, prostanoids, hydrogen sulphide and neurotransmitters, as well as mechanical protection. This article aims to highlight and compare the dual role of PVAT in the development and progression of coronary atherosclerosis, as well as in increased graft patency. Different deleterious and protective mechanisms of PVAT are also discussed and the inside-outside signalling paradigm of atherosclerosis development re-evaluated. The bidirectional communication between the arterial and venous wall and their surrounding PVAT, where signals originating from the vascular wall or lumen can affect PVAT phenotype, has been shown to be very complex. Moreover, signals from PVAT also influence the structure and function of the vascular wall in a paracrine manner. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- M S Fernández-Alfonso
- Instituto Pluridisciplinar and Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - M Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - I Aranguez
- Instituto Pluridisciplinar and Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - D Souza
- Department of Cardiothoracic and Vascular Surgery and University Health Care Research Center, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - M Dreifaldt
- Department of Cardiothoracic and Vascular Surgery and University Health Care Research Center, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - B Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - M R Dashwood
- Royal Free Hospital Campus, University College Medical School, London, UK
| |
Collapse
|
27
|
Iacobellis G, Mohseni M, Bianco SD, Banga PK. Liraglutide causes large and rapid epicardial fat reduction. Obesity (Silver Spring) 2017; 25:311-316. [PMID: 28124506 DOI: 10.1002/oby.21718] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Epicardial adipose tissue (EAT), the visceral fat depot of the heart, is a modifiable cardiovascular risk factor and emerging therapeutic target. Liraglutide, an analog of glucagon-like peptide-1, is indicated for the treatment of type 2 diabetes mellitus. Liraglutide has recently been shown to reduce cardiovascular risk. Nevertheless, whether liraglutide could reduce EAT is unknown. METHODS To test the hypothesis, a 6-month randomized, open-label, controlled study was performed in 95 type 2 diabetic subjects with body mass index (BMI) ≥27 kg/m2 and hemoglobinA1c ≤8% on metformin monotherapy. Individuals were randomized in two groups to receive additional liraglutide up to 1.8 mg s.c. once daily (n = 54) or to remain on metformin up to 1,000 mg twice daily (n = 41). Ultrasound-measured EAT thickness was measured at baseline and at 3- and 6-month follow-ups. RESULTS In the liraglutide group, EAT decreased from 9.6 ± 2 to 6.8 ± 1.5 and 6.2 ± 1.5 mm (P < 0.001), accounting for a -29% and -36% of reduction at 3 and 6 months, respectively, whereas there was no EAT reduction in the metformin group; BMI and hemoglobinA1c improved only in the liraglutide group after 6 months. CONCLUSIONS Liraglutide causes a substantial and rapid EAT reduction. Liraglutide cardiometabolic effects may be EAT-mediated.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Mahshid Mohseni
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Suzy D Bianco
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Pritisheel K Banga
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
28
|
Liberale L, Bonaventura A, Vecchiè A, Casula M, Dallegri F, Montecucco F, Carbone F. The Role of Adipocytokines in Coronary Atherosclerosis. Curr Atheroscler Rep 2017; 19:10. [PMID: 28185154 DOI: 10.1007/s11883-017-0644-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to overview the pathophysiological role of adipocytokines in atherogenesis, focusing on their potential role as biomarkers of coronary disease. RECENT FINDINGS Several lines of evidence indicated adipose tissue not only as depot but rather as an endocrine organ. In this context, the balance between pro- and anti-inflammatory adipocytokines has been shown to critically regulate vascular homeostasis in both physiological and pathophysiological conditions. Overweight and obesity are characterized by dysfunctional adipose tissue and then the prevalence of pro-inflammatory mediators, with a detrimental effect on vascular health. As opposite to adiponectin, pro-inflammatory adipocytokines, such as leptin and resistin, promote endothelial dysfunction and inflammatory processes involved in atherosclerotic plaque progression and vulnerability. Therefore, many adipocytokines have been investigated as potential biomarkers of cardiovascular (CV) risk, but their role has not yet been clearly established. Furthermore, the perivascular adipose tissue recently emerged as a critical modulator of atherosclerotic processes, due to the close interaction with the underlying vascular tissue. The ongoing discovery of new adipocytokines and the complex pathophysiological role of the different adipose tissue depots strongly contribute to define the complexity of adipocytokines network. Understanding those complex interactions may allow determining new potential biomarkers of CV risk and potential therapeutic targets.
Collapse
Affiliation(s)
- Luca Liberale
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Alessandra Vecchiè
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Matteo Casula
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Franco Dallegri
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa and IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 10 Largo Benzi, 16132, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 Viale Benedetto XV, 16132, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
| |
Collapse
|
29
|
Akoumianakis I, Tarun A, Antoniades C. Perivascular adipose tissue as a regulator of vascular disease pathogenesis: identifying novel therapeutic targets. Br J Pharmacol 2016; 174:3411-3424. [PMID: 27976387 DOI: 10.1111/bph.13666] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/04/2016] [Accepted: 10/28/2016] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue (AT) is an active endocrine organ with the ability to dynamically secrete a wide range of adipocytokines. Importantly, its secretory profile is altered in various cardiovascular disease states. AT surrounding vessels, or perivascular AT (PVAT), is recognized in particular as an important local regulator of vascular function and dysfunction. Specifically, PVAT has the ability to sense vascular paracrine signals and respond by secreting a variety of vasoactive adipocytokines. Due to the crucial role of PVAT in regulating many aspects of vascular biology, it may constitute a novel therapeutic target for the prevention and treatment of vascular disease pathogenesis. Signalling pathways in PVAT, such as those using adiponectin, H2 S, glucagon-like peptide 1 or pro-inflammatory cytokines, are among the potential novel pharmacological therapeutic targets of PVAT. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| | - Akansha Tarun
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| |
Collapse
|
30
|
Body composition: Where and when. Eur J Radiol 2016; 85:1456-60. [DOI: 10.1016/j.ejrad.2015.10.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 10/19/2015] [Accepted: 10/27/2015] [Indexed: 11/20/2022]
|
31
|
Chirumbolo S. Heart adipose tissue and sudden cardiac death in middle-aged men. Int J Cardiol 2016; 214:362-3. [DOI: 10.1016/j.ijcard.2016.03.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/19/2016] [Indexed: 10/22/2022]
|
32
|
Alman AC, Jacobs DR, Lewis CE, Snell-Bergeon JK, Carnethon MR, Terry JG, Goff DC, Ding J, Carr JJ. Higher pericardial adiposity is associated with prevalent diabetes: The Coronary Artery Risk Development in Young Adults study. Nutr Metab Cardiovasc Dis 2016; 26:326-32. [PMID: 26803596 PMCID: PMC4823150 DOI: 10.1016/j.numecd.2015.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/29/2015] [Accepted: 12/17/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Pericardial adipose tissue (PAT) is located on both sides of the pericardium. We tested whether PAT was associated with prevalent diabetes at the year 25 exam of the Coronary Artery Risk Development in Young Adults (CARDIA) study. METHODS AND RESULTS The CARDIA Year 25 exam (2010-2011) included complete data for all covariates on 3107 participants. Prevalent diabetes (n = 436) was defined as high fasting (≥126 mg/dl) or 2-h postload glucose (≥200 mg/dl) or HbA1c (≥6.5%) or use of diabetes medications. Volume of PAT was measured from computed tomographic scans. Logistic regression was performed to examine the relationship between quartiles of PAT and diabetes. In regression models adjusted for field center, sex, race, age, systolic blood pressure, total cholesterol, log triglycerides, and treatment with blood pressure and cholesterol lowering medication, PAT volume in the 4th quartile was significantly associated with diabetes status after adjustment for BMI (OR 2.57, 95% CI 1.66, 3.98) or visceral adipose tissue (OR 2.08, 95% CI 1.32, 3.29). PAT volume in the 2nd and 3rd quartiles was not significantly associated with diabetes status relative to the first quartile. CONCLUSIONS Metabolically active pericardial adipose tissue is associated with prevalent diabetes only at higher volumes independent of overall obesity.
Collapse
Affiliation(s)
- A C Alman
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL 33612-3805, USA.
| | - D R Jacobs
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - C E Lewis
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J K Snell-Bergeon
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M R Carnethon
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - J G Terry
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - D C Goff
- Colorado School of Public Health, Aurora, CO, USA
| | - J Ding
- Sticht Center on Aging, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - J J Carr
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
33
|
Iacobellis G. Epicardial fat: a new cardiovascular therapeutic target. Curr Opin Pharmacol 2016; 27:13-8. [PMID: 26848943 DOI: 10.1016/j.coph.2016.01.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/14/2016] [Accepted: 01/18/2016] [Indexed: 01/15/2023]
Abstract
Epicardial fat is the visceral fat depot of the heart. Given its rapid metabolism, organ fat specificity and simple objective measurability, epicardial fat can serve as target for pharmaceutical agents targeting the adipose tissue. Epicardial fat has shown to significantly respond to thiazolidinediones, glucagon like peptide 1 receptor agonists, dipeptidyl peptidase-4 inhibitors and statins. Epicardial fat may represent a measurable risk factor and modifiable therapeutic target. Targeted pharmaceutical interventions may allow the epicardial fat to resume its physiological role. A drug-induced browning effect on epicardial fat suggests the development of pharmacological strategies to increase energy consumption. The potential of modulating the epicardial fat transcriptome with targeted pharmacological agents can open new avenues in the pharmacotherapy of cardio-metabolic diseases.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
34
|
Bays HE, Jones PH, Jacobson TA, Cohen DE, Orringer CE, Kothari S, Azagury DE, Morton J, Nguyen NT, Westman EC, Horn DB, Scinta W, Primack C. Lipids and bariatric procedures part 1 of 2: Scientific statement from the National Lipid Association, American Society for Metabolic and Bariatric Surgery, and Obesity Medicine Association: FULL REPORT. J Clin Lipidol 2016; 10:33-57. [DOI: 10.1016/j.jacl.2015.12.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023]
|
35
|
Liao PH, Kuo WW, Kuo CH, Yeh YL, Shen CY, Chen YH, Chen RJ, Padma VV, Chen YH, Huang CY. Lactobacillus reuteri GMNL-263 reduces hyperlipidaemia and the heart failure process in high-calorie diet-fed induced heart dysfunction in rats. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|