1
|
Prajapati R, Qin T, Connelly KA, Merdad A, Chow CM, Leong-Poi H, Ong G. Echocardiographic Assessment of Cardiac Remodeling According to Obesity Class. Am J Cardiol 2025; 236:34-41. [PMID: 39505229 DOI: 10.1016/j.amjcard.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024]
Abstract
Evidence supports the existence of cardiac remodeling in obesity; however, no standard diagnostic criteria has been proposed or validated. This study aimed to identify echocardiographic features of cardiac remodeling according to obesity class and assess the effect of nonsurgical weight loss on cardiac structure and function. A total of 120 patients were divided according to their obesity class (group 1: body mass index [BMI] 18.5 to 24.9, group 2: 25 to 29.9, group 3: 30 to 39.9, and group 4: >40) and underwent cross-sectional transthoracic echocardiography. Echocardiographic parameters of cardiac chamber quantification and function were compared among the 4 groups. Echocardiographic parameters were compared before and after nonsurgical weight loss in a subgroup of patients. Overall, there was an incremental increase in left ventricular (LV), left atrial (LA), and right ventricular dimensions, LV mass (LVM), and LV stroke volume (all p <0.0001) across the obesity classes. There was no significant difference in LV ejection fraction or right ventricular systolic function, as assessed by tricuspid annular plane systolic excursion; however, there was a significant decrease in global longitudinal strain (BMI 18.5 to 24.9: 22.8 ± 1.7%, BMI 25 to 29.9: 22.0 ± 1.4%, BMI 30 to 39.9: 20.8 ± 1.1%, BMI >40: 20.6 ± 1.3%, p <0.0001) and LA strain (BMI 18.5 to 24.9: 37.7 ± 2.3%, BMI 25 to 29.9: 32.8 ± 2.1%, BMI 30 to 39.9: 31.5 ± 1.8%, BMI >40: 29.0 ± 2.8%, p <0.0001). Allometric height-indexed LV and LA dimensions increased with increasing BMI class (p <0.0001). Echocardiographic parameters did not change significantly after nonsurgical weight loss. In conclusion, echocardiographic features can be described according to obesity class. Allometric height indexation may better reflect cardiac remodeling in obesity than body surface area indexation. Nonsurgical weight loss was not associated with significant changes in cardiac chamber dimensions and function.
Collapse
Affiliation(s)
- Rahil Prajapati
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tingting Qin
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Ultrasound, Affiliated hospital of Jining Medical University, Jining, China
| | - Kim A Connelly
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Anas Merdad
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Chi-Ming Chow
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Howard Leong-Poi
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Geraldine Ong
- Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Xi Z, Shu L, Xiao L, Fang X, Dai M, Wang J, Wu Y, Zhang J, Bao M. Macrophage NLRP3 inflammasome mediates the effects of sympathetic nerve on cardiac remodeling in obese rats. Mol Cell Endocrinol 2025; 596:112417. [PMID: 39557185 DOI: 10.1016/j.mce.2024.112417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Obesity-associated cardiac remodeling is characterized by cardiac sympathetic nerve over-activation and pro-inflammatory macrophage infiltration. We identified norepinephrine (NE), a sympathetic neurotransmitter, as a pro-inflammatory effector to activate macrophage NLRP3 inflammasome, which contributed to cardiac inflammation. In vivo, Sprague-Dawley (SD) rats were fed a high-fat diet (HFD) for 12 weeks to establish obese rat models. Obese rats exhibited marked cardiac hypertrophy compared to normal rats. The expression of NLRP3 and interleukin (IL)-1β was upregulated, accompanied by CD68+NLRP3+ macrophage infiltration in the hearts of the obese rats. The obese rats also showed increased sympathetic nerve activity. β-adrenergic receptor (AR) inhibition mitigated these changes. In vitro, sympathetic neurotransmitter NE significantly exacerbated palmitic acid (PA)-induced macrophage polarization toward pro-inflammatory type and NLRP3 inflammasome activation in THP-1 macrophages. It was further found that the pro-inflammatory role of NE is dependent on the activation of protein kinase A (PKA) and subsequently inhibition of β-arrestin2, which is an important regulator of the nuclear factor-kappa B (NF-κB) pathway. This study identifies the neuro-immune axis as an important mediator in obesity-associated cardiac remodeling. Targeting the neuro-immune system may open therapeutic opportunities for the treatment of cardiac remodeling in obesity.
Collapse
Affiliation(s)
- Zhaoqing Xi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Ling Shu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Lingling Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xuesheng Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jing Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China; State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China
| | - Yuan Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Junxia Zhang
- Department of Endocrinology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei, 430050, China.
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
3
|
Cifuentes M, Verdejo HE, Castro PF, Corvalan AH, Ferreccio C, Quest AFG, Kogan MJ, Lavandero S. Low-Grade Chronic Inflammation: a Shared Mechanism for Chronic Diseases. Physiology (Bethesda) 2025; 40:0. [PMID: 39078396 DOI: 10.1152/physiol.00021.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
Inflammation is an important physiological response of the organism to restore homeostasis upon pathogenic or damaging stimuli. However, the persistence of the harmful trigger or a deficient resolution of the process can evolve into a state of low-grade, chronic inflammation. This condition is strongly associated with the development of several increasingly prevalent and serious chronic conditions, such as obesity, cancer, and cardiovascular diseases, elevating overall morbidity and mortality worldwide. The current pandemic of chronic diseases underscores the need to address chronic inflammation, its pathogenic mechanisms, and potential preventive measures to limit its current widespread impact. The present review discusses the current knowledge and research gaps regarding the association between low-grade chronic inflammation and chronic diseases, focusing on obesity, cardiovascular diseases, digestive diseases, and cancer. We examine the state of the art in selected aspects of the topic and propose future directions and approaches for the field.
Collapse
Affiliation(s)
- Mariana Cifuentes
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- OMEGA Laboratory, Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Division of Cardiovascular Diseases, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Division of Cardiovascular Diseases, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Alejandro H Corvalan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Hematology and Oncology, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Catterina Ferreccio
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Public Health, Facultad Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomedicas (ICBM), Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Department of Pharmacological & Toxicological Chemistry, Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Facultad Medicina & Instituto de Nutricion y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomedicas (ICBM), Facultad Medicina, Universidad de Chile, Santiago, Chile
- Department of Biochemistry & Molecular Biology, Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
4
|
Hua F, Sun J, Shi M, Mei R, Song Z, Liu J, Zhang M. Macrophage-derived extracellular vesicles transfer mitochondria to adipocytes and promote adipocyte-myofibroblast transition in epidural fibrosis. NPJ Regen Med 2024; 9:43. [PMID: 39738050 DOI: 10.1038/s41536-024-00388-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025] Open
Abstract
Epidural fibrosis post laminectomy is the leading cause of failed back surgery syndrome. Little is known about the role and mechanisms of adipose tissues in epidural fibrosis. Here, we found that obese patients were more likely to develop epidural fibrosis after spine surgery. Similarly, obesity led to more progressive epidural fibrosis in a mouse model of laminectomy. Adipocyte-myofibroblast transition (AMT) occurs in epidural scarring. Mechanistically, large extracellular vesicles (EVs) from M2-type macrophages transfer mitochondria into adipocytes and promote AMT by activating the TGF-β and PAI-1 pathways. Blocking the PAI-1 pathway significantly attenuated the transition of adipocytes into myofibroblasts. We conclude that large EVs from macrophages transfer mitochondria to promote AMT in epidural fibrosis.
Collapse
Affiliation(s)
- Feng Hua
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinpeng Sun
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mohan Shi
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Mei
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zeyuan Song
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Jiangsu Province Engineering Research Center of Antibody Drug, Department of Immunology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Wang N, Jia X, Fan Z, Yang C, Wang Y, Fan J, Zhao C, Yang Y, Shi X. Role of Life's Essential 8 score in mediating socioeconomic status in the incidence of atrial fibrillation and heart failure:a population-based cohort study. Hellenic J Cardiol 2024:S1109-9666(24)00269-0. [PMID: 39742967 DOI: 10.1016/j.hjc.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE To assess whether the Life's Essential 8 (LE8) score mediates the association of socioeconomic status (SES) with atrial fibrillation (AF) and heart failure (HF). METHODS A total of 236,754 participants from the UK Biobank were included. SES was determined based on household income, education attainment, and employment status using latent class analysis. Cox regression was utilized to explore the association of SES with AF and HF after adjusting for age, sex, ethnicity, and alcohol status. Counterfactual mediation analysis was employed to calculate the mediation proportion of LE8 score. Stratified analysis was conducted based on age and sex. RESULTS With a median of 13.61 years follow-up, 14,635 cases of AF and 6,878 cases of HF were documented. The HR (95% CI) of the total effect of SES on AF was 1.43 (1.36, 1.48). The indirect effect mediated by the LE8 score was 1.14 (1.13, 1.15), with mediation proportion was 40.84 (36.97, 47.01) %. The total effect of SES on HF was 2.44 (2.26, 2.59). The indirect effect was 1.28 (1.25, 1.29), with mediation proportion was 36.77 (34.59, 39.06) %. The mediation proportion was greater for AF in age < 60 years compared to age ≥ 60 years, and it was also higher in males than females for both AF and HF. CONCLUSIONS Approximately one-third of the socioeconomic inequalities in AF and HF could be explained by the LE8 score. These findings highlighted the importance of integrating cardiovascular health promotion into public health policies aimed at mitigating socioeconomic health inequalities.
Collapse
Affiliation(s)
- Nana Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xiaocan Jia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhixing Fan
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University and Yichang Central People's Hospital, Yichang 443003, Hubei, China
| | - Chaojun Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuping Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jingwen Fan
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Chenyu Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Xuezhong Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
6
|
Lolescu BA, Furdui-Lința AV, Ilie CA, Sturza A, Zară F, Muntean DM, Blidișel A, Crețu OM. Adipose tissue as target of environmental toxicants: focus on mitochondrial dysfunction and oxidative inflammation in metabolic dysfunction-associated steatotic liver disease. Mol Cell Biochem 2024:10.1007/s11010-024-05165-z. [PMID: 39704874 DOI: 10.1007/s11010-024-05165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024]
Abstract
Obesity, diabetes, and their cardiovascular and hepatic comorbidities are alarming public health issues of the twenty-first century, which share mitochondrial dysfunction, oxidative stress, and chronic inflammation as common pathophysiological mechanisms. An increasing body of evidence links the combined exposure to multiple environmental toxicants with the occurrence and severity of metabolic diseases. Endocrine disruptors (EDs) are ubiquitous chemicals or mixtures with persistent deleterious effects on the living organisms beyond the endocrine system impairment; in particular, those known as metabolism-disrupting chemicals (MDCs), increase the risk of the metabolic pathologies in adult organism or its progeny. Being largely lipophilic, MDCs mainly target the adipose tissue and elicit mitochondrial dysfunction by interfering with mitochondrial bioenergetics, biogenesis, dynamics and/or other functions. Plastics, when broken down into micro- and nano-plastics (MNPs), have been detected in several human tissues, including the liver. The harmful interplay between inflammatory and redox processes, which mutually interact in a positive feed-back loop, hence the term oxidative inflammation ("OxInflammation"), occurs both at systemic and organ level. In both liver and adipose tissue, oxinflammation contributes to the progression of the metabolic dysfunction-associated steatotic liver disease (MASLD). Moreover, it has been reported that individuals with MASLD may be more susceptible to the harmful effects of toxicants (mainly, those related to mitochondria) and that chronic exposure to EDs/MDCs or MNPs may play a role in the development of the disease. While liver has been systematically investigated as major target organ for ambient chemicals, surprisingly, less information is available in the literature with respect to the adipose tissue. In this narrative review, we delve into the current literature on the most studied environmental toxicants (bisphenols, polychlorinated biphenyls, phthalates, tolylfluanid and tributyltin, per-fluoroalkyl and polyfluoroalkyl substances, heavy metals and MNPs), summarize their deleterious effects on adipose tissue, and address the role of dysregulated mitochondria and oxinflammation, particularly in the setting of MASLD.
Collapse
Affiliation(s)
- Bogdan A Lolescu
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adina V Furdui-Lința
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Cosmin A Ilie
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Public Health & Sanitary Management, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adrian Sturza
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Flavia Zară
- Department II Microscopic Morphology-Chair of Histology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department of Pathology, Timisoara Municipal Emergency Clinical Hospital, Timișoara, Romania
| | - Danina M Muntean
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Alexandru Blidișel
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania.
| | - Octavian M Crețu
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania
| |
Collapse
|
7
|
Schmidt CE, Müller HD. Myocardial Fibrosis in Diabetic Cardiomyopathy: Mechanisms, Implications, and Therapeutic Perspectives. Curr Probl Cardiol 2024:102976. [PMID: 39706391 DOI: 10.1016/j.cpcardiol.2024.102976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Diabetic cardiomyopathy (DCM) represents a distinct form of heart disease characterized by structural and functional alterations in the myocardium, occurring in the absence of other cardiac conditions. This review delves into the pathophysiological mechanisms underlying myocardial fibrosis in DCM, highlighting the pivotal role of fibroblast transdifferentiation into myofibroblasts. We examine the interplay between hyperglycemia, immune cell activation, and neurohumoral signaling pathways, with a particular focus on the transforming growth factor-beta (TGF-β) signaling cascade and its contributions to collagen deposition and cardiac dysfunction. Despite significant advancements in understanding the cellular and molecular mechanisms of DCM, critical gaps remain in elucidating the precise regulatory networks involved in fibroblast activation and the role of microRNAs in these processes. By providing a comprehensive overview of current knowledge, this review aims to identify potential therapeutic targets to mitigate myocardial fibrosis and improve clinical outcomes in diabetic patients. Ultimately, addressing these gaps will pave the way for novel therapeutic strategies that can enhance heart function and reduce the burden of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Clara Elisabeth Schmidt
- Bioanalytical Lab, Meso Scale Discovery, Rockville, MD 20850-3173, USA; Department of Pediatric Endocrinology and Rheumatology, Institute of Pediatrics, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Hans Dietrich Müller
- Department of Pediatric Endocrinology and Rheumatology, Institute of Pediatrics, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| |
Collapse
|
8
|
Zhao CZ, Ding HM, Hu ZQ, Zhou L, Du YQ, Zhou P, Wang L. Exploring the mechanism of Ling-Gui-Zhu-Gan decoction in metabolic cardiomyopathy via inhibiting ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156234. [PMID: 39547098 DOI: 10.1016/j.phymed.2024.156234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/14/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study was to investigate the mechanism of Ling-Gui-Zhu-Gan decoction (LGZGD) in regulating lipid metabolism and thus inhibiting ferroptosis. METHODS UPLC for the determination of the main chemical composition of LGZGD. A HF-induced rat model of metabolic cardiomyopathy was established. Echocardiography was used to detect cardiac function. Serum lipid levels, myocardial injury markers, and lipid peroxidation levels were detected. Pathological changes were detected. Lipid deposition was assessed by oil red O, and the mitochondrial ultrastructure was observed by electron microscopy. Mechanistically, PLIN5, CD36, ATGL, GPX4, ACSL4, FPN1, DRP1, MFF, FIS1, and OPA1 expressions were examined. After PA-induced H9c2 cells established, apoptosis, myocardial injury markers, and lipid peroxidation levels were detected and lipid deposition levels were assessed. The expressions of PLIN5, CD36, ATGL, GPX4, ACSL4 and FPN1 were detected. H9c2 cardiomyocytes with transient knockdown of PLIN5 and overexpression of PLIN5 were constructed and treated with drug administration and modeling, and the apoptosis level was detected by flow cytometry, the levels of lipid peroxidation and ROS were detected by fluorescence, and the protein and gene expressions of ACSL4 and GPX4 were detected. Results The main active components of LGZGD were liquiritin, isoliquiritin, cinnamic acid, cinnamaldehyde, glycyrrhizic acid, and atractylenolide III. LGZGD significantly improved cardiac dysfunction, lowered lipid level and lipid deposition, reduced CK, NT-proBNP and MDA levels, restored SOD levels, and improved inflammatory cell infiltration as well as collagen fiber deposition. LGZGD decreased the expression of PLIN5, CD36, ACSL4, and increased the expression of ATGL, GPX4, and FPN1. LGZGD also decreased the gene expression of DRP1, MFF, FIS1, and increased OPA1 expression. LGZGD significantly ameliorated PA-induced apoptosis, decreased lipid deposition, lowered lipid peroxidation levels and CK level, decreased PLIN5, CD36, and ACSL4 expressions, and increased ATGL, GPX4, and FPN1 expressions. LGZGD reversed cardiomyocyte injury aggravated by transient knockdown of PLIN5, decreased apoptosis levels, lipid peroxidation levels, ROS levels, and ACSL4 expressions, and increased GPX4 expression. LGZGD enhanced cardiomyocyte protection after overexpression of PLIN5, reduced apoptosis levels, lipid peroxidation level and ROS level, decreased ACSL4 expression, and increased GPX4 expression. CONCLUSION PLIN5 interferes with lipid peroxidation, regulates mitochondrial function, and inhibits HF-induced ferroptosis in cardiomyocytes. LGZGD ameliorates impairment of cardiac structural function in model rats through PLIN5-mediated ferroptosis pathway, and has the effect of preventing metabolic cardiomyopathy.
Collapse
Affiliation(s)
- Chuan-Zhi Zhao
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Hui-Min Ding
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zi-Qing Hu
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Lan Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yong-Qin Du
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Peng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China; Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China.
| | - Liang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China; Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China.
| |
Collapse
|
9
|
Lu C, Gao C, Wei J, Dong D, Sun M. SIRT1-FOXOs signaling pathway: A potential target for attenuating cardiomyopathy. Cell Signal 2024; 124:111409. [PMID: 39277092 DOI: 10.1016/j.cellsig.2024.111409] [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: 06/12/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Cardiomyopathy constitutes a global health burden. It refers to myocardial injury that causes alterations in cardiac structure and function, ultimately leading to heart failure. Currently, there is no definitive treatment for cardiomyopathy. This is because existing treatments primarily focus on drug interventions to attenuate symptoms rather than addressing the underlying causes of the disease. Notably, the cardiomyocyte loss is one of the key risk factors for cardiomyopathy. This loss can occur through various mechanisms such as metabolic disturbances, cardiac stress (e.g., oxidative stress), apoptosis as well as cell death resulting from disorders in autophagic flux, etc. Sirtuins (SIRTs) are categorized as class III histone deacetylases, with their enzyme activity primarily reliant on the substrate nicotinamide adenine dinucleotide (NAD (+)). Among them, Sirtuin 1 (SIRT1) is the most intensively studied in the cardiovascular system. Forkhead O transcription factors (FOXOs) are the downstream effectors of SIRT1. Several reports have shown that SIRT1 can form a signaling pathway with FOXOs in myocardial tissue, and this pathway plays a key regulatory role in cell loss. Thus, this review describes the basic mechanism of SIRT1-FOXOs in inhibiting cardiomyocyte loss and its favorable role in cardiomyopathy. Additionally, we summarized the SIRT1-FOXOs related regulation factor and prospects the SIRT1-FOXOs potential clinical application, which provide reference for the development of cardiomyopathy treatment.
Collapse
Affiliation(s)
- Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China.
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China.
| |
Collapse
|
10
|
Jose A, Fernando JJ, Kienesberger PC. Lysophosphatidic acid metabolism and signaling in heart disease. Can J Physiol Pharmacol 2024; 102:685-696. [PMID: 38968609 DOI: 10.1139/cjpp-2024-0077] [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] [Indexed: 07/07/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that is mainly produced by the secreted lysophospholipase D, autotaxin (ATX), and signals through at least six G protein-coupled receptors (LPA1-6). Extracellular LPA is degraded through lipid phosphate phosphatases (LPP1, LPP2, and LPP3) at the plasmamembrane, terminating LPA receptor signaling. The ATX-LPA-LPP3 pathway is critically involved in a wide range of physiological processes, including cell survival, migration, proliferation, angiogenesis, and organismal development. Similarly, dysregulation of this pathway has been linked to many pathological processes, including cardiovascular disease. This review summarizes and interprets current literature examining the regulation and role of the ATX-LPA-LPP3 axis in heart disease. Specifically, the contribution of altered LPA metabolism via ATX and LPP3 and resulting changes to LPA receptor signaling in obesity cardiomyopathy, cardiac mitochondrial dysfunction, myocardial infarction/ischemia-reperfusion injury, hypertrophic cardiomyopathy, and aortic valve stenosis is discussed.
Collapse
Affiliation(s)
- Anu Jose
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jeffy J Fernando
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| |
Collapse
|
11
|
Ajoolabady A, Pratico D, Dunn WB, Lip GYH, Ren J. Metabolomics: Implication in cardiovascular research and diseases. Obes Rev 2024; 25:e13825. [PMID: 39370721 DOI: 10.1111/obr.13825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 10/08/2024]
Abstract
Cellular metabolism influences all aspects of cellular function and is crucial for overall organismal health. Metabolic disorders related to cardiovascular health can lead to cardiovascular diseases (CVDs). Moreover, associated comorbidities may also damage cardiovascular metabolism, exacerbating CVD and perpetuating a vicious cycle. Given the prominent role of metabolic alterations in CVD, metabolomics has emerged as an imperative technique enabling a comprehensive assessment of metabolites and metabolic architecture within the body. Metabolite profile and metabolic pathways help to deepen and broaden our understanding of complex genomic landscape and pathophysiology of CVD. Here in this review, we aim to overview the experimental and clinical applications of metabolomics in pathogenesis, diagnosis, prognosis, and management of various CVD plus future perspectives and limitations.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Warwick B Dunn
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
12
|
Xu Z, Ma Z, Zhao X, Zhang B. Aerobic exercise mitigates high-fat diet-induced cardiac dysfunction, pyroptosis, and inflammation by inhibiting STING-NLRP3 signaling pathway. Mol Cell Biochem 2024; 479:3459-3470. [PMID: 38388792 DOI: 10.1007/s11010-024-04950-0] [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: 01/02/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
Obesity has been identified as an independent risk factor for cardiovascular disease. Recent reports have highlighted the significance of stimulator of interferon genes (STING)-NOD-like receptor protein 3 (NLRP3) signaling pathway mediated pyroptosis, and inflammation in cardiovascular disease. Previous studies have demonstrated that exercise training effectively prevents cardiac pyroptosis and inflammation in high-fat diet (HFD)-fed mice. However, it is currently unknown whether exercise reduces pyroptosis and inflammation in obese hearts by targeting the STING-NLRP3 signaling pathway. We investigated the impact of an 8-week aerobic exercise regimen on cardiac function, pyroptosis, inflammation, and the STING-NLRP3 signaling pathway in HFD-induced obese mice. Additionally, to explore the underlying mechanism of STING in exercise-mediated cardioprotection, we administered intraperitoneal injections of the STING agonist diABZI to the mice. Furthermore, to investigate the role of the STING-NLRP3 signaling pathway in HFD-induced cardiac dysfunction, we administered adeno-associated virus 9 (AAV9) encoding shRNA targeting STING (shRNA-STING) via tail vein injection to knockdown STING expression specifically in mouse hearts. After one week of AAV9 injection, we intraperitoneally injected nigericin as an NLRP3 agonist. We first found that aerobic exercise effectively suppressed HFD-mediated upregulation of STING and NLRP3 in the hearts. Moreover, we demonstrated that the protective effect of aerobic exercise in HFD-induced cardiac dysfunction, pyroptosis, and inflammation was impaired by stimulating the STING pathway using diABZI. Additionally, activation of the NLRP3 with nigericin abolished the ameliorative effect of STING deficiency in HFD-induced cardiac dysfunction, pyroptosis, and inflammation. Based on these findings, we concluded that 8-week aerobic exercise alleviates HFD-induced cardiac dysfunction, pyroptosis, and inflammation by targeting STING-NLRP3 signaling pathway. Inhibition of STING-NLRP3 signaling pathway may serve as a promising therapeutic strategy against obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Zujie Xu
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China.
| | - Zheying Ma
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China
| | - Xiaoqin Zhao
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China
| | - Bing Zhang
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
13
|
Zeng X, Lian X, Wang Y, Shang X, Yu H. Association of childhood-adulthood body size trajectories with risk of micro- and macrovascular complications among individuals with type 2 diabetes: a prospective study. Diabetol Metab Syndr 2024; 16:289. [PMID: 39609926 PMCID: PMC11605916 DOI: 10.1186/s13098-024-01499-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
AIMS This study aimed to investigate the association between childhood-adulthood body size trajectories and the risk of micro- and macrovascular complications in individuals with type 2 diabetes (T2D) using data from the UK Biobank. METHODS Childhood body size (average, thinner, and plumper) was self-reported for age 10, and adulthood body size (normal weight, overweight, obesity) was measured using body mass index at baseline. We defined nine body size trajectories by combining childhood and adulthood body size categories. Cox regression models were used to assess the association between these trajectories and the risk of diabetic complications. RESULTS Among 22,123 participants with T2D, 4,693 developed microvascular complications, and 3,640 developed macrovascular complications. Compared to individuals who maintained a normal body size from childhood to adulthood (the Average-Normal weight group), those with a high body size trajectory from childhood to adulthood (the Plumper-Obesity group) showed the highest risk for microvascular complications (HR 1.55; 95% CI: 1.31, 1.83), diabetic neuropathy (HR 2.18; 95% CI: 1.49, 3.21), diabetic nephropathy (HR 1.79; 95% CI: 1.45, 2.21), macrovascular complications (HR 1.30; 95% CI: 1.09, 1.55), and ischemic heart disease (HR 1.51; 95% CI: 1.23, 1.86). In contrast, individuals who were plumper in childhood but maintained a normal weight in adulthood did not show an increased risk of these complications. CONCLUSIONS A persistent high body size trajectory from childhood to adulthood is associated with the greatest risk of both micro- and macrovascular complications in individuals with T2D, whereas those who were plumper in childhood but achieved a normal weight in adulthood did not show an increased risk of these complications. These findings underscore the importance of weight management from childhood and maintaining a healthy weight throughout adulthood to reduce the risk of diabetic vascular complications in those with T2D.
Collapse
Affiliation(s)
- Xiaomin Zeng
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Eye Institute, Southern Medical University, Guangzhou, China
| | - Xingji Lian
- Department of Geriatrics, Guangzhou First People's Hospital, National Key Clinic Specialty, Guangzhou Medical University, Guangzhou, China
| | - Yaxin Wang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Eye Institute, Southern Medical University, Guangzhou, China
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Eye Institute, Southern Medical University, Guangzhou, China.
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Eye Institute, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
14
|
Liu SQ, Xie SY, Zhang T, Zhang H, Chen MY, Xing Y, Zhao N, Li L, Chen S, Wang SS, Zeng XF, Deng W, Tang QZ. Impeding Nucleotide-Binding Oligomerization Domain-Like Receptor 3 Inflammasome Ameliorates Cardiac Remodeling and Dysfunction in Obesity-Associated Cardiomyopathy. J Am Heart Assoc 2024:e035234. [PMID: 39604027 DOI: 10.1161/jaha.124.035234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Inflammation and metabolic disturbances are key culprits in the pathogenesis of obesity-associated cardiomyopathy. The NLRP3 (nucleotide-binding oligomerization domain-like receptor 3) inflammasome mediates the release of the proinflammatory cytokines IL-1β (interleukin-1β) and IL-18 by activating caspase-1, which is strongly implicated in metabolic disturbances. We here sought to determine whether NLRP3 inflammasome inhibition could ameliorate obesity cardiomyopathy and if so, to further explore its underlying mechanisms. METHODS AND RESULTS Male mice were fed a high-fat diet for 24 weeks to induce obesity cardiomyopathy. MCC950 was used to inhibit NLRP3 inflammasome activation. Recombinant adeno-associated virus serotype 9 encoding TXNIP (thioredoxin-interacting protein) under cTnT (cardiac troponin T) promoter and the mitochondrial-targeted antioxidant MitoTEMPO were injected into obese mice to investigate the specific mechanism. To mimic obesity cardiomyopathy in vitro, neonatal rat ventricular myocytes transfected with the small interfering RNA against TXNIP were incubated with 400 μmol palmitic acid for 24 hours. NLRP3 inflammasome was significantly increased in obese hearts. NLRP3 inflammasome inhibition by NLRP3 deletion or MCC950 prevented obesity-induced cardiac systolic and diastolic dysfunction, myocardial hypertrophy and fibrosis, and excessive lipid accumulation in male mice. Conversely, TXNIP overexpression worsened obesity-associated cardiomyopathy. Similarly, MCC950 treatment or TXNIP knockdown reduced palmitic acid-induced NLRP3 inflammasome activation and lipid storage. Mechanistically, abnormal NF-κB (nuclear factor kappa B) pathway activation, increased mitochondrial reactive oxygen species, and elevated TXNIP levels led to excessive NLRP3 inflammasome activation. CONCLUSIONS Our study confirms that aberrant NLRP3 inflammasome activation in cardiomyocytes worsens obesity-associated cardiomyopathy and implicates inhibition of NLRP3 inflammasome as a potent therapeutic approach for obesity cardiomyopathy.
Collapse
Affiliation(s)
- Shi-Qiang Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Sai-Yang Xie
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Tong Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Heng Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Meng-Ya Chen
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Yun Xing
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Nan Zhao
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Lanlan Li
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Sha-Sha Wang
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Xiao-Feng Zeng
- Cardiovascular Research Institute of Wuhan University Wuhan PR China
| | - Wei Deng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| | - Qi-Zhu Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan PR China
| |
Collapse
|
15
|
Museedi AS, Samson R, Le Jemtel TH. Menopause, epicardial adiposity and preserved ejection fraction heart failure. Int J Cardiol 2024; 415:132478. [PMID: 39179034 DOI: 10.1016/j.ijcard.2024.132478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Postmenopausal women are overrepresented in the preserved ejection heart failure population. Expansion of visceral and epicardial adipose tissue during the menopause transition leads to local and low-grade systemic inflammation that in turn contributes to left ventricular concentric remodeling, diastolic dysfunction and the development and progression of preserved ejection fraction. In contrast to visceral adipose tissue imaging, epicardial adipose tissue can be inexpensively imaged on low radiation coronary calcium score computerized tomography examination. The menopause transition provides a unique time frame to evaluate the contribution of epicardial adipose tissue expansion to the pathogenesis of preserved ejection heart failure.
Collapse
Affiliation(s)
- Abdulrahman S Museedi
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States of America
| | - Rohan Samson
- Advanced Heart Failure Therapies Program, University of Louisville Health-Jewish Hospital, 201 Abraham Flexner Way, Suite 1001, Louisville, KY 40202, United States of America
| | - Thierry H Le Jemtel
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States of America.
| |
Collapse
|
16
|
Chen D, Shi J, Wu Y, Miao L, Wang Z, Wang Y, Xu S, Lou Y. Dapagliflozin alleviates high-fat-induced obesity cardiomyopathy by inhibiting ferroptosis. ESC Heart Fail 2024. [PMID: 39523867 DOI: 10.1002/ehf2.15150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/10/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
AIM Dapagliflozin (Dapa) is a novel hypoglycaemic agent with multiple cardiovascular protective effects, and it is widely used in treatment of heart failure patients, but whether it can improve obese phenotype of heart failure and its mechanism is still unclear. Ferroptosis is an iron dependent form of cell death and has been proved to be an important role in heart failure. The aim of this study is to determine whether Dapa improves obesity-related heart failure by regulating ferroptosis in high-fat diet rats. METHODS AND RESULTS Male SD rats were fed a high-fat diet for 12 weeks and confirmed of obese heart failure by metabolic parameters and cardiac ultrasound. Being overweight by 20% compared with the normal group, with elevated systolic blood pressure and abnormal levels of insulin and blood lipid (TG and LDL-c), is recognized as obesity. The obese rats with reduced EF, FS, and E/A shown on ultrasound are defined as the obese heart failure (OHF) group. Histological tests confirmed the more pronounced cardiac fibrosis, mitochondrial volume and collagen deposition in OHF group. Dapa treatment effectively reduced body weight, INS, ISI/IRI index, TG and HDL-C levels (P < 0.05). Also, Dapa administration can slightly decrease the SBP and DBP levels; however, there was no statistical difference among those four groups. Furthermore, Dapa treatment can significantly improve high-fat induced systolic and diastolic dysfunction via regulating cardiac histological abnormalities, including less obvious mitochondrial swelling, muscle fibre dissolution and collagen deposition. Additionally, genes from the OHF group were used by GO enrichment analysis, and it shows that ferroptosis metabolic pathway participated in the development of obese phenotype of heart failure. More importantly, Dapa significantly inhibited Fe2+ and MDA levels (P < 0.05), but augmented GSH content (P < 0.05). In addition, the mRNAs and protein expression of some important regulators of ferroptosis, like GPX4, SLC7A11, FTH1 and FPN1, were all decreased after Dapa intervention. CONCLUSION Dapa improved high-fat induced obese cardiac dysfunction via regulating ferroptosis pathway.
Collapse
Affiliation(s)
- Di Chen
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiahao Shi
- Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yue Wu
- Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lizhu Miao
- Yancheng No. 1 People's Hospital, Yancheng, China
| | - Zilin Wang
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yixuan Wang
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Siwei Xu
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Yu Lou
- The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Su S, Quan C, Chen Q, Wang R, Du Q, Zhu S, Li M, Yang X, Rong P, Chen J, Bai Y, Zheng W, Feng W, Liu M, Xie B, Ouyang K, Shi YS, Lan F, Zhang X, Xiao R, Chen X, Wang HY, Chen S. AS160 is a lipid-responsive regulator of cardiac Ca 2+ homeostasis by controlling lysophosphatidylinositol metabolism and signaling. Nat Commun 2024; 15:9602. [PMID: 39505896 PMCID: PMC11542008 DOI: 10.1038/s41467-024-54031-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
The obese heart undergoes metabolic remodeling and exhibits impaired calcium (Ca2+) homeostasis, which are two critical assaults leading to cardiac dysfunction. The molecular mechanisms underlying these alterations in obese heart are not well understood. Here, we show that the Rab-GTPase activating protein AS160 is a lipid-responsive regulator of Ca2+ homeostasis through governing lysophosphatidylinositol metabolism and signaling. Palmitic acid/high fat diet inhibits AS160 activity through phosphorylation by NEK6, which consequently activates its downstream target Rab8a. Inactivation of AS160 in cardiomyocytes elevates cytosolic Ca2+ that subsequently impairs cardiac contractility. Mechanistically, Rab8a downstream of AS160 interacts with DDHD1 to increase lysophosphatidylinositol metabolism and signaling that leads to Ca2+ release from sarcoplasmic reticulum. Inactivation of NEK6 prevents inhibition of AS160 by palmitic acid/high fat diet, and alleviates cardiac dysfunction in high fat diet-fed mice. Together, our findings reveal a regulatory mechanism governing metabolic remodeling and Ca2+ homeostasis in obese heart, and have therapeutic implications to combat obesity cardiomyopathy.
Collapse
Affiliation(s)
- Shu Su
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Chao Quan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ruizhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qian Du
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Sangsang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Min Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Xinyu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yingyu Bai
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen Zheng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Minjun Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Ruiping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Xiongwen Chen
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Nanjing, China.
| |
Collapse
|
18
|
Comella F, Aragón-Herrera A, Pirozzi C, Feijóo-Bandin S, Lama A, Opallo N, Melini S, Del Piano F, Gualillo O, Meli R, Mattace Raso G, Lago F. Oleoylethanolamide mitigates cardiometabolic disruption secondary to obesity induced by high-fat diet in mice. Life Sci 2024; 359:123226. [PMID: 39515418 DOI: 10.1016/j.lfs.2024.123226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic lipid overnutrition has been demonstrated to promote cardiac dysfunction resulting from metabolic derangement, inflammation, and fibrosis. Oleoylethanolamide (OEA), an endogenous peroxisome proliferator activating receptor (PPAR)-α agonist, has been extensively studied for its metabolic properties. The aim of this study was to determine if OEA has beneficial effects on high-fat diet (HFD)-induced cardiac disruption in obese mice, focusing on the underlying pathological mechanisms. OEA treatment restores the metabolic pattern, improving serum glycaemic and lipid profile. OEA also reduces heart weight and serum creatine kinase-myocardial band (CK-MB), a marker of cardiac damage. Accordingly, OEA modulates cardiac metabolism, increasing insulin signaling and reducing lipid accumulation. OEA increases AMPK and AKT phosphorylation, converging in the rise of AS160 activation and glucose transporter (GLUT)4 protein level. Moreover, OEA reduces the transcription of the cardiac fatty acid transporter CD36 and fatty acid synthase and increases PPAR-α mRNA levels. Adiponectin and meteorite-like protein transcription levels were significantly reduced by OEA in HFD mice, as well as those of inflammatory cytokines and pro-fibrotic markers. An increased autophagic process was also shown, contributing to OEA's cardioprotective effects. Metabolomic analyses of cardiac tissue revealed the modulation of different lipids, including triglycerides, glycerophospholipids and sphingomyelins by OEA treatment. In vitro experiments on HL-1 cardiomyocytes showed OEA's capability in reducing inflammation and fibrosis following palmitate challenge, demonstrating a direct activity of OEA on cardiac cells, mainly mediated by PPAR-α activation. Our results indicate OEA as a potential therapeutic to restrain cardiac damage associated with metabolic disorders.
Collapse
Affiliation(s)
- Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Sandra Feijóo-Bandin
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Stefania Melini
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II, 80100 Naples, Italy
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain; International PhD School, University of Santiago de Compostela (EDIUS), Santiago de Compostela 15706, Spain
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy.
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
19
|
Yerra VG, Connelly KA. Extrarenal Benefits of SGLT2 Inhibitors in the Treatment of Cardiomyopathies. Physiology (Bethesda) 2024; 39:0. [PMID: 38888433 DOI: 10.1152/physiol.00008.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have emerged as pivotal medications for heart failure, demonstrating remarkable cardiovascular benefits extending beyond their glucose-lowering effects. The unexpected cardiovascular advantages have intrigued and prompted the scientific community to delve into the mechanistic underpinnings of these novel actions. Preclinical studies have generated many mechanistic theories, ranging from their renal and extrarenal effects to potential direct actions on cardiac muscle cells, to elucidate the mechanisms linking these drugs to clinical cardiovascular outcomes. Despite the strengths and limitations of each theory, many await validation in human studies. Furthermore, whether SGLT2 inhibitors confer therapeutic benefits in specific subsets of cardiomyopathies akin to their efficacy in other heart failure populations remains unclear. By examining the shared pathological features between heart failure resulting from vascular diseases and other causes of cardiomyopathy, certain specific molecular actions of SGLT2 inhibitors (particularly those targeting cardiomyocytes) would support the concept that these medications will yield therapeutic benefits across a broad range of cardiomyopathies. This article aims to discuss the important mechanisms of SGLT2 inhibitors and their implications in hypertrophic and dilated cardiomyopathies. Furthermore, we offer insights into future research directions for SGLT2 inhibitor studies, which hold the potential to further elucidate the proposed biological mechanisms in greater detail.
Collapse
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| |
Collapse
|
20
|
Aleman R, Napoli F, Jamroz T, Baran DA, Sheffield C, Navia J, Rosenthal RJ, Brozzi NA. The dual prevalence of advanced degrees of obesity and heart failure: a study from the National Inpatient Sample database. Surg Obes Relat Dis 2024; 20:1090-1097. [PMID: 38876939 DOI: 10.1016/j.soard.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND National prevalence rates for obesity and heart failure (HF) have been steadily increasing, which predisposes patients to higher morbidity and mortality rates. OBJECTIVES The purpose of this study was to evaluate the prevalence of HF stages in hospitalized patients according to their body mass index (BMI). SETTING Academic institution. METHODS National Inpatient Sample data from 2016 to 2018 were examined to identify patients with obesity, HF (presence or absence of advanced HF [AHF]), and cardiogenic shock (CS). The proportion of hospital admissions was determined for each category on the basis of the presence of AHF with/without CS. A comparative analysis was performed between patients with and without AHF, and multivariate logistic regression analysis was performed for the event of AHF. The same analyses were performed for the event of CS. RESULTS A total of 3,354,970 hospital admissions were identified. The prevalence of hospital admissions with a diagnosis of AHF and class III obesity and a diagnosis of CS and class III obesity was 21% and .5%, respectively. The prevalence of AHF and other classes of BMI and CS and other classes of BMI was 17% and .5%, respectively. The univariate analysis showed that there were significant variations in 10 factors between hospital admissions with/without the diagnosis of both AHF and CS. Statistical analyses indicated the following findings: Hospitalized patients in higher obesity groups are more likely to have AHF, and they are less likely to have CS compared with those with a BMI of ≤29.9. CONCLUSIONS This study revealed that the prevalence of AHF was significantly higher in hospitalized patients with class III obesity. These findings have implications for clinical management, and it can be inferred that these patients are less likely to receive advanced cardiac replacement therapies and might benefit from innovative approaches to address severe dual morbidity.
Collapse
Affiliation(s)
- Rene Aleman
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Federico Napoli
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Tatiana Jamroz
- Department of Anesthesiology, Weston Hospital, Weston, Florida
| | - David A Baran
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Cedric Sheffield
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Jose Navia
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Raul J Rosenthal
- The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Nicolas A Brozzi
- Heart, Vascular & Thoracic Institute, Cleveland Clinic Florida, Weston, Florida.
| |
Collapse
|
21
|
Kaimori R, Nishida H, Tamura M, Kuroki K, Murata K, Kawamura K, Mori S, Daa T. Obesity cardiomyopathy could contribute to sudden cardiac death: a Japanese epidemiological morphological study. Cardiovasc Diabetol 2024; 23:378. [PMID: 39449032 PMCID: PMC11520142 DOI: 10.1186/s12933-024-02456-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND We aimed to clarify the existence and pathological features of obesity cardiomyopathy (OCM) in Japan using our series of autopsy cases. METHODS In this retrospective autopsy study, OCM was defined as cardiac hypertrophy (≥ 400 g in men, ≥ 320 g in women) of unknown aetiology in individuals with obesity (body mass index [BMI] ≥ 25 kg/m2 according to the Japanese definition of obesity). We compared cases of OCM with those with obesity without cardiac hypertrophy (OB) and normal weight without cardiac hypertrophy (normal control). Macroscopically, heart weight and cardiac parameters, including epicardial adipose tissue, were measured. Fibrosis, cardiomyocyte diameter, and adipose tissue infiltration were analysed microscopically. RESULTS Of the 294 cases, we identified 19 cases of OCM (6.5%) and compared them with the OB and normal control groups. Patients with OCM were slightly younger than non-OCM patients (p = 0.081). The median heart weight was significantly heavier in OCM cases than in OB cases (435 g, interquartile range [IQR] 408-515 g vs. 360 g, IQR 341-385 g). Macroscopically, OCM hearts had a "globoid" appearance with a thickened right ventricular outflow tract. Some OCM cases showed focal interstitial fibrosis in the left ventricle. Approximately half the OCM cases were diagnosed with sudden cardiac death (SCD), with significant differences. CONCLUSIONS The prevalence of OCM may be higher than expected in Japan, and this may be a specific pathological finding. Given that approximately half the cases of OCM were due to SCD, OCM may cause SCD, emphasizing the need to recognise and diagnose OCM.
Collapse
Affiliation(s)
- Ryo Kaimori
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan.
- Department of Diagnostic Pathology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan.
| | - Haruto Nishida
- Department of Diagnostic Pathology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan.
| | - Mari Tamura
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Kohji Kuroki
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Kumi Murata
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Kazuhiro Kawamura
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
- Department of Diagnostic Pathology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Shinjiro Mori
- Department of Forensic Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Tsutomu Daa
- Department of Diagnostic Pathology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| |
Collapse
|
22
|
Zhuang Z, Zhu Y, Tao J, Liu Y, Lin J, Yang C, Dong C, Qin X, Li Q, Reiter RJ, Wang G, Pei Z, Ren J. UCF101 Rescues against Diabetes-Evoked Cardiac Remodeling and Contractile Anomalies through AMP-Activated Protein Kinase-Mediated Induction of Mitophagy. Pharmacology 2024:1-14. [PMID: 39413739 DOI: 10.1159/000541569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Diabetes mellitus is known to provoke devastating anomalies in myocardial structure and function, while effective therapeutic regimen is still lacking. The selective protease inhibitor UCF101 (5-[5-(2-nitrophenyl) furfuryl iodine]-1,3-diphenyl-2-thiobarbituric acid) has been shown to fend off ischemic heart injury, although its impact on diabetic cardiomyopathy remains elusive. METHODS Our present work was conducted to examine the effect of UCF101 on experimental diabetes-evoked cardiac geometric and functional abnormalities as well as mechanisms involved. Adult mice were made diabetic using streptozotocin (STZ, 50 mg/kg, i.p., for 5 days) while receiving UCF101 (7.15 mg/kg, i.p.). RESULTS STZ evoked cardiac hypertrophy, interstitial fibrosis, mitochondrial ultrastructural damage, oxidative stress, dampened autophagy (LC3B, Beclin 1, elevated p62), mitophagy (FUNDC1 and Parkin with upregulated TOM20), increased left ventricular end systolic diameter, reduced fractional shortening, ejection fraction, cardiomyocyte shortening capacity, velocities of shortening/re-lengthening, and rise in intracellular Ca2+ in conjunction with elongated diastole and intracellular Ca2+ removal, the responses were overtly reconciled by UCF101 with little effects from UCF101 itself. Levels of cell injury markers Omi/HtrA2, TNFα, and stress signaling (JNK, ERK, p38) were overtly enhanced along with compromised phosphorylation of cellular fuel AMP-activated protein kinase (AMPK) (Thr172) and cell survival molecule GSK3β, as well as downregulated SERCA2a and elevated phospholamban, the effect was reversed by UCF101 (except for SERCA2a). AMPK knockout, pharmacological inhibition, the mitophagy inhibitor liensinine, and parkin knockout nullified UCF101-offered cardioprotection in diabetes. UCF101 reversed STZ-induced upregulation in the AMPK degrading enzymes PP2A and PP2C. CONCLUSION These findings suggest that UCF101 rescues diabetes-mediated alterations in cardiac structure and function, likely through AMPK-mediated regulation of mitophagy.
Collapse
Affiliation(s)
- Zhiqiang Zhuang
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuxi Zhu
- The Second Department of Cardiology, Nanchang City Renmin Hospital, Nanchang, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yandong Liu
- The Second Department of Cardiology, Nanchang City Renmin Hospital, Nanchang, China
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Chunjie Yang
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Chule Dong
- Department of Clinical Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xing Qin
- Department of Cardiology, Air Force Medical University, Xi'an, China
| | - Qun Li
- Guangzhou Cellprotek Pharmaceutical Co., Ltd, Guangzhou, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas, USA
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, China
| | - Zhaohui Pei
- The Second Department of Cardiology, Nanchang City Renmin Hospital, Nanchang, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
23
|
Okon IA, Beshel JA, Okorocha AE, Eze EE, Owu DU. Blood pressure and ECG variables of healthy young males and females participating in moderate aerobic exercise. J Bodyw Mov Ther 2024; 40:965-970. [PMID: 39593704 DOI: 10.1016/j.jbmt.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/03/2024] [Accepted: 07/07/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION Changes in blood pressure and electrocardiogram are important factors that determine exercise testing. This study investigated blood pressure and electrocardiogram (ECG) changes in healthy young adults after performing acute moderate aerobic exercise protocols. METHODS Forty young healthy untrained non-athletes, twenty males and twenty females (age, 25 ± 5.6 years; body weight, 65 ± 4.0 kg; body height, 176.9 ± 2.5 cm) were recruited for the study. The exercise regimen was acute moderate exercise for 20 min on a treadmill consistently for 14 days daily at the speed of 13 km/h. The body weight, blood pressure, and electrocardiograph were measured before and after exercise. RESULTS There was a significant reduction (P < 0.05) in body weight (59 ± 3.2 kg) of female participants after 14 days of aerobic exercise relative to their baseline mean value (63 ± 2.9 kg). The systolic blood pressure decreased (P < 0.05) in males (117 ± 1.1 mmHg) and females (117 ± 1.0 mmHg) when compared to the mean baseline values in males (127 ± 1.3 mmHg) and females (128 ±0 .3 mmHg). The diastolic blood pressure also decreased (P < 0.05) in male (71 ± 0.88 mmHg) participants after exercise when compared to the baseline mean values (79 ± 1.2 mmHg) while there was no change in diastolic blood pressure of females. The ECG parameters remained unchanged, while the heart rate (75 ± 1.3 beats/min) increased (P < 0.05) after exercise in all participants relative to the baseline (69 ± 2 beats/min). CONCLUSION The results suggest that moderate aerobic exercise normalized blood pressure and electrical activity of the heart while reducing heart rate after 14 days of consistent aerobic exercise in healthy individuals.
Collapse
Affiliation(s)
- Idara Asuquo Okon
- Department of Physiology, Kampala International University, Western Campus, Bushenyi, Uganda; Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria.
| | - Justin Atiang Beshel
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Albert E Okorocha
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Elijah Ekene Eze
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Daniel U Owu
- Department of Physiology, Kampala International University, Western Campus, Bushenyi, Uganda; Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
24
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in obesity: Lessons from the heart and other tissues. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119814. [PMID: 39128598 DOI: 10.1016/j.bbamcr.2024.119814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Obesity causes a range of tissue dysfunctions that increases the risk for morbidity and mortality. Protein kinase D (PKD) represents a family of stress-activated intracellular signalling proteins that regulate essential processes such as cell proliferation and differentiation, cell survival, and exocytosis. Evidence suggests that PKD regulates the cellular adaptations to the obese environment in metabolically important tissues and drives the development of a variety of diseases. This review explores the role that PKD plays in tissue dysfunction in obesity, with special consideration of the development of obesity-mediated cardiomyopathy, a distinct cardiovascular disease that occurs in the absence of common comorbidities and leads to eventual heart failure and death. The downstream mechanisms mediated by PKD that could contribute to dysfunctions observed in the heart and other metabolically important tissues in obesity, and the predicted cell types involved are discussed to suggest potential targets for the development of therapeutics against obesity-related disease.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia; The Fralin Biomedical Research Institute at Virginia Tech Carilion, Centre for Vascular and Heart Research, Roanoke, VA, USA.
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia.
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia.
| |
Collapse
|
25
|
Caliskan B, Inanir M. SPECT myocardial perfusion imaging for the evaluation of left ventricular mechanical dyssynchrony in obese patients. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:1037-1043. [PMID: 39031559 DOI: 10.1002/jcu.23752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 07/22/2024]
Abstract
OBJECTIVE Left ventricular dyssynchrony (LVD), the loss of coordinated contraction in the left ventricle, is an early sign of heart failure. LVD can be assessed using phase analysis techniques with gated single-photon emission computed tomography (SPECT) myocardial perfusion imaging (MPI). This study aimed to investigate the impact of obesity on LVD through phase analysis. METHODS We retrospectively enrolled 152 obese patients and 80 age- and sex-matched nonobese patients who showed normal myocardial perfusion and normal left ventricular ejection fraction (LVEF) on MPI. Phase standard deviation (PSD) and phase histogram bandwidth (PBW), as phase analysis parameters, were compared between patients with and without obesity. RESULTS Although PSD values were within the normal range (cut-off value >23) for both groups, the PSD values of obese patients were higher than those of the nonobese (20.49 ± 8.66 vs. 14.81 ± 4.93; p < 0.05). PBW values of obese patients were statistically significantly higher than those of the nonobese (57.03 ± 23.17 vs. 41.40 ± 9.96; p < 0.05). The PBW values of obese patients exceeded the normal limits (cut-off value >49). A weak positive correlation was observed between body mass index (BMI) and PBW values in obese patients (r = 0.181, p < 0.05). In patients of normal weight, no correlations were found between BMI and phase analysis parameters. CONCLUSION LVD may develop in obese patients, even when myocardial perfusion and ejection fraction are preserved. The use of phase analysis with gated SPECT could be an additional finding improving the early detection of left ventricular dyssynchrony in obese patients.
Collapse
Affiliation(s)
- Billur Caliskan
- Department of Nuclear Medicine, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| | - Mehmet Inanir
- Department of Cardiology, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| |
Collapse
|
26
|
Luo Y, Ye T, Tian H, Song H, Kan C, Han F, Hou N, Sun X, Zhang J. Empagliflozin alleviates obesity-related cardiac dysfunction via the activation of SIRT3-mediated autophagosome formation. Lipids Health Dis 2024; 23:308. [PMID: 39334359 PMCID: PMC11430456 DOI: 10.1186/s12944-024-02293-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Empagliflozin (EMPA) has demonstrated efficacy in providing cardiovascular benefits in metabolic diseases. However, the direct effect of EMPA on autophagy in obesity-related cardiac dysfunction remains unclear. Therefore, this study aimed to determine changes in cardiac autophagy during diet-induced obesity and clarify the exact mechanism by which EMPA regulates autophagic pathways. METHODS Male C57BL/6J mice were fed a 12-week high-fat diet (HFD) followed by 8 weeks of EMPA treatment. Body composition analysis and echocardiography were performed to evaluate metabolic alterations and cardiac function. Histological and immunofluorescence staining was used to evaluate potential enhancements in myocardial structure and biological function. Additionally, H9c2 cells were transfected with small interfering RNA targeting sirtuin 3 (SIRT3) and further treated with palmitic acid (PA) with or without EMPA. Autophagy-related targets were analyzed by western blotting and RT‒qPCR. RESULTS EMPA administration effectively ameliorated metabolic disorders and cardiac diastolic dysfunction in HFD-fed mice. EMPA prevented obesity-induced myocardial hypertrophy, fibrosis, and inflammation through the activation of SIRT3-mediated autophagosome formation. The upregulation of SIRT3 triggered by EMPA promoted the initiation of autophagy by activating AMP-activated protein kinase (AMPK) and Beclin1. Furthermore, activated SIRT3 contributed to the elongation of autophagosomes through autophagy-related 4B cysteine peptidase (ATG4B) and autophagy-related 5 (ATG5). CONCLUSIONS EMPA promotes SIRT3-mediated autophagosome formation to alleviate damage to the cardiac structure and function of obese mice. Activated SIRT3 initiates autophagy through AMPK/Beclin1 and further stimulates elongation of the autophagosome membrane via ATG4B/ATG5. These results provide a new explanation for the cardioprotective benefits of EMPA in obesity.
Collapse
Affiliation(s)
- Youhong Luo
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Tongtong Ye
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Hongzhan Tian
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Hongwei Song
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China.
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China.
| |
Collapse
|
27
|
Guo B, Zhang F, Yin Y, Ning X, Zhang Z, Meng Q, Yang Z, Jiang W, Liu M, Wang Y, Sun L, Yu L, Mu N. Post-translational modifications of pyruvate dehydrogenase complex in cardiovascular disease. iScience 2024; 27:110633. [PMID: 39224515 PMCID: PMC11367490 DOI: 10.1016/j.isci.2024.110633] [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] [Indexed: 09/04/2024] Open
Abstract
Pyruvate dehydrogenase complex (PDC) is a crucial enzyme that connects glycolysis and the tricarboxylic acid (TCA) cycle pathway. It plays an essential role in regulating glucose metabolism for energy production by catalyzing the oxidative decarboxylation of pyruvate to acetyl coenzyme A. Importantly, the activity of PDC is regulated through post-translational modifications (PTMs), phosphorylation, acetylation, and O-GlcNAcylation. These PTMs have significant effects on PDC activity under both physiological and pathophysiological conditions, making them potential targets for metabolism-related diseases. This review specifically focuses on the PTMs of PDC in cardiovascular diseases (CVDs) such as myocardial ischemia/reperfusion injury, diabetic cardiomyopathy, obesity-related cardiomyopathy, heart failure (HF), and vascular diseases. The findings from this review offer theoretical references for the diagnosis, treatment, and prognosis of CVD.
Collapse
Affiliation(s)
- Bo Guo
- Department of Pharmacy, Northwest Woman’s and Children’s Hospital, Xi’an, China
| | - Fujiao Zhang
- College of Life Sciences, Northwest University, Xi’an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Xingmin Ning
- College of Life Sciences, Northwest University, Xi’an, China
| | - Zihui Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Qinglei Meng
- College of Life Sciences, Yan’an University, Yan’an, China
| | - Ziqi Yang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Lijuan Sun
- Eye Institute of Chinese PLA and Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
28
|
Belin MAF, Vieira TA, Grandini NA, Siqueira JS, Palacio TLN, Cruzeiro J, Sormani LE, Tanganini MD, Barbosa GS, Gregolin CS, de Campos DHS, Bazan SGZ, Minatel IO, Lima GPP, Correa CR. Cardiac biogenic amine profile and its relationship with parameters of cardiovascular disease in obesity. Vascul Pharmacol 2024; 156:107412. [PMID: 39033868 DOI: 10.1016/j.vph.2024.107412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
AIMS To identify the cardiac biogenic amine profile of obese rats and associate these compounds with parameters of cardiovascular disease. MAIN METHODS Wistar rats (n = 20) were randomly distributed into two groups: control and obese. Obesity was induced by a high-sugar fat diet. Biochemical parameters were evaluated. Doppler Echocardiography and systolic blood pressure; interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α), protein carbonylation, ferric reducing antioxidant power (FRAP), and catalase activity were measured in cardiac tissue. HPLC evaluated the cardiac biogenic profile. Data were compared using the Student's T or Mann-Whitney tests and Spearman's correlation at 5% significance. The principal component analysis (PCA) was performed. KEY FINDINGS Obesity generated hypertension, cardiac remodeling and dysfunction, and imbalanced all biochemical, inflammatory, and oxidative markers (p < 0.001). Eight biogenic amines were found in cardiac tissue. Obesity increased serotonin and decreased agmatine, putrescine, cadaverine, and spermidine. Serotonin (r = 0.534 to 0.808) was strong and positively correlated with obesity, biochemical parameters, cardiac inflammation, oxidative stress, hypertension, cardiac remodeling, and dysfunction (p < 0.001). Spermidine (r = -0.560 to -0.680), putrescine (r = -0.532 to -0.805), cadaverine (r = -0.534 to -0.860), and agmatine (r = -0.579 to -0.884) were inversely correlated with the same parameters (p < 0.001). PCA allowed for distinguishing the control and obese groups. SIGNIFICANCE There are strong correlations between cardiac biogenic amine levels, cardiac remodeling, and dysfunction resulting from obesity. CONCLUSION There is an association between cardiac biogenic amines and cardiovascular disease in obesity. In addition, agmatine, putrescine, cadaverine, and, mainly, serotonin may be new biomarkers for cardiovascular health in obesity and help to improve the diagnosis and treatment of CVD resulting or not from obesity. However, more research is needed to support this conclusion.
Collapse
Affiliation(s)
| | - Taynara Aparecida Vieira
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Núbia Alves Grandini
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Juliana Silva Siqueira
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | | | - Jordanna Cruzeiro
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Luis Eduardo Sormani
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Murilo Dalarme Tanganini
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Gabriela Souza Barbosa
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Cristina Schmitt Gregolin
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | | | | | - Igor Otávio Minatel
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Giuseppina Pace Pereira Lima
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Camila Renata Correa
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil.
| |
Collapse
|
29
|
Kaur S, Khullar N, Navik U, Bali A, Bhatti GK, Bhatti JS. Multifaceted role of dynamin-related protein 1 in cardiovascular disease: From mitochondrial fission to therapeutic interventions. Mitochondrion 2024; 78:101904. [PMID: 38763184 DOI: 10.1016/j.mito.2024.101904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Mitochondria are central to cellular energy production and metabolic regulation, particularly in cardiomyocytes. These organelles constantly undergo cycles of fusion and fission, orchestrated by key proteins like Dynamin-related Protein 1 (Drp-1). This review focuses on the intricate roles of Drp-1 in regulating mitochondrial dynamics, its implications in cardiovascular health, and particularly in myocardial infarction. Drp-1 is not merely a mediator of mitochondrial fission; it also plays pivotal roles in autophagy, mitophagy, apoptosis, and necrosis in cardiac cells. This multifaceted functionality is often modulated through various post-translational alterations, and Drp-1's interaction with intracellular calcium (Ca2 + ) adds another layer of complexity. We also explore the pathological consequences of Drp-1 dysregulation, including increased reactive oxygen species (ROS) production and endothelial dysfunction. Furthermore, this review delves into the potential therapeutic interventions targeting Drp-1 to modulate mitochondrial dynamics and improve cardiovascular outcomes. We highlight recent findings on the interaction between Drp-1 and sirtuin-3 and suggest that understanding this interaction may open new avenues for therapeutically modulating endothelial cells, fibroblasts, and cardiomyocytes. As the cardiovascular system increasingly becomes the focal point of aging and chronic disease research, understanding the nuances of Drp-1's functionality can lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India.
| |
Collapse
|
30
|
Seferović PM, Paulus WJ, Rosano G, Polovina M, Petrie MC, Jhund PS, Tschöpe C, Sattar N, Piepoli M, Papp Z, Standl E, Mamas MA, Valensi P, Linhart A, Lalić N, Ceriello A, Döhner W, Ristić A, Milinković I, Seferović J, Cosentino F, Metra M, Coats AJS. Diabetic myocardial disorder. A clinical consensus statement of the Heart Failure Association of the ESC and the ESC Working Group on Myocardial & Pericardial Diseases. Eur J Heart Fail 2024; 26:1893-1903. [PMID: 38896048 DOI: 10.1002/ejhf.3347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
The association between type 2 diabetes mellitus (T2DM) and heart failure (HF) has been firmly established; however, the entity of diabetic myocardial disorder (previously called diabetic cardiomyopathy) remains a matter of debate. Diabetic myocardial disorder was originally described as the occurrence of myocardial structural/functional abnormalities associated with T2DM in the absence of coronary heart disease, hypertension and/or obesity. However, supporting evidence has been derived from experimental and small clinical studies. Only a minority of T2DM patients are recognized as having this condition in the absence of contributing factors, thereby limiting its clinical utility. Therefore, this concept is increasingly being viewed along the evolving HF trajectory, where patients with T2DM and asymptomatic structural/functional cardiac abnormalities could be considered as having pre-HF. The importance of recognizing this stage has gained interest due to the potential for current treatments to halt or delay the progression to overt HF in some patients. This document is an expert consensus statement of the Heart Failure Association of the ESC and the ESC Working Group on Myocardial & Pericardial Diseases. It summarizes contemporary understanding of the association between T2DM and HF and discuses current knowledge and uncertainties about diabetic myocardial disorder that deserve future research. It also proposes a new definition, whereby diabetic myocardial disorder is defined as systolic and/or diastolic myocardial dysfunction in the presence of diabetes. Diabetes is rarely exclusively responsible for myocardial dysfunction, but usually acts in association with obesity, arterial hypertension, chronic kidney disease and/or coronary artery disease, causing additive myocardial impairment.
Collapse
Affiliation(s)
- Petar M Seferović
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Walter J Paulus
- Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Giuseppe Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, Italy
| | - Marija Polovina
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Pardeep S Jhund
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Carsten Tschöpe
- Berlin Institute of Health at Charité - Center for Regenerative Therapies, Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology (CVK) and German Centre for Cardiovascular Research (DZHK)- Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Massimo Piepoli
- Cardiology University Department, RCCS Policlinico San Donato, San Donato Milanese, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eberhard Standl
- Diabetes Research Group e.V. at Munich Helmholtz Center, Munich, Germany
| | - Mamas A Mamas
- Cardiovascular Research Group, Keele University, Keele, UK
| | - Paul Valensi
- Polyclinique d'Aubervilliers, Aubervilliers, and Paris Nord University, Bobigny, France
| | - Ales Linhart
- Department of Internal Medicine, School of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Nebojša Lalić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
- Department of Endocrinology, University Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Wolfram Döhner
- Berlin Institute of Health at Charité - Center for Regenerative Therapies, Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology (CVK) and German Centre for Cardiovascular Research (DZHK)- Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin (CSB), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Arsen Ristić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Ivan Milinković
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelena Seferović
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Endocrinology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | |
Collapse
|
31
|
Zhang L, Han H, Xu A, Sathe A, Fu S, Zhao J, Cai W, Yang Y, Liu J, Bai H, Ben J, Zhu X, Li X, Yang Q, Wang Z, Gu Y, Xing C, Schiattarella GG, Cheng SY, Zhang H, Chen Q. Lysozyme 1 Inflamed CCR2 + Macrophages Promote Obesity-Induced Cardiac Dysfunction. Circ Res 2024; 135:596-613. [PMID: 39056179 DOI: 10.1161/circresaha.124.324106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Macrophages are key players in obesity-associated cardiovascular diseases, which are marked by inflammatory and immune alterations. However, the pathophysiological mechanisms underlying macrophage's role in obesity-induced cardiac inflammation are incompletely understood. Our study aimed to identify the key macrophage population involved in obesity-induced cardiac dysfunction and investigate the molecular mechanism that contributes to the inflammatory response. METHODS In this study, we used single-cell RNA-sequencing analysis of Cd45+CD11b+F4/80+ cardiac macrophages to explore the heterogeneity of cardiac macrophages. The CCR2+ (C-C chemokine receptor 2) macrophages were specifically removed by a dual recombinase approach, and the macrophage CCR2 was deleted to investigate their functions. We also performed cleavage under target and tagmentation analysis, chromatin immunoprecipitation-polymerase chain reaction, luciferase assay, and macrophage-specific lentivirus transfection to define the impact of lysozyme C in macrophages on obesity-induced inflammation. RESULTS We find that the Ccr2 cluster undergoes a functional transition from homeostatic maintenance to proinflammation. Our data highlight specific changes in macrophage behavior during cardiac dysfunction under metabolic challenge. Consistently, inducible ablation of CCR2+CX3CR1+ macrophages or selective deletion of macrophage CCR2 prevents obesity-induced cardiac dysfunction. At the mechanistic level, we demonstrate that the obesity-induced functional shift of CCR2-expressing macrophages is mediated by the CCR2/activating transcription factor 3/lysozyme 1/NF-κB (nuclear factor kappa B) signaling. Finally, we uncover a noncanonical role for lysozyme 1 as a transcription activator, binding to the RelA promoter, driving NF-κB signaling, and strongly promoting inflammation and cardiac dysfunction in obesity. CONCLUSIONS Our findings suggest that lysozyme 1 may represent a potential target for the diagnosis of obesity-induced inflammation and the treatment of obesity-induced heart disease.
Collapse
Affiliation(s)
- Lai Zhang
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, China (L.Z.)
| | - Huian Han
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Andi Xu
- Department of Pathology, Nanjing Drum Tower Hospital, China (A.X.)
| | - Adwait Sathe
- Eugene McDermott Center for Human Growth and Development (A.S., C.X.), University of Texas Southwestern Medical Center, Dallas
| | - Siying Fu
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Jiaqi Zhao
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Wenhan Cai
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Yaqing Yang
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Jinting Liu
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Hui Bai
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Jingjing Ben
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Xudong Zhu
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Xiaoyu Li
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Qing Yang
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Zidun Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, China (Z.W.)
| | - Yayun Gu
- State Key Laboratory of Reproductive Medicine (Y.G.), Nanjing Medical University, Jiangsu, China
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development (A.S., C.X.), University of Texas Southwestern Medical Center, Dallas
- Department of Bioinformatics (C.X.), University of Texas Southwestern Medical Center, Dallas
- Department of Population and Data Sciences (C.X.), University of Texas Southwestern Medical Center, Dallas
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité - Universitätsmedizin Berlin, Germany (G.G.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany (G.G.S.)
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (G.G.S.)
| | - Steven Yan Cheng
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Hanwen Zhang
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| | - Qi Chen
- Department of Pathophysiology (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases (L.Z., H.H., S.F., J.Z., W.C., Y.Y., J.L., H.B., J.B., X.Z., X.L., Q.Y., S.Y.C., H.Z., Q.C.), Nanjing Medical University, Jiangsu, China
| |
Collapse
|
32
|
Cui X, Anatolevna ST, Wang Y. Deciphering Blood Flow Restriction Training to Aid Lipid Lowering in Obese College Students through Untargeted Metabolomics. Metabolites 2024; 14:433. [PMID: 39195529 DOI: 10.3390/metabo14080433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
(1) Objective: The aim of this study was to observe the lipid-lowering effects of blood flow restriction training (BFR) combined with moderate-intensity continuous training (MICT) in obese college students by observing lipid-lowering hormones and untargeted metabolomics. (2) Methods: In this study, 14 obese college students were convened into three groups-MICT, MICT+BFR, and high-intensity interval training (HIIT)-for a crossover experiment. Blood was drawn before and after exercise for the analysis of lipolytic agents and untargeted metabolomics. The study used a paired t-test and ANOVA for statistical analyses. (3) Results: The lipolytic agent results showed that MICT+BFR was superior to the other two groups in terms of two agents (p = 0.000 and p = 0.003), namely, GH and IL-6 (difference between before and after testing: 10,986.51 ± 5601.84 and 2.42 ± 2.49, respectively), and HIIT was superior to the other two groups in terms of one agent (p = 0.000), i.e., EPI (22.81 ± 16.12). No advantage was observed for MICT. The metabolomics results showed that, compared to MICT, MICT+BFR was associated with the upregulated expression of xanthine, succinate, lactate, N-lactoylphenylalanine, citrate, ureido acid, and myristic acid after exercise, with the possibility of the involvement of the citric acid cycle, alanine, aspartic acid, glutamate metabolism, butyric acid metabolism, and the histidylate metabolism pathway. (4) Conclusions: The superior lipid-lowering effect of MICT+BFR over MICT in a group of obese college students may be due to the stronger activation of GH and IL-6 agents, with the citric acid cycle and alanine, aspartate, and glutamate metabolic pathways being associated with this type of exercise.
Collapse
Affiliation(s)
- Xianyou Cui
- Zhejiang Guang Sha Vocational and Technical University of Construction, No.1 Guangfu East Street, Dongyang 322103, China
- Moscow State Academy of Physical Education, Liubertsy District, Malakhovka, Shosseynaya St. 33, 140030 Moscow, Russia
| | - Sidorenko Tatiana Anatolevna
- Moscow State Academy of Physical Education, Liubertsy District, Malakhovka, Shosseynaya St. 33, 140030 Moscow, Russia
- Ryazan State University Named for S. A. Yesenin, St. Svobody, 46, 390000 Ryazan, Russia
| | - Yu Wang
- Moscow State University of Sport and Tourism, Kirovogradskaya Street, 21, Building 1 (South Campus), 117519 Moscow, Russia
| |
Collapse
|
33
|
Meddeb M, Koleini N, Binek A, Keykhaei M, Darehgazani R, Kwon S, Aboaf C, Margulies KB, Bedi KC, Lehar M, Sharma K, Hahn VS, Van Eyk JE, Drachenberg CI, Kass DA. Myocardial ultrastructure of human heart failure with preserved ejection fraction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:907-914. [PMID: 39196036 PMCID: PMC11498130 DOI: 10.1038/s44161-024-00516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/26/2024] [Indexed: 08/29/2024]
Abstract
Over half of patients with heart failure have a preserved ejection fraction (>50%, called HFpEF), a syndrome with substantial morbidity/mortality and few effective therapies1. Its dominant comorbidity is now obesity, which worsens disease and prognosis1-3. Myocardial data from patients with morbid obesity and HFpEF show depressed myocyte calcium-stimulated tension4 and disrupted gene expression of mitochondrial and lipid metabolic pathways5,6, abnormalities shared by human HF with a reduced EF but less so in HFpEF without severe obesity. The impact of severe obesity on human HFpEF myocardial ultrastructure remains unexplored. Here we assessed the myocardial ultrastructure in septal biopsies from patients with HFpEF using transmission electron microscopy. We observed sarcomere disruption and sarcolysis, mitochondrial swelling with cristae separation and dissolution and lipid droplet accumulation that was more prominent in the most obese patients with HFpEF and not dependent on comorbid diabetes. Myocardial proteomics revealed associated reduction in fatty acid uptake, processing and oxidation and mitochondrial respiration proteins, particularly in very obese patients with HFpEF.
Collapse
Grants
- R01 HL149891 NHLBI NIH HHS
- HL166565-01 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL007227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL149891 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL007227 NHLBI NIH HHS
- R35 HL166565 NHLBI NIH HHS
- 20SRG35490443 American Heart Association (American Heart Association, Inc.)
- 23POST1026402 American Heart Association (American Heart Association, Inc.)
- R35 HL135827 NHLBI NIH HHS
- HL155346 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL166565 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL135827 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL155346 NHLBI NIH HHS
- K23HL166770 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K23 HL166770 NHLBI NIH HHS
- 16SFRN28620000 American Heart Association (American Heart Association, Inc.)
- L30 HL138884 NHLBI NIH HHS
Collapse
Affiliation(s)
- Mariam Meddeb
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Navid Koleini
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohammad Keykhaei
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Reyhane Darehgazani
- Department of Biological Sciences, University of Maryland, Baltimore, MD, USA
| | - Seoyoung Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Celia Aboaf
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ken C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamed Lehar
- Department of Anesthesia, Johns Hopkins University, Baltimore, MD, USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
34
|
Blackwood SJ, Tischer D, van de Ven MPF, Pontén M, Edman S, Horwath O, Apró W, Röja J, Ekblom MM, Moberg M, Katz A. Elevated heart rate and decreased muscle endothelial nitric oxide synthase in early development of insulin resistance. Am J Physiol Endocrinol Metab 2024; 327:E172-E182. [PMID: 38836779 DOI: 10.1152/ajpendo.00148.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Insulin resistance (IR) is a risk factor for the development of several major metabolic diseases. Muscle fiber composition is established early in life and is associated with insulin sensitivity. Hence, muscle fiber composition was used to identify early defects in the development of IR in healthy young individuals in the absence of clinical manifestations. Biopsies were obtained from the thigh muscle, followed by an intravenous glucose tolerance test. Indices of insulin action were calculated and cardiovascular measurements, analyses of blood and muscle were performed. Whole body insulin sensitivity (SIgalvin) was positively related to expression of type I muscle fibers (r = 0.49; P < 0.001) and negatively related to resting heart rate (HR, r = -0.39; P < 0.001), which was also negatively related to expression of type I muscle fibers (r = -0.41; P < 0.001). Muscle protein expression of endothelial nitric oxide synthase (eNOS), whose activation results in vasodilation, was measured in two subsets of subjects expressing a high percentage of type I fibers (59 ± 6%; HR = 57 ± 9 beats/min; SIgalvin = 1.8 ± 0.7 units) or low percentage of type I fibers (30 ± 6%; HR = 71 ± 11; SIgalvin = 0.8 ± 0.3 units; P < 0.001 for all variables vs. first group). eNOS expression was 1) higher in subjects with high type I expression; 2) almost twofold higher in pools of type I versus II fibers; 3) only detected in capillaries surrounding muscle fibers; and 4) linearly associated with SIgalvin. These data demonstrate that an altered function of the autonomic nervous system and a compromised capacity for vasodilation in the microvasculature occur early in the development of IR.NEW & NOTEWORTHY Insulin resistance (IR) is a risk factor for the development of several metabolic diseases. In healthy young individuals, an elevated heart rate (HR) correlates with low insulin sensitivity and high expression of type II skeletal muscle fibers, which express low levels of endothelial nitric oxide synthase (eNOS) and, hence, a limited capacity to induce vasodilation in response to insulin. Early targeting of the autonomic nervous system and microvasculature may attenuate development of diseases stemming from insulin resistance.
Collapse
Affiliation(s)
- Sarah J Blackwood
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Dominik Tischer
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Myrthe P F van de Ven
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Marjan Pontén
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Sebastian Edman
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Oscar Horwath
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - William Apró
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Julia Röja
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Maria M Ekblom
- Department of Physical Activity and Health, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Moberg
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Abram Katz
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| |
Collapse
|
35
|
Gonzalez K, Merlin AC, Roye E, Ju B, Lee Y, Chicco AJ, Chung E. Voluntary Wheel Running Reduces Cardiometabolic Risks in Female Offspring Exposed to Lifelong High-Fat, High-Sucrose Diet. Med Sci Sports Exerc 2024; 56:1378-1389. [PMID: 38595204 PMCID: PMC11250925 DOI: 10.1249/mss.0000000000003443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
PURPOSE Maternal and postnatal overnutrition has been linked to an increased risk of cardiometabolic diseases in offspring. This study investigated the impact of adult-onset voluntary wheel running to counteract cardiometabolic risks in female offspring exposed to a life-long high-fat, high-sucrose (HFHS) diet. METHODS Dams were fed either an HFHS or a low-fat, low-sucrose (LFLS) diet starting from 8 wk before pregnancy and continuing throughout gestation and lactation. Offspring followed their mothers' diets. At 15 wk of age, they were divided into sedentary (Sed) or voluntary wheel running (Ex) groups, resulting in four groups: LFLS/Sed ( n = 10), LFLS/Ex ( n = 5), HFHS/Sed ( n = 6), HFHS/Ex ( n = 5). Cardiac function was assessed at 25 wk, with tissue collection at 26 wk for mitochondrial respiratory function and protein analysis. Data were analyzed using two-way ANOVA. RESULTS Although maternal HFHS diet did not affect the offspring's body weight at weaning, continuous HFHS feeding postweaning resulted in increased body weight and adiposity, irrespective of the exercise regimen. HFHS/Sed offspring showed increased left ventricular wall thickness and elevated expression of enzymes involved in fatty acid transport (CD36, FABP3), lipogenesis (DGAT), glucose transport (GLUT4), oxidative stress (protein carbonyls, nitrotyrosine), and early senescence markers (p16, p21). Their cardiac mitochondria displayed lower oxidative phosphorylation (OXPHOS) efficiency and reduced expression of OXPHOS complexes and fatty acid metabolism enzymes (ACSL5, CPT1B). However, HFHS/Ex offspring mitigated these effects, aligning more with LFLS/Sed offspring. CONCLUSIONS Adult-onset voluntary wheel running effectively counteracts the detrimental cardiac effects of a lifelong HFHS diet, improving mitochondrial efficiency, reducing oxidative stress, and preventing early senescence. This underscores the significant role of physical activity in mitigating diet-induced cardiometabolic risks.
Collapse
Affiliation(s)
- Kassandra Gonzalez
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| | - Andrea Chiñas Merlin
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
- Biomedical Engineering, Tecnologico de Monterrey, Campus Monterrey, MEXICO
| | - Erin Roye
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| | - Beomsoo Ju
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, University of West Florida, Pensacola, FL
| | - Youngil Lee
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, University of West Florida, Pensacola, FL
| | - Adam J. Chicco
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Eunhee Chung
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| |
Collapse
|
36
|
Li FJ, Abudureyimu M, Zhang ZH, Tao J, Ceylan AF, Lin J, Yu W, Reiter RJ, Ashrafizadeh M, Guo J, Ren J. Inhibition of ER stress using tauroursodeoxycholic acid rescues obesity-evoked cardiac remodeling and contractile anomalies through regulation of ferroptosis. Chem Biol Interact 2024; 398:111104. [PMID: 38906502 DOI: 10.1016/j.cbi.2024.111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
Interrupted ER homeostasis contributes to the etiology of obesity cardiomyopathy although it remains elusive how ER stress evokes cardiac anomalies in obesity. Our study evaluated the impact of ER stress inhibition on cardiac anomalies in obesity. Lean and ob/ob obese mice received chemical ER chaperone tauroursodeoxycholic acid (TUDCA, 50 mg/kg/d, p.o.) for 35 days prior to evaluation of glucose sensitivity, echocardiographic, myocardial geometric, cardiomyocyte mechanical and subcellular Ca2+ property, mitochondrial integrity, oxidative stress, apoptosis, and ferroptosis. Intracellular Ca2+ governing domains including sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) were monitored by45Ca2+uptake and immunoblotting. Our results noted that TUDCA alleviated myocardial remodeling (fibrosis, hypertrophy, enlarged LVESD), echocardiographic anomalies (compromised fractional shortening and ejection fraction), cardiomyocyte contractile dysfunction (amplitude and velocity of cell shortening, relengthening time) and intracellular Ca2+ anomalies (compromised subcellular Ca2+ release, clearance and SERCA function), mitochondrial damage (collapsed membrane potential, downregulated mitochondrial elements and ultrastructural alteration), ER stress (GRP78, eIF2α and ATF4), oxidative stress, apoptosis and ferroptosis [downregulated SLC7A11, GPx4 and upregulated transferrin receptor (TFRC)] without affecting global glucose sensitivity and serum Fe2+ in obese mice. Obesity-evoked change in HSP90, phospholamban and Na+-Ca2+ exchanger was spared by the chemical ER chaperone. Moreover, in vitro results noted that TUDCA, PERK inhibitor GSK2606414, TFRC neutralizing antibody and ferroptosis inhibitor LIP1 mitigated palmitic acid-elicited changes in lipid peroxidation and mechanical function. Our findings favored a role for ferroptosis in obesity cardiomyopathy downstream of ER stress.
Collapse
Affiliation(s)
- Feng-Juan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China
| | - Zeng-Hui Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Asli F Ceylan
- Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Medical Pharmacology, Bilkent, Ankara, Turkey
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Wei Yu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Milad Ashrafizadeh
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jun Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
37
|
Zhang J, Wang J, Xu D, Gui Y, Bai F, Huo Y, Cao L, Gui Y. Promoting Glutathione Synthesis: A Possibility for Treating Cardiomyopathy Induced by a Maternal Western Diet. Nutrients 2024; 16:2520. [PMID: 39125400 PMCID: PMC11313981 DOI: 10.3390/nu16152520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The adverse effects of a Western diet on obesity and diabetes among reproductive-aged women pose a significant threat to the cardiovascular health of their offspring. Given the crucial role of glutathione metabolism and glutathione-related antioxidant defense systems in cardiovascular diseases through scavenging ROS and maintaining redox homeostasis, further exploration of their specific influence is imperative to develop therapeutic strategies for cardiomyopathy induced by a maternal Western diet. METHODS We developed a prenatal maternal Western diet exposure model in C57/B6 mice to investigate cardiac morphology and function through histological analysis and echocardiography. RNA sequencing and analysis were utilized to elucidate the mechanisms underlying the impact of a maternal Western diet and N-acetylcysteine treatment on cardiomyopathy. Additionally, ELISAs, transmission electron microscopy, and flow cytometry were employed to assess the antioxidant defense system and mitochondrial ROS levels in progenitor cardiomyocytes. RESULTS N-acetylcysteine significantly mitigated cardiomyocyte hypertrophy, myocardial interstitial fibrosis, collagen type I accumulation, and left ventricular remodeling induced by a maternal Western diet, particularly in male offspring. Furthermore, N-acetylcysteine reversed the increase in apoptosis and the increase in the β/α-MyHC ratio in the myocardium of offspring that results from a maternal Western diet. RNA sequencing and GSEA revealed that the beneficial effects of N-acetylcysteine were linked to its ability to modulate oxidative phosphorylation pathways. Additionally, N-acetylcysteine treatment during pregnancy can markedly elevate glutathione levels, augment glutathione peroxidase (GPx) activity, and mitigate the accumulation of mitochondrial ROS caused by a maternal Western diet. CONCLUSIONS N-acetylcysteine mitigated cardiomyopathy induced by a maternal Western diet by bolstering glutathione synthesis and enhancing GPx activity, thereby scavenging mitochondrial ROS and modulating oxidative phosphorylation pathways.
Collapse
Affiliation(s)
- Jialing Zhang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China; (J.Z.)
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
| | - Jiayu Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China; (J.Z.)
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
| | - Da Xu
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Yiting Gui
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Fan Bai
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Yu Huo
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Li Cao
- Ultrasound Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Yonghao Gui
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
38
|
Musale V, Murdoch CE, Banah AK, Hasib A, Hennayake CK, Dong B, Lang CC, Wasserman DH, Kang L. Limiting extracellular matrix expansion in diet-induced obese mice reduces cardiac insulin resistance and prevents myocardial remodelling. Mol Metab 2024; 86:101970. [PMID: 38908792 PMCID: PMC11334943 DOI: 10.1016/j.molmet.2024.101970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/16/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024] Open
Abstract
OBJECTIVE Obesity increases deposition of extracellular matrix (ECM) components of cardiac tissue. Since obesity aggregates with insulin resistance and heart disease, it is imperative to determine whether the increased ECM deposition contributes to this disease cluster. The hypotheses tested in this study were that in cardiac tissue of obese mice i) increased deposition of ECM components (collagens and hyaluronan) contributes to cardiac insulin resistance and that a reduction in these components improves cardiac insulin action and ii) reducing excess collagens and hyaluronan mitigates obesity-associated cardiac dysfunction. METHODS Genetic and pharmacological approaches that manipulated collagen and hyaluronan contents were employed in obese C57BL/6 mice fed a high fat (HF) diet. Cardiac insulin sensitivity was measured by hyperinsulinemic-euglycemic clamp and cardiac function was measured by pressure-volume loop analysis in vivo. RESULTS We demonstrated a tight association between increased ECM deposition with cardiac insulin resistance. Increased collagen deposition by genetic deletion of matrix metalloproteinase 9 (MMP9) exacerbated cardiac insulin resistance and pirfenidone, a clinically available anti-fibrotic medication which inhibits collagen expression, improved cardiac insulin resistance in obese mice. Furthermore, decreased hyaluronan deposition by treatment with PEGylated human recombinant hyaluronidase PH20 (PEGPH20) improved cardiac insulin resistance in obese mice. These relationships corresponded to functional changes in the heart. Both PEGPH20 and pirfenidone treatment in obese mice ameliorated HF diet-induced abnormal myocardial remodelling. CONCLUSION Our results provide important new insights into the role of ECM deposition in the pathogenesis of cardiac insulin resistance and associated dysfunction in obesity of distinct mouse models. These findings support the novel therapeutic potential of targeting early cardiac ECM abnormalities in the prevention and treatment of obesity-related cardiovascular complications.
Collapse
Affiliation(s)
- Vishal Musale
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Colin E. Murdoch
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Ayman K. Banah
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Annie Hasib
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Chandani K. Hennayake
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, Jinan, China
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - David H. Wasserman
- Department of Molecular Physiology and Biophysics, Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN, USA
| | - Li Kang
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
39
|
Gherbesi E, Faggiano A, Sala C, Carugo S, Grassi G, Tadic M, Cuspidi C. Left ventricular systolic dysfunction in obesity: a meta-analysis of speckle tracking echocardiographic studies. J Hypertens 2024; 42:1449-1459. [PMID: 38780168 DOI: 10.1097/hjh.0000000000003761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Obesity is a risk factor for left ventricular hypertrophy (LVH) and diastolic dysfunction. Available evidence on impaired myocardial deformation in obese patients without apparent systolic dysfunction assessed by LV ejection fraction (LVEF) is based on single studies. The aim of the present meta-analysis was to provide a comprehensive and updated information on this issue. METHODS The PubMed, OVID-MEDLINE, and Cochrane library databases were analysed to search English-language articles published from the inception up to 31 December 2023. Studies were identified by using MeSH terms and crossing the following search items: ' myocardial strain', 'left ventricular mechanics', 'longitudinal global strain', 'speckle tracking echocardiography', 'systolic dysfunction', 'left ventricular ejection fraction', and 'obesity'. RESULTS Twenty-four studies including 5792 obese and 5518 nonobese individuals from different clinical settings were considered for the analysis. LV global longitudinal strain (GLS) was significantly impaired in the obese group [standard means difference (SMD): -0.86 ± 0.08; confidence interval (CI) -1.02 to -0.69, P < 0.0001] and this was paralleled by a significant difference in pooled LVEF between obese and controls (SMD -0.27 ± 0.06; CI -0.40 to -0.15, P < 0.0001). Unlike GLS, however, the majority of the selected studies failed to show statistically significant differences in LVEF. Furthermore, in patients with advanced obesity (BMI > 35 kg/m 2 , data from six studies), LV systolic dysfunction was more significantly detected by GLS (SMD -1.24 ± 0.19, CI -1.61/-0.87, P < 0.0001) than by LVEF (SMD -0.54 ± 0.27, CI -1.07 to -0.01, P = 0.046). CONCLUSION The present meta-analysis suggests that GLS may unmask systolic dysfunction often undetected by conventional LVEF in the obese setting; thus, this parameter should be incorporated into routine work-up aimed to identify obesity-mediated subclinical cardiac damage.
Collapse
Affiliation(s)
- Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico
| | - Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico
- Department of Clinical Sciences and Community Health, University of Milano
| | - Carla Sala
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico
- Department of Clinical Sciences and Community Health, University of Milano
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico
- Department of Clinical Sciences and Community Health, University of Milano
| | - Guido Grassi
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Marijana Tadic
- University Heart Center Ulm, University Ulm, Albert-Einstein Allee, Ulm, Germany
| | - Cesare Cuspidi
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
40
|
Han Q, Yu Y, Liu X, Guo Y, Shi J, Xue Y, Li Y. The Role of Endothelial Cell Mitophagy in Age-Related Cardiovascular Diseases. Aging Dis 2024:AD.2024.0788. [PMID: 39122456 DOI: 10.14336/ad.2024.0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Aging is a major risk factor for cardiovascular diseases (CVD), and mitochondrial autophagy impairment is considered a significant physiological change associated with aging. Endothelial cells play a crucial role in maintaining vascular homeostasis and function, participating in various physiological processes such as regulating vascular tone, coagulation, angiogenesis, and inflammatory responses. As aging progresses, mitochondrial autophagy impairment in endothelial cells worsens, leading to the development of numerous cardiovascular diseases. Therefore, regulating mitochondrial autophagy in endothelial cells is vital for preventing and treating age-related cardiovascular diseases. However, there is currently a lack of systematic reviews in this area. To address this gap, we have written this review to provide new research and therapeutic strategies for managing aging and age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghong Guo
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Marina Arroyo M, Ramírez Gallegos I, López-González ÁA, Vicente-Herrero MT, Vallejos D, Sastre-Alzamora T, Ramírez Manent JI. Usefulness of the ECORE-BF Scale to Determine Atherogenic Risk in 386,924 Spanish Workers. Nutrients 2024; 16:2434. [PMID: 39125315 PMCID: PMC11314428 DOI: 10.3390/nu16152434] [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: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are the leading cause of death worldwide. Obesity and atherosclerosis are considered risk factors for this pathology. There are multiple methods to evaluate obesity, in the same way as there are different formulas to determine atherogenic risk. Since both pathologies are closely related, the objective of our work was to evaluate whether the ECORE-BF scale is capable of predicting atherogenic risk. METHODS Observational, descriptive, and cross-sectional study in which 386,924 workers from several autonomous communities in Spain participated. The association between the ECORE-BF scale and five atherogenic risk indices was evaluated. The relationship between variables was assessed using the chi-square test and Student's t test in independent samples. Multivariate analysis was performed with the multinomial logistic regression test, calculating the odds ratio and 95% confidence intervals, with the Hosmer-Lemeshow goodness-of-fit test. ROC curves established the cut-off points for moderate and high vascular age and determined the Youden index. RESULTS The mean values of the ECORE-BF scale were higher in individuals with atherogenic dyslipidemia and the lipid triad, as well as in those with elevated values of the three atherogenic indices studied, with p <0.001 in all cases. As atherogenic risk increased across the five evaluated scales, the prevalence of obesity also significantly increased, with p <0.001 in all cases. In the ROC curve analysis, the AUCs for atherogenic dyslipidemia and the lipid triad were above 0.75, indicating a good association between these scales and the ECORE-BF. Although the Youden indices were not exceedingly high, they were around 0.5. CONCLUSIONS There is a good association between atherogenic risk scales, atherogenic dyslipidemia, and lipid triad, and the ECORE-BF scale. The ECORE-BF scale can be a useful and quick tool to evaluate atherogenic risk in primary care and occupational medicine consultations without the need for blood tests.
Collapse
Affiliation(s)
- Marta Marina Arroyo
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
| | - Ignacio Ramírez Gallegos
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
| | - Ángel Arturo López-González
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
- Faculty of Dentistry, ADEMA University School, 07010 Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària de les Illes Balears (IDISBA), Health Research Institute of the Balearic Islands, 07010 Palma, Balearic Islands, Spain
- Health Service of the Balearic Islands, 07010 Palma, Balearic Islands, Spain
| | - María Teófila Vicente-Herrero
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
| | - Daniela Vallejos
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
| | - Tomás Sastre-Alzamora
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
| | - José Ignacio Ramírez Manent
- Research ADEMA SALUD Group, University Institute for Research in Health Sciences (IUNICS), 07010 Palma, Balearic Islands, Spain; (M.M.A.); (I.R.G.); (M.T.V.-H.); (D.V.); (T.S.-A.); (J.I.R.M.)
- Institut d’Investigació Sanitària de les Illes Balears (IDISBA), Health Research Institute of the Balearic Islands, 07010 Palma, Balearic Islands, Spain
- Health Service of the Balearic Islands, 07010 Palma, Balearic Islands, Spain
- Faculty of Medicine, University of the Balearic Islands, 07010 Palma, Balearic Islands, Spain
| |
Collapse
|
42
|
Qin R, Xiang J, Zou L, Xiang G, Xiang H. Potential Associations Between Vitamin Intake and Leukemia: A Cross-Sectional Study. Nutr Cancer 2024; 76:1001-1007. [PMID: 39049206 DOI: 10.1080/01635581.2024.2383334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
The present study assessed potential associations between vitamin intake and leukemia in a national sample of adults in the United States. A total of 5520 participants were included in this cross-sectional study to investigate the relationship between vitamin intake (including vitamins A, C, D, and E) and leukemia. Results revealed negative associations between vitamin C and E intake and leukemia, whereas associations between vitamin A and D and leukemia were not statistically significant. For vitamin C, compared with the first tertile, the odds ratio (OR) and corresponding 95% confidential interval (CI) was 0.90 (0.75-0.95) for the second tertile and 0.82 (0.61-0.90) for the third tertile (p < 0.01). For vitamin E, compared with the first tertile, the OR and 95% CI was 0.92 (0.80-0.96) for the second tertile and 0.86 (0.71-0.92) for the third tertile (p < 0.01). Furthermore, the inverse relationship between intake of vitamins C and E and leukemia were more evident for individuals ≥60 years of age and those with a body mass index >30 kg/m2. Results of this study provide evidence suggesting that intake of vitamin C and E intake may decrease the prevalence of leukemia; however, further large-scale prospective cohort studies are needed to verify these findings.
Collapse
Affiliation(s)
- Rui Qin
- Department of Hematology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, P. R. China
| | - Jinping Xiang
- Department of Hematology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, P. R. China
| | - Luping Zou
- Department of Hematology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, P. R. China
| | - Guoqiang Xiang
- Department of Hematology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, P. R. China
| | - Hang Xiang
- Department of Hematology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, P. R. China
| |
Collapse
|
43
|
Wang HB, Xiao X, Dai W, Cui Y, Li WM, Peng R, Hu L, Wang ST. Dispel some mist on circulating biopterins: measurement, physiological interval and pathophysiological implication. Metabolomics 2024; 20:74. [PMID: 38980520 DOI: 10.1007/s11306-024-02137-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND AND AIMS Biopterins, including tetrahydrobiopterin (BH4), dihydrobiopterin (BH2), and biopterin (B), were crucial enzyme cofactors in vivo. Despite their recognized clinical significance, there remain notable research gaps and controversies surrounding experimental outcomes. This study aims to clarify the biopterins-related issues, including analytical art, physiological intervals, and pathophysiological implications. MATERIALS AND METHODS A novel LC-MS/MS method was developed to comprehensively profile biopterins in plasma, utilizing chemical derivatization and cold-induced phase separation. Subsequently, apparently healthy individuals were enrolled to investigate the physiological ranges. And the relationships between biopterins and biochemical indicators were analyzed to explore the pathophysiological implications. RESULTS The developed method was validated as reliable for detecting biopterins across the entire physiological range. Timely anti-oxidation was found to be essential for accurate assessment of biopterins. The observed overall mean ± SDs levels were 3.51 ± 0.94, 1.54 ± 0.48, 2.45 ± 0.84 and 5.05 ± 1.14 ng/mL for BH4, BH2, BH4/BH2 and total biopterins. The status of biopterins showed interesting correlations with age, gender, hyperuricemia and overweight. CONCLUSION In conjunction with proper anti-oxidation, the newly developed method enables accurate determination of biopterins status in plasma. The observed physiological intervals and pathophysiological implications provide fundamental yet inspiring support for further clinical researches.
Collapse
Affiliation(s)
- Hai-Bo Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen Dai
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wan-Man Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Medical Laboratory, Liuzhou Municipal Liutie Central Hospital, Liuzhou, 545000, China
| | - Rui Peng
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liu Hu
- Physical Examination Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shao-Ting Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
44
|
Yaghoobi A, Rezaee M, Hedayati N, Keshavarzmotamed A, Khalilzad MA, Russel R, Asemi Z, Rajabi Moghadam H, Mafi A. Insight into the cardioprotective effects of melatonin: shining a spotlight on intercellular Sirt signaling communication. Mol Cell Biochem 2024:10.1007/s11010-024-05002-3. [PMID: 38980593 DOI: 10.1007/s11010-024-05002-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/25/2024] [Indexed: 07/10/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and illness worldwide. While there have been advancements in the treatment of CVDs using medication and medical procedures, these conventional methods have limited effectiveness in halting the progression of heart diseases to complete heart failure. However, in recent years, the hormone melatonin has shown promise as a protective agent for the heart. Melatonin, which is secreted by the pineal gland and regulates our sleep-wake cycle, plays a role in various biological processes including oxidative stress, mitochondrial function, and cell death. The Sirtuin (Sirt) family of proteins has gained attention for their involvement in many cellular functions related to heart health. It has been well established that melatonin activates the Sirt signaling pathways, leading to several beneficial effects on the heart. These include preserving mitochondrial function, reducing oxidative stress, decreasing inflammation, preventing cell death, and regulating autophagy in cardiac cells. Therefore, melatonin could play crucial roles in ameliorating various cardiovascular pathologies, such as sepsis, drug toxicity-induced myocardial injury, myocardial ischemia-reperfusion injury, hypertension, heart failure, and diabetic cardiomyopathy. These effects may be partly attributed to the modulation of different Sirt family members by melatonin. This review summarizes the existing body of literature highlighting the cardioprotective effects of melatonin, specifically the ones including modulation of Sirt signaling pathways. Also, we discuss the potential use of melatonin-Sirt interactions as a forthcoming therapeutic target for managing and preventing CVDs.
Collapse
Affiliation(s)
- Alireza Yaghoobi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | | | | | - Reitel Russel
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hasan Rajabi Moghadam
- Department of Cardiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
45
|
Miao Y, Wang Y, Wang Y, Yan P, Chen Z, Wan Q. The Association Between Triglyceride-Glucose Index and Its Combination with Obesity Indicators and Lower Extremity Peripheral Artery Disease in Patients with Type 2 Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2024; 17:2607-2617. [PMID: 38953012 PMCID: PMC11216433 DOI: 10.2147/dmso.s469692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
Background Lower extremity peripheral artery disease (LEAD) is a significant chronic complication of type 2 diabetes mellitus (T2DM) that significantly contributes to disability and mortality. The subtle presentation of LEAD symptoms often leads to underrecognition and misdiagnosis. Therefore, identifying simple and effective evaluation indicators is essential for the early detection and management of LEAD. Insulin resistance is closely associated with diabetes and its complications. However, the specific relationship between insulin resistance-measured by the triglyceride-glucose (TyG) index-and obesity indicators in relation to LEAD remains unclear. Objective This study aims to investigate the association between the TyG index and its combination with obesity indicators in participants with T2DM and LEAD. Methods We performed a univariate analysis on 3176 T2DM patients to identify risk factors for LEAD. Patients were then divided into quartiles based on the TyG index combined with various obesity indicators. The chi-square test was used to compare the prevalence of LEAD across these groups. Logistic regression analysis was conducted to examine the association between the TyG index, in combination with different obesity indicators, and the occurrence of LEAD. Finally, we assessed the predictive ability of the TyG index combined with obesity indicators for LEAD by comparing the area under the ROC curve (AUC). Results The study included 3176 T2DM patients (1691 males and 1485 females) with a mean age of 56.16±10.60 years. Among them, 106 individuals had LEAD. The prevalence of LEAD varied significantly across quartiles of the TyG index, TyG-WC, and TyG-WHR (Q4 > Q3 > Q2 > Q1; P < 0.05). Multiple logistic regression analysis showed that the TyG index, TyG-WC, and TyG-WHR were positively associated with the risk of LEAD in T2DM patients. ROC curve analysis identified the best cutoff values for predicting LEAD: 9.8059 for the TyG index (sensitivity: 49.1%, specificity: 67.9%, AUC: 0.583), 808.8397 for TyG-WC (sensitivity: 70.8%, specificity: 47.8%, AUC: 0.603), and 8.8543 for TyG-WHR (sensitivity: 75.5%, specificity: 44.6%, AUC: 0.607). Conclusion In T2DM patients, the TyG index, TyG-WHR, and TyG-WC are positively associated with the occurrence of LEAD. TyG-WHR and TyG-WC exhibit a stronger correlation with LEAD compared to the TyG index alone, indicating their superior diagnostic value.
Collapse
Affiliation(s)
- Ying Miao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Yu Wang
- Department of Cardiology, Luzhou People’s Hospital, Luzhou, People’s Republic of China
| | - Yi Wang
- Department of Pharmacy, Luzhou People’s Hospital, Luzhou, People’s Republic of China
| | - Pijun Yan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhuang Chen
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qin Wan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
46
|
Fuentevilla-Alvarez G, Soto ME, Robles-Herrera GJ, Vargas-Alarcón G, Sámano R, Meza-Toledo SE, Huesca-Gómez C, Gamboa R. Analysis of Circulating miRNA Expression Profiles in Type 2 Diabetes Patients with Diabetic Foot Complications. Int J Mol Sci 2024; 25:7078. [PMID: 39000190 PMCID: PMC11241130 DOI: 10.3390/ijms25137078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with various complications, including diabetic foot, which can lead to significant morbidity and mortality. Non-healing foot ulcers in diabetic patients are a major risk factor for infections and amputations. Despite conventional treatments, which have limited efficacy, there is a need for more effective therapies. MicroRNAs (miRs) are small non-coding RNAs that play a role in gene expression and have been implicated in diabetic wound healing. miR expression was analyzed through RT-qPCR in 41 diabetic foot Mexican patients and 50 controls. Diabetic foot patients showed significant increases in plasma levels of miR-17-5p (p = 0.001), miR-191-5p (p = 0.001), let-7e-5p (p = 0.001), and miR-33a-5p (p = 0.005) when compared to controls. Elevated levels of miR-17, miR-191, and miR-121 correlated with higher glucose levels in patients with diabetic foot ulcers (r = 0.30, p = 0.004; r = 0.25, p = 0.01; and r = 0.21, p = 0.05, respectively). Levels of miR-17 showed the highest diagnostic potential (AUC 0.903, p = 0.0001). These findings underscore the possible role of these miRs in developing diabetes complications. Our study suggests that high miR-17, miR-191, and miR-121 expression is strongly associated with higher glucose levels and the development of diabetic foot ulcers.
Collapse
Affiliation(s)
- Giovanny Fuentevilla-Alvarez
- Endocrinology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico;
| | - María Elena Soto
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (M.E.S.); (G.V.-A.)
- Cardiovascular Line in American British Cowdary (ABC) Medical Center, Sur 136 No. 116 Col. Las Américas, Mexico City 01120, Mexico
| | - Gustavo Jaziel Robles-Herrera
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| | - Gilberto Vargas-Alarcón
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (M.E.S.); (G.V.-A.)
| | - Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico;
| | - Sergio Enrique Meza-Toledo
- Biochemistry Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico;
| | - Claudia Huesca-Gómez
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| | - Ricardo Gamboa
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| |
Collapse
|
47
|
Qu H, Liu X, Zhu J, He N, He Q, Zhang L, Wang Y, Gong X, Xiong X, Liu J, Wang C, Yang G, Yang Q, Luo G, Zhu Z, Zheng Y, Zheng H. Mitochondrial glycerol 3-phosphate dehydrogenase deficiency exacerbates lipotoxic cardiomyopathy. iScience 2024; 27:109796. [PMID: 38832016 PMCID: PMC11145339 DOI: 10.1016/j.isci.2024.109796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 03/21/2024] [Accepted: 04/18/2024] [Indexed: 06/05/2024] Open
Abstract
Metabolic diseases such as obesity and diabetes induce lipotoxic cardiomyopathy, which is characterized by myocardial lipid accumulation, dysfunction, hypertrophy, fibrosis and mitochondrial dysfunction. Here, we identify that mitochondrial glycerol 3-phosphate dehydrogenase (mGPDH) is a pivotal regulator of cardiac fatty acid metabolism and function in the setting of lipotoxic cardiomyopathy. Cardiomyocyte-specific deletion of mGPDH promotes high-fat diet induced cardiac dysfunction, pathological hypertrophy, myocardial fibrosis, and lipid accumulation. Mechanically, mGPDH deficiency inhibits the expression of desuccinylase SIRT5, and in turn, the hypersuccinylates majority of enzymes in the fatty acid oxidation (FAO) cycle and promotes the degradation of these enzymes. Moreover, manipulating SIRT5 abolishes the effects of mGPDH ablation or overexpression on cardiac function. Finally, restoration of mGPDH improves lipid accumulation and cardiomyopathy in both diet-induced and genetic obese mouse models. Thus, our study indicates that targeting mGPDH could be a promising strategy for lipotoxic cardiomyopathy in the context of obesity and diabetes.
Collapse
Affiliation(s)
- Hua Qu
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiufei Liu
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jiaran Zhu
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Niexia He
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qingshan He
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Linlin Zhang
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yuren Wang
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaoli Gong
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xin Xiong
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jinbo Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingwu Yang
- Department of Neurology, the Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Gang Luo
- Department of Orthopedics, the Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, the Third Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yi Zheng
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
48
|
Chen W, Rams D, Zając M, Albalawi R, Atkinson AJ, Aminu AJ, Mazur M, Hołda MK, Walocha J, Gil K, Kuniewicz M, Dobrzynski H. Morphology of human sinoatrial node and its surrounding right atrial muscle in the global obesity pandemic-does fat matter? Front Med (Lausanne) 2024; 11:1415065. [PMID: 38966523 PMCID: PMC11223615 DOI: 10.3389/fmed.2024.1415065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction The sinus node (SN) is the main pacemaker site of the heart, located in the upper right atrium at the junction of the superior vena cava and right atrium. The precise morphology of the SN in the human heart remains relatively unclear especially the SN microscopical anatomy in the hearts of aged and obese individuals. In this study, the histology of the SN with surrounding right atrial (RA) muscle was analyzed from young non-obese, aged non-obese, aged obese and young obese individuals. The impacts of aging and obesity on fibrosis, apoptosis and cellular hypertrophy were investigated in the SN and RA. Moreover, the impact of obesity on P wave morphology in ECG was also analyzed to determine the speed and conduction of the impulse generated by the SN. Methods Human SN/RA specimens were dissected from 23 post-mortem hearts (preserved in 4% formaldehyde solution), under Polish local ethical rules. The SN/RA tissue blocks were embedded in paraffin and histologically stained with Masson's Trichrome. High and low-magnification images were taken, and analysis was done for appropriate statistical tests on Prism (GraphPad, USA). 12-lead ECGs from 14 patients under Polish local ethical rules were obtained. The P wave morphologies from lead II, lead III and lead aVF were analyzed. Results Compared to the surrounding RA, the SN in all four groups has significantly more connective tissue (P ≤ 0.05) (young non-obese individuals, aged non-obese individuals, aged obese individuals and young obese individuals) and significantly smaller nodal cells (P ≤ 0.05) (young non-obese individuals, aged non-obese individuals, aged obese individuals, young obese individuals). In aging, overall, there was a significant increase in fibrosis, apoptosis, and cellular hypertrophy in the SN (P ≤ 0.05) and RA (P ≤ 0.05). Obesity did not further exacerbate fibrosis but caused a further increase in cellular hypertrophy (SN P ≤ 0.05, RA P ≤ 0.05), especially in young obese individuals. However, there was more infiltrating fat within the SN and RA bundles in obesity. Compared to the young non-obese individuals, the young obese individuals showed decreased P wave amplitude and P wave slope in aVF lead. Discussion Aging and obesity are two risk factors for extensive fibrosis and cellular hypertrophy in SN and RA. Obesity exacerbates the morphological alterations, especially hypertrophy of nodal and atrial myocytes. These morphological alterations might lead to functional alterations and eventually cause cardiovascular diseases, such as SN dysfunction, atrial fibrillation, bradycardia, and heart failure.
Collapse
Affiliation(s)
- Weixuan Chen
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Daniel Rams
- Department of Anatomy, Jagiellonian University, Kraków, Poland
| | - Maciej Zając
- Department of Anatomy, Jagiellonian University, Kraków, Poland
| | - Raghad Albalawi
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew J. Atkinson
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Abimbola J. Aminu
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Mateusz K. Hołda
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- Heart Embryology and Anatomy Research Team, Department of Anatomy, Jagiellonian University Medical College, Kraków, Poland
| | - Jerzy Walocha
- Department of Anatomy, Jagiellonian University, Kraków, Poland
| | - Krzysztof Gil
- Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland
| | - Marcin Kuniewicz
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- Department of Anatomy, Jagiellonian University, Kraków, Poland
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- Department of Anatomy, Jagiellonian University, Kraków, Poland
| |
Collapse
|
49
|
Mao S, Qian G, Xiao K, Xu H, Zhou H, Guo X. Study on the relationship between body mass index and blood pressure indices in children aged 7-17 during COVID-19. Front Public Health 2024; 12:1409214. [PMID: 38962763 PMCID: PMC11220196 DOI: 10.3389/fpubh.2024.1409214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/27/2024] [Indexed: 07/05/2024] Open
Abstract
Background To explore the relationship between body mass index (BMI), age, sex, and blood pressure (systolic blood pressure, SBP; diastolic blood pressure, DBP) in children during COVID-19, providing reference for the prevention and screening of hypertension in children. Methods This study adopted a large-scale cross-sectional design to investigate the association between BMI and blood pressure in 7-17-year-old students in City N, China, during COVID-19. Thirty-six primary and secondary schools in City N were sampled using a stratified cluster sampling method. A total of 11,433 students aged 7-17 years in City N, China, were selected for blood pressure (Diastolic blood pressure, DBP, Systolic blood pressure, SBP), height, and weight, Resting heart rate (RHR), chest circumference, measurements, and the study was written using the STROBE checklist. Data analysis was conducted using SPSS 26.0, calculating the mean and standard deviation of BMI and blood pressure for male and female students in different age groups. Regression analysis was employed to explore the impact of BMI, age, and sex on SBP and DBP, and predictive models were established. The model fit was evaluated using the model R2. Results The study included 11,287 primary and secondary school students, comprising 5,649 boys and 5,638 girls. It was found that with increasing age, BMI and blood pressure of boys and girls generally increased. There were significant differences in blood pressure levels between boys and girls in different age groups. In regression models, LC, Age, BMI, and chest circumference show significant positive linear relationships with SBP and DBP in adolescents, while RHR exhibits a negative linear relationship with SBP. These factors were individually incorporated into a stratified regression model, significantly enhancing the model's explanatory power. After including factors such as Age, Gender, and BMI, the adjusted R2 value showed a significant improvement, with Age and BMI identified as key predictive factors for SBP and DBP. The robustness and predictive accuracy of the model were further examined through K-fold cross-validation and independent sample validation methods. The validation results indicate that the model has a high accuracy and explanatory power in predicting blood pressure in children of different weight levels, especially among obese children, where the prediction accuracy is highest. Conclusion During COVID-19, age, sex, and BMI significantly influence blood pressure in children aged 7-17 years, and predictive models for SBP and DBP were established. This model helps predict blood pressure in children and reduce the risk of cardiovascular diseases. Confirmation of factors such as sex, age, and BMI provide a basis for personalized health plans for children, especially during large-scale infectious diseases, providing guidance for addressing health challenges and promoting the health and well-being of children.
Collapse
Affiliation(s)
- SuJie Mao
- Graduate Development, Harbin Sport University, Harbin, Heilongjiang, China
| | - GuoPing Qian
- Faculty of Sports Medicine, Gdansk University of Sport, Gdańsk, Poland
| | - KaiWen Xiao
- Discipline Development Office, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Hong Xu
- College of Sports and Health, Sangmyung University, Seoul, Republic of Korea
| | - Hao Zhou
- Teaching Evaluation Center, Nanjing Police University, Nanjing, Jiangsu, China
| | - XiuJin Guo
- Discipline Development Office, Nanjing Sport Institute, Nanjing, Jiangsu, China
| |
Collapse
|
50
|
Omidi F, Nasiri MJ, Sadeghi S. Impact of Obesity on Cardiac Volumes and Left Ventricular Diameter: A Cross-Sectional Study in an Iranian Heart Center. Adv Med 2024; 2024:7038875. [PMID: 38899165 PMCID: PMC11186677 DOI: 10.1155/2024/7038875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/14/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Obesity, a pressing global health issue worldwide, contributes to risk factors such as hypertension and dyslipidemia, creating an unfavorable cardiovascular environment and increasing the likelihood of adverse cardiac events. His study aims to assess the impact of obesity on various cardiovascular parameters. Methods A cross-sectional analysis was conducted at a Heart Center, focusing on adults admitted for suspected heart diseases. The dataset included information on demographics, clinical history, laboratory results, and echocardiography. Descriptive analysis and multiple linear regression were employed using IBM SPSS Statistics version 26. Results The study of 105 individuals with suspected heart diseases revealed prevalent health factors such as hypertension (47.6%) and hyperlipidemia (61%). Body mass index (BMI) averaged 30, indicating a trend toward overweight. Obesity significantly associated with higher systolic blood pressure (SBP, p=0.005) and diastolic blood pressure (DBP, p=0.002), larger cardiac volumes (end-diastolic volume, EDV, p=0.013; end-systolic volume, ESV, p=0.040), and a marginally significant influence on left ventricular end-diastolic diameter (LVEDD, p=0.068). No significant associations were found with left ventricular end-systolic diameter (LVEDS), heart rate (HR), or ejection fraction (EF). Conclusions Our study highlights a significant association between obesity and elevated blood pressure, larger cardiac volumes, and a marginal impact on left ventricular end-diastolic diameter. While caution is needed in inferring causation due to the study's cross-sectional nature, these findings underscore the importance of addressing obesity as a potential risk factor for adverse cardiovascular outcomes. Further investigations are warranted to enhance our understanding of the complex interplay between obesity and cardiovascular health.
Collapse
Affiliation(s)
- Fatemeh Omidi
- Department of Cardiology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheila Sadeghi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|