1
|
Demailly Z, Tamion F, Besnier E, Bekri S, Tebani A. Understanding metabolic remodeling in shock through metabolomics lenses. Mol Cell Endocrinol 2025; 600:112491. [PMID: 39961415 DOI: 10.1016/j.mce.2025.112491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
The management of shock in critical care must transition from a predominantly hemodynamic approach to one that comprehensively addresses the biological intricacies of this complex multisystemic syndrome. A thorough understanding of the metabolic mechanisms involved in shock is pivotal for precise patient phenotyping and accurate risk stratification. Metabolomics, an emerging "-omics" approach, offers a powerful tool for unraveling the molecular underpinnings of shock. By analyzing the metabolic pathways within the cardiovascular system, metabolomics can elucidate the diverse mechanisms leading to circulatory insufficiency. This approach holds significant promise for identifying clinically actionable diagnostic and prognostic biomarkers, which can enhance individualized patient management and potentially prevent the progression to multi-organ failure. Improved insight into the metabolic alterations in shock may pave the way for novel therapeutic strategies and more targeted treatments, ultimately improving patient outcomes in critical care settings. This work provides a comprehensive overview of metabolomic investigations in shock, focusing on septic shock and the main metabolic pathways involved in cardiac and vascular dysfunction.
Collapse
Affiliation(s)
- Zoé Demailly
- Medical ICU, Rouen University Hospital, Rouen, France; INSERM U1096, University of Rouen, Rouen, France; Department of Anesthesiology, Critical Care and Perioperative Medicine, Rouen University Hospital, Rouen, France.
| | - Fabienne Tamion
- Medical ICU, Rouen University Hospital, Rouen, France; INSERM U1096, University of Rouen, Rouen, France
| | - Emmanuel Besnier
- INSERM U1096, University of Rouen, Rouen, France; Department of Anesthesiology, Critical Care and Perioperative Medicine, Rouen University Hospital, Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, U1245, CHUROUEN, Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Abdellah Tebani
- Normandie Univ, UNIROUEN, U1245, CHUROUEN, Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| |
Collapse
|
2
|
Chen D, Xing ZX, Li SP, Lu T, Wang JX, Wu YX, Pang QF. Preconception maternal hyperoxia exposure causes cardiac insufficiency through induction of mitochondrial toxicity in mice offspring. Reprod Toxicol 2025; 133:108864. [PMID: 39988061 DOI: 10.1016/j.reprotox.2025.108864] [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: 09/18/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Although essential, excessive oxygen is toxic. The adverse effects of maternal hyperoxygenation have recently garnered attention. However, the potential toxicity of maternal hyperoxia exposure before pregnancy and its effects on offspring development remain unclear. This study aims to investigate the cardiac developmental toxicity of maternal pre-pregnancy hyperoxia exposure on the offspring. Our findings reveal that preconception maternal hyperoxia exposure leads to growth retardation, cardiac insufficiency, and remodeling in both male and female offspring. Additionally, maternal pre-pregnancy hyperoxia exposure induces mitochondrial damage characterized by reduced oxidative phosphorylation, inhibited tricarboxylic acid (TCA) cycle, and decreased ATP production in the cardiac tissues of offspring mice. Supplementation of sodium propionate during lactation significantly improves growth retardation, mitigates metabolic remodeling, and partially restores cardiac function in hyperoxia-exposed offspring. In conclusion, our study suggests that maternal hyperoxia exposure before pregnancy leads to cardiac insufficiency in murine offspring. These findings may have important implications for mitigating maternal high oxygen toxicity on offspring development and disease risk, especially the cardiotoxic effects of hyperoxia on offspring development.
Collapse
Affiliation(s)
- Dan Chen
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China.
| | - Zhi-Xuan Xing
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China
| | - Sheng-Peng Li
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China
| | - Tao Lu
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China
| | - Jia-Xin Wang
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China
| | - Ya-Xian Wu
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China
| | - Qing-Feng Pang
- A Department of physiopathology, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu Province 214122, China.
| |
Collapse
|
3
|
Lian H, Ren Q, Liu W, Zhang R, Zou X, Zhang S, Luo Y, Deng W, Wang Q, Qi L, Li Y, Wang W, Zhong L, Zhang P, Guo C, Li L, Li Y, Ba T, Yang C, Huo L, Wang Y, Li C, Hao D, Zhang Y, Xu Y, Wang F, Wang X, Zhang F, Gong S, Yang W, Han X, Ji L. Cardiovascular abnormalities already occurred in newly-diagnosed patients with early-onset type 2 diabetes. Cardiovasc Diabetol 2025; 24:140. [PMID: 40140837 PMCID: PMC11948644 DOI: 10.1186/s12933-025-02665-0] [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: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The prevalence of early-onset type 2 diabetes (EOD) is rapidly increasing. This study intends to screen for early cardiovascular abnormalities in patients newly diagnosed with EOD and evaluate the cardiovascular risk across cluster phenotypes. METHOD A total of 400 patients ≤ 40 years old with newly diagnosed type 2 diabetes were enrolled from the START cohort (the Study of The newly diAgnosed eaRly onset diabeTes). Cluster classification was performed using the K-means method based on age, BMI, HbA1c, HOMA2-β, HOMA2-IR, and GAD antibodies. Echocardiography and carotid ultrasound were performed within 3 months of diabetes diagnosis. Carotid ultrasound abnormalities included intimal thickening and plaque formation, while echocardiography assessed changes in cardiac structure and systolic/diastolic function. Cluster-specific partitioned polygenic scores (pPS) were used to validate our findings from a genetic perspective. RESULT Carotid artery abnormalities were detected in 26.3% of patients, and echocardiography abnormalities were observed in 20.0%. Patients with severe insulin resistant diabetes (SIRD) had the highest incidence of carotid artery abnormality (40.0%). After adjusting for relevant risk factors, fasting C-peptide levels were significantly associated with a 1.247-fold increase in the risk of carotid artery abnormalities. Left atrial enlargement was more prevalent in the SIRD (16.7%) and mild obesity-related diabetes (MOD) (18.5%) classifications. A high proportion of patients with SIRD had abnormal left ventricular geometry (36.1%). Increases in BMI, fasting C-peptide level and HOMA2IR were accompanied by a further increase in left atrial enlargement risk by 1.136-, 1.781- and 1.687-fold respectively. The pPS for lipodystrophy was higher in the EOD group with plaque formation, and showed a significant linear correlation with the ratio of the left atrial anteroposterior diameter to body surface area (LAAP/BSA) (R = 0.344, p < 0.001). CONCLUSION Heart and carotid artery abnormalities are common in patients with early-onset T2DM at the time of diagnosis. Patients with obesity and insulin resistance are at higher risk for cardiovascular abnormalities. Cluster classification based on clinical characteristics enables more accurate identification of patients at increased risk of cardiovascular complications at an early stage.
Collapse
Affiliation(s)
- Hong Lian
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Qian Ren
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Wei Liu
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Rui Zhang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Xiantong Zou
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Simin Zhang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Yingying Luo
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Wei Deng
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing, 100035, People's Republic of China
| | - Qiuping Wang
- Department of Endocrinology, Bejing Fangshan District Liangxiang Hospital, Beijing, 102400, People's Republic of China
| | - Lin Qi
- Department of Endocrinology, Bejing Yanhua Hospital, Beijing, 102500, People's Republic of China
| | - Yufeng Li
- Department of Endocrinology, Beijing Pinggu Hospital, Beijing, 101299, People's Republic of China
| | - Wenbo Wang
- Department of Endocrinology, Beijing Univesity Shougang Hospital, Beijing, 100144, People's Republic of China
| | - Liyong Zhong
- Department of Endocrinology, Capital Medical University Beijing Tiantan Hospital, Beijing, 100050, People's Republic of China
| | - Pengkai Zhang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Chengcheng Guo
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Li Li
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Yating Li
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Tianhao Ba
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Chaochao Yang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Lili Huo
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing, 100035, People's Republic of China
| | - Yan'ai Wang
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing, 100035, People's Republic of China
| | - Chunxia Li
- Department of Endocrinology, Bejing Fangshan District Liangxiang Hospital, Beijing, 102400, People's Republic of China
| | - Dejun Hao
- Department of Endocrinology, Bejing Yanhua Hospital, Beijing, 102500, People's Republic of China
| | - Yajing Zhang
- Department of Endocrinology, Beijing Pinggu Hospital, Beijing, 101299, People's Republic of China
| | - Yan Xu
- Department of Endocrinology, Beijing Univesity Shougang Hospital, Beijing, 100144, People's Republic of China
| | - Fang Wang
- Department of Endocrinology, Capital Medical University Beijing Tiantan Hospital, Beijing, 100050, People's Republic of China
| | - Xiangqing Wang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Fang Zhang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Siqian Gong
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Wenjia Yang
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Xueyao Han
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, People's Republic of China.
| | - Linong Ji
- Department of Endocrinology, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
- Peking University Diabetes Centre, Beijing, 100191, People's Republic of China.
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, People's Republic of China.
| |
Collapse
|
4
|
Dunzhu D, Han G, Shanshan Q, Li S, Yang J, He J, Gou S, Dong G, Jiang C, Hou J. The role of Perilipin 5 in pathological myocardial remodeling. Front Pharmacol 2025; 16:1526494. [PMID: 40166465 PMCID: PMC11955653 DOI: 10.3389/fphar.2025.1526494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Pathological cardiac remodeling (REM), caused by various pathological factors and characterized by changes in cardiac structure and geometry, is strongly associated with heart failure (HF). It damages cardiac tissue, alters energy metabolism, increases oxidative stress, and cause matrix metalloproteinase activation, cardiomyocyte hypertrophy, and interstitial fibrosis, leading to HF. REM determines the outcome of cardiovascular disease. Current treatments have limitations. REM is associated with cardiac energetic remodeling, and modulation of metabolic substrates may slow down the disease. Perilipin 5 (Plin5), positioned as a structural protein located on the surface of lipid droplets (LDs), is abundant in tissues and cells that rely on mitochondrial β-oxidation for energy production. It is the most recently identified member of the perilipin protein (PAT) family, with a notable enrichment in the cardiac muscle. Emerging evidence highlights the critical role of intracellular LD in the regulation of energy metabolism, with metabolic disruptions of LD being directly correlated with the incidence of metabolic disease. As a key barrier to LD, Plin5 is instrumental in controlling the catabolism of LD and regulating the metabolism and transport of fatty acids (FAs). As a protectant against excessive β-oxidation of free fatty acids (FFAs), Plin5 acts to isolate and neutralize overly oxidized fatty acids, thereby shielding the heart from myocardial remodeling instigated by a variety of etiological factors. This protective mechanism helps to ameliorate the progression of persistent and detrimental myocardial remodeling, which can otherwise lead to the development of severe heart failure. This systematic review attempts to delineate the metabolic disorders associated with pathological cardiac remodeling, focusing on the properties and regulatory mechanisms of Plin5. By synthesising current literature, it investigates the pivotal role of Plin5 in modulating the distinctive attributes, initiating factors, and molecular signaling networks underpinning pathological cardiac remodeling.
Collapse
Affiliation(s)
| | - Gao Han
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Qin Shanshan
- School of Medicine, Tibet University, Lhasa, China
| | - Shangshi Li
- The Department of High Mountain Sickness, The General Hospital of Xizang Military Area Command, Xizang, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jian He
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Siyu Gou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Gang Dong
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Chunrong Jiang
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Jun Hou
- The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
5
|
Hørsdal OK, Ellegaard MS, Larsen AM, Guldbrandsen H, Moeslund N, Møller JE, Helgestad OKL, Ravn HB, Wiggers H, Nielsen R, Gopalasingam N, Berg-Hansen K. Lactate infusion improves cardiac function in a porcine model of ischemic cardiogenic shock. Crit Care 2025; 29:113. [PMID: 40083003 PMCID: PMC11907994 DOI: 10.1186/s13054-025-05346-2] [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: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Cardiogenic shock (CS) is associated with high mortality and medical therapies have failed to improve survival. Treatment with lactate is associated with improved cardiac function which may benefit this condition. Comprehensive hemodynamic assessment of lactate administration in CS is lacking, and the mechanisms underlying the cardiovascular effects of lactate in CS have not yet been elucidated. In this study we aimed to study the cardiovascular and cardiometabolic effects of treatment with lactate in experimental ischemic CS. METHODS In a randomized, blinded design, 20 female pigs (60 kg) were studied. Left main coronary artery microsphere injections were used to cause CS, defined as a 30% reduction in CO or mixed venous saturation (SvO2). Subjects were randomized to receive either intravenous exogenous lactate or euvolemic, equimolar saline (control) for 180 min. Positive inotropic control with dobutamine was administered on top of ongoing treatment after 180 min. Extensive hemodynamic measurements were obtained from pulmonary artery and left ventricular (LV) pressure-volume catheterization. Furthermore, endomyocardial biopsies were analyzed for mitochondrial function and arterial, renal vein, and coronary sinus blood samples were collected. The primary endpoint was change in CO during 180 min of treatment. RESULTS Arterial lactate levels increased from 2.4 ± 1.1 to 7.7 ± 1.1 mmol/L (P < 0.001) during lactate infusion. CO increased by 0.7 L/min (P < 0.001) compared with control, due to increased stroke volume (P = 0.003). Notably, heart rate and mean arterial pressure did not differ significantly between treatments. End-systolic elastance (load independent contractility) was enhanced during lactate infusion (P = 0.048), together with LV ejection fraction (P = 0.009) and dP/dt(max) (P = 0.041). Arterial elastance (afterload) did not differ significantly (P = 0.12). This resulted in improved ventriculo-arterial coupling efficiency (P = 0.012). Cardiac mechanical efficiency (P = 0.003), diuresis (P = 0.016), and SvO2 (P = 0.018) were increased during lactate infusion. Myocardial mitochondrial complex I respiration was enhanced during lactate infusion compared with control (P = 0.04). Concomitant administration of dobutamine on top of lactate resulted in further hemodynamic improvements compared with control. CONCLUSIONS Lactate infusion improved cardiac function and myocardial mitochondrial respiration in a porcine model of CS. The hemodynamic effects included increased CO mediated through stroke volume increase. These favorable cardiovascular effects may benefit patients with CS.
Collapse
Affiliation(s)
- Oskar Kjærgaard Hørsdal
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark.
| | - Mark Stoltenberg Ellegaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander Møller Larsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Halvor Guldbrandsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Niels Moeslund
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Heart-, Lung-, and Vascular Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob Eifer Møller
- Heart Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Ole Kristian Lerche Helgestad
- Department of Cardiology, Odense University Hospital, Odense, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
| | - Hanne Berg Ravn
- Department of Anesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henrik Wiggers
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Roni Nielsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Nigopan Gopalasingam
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
- Department of Cardiology, Gødstrup Hospital, Gødstrup, Denmark
| | - Kristoffer Berg-Hansen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus, Denmark
| |
Collapse
|
6
|
Gu S, Kopecky BJ, Peña B, Vagnozzi RJ, Lahm T. Sex-dependent Pathophysiology and Therapeutic Considerations in Right Heart Disease. Can J Cardiol 2025:S0828-282X(25)00178-3. [PMID: 40054579 DOI: 10.1016/j.cjca.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Right ventricular (RV) adaptation to the increased afterload in the setting of pulmonary hypertension (PH) and other cardiac and pulmonary vascular conditions is a major determinant of survival. Although the RV remains understudied and less well understood than the left ventricle, recent advances have been made in understanding the function and biology of the RV in health and in disease, particularly in PH. RV adaptation in PH exhibits significant sexual dimorphisms in pathophysiology, adaptation, and outcomes. Despite a higher incidence of PH, women consistently demonstrate better RV adaptation and survival rates in the setting of increased RV afterload compared with men. Sexual dimorphisms extend to therapy responsiveness, with women benefiting more from certain pulmonary vasodilators and exhibiting superior RV recovery. In this review we discuss the current literature on sexual dimorphisms in RV structure, function, and molecular pathways in health and disease, as well as in RV-specific clinical manifestations, treatments, and outcomes in PH. Sex steroid-mediated effects as well as emerging studies on sex steroid-independent effects are reviewed. In general, sex steroids such as 17β-estradiol and dehydroepiandrosterone exert RV-protective effects. In contrast, testosterone negatively impacts RV structure and function. Emerging evidence highlights the influence of nonhormonal genetic determinants, such as BMPR1A and DMRT2 loci, which are associated with better RV function in women. A better understanding of the interplay between sex hormones, genetic factors, and RV biology is crucial for advancing and developing RV-directed therapies for patients of either sex.
Collapse
Affiliation(s)
- Sue Gu
- Cardio Vascular Pulmonary Research Laboratory, University of Colorado School of Medicine, Aurora, Colorado, USA; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Benjamin J Kopecky
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, College of Engineering, Design and Computing, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ronald J Vagnozzi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA.
| |
Collapse
|
7
|
Crabtree CD, Pan Y, Chandrasekaran P, Wang Y, Scandling D, Bedell T, Stoner J, Decker D, Kackley M, Robinson B, Buga A, Han Y, Volek J, Simonetti OP. A ketogenic-promoting beverage acutely elevates cardiac function and myocardial blood flow compared to placebo in adults: A cardiac MRI investigation. Physiol Rep 2025; 13:e70208. [PMID: 40102665 PMCID: PMC11919629 DOI: 10.14814/phy2.70208] [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: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 03/20/2025] Open
Abstract
Increasing evidence suggests cardiac function improves in healthy and failing hearts alongside circulating ketones (1-4 mM). This study characterized cardiac function and blood flow responses to a ketogenic beverage compared to a volume/calorie matched placebo with repeated imaging over 120 min. This was a two-group, placebo-controlled, acute cardiac imaging study. Adults without cardiac abnormalities underwent baseline cardiac MRI including quantitative myocardial perfusion to measure myocardial blood flow (MBF). Subjects consumed 50 g of a ketogenic-promoting beverage [bis-hexanoyl R-1-3-butanediol (BH-BD)] (BH-BD; n = 11) or a calorically/volume-matched lipid-based placebo (PL; n = 10) with cardiac MRI every 15-30 min. Following 120 min, subjects underwent a final scan including MBF measurement. R-BHB and glucose were measured at every timepoint. 120 min following BH-BD consumption, R-BHB reached 2.1 mM. Cardiac output (CO) was elevated compared to PL (p < 0.05) and increased +31% 120 min after BH-BD ingestion (p < 0.001). CO elevation was due to increased stroke volume (+11%; p = 0.02) and heart rate (+22%; p < 0.001). MBF increased 29% from baseline (p < 0.001). PL did not induce differences in cardiac parameters. 50 g BH-BD ingestion achieves exogenous ketosis and is associated with elevated MBF and CO providing evidence supporting their use as a therapeutic clinical agent.
Collapse
Affiliation(s)
- Christopher D Crabtree
- Department of Radiology, The Ohio State University, Columbus, Ohio, USA
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Yue Pan
- Department of Radiology, The Ohio State University, Columbus, Ohio, USA
| | | | - Yuwei Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Debbie Scandling
- Department of Radiology, The Ohio State University, Columbus, Ohio, USA
| | - Teryn Bedell
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Justen Stoner
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Drew Decker
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Madison Kackley
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Bradley Robinson
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Alex Buga
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Yuchi Han
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jeff Volek
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Orlando P Simonetti
- Department of Radiology, The Ohio State University, Columbus, Ohio, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
8
|
Rodríguez-Rodríguez R, Baena M, Zagmutt S, Paraiso WK, Reguera AC, Fadó R, Casals N. International Union of Basic and Clinical Pharmacology: Fundamental insights and clinical relevance regarding the carnitine palmitoyltransferase family of enzymes. Pharmacol Rev 2025; 77:100051. [PMID: 40106976 DOI: 10.1016/j.pharmr.2025.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The carnitine palmitoyltransferases (CPTs) play a key role in controlling the oxidation of long-chain fatty acids and are potential therapeutic targets for diseases with a strong metabolic component, such as obesity, diabetes, and cancer. Four distinct proteins are CPT1A, CPT1B, CPT1C, and CPT2, differing in tissue expression and catalytic activity. CPT1s are finely regulated by malonyl-CoA, a metabolite whose intracellular levels reflect the cell's nutritional state. Although CPT1C does not exhibit significant catalytic activity, it is capable of modulating the functioning of other neuronal proteins. Structurally, all CPTs share a Y-shaped catalytic tunnel that allows the entry of 2 substrates and accommodation of the acyl group in a hydrophobic pocket. Several molecules targeting these enzymes have been described, some showing potential in normalizing blood glucose levels in diabetic patients, and others that, through a central mechanism, are anorexigenic and enhance energy expenditure. However, given the critical roles that CPTs play in certain tissues, such as the heart, liver, and brain, it is essential to fully understand the differences between the various isoforms. We analyze in detail the structure of these proteins, their cellular and physiological functions, and their potential as therapeutic targets in diseases such as obesity, diabetes, and cancer. We also describe drugs identified to date as having inhibitory or activating capabilities for these proteins. This knowledge will support the design of new drugs specific to each isoform, and the development of nanomedicines that can selectively target particular tissues or cells. SIGNIFICANCE STATEMENT: Carnitine palmitoyltransferase (CPT) proteins, as gatekeepers of fatty acid oxidation, have great potential as pharmacological targets to treat metabolic diseases like obesity, diabetes, and cancer. In recent years, significant progress has been made in understanding the 3-dimensional structure of CPTs and their pathophysiological functions. A deeper understanding of the differences between the various CPT family members will enable the design of selective drugs and therapeutic approaches with fewer side effects.
Collapse
Affiliation(s)
- Rosalía Rodríguez-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Baena
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Sebastián Zagmutt
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - West Kristian Paraiso
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Ana Cristina Reguera
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Heimerl M, Erschow S, Müller-Olling M, Manstein DJ, Decher N, Kauferstein S, Jenewein T, Pich A, Ricke-Hoch M, Hilfiker-Kleiner D. Cardiac dysfunction related to cardiac mRNA and protein traffic impairment due to reduced unconventional motor protein myosin-5b expression. Eur Heart J 2025:ehaf047. [PMID: 39969162 DOI: 10.1093/eurheartj/ehaf047] [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: 06/17/2024] [Revised: 09/09/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND AND AIMS The present study analysed the expression patterns of class-5 myosin motor proteins (MYO5a, b, and c) in the heart with a specific focus on the role of MYO5b. METHODS RNA-sequencing, quantitative real-time polymerase chain reaction, immunohistochemistry, Western blot, immunoprecipitation, and proteomics were performed in mice and human tissues. Functional analyses were performed in mice with a cardiac-specific knockout (KO) of MYO5b (αMHC-Cretg/-; MYO5bflox/flox), wild-type (WT) (MYO5bflox/flox), and αMHC-Cretg/- mice and in isolated adult cardiomyocytes. Next-generation sequencing screened for MYO5B gene variants in a cohort of sudden cardiac death in the young/sudden infant death syndrome patients. RESULTS The expression of MYO5b, but not MYO5a or c, increased during postnatal cardiomyocyte maturation. Myosin-5b was reduced in end-stage failing human hearts and infarcted murine hearts. Heterozygous rare and likely pathogenic missense MYO5B gene variants (n = 6) were identified in three patients of a cohort of young patients (n = 95) who died of sudden cardiac death in the young/sudden infant death syndrome. MYO5b-KO mice revealed impaired electric conductance and metabolism, developed sarcomeric disarrangement, heart failure and death with altered mRNA levels for genes involved in sarcomere organization, fatty acid and glucose metabolism, ion channel sub-units, and Ca2+-homeostasis prior to heart failure. In cardiomyocytes, myosin-5b is associated with mitochondrial and ribosomal proteins. Myosin-5b-associated ribonucleoprotein particles (RNPs) contained mRNAs of sarcomeric, metabolic, cytoskeletal, and ion channel proteins. CONCLUSIONS MYO5b is the major MYO5 gene expressed in postnatal cardiomyocytes where it transports vesicles, proteins, and multi-protein complexes. Among these are mRNA/RNP complexes affecting electric conductance, sarcomere homeostasis, cell metabolism, and cytoskeletal organization. Impairment in MYO5b expression and function promotes cardiac dysfunction, heart failure, and death.
Collapse
Affiliation(s)
- Maren Heimerl
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Sergej Erschow
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Mirco Müller-Olling
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for Medical Research, Carl-Neuberg Str. 1, Hannover 30625, Germany
- Division for Structural Biochemistry, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Niels Decher
- Department of Vegetative Physiology and Center for Mind, Brain and Behavior (CMBB), Medical Faculty, Philipps University Marburg, Deutschausstrasse 1-2, Marburg 35037, Germany
| | - Silke Kauferstein
- Institute of Legal Medicine, Goethe University Frankfurt, University Hospital, Kennedyallee 104, Frankfurt am Main 60598, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK, German Centre for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt am Main 60598, Germany
| | - Tina Jenewein
- Institute of Legal Medicine, Goethe University Frankfurt, University Hospital, Kennedyallee 104, Frankfurt am Main 60598, Germany
| | - Andreas Pich
- Core Facility Proteomics, Institute of Toxicology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
- Department of Cardiovascular Complications of Oncologic Therapies, Medical Faculty of the Phillipps University Marburg, Baldingerstraße, Marburg 35032, Germany
| |
Collapse
|
10
|
Wan T, Liang Y, Wei T, Chen Z, Li Y. Targeting Lactic Acid Modification in Ischemic Heart Diseases: Novel Therapeutics and Mechanism. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10593-3. [PMID: 39920549 DOI: 10.1007/s12265-025-10593-3] [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: 09/13/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Ischemic heart disease (IHD), especially acute myocardial infarction (AMI), has a high mortality rate and poses a great threat to human health. When myocardial infarction occurs, the structure and function of the myocardium are significantly damaged, and its metabolisms switch from oxidative phosphorylation to glycolysis, producing lactate. Lactylation, as a newly discovered post-translational modification (PMT) in recent years, is involved in the regulation of gene expression, and cell proliferation. Emerging studies have revealed that lactate and lactylation modifications participate in inflammation and cardiac repair, and play an important role in cardiovascular diseases, such as myocardial infarction, myocardial fibrosis, and heart failure. Therefore, in this review, we discuss how glucose metabolism, glycolytic end-product lactate, and lactylation potentially interact with pathological processes, including inflammation, cardiac fibrosis, and heart failure. And targeting glycolysis and lactylation modification could provide a promising future for cardiovascular diseases.
Collapse
Affiliation(s)
- Tangjiang Wan
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China
| | - Yucheng Liang
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China
| | - Tianwen Wei
- Department of Cardiovascular Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zijie Chen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Fudan University, Shanghai, China
| | - Yafei Li
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China.
| |
Collapse
|
11
|
Mou T, Meng J, Lin C, Xie X, Hsu B, Zhang X. XTR003, a fatty acid metabolism PET tracer: A phase I study to evaluate the safety, biodistribution, radiation dosimetry, and pharmacokinetics in healthy volunteers. J Nucl Cardiol 2025:102144. [PMID: 39923833 DOI: 10.1016/j.nuclcard.2025.102144] [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: 03/26/2024] [Revised: 01/04/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE XTR003 is a novel 18F-labeled fatty acid PET tracer to image myocardial fatty acid metabolism that can be potentially used to assess myocardial viability for ischemic heart disease. This Phase I study evaluated its safety, biodistribution, radiation dosimetry, and pharmacokinetics. METHODS Ten healthy Chinese volunteers (mean age of 28.4 ± 4.6 years, 3 females) were intravenously injected with XTR003 (296-370 MBq) at rest and monitored for adverse events on the day of injection and follow-up days. Multiple whole-body PET images were acquired within 290 minutes and processed to investigate the biodistribution and radiation dosimetry. Whole blood, plasma, and urine were collected simultaneously for 420 minutes to evaluate the pharmacokinetics with the measurement of radioactivity. RESULTS Only two treatment-related adverse events occurred with no severe adverse effects. After tracer injection, XTR003 in the plasma peaked at 2.883 minutes as .0108235% of injected dose per gram (%ID/g) and reduced to the minimum at 30 minutes. The 0-20 minutes whole-body PET images indicated that both heart and liver were two critical organs with the highest percentage of injected dose (%ID) (4.37 ± .66 and 48.76 ± 4.17 %ID). Specifically, XTR003 demonstrated high initial uptake in the heart, with sustained retention for up to 290 minutes (standardized uptake value: 6.50 ± 2.54 at 0-20 minutes and 5.89 ± 2.18 at 270-290 minutes). The whole-body effective radiation dose was 17 μSv/MBq. The cumulative urinary excretion was 9.009%. CONCLUSIONS XTR003, as an18F-labeled radiotracer, was safe and well-tolerated. The rapid uptake and prolonged retention of XTR003 in the heart show promise for evaluating myocardial fatty acid metabolism. The Phase II clinical trial to explore the efficacy of XTR003 for detecting myocardial viability should be warranted. TRAIL REGISTRATION NUMBER ClinicalTrials.gov Identifier: NCT05136391.
Collapse
Affiliation(s)
- Tiantian Mou
- Department of Nuclear Medicine, Laboratory for Molecular Imaging, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jingjing Meng
- Department of Nuclear Medicine, Laboratory for Molecular Imaging, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengyu Lin
- Sinotau Pharmaceutical Group, Beijing, China
| | - Xiaofen Xie
- Department of Nuclear Medicine, Laboratory for Molecular Imaging, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bailing Hsu
- Nuclear Science and Engineering Institute, University of Missouri-Columbia, Columbia, MO, USA.
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Laboratory for Molecular Imaging, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Li H, Cheng Z, Wu D, Hu Q. Nitric oxide and mitochondrial function in cardiovascular diseases. Nitric Oxide 2025; 154:42-50. [PMID: 39577487 DOI: 10.1016/j.niox.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/11/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Nitric oxide (NO) has been highlighted as an important factor in cardiovascular system. As a signaling molecule in the cardiovascular system, NO can relax blood vessels, lower blood pressure, and prevent platelet aggregation. Mitochondria serve as a central hub for cellular metabolism and intracellular signaling, and their dysfunction can lead to a variety of diseases. Accumulating evidence suggests that NO can act as a regulator of mitochondria, affecting mitochondrial function and cellular activity, which in turn mediates the onset and progression of disease. However, there is a lack of comprehensive understanding of how NO regulates mitochondrial function in the cardiovascular system. This review aims to summarize the regulation of mitochondrial function by nitric oxide in cardiovascular related diseases, as well as the multifaceted and complex roles of NO in the cardiovascular system. Understanding the mechanism of NO mediated mitochondrial function can provide new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoqi Li
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zijie Cheng
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
13
|
Kim Y, Kim D, Kim J, Yun M, Kang ES. Association between appendicular skeletal muscle mass and myocardial glucose uptake measured by 18F-FDG PET. ESC Heart Fail 2025; 12:467-476. [PMID: 39344859 PMCID: PMC11769618 DOI: 10.1002/ehf2.15086] [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: 04/30/2024] [Revised: 08/18/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Low muscle mass is associated with high insulin resistance and an increased risk of cardiovascular disease. This study aims to determine whether low muscle mass affects the alterations in myocardial substrate metabolism that are associated with the development of cardiovascular disease. METHOD The study included 299 individuals (182 men and 117 women) who underwent examination at the Severance Health Check-up Center between January 2018 and February 2019. Myocardial glucose uptake was assessed using [18F]-fluorodeoxyglucose-positron emission tomography (18F-FDG PET/CT) scanning. Direct segmental bioimpedance analysis was used to measure appendicular skeletal muscle mass (ASM). RESULTS We analysed men and women separately owing to sex-related body composition differences. ASM/Ht2 was significantly positively correlated with myocardial glucose uptake measured by 18F-FDG PET/CT [ln (SUVheart/liver)] only in men (r = 0.154, P = 0.038 in men; r = -0.042, P = 0.652 in women, respectively). In men, myocardial glucose uptake was significantly associated with ASM/Ht2 even after adjusting for multiple confounders in a multivariable linear regression model (standardized β = 0.397, P = 0.004, in men; β = - 0.051, P = 0.698, in women). In women, age (β = -0.424 P = 0.029) was independent determinants of myocardial glucose uptake. CONCLUSIONS In men, ASM was strongly associated with myocardial glucose uptake as measured by 18F-FDG PET/CT. In women, age was significantly correlated with myocardial substrate utilization, but not with ASM.
Collapse
Affiliation(s)
- Young‐eun Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Anam HospitalKorea University College of MedicineSeoulRepublic of Korea
| | - Dongwoo Kim
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Jiwon Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Department of Internal Medicine, National Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Mijin Yun
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
14
|
Yang F, He Y, Zhao L, Huang J, Du F, Tian S, Zhang Y, Liu X, Chen B, Ge J, Jiang Z. Leptin drives glucose metabolism to promote cardiac protection via OPA1-mediated HDAC5 translocation and Glut4 transcription. Funct Integr Genomics 2025; 25:28. [PMID: 39875704 PMCID: PMC11774999 DOI: 10.1007/s10142-024-01515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025]
Abstract
Metabolic reprogramming, the shifting from fatty acid oxidation to glucose utilization, improves cardiac function as heart failure (HF) progresses. Leptin plays an essential role in regulating glucose metabolism. However, the crosstalk between leptin and metabolic reprogramming is poorly understood. We tested the hypothesis that leptin improves cardiac function after myocardial infarction via enhancing glucose metabolism. In the isoproterenol (ISO)-induced heart failure model in vitro, H9c2 cell apoptosis was assessed by the TUNEL and Annexin V/PI staining assay. Leptin-mediated mitochondrial fusion was performed via TEM, and glucose oxidation was explored, as well as the ECAR, OCR, and protein expression of the vital metabolic enzymes. By blocking OPA1 expression or HDAC5 inhibition, the mitochondrial dynamic and glucose metabolic were detected to evaluate the role of OPA1 and HDAC5 in leptin-stimulated glucose metabolism. In the mouse model of HF in vivo, intraperitoneal leptin administration appreciably increased glucose oxidation and preserved cardiac function 56 days after coronary artery ligation. In vitro, we identified the OPA1-dependent HDAC5 nucleus export as a crucial process in boosting glucose utilization by activating MEF2 to upregulate Glut4 expression using the RNA interference technique in H9c2 cells. In vivo, leptin promotes glucose utilization and confers heart functional and survival benefits in chronic ischemic HF. The current study provided a novel insight into the role of leptin in metabolic reprogramming and revealed potential therapeutic targets for chronic HF.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Youfu He
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Ling Zhao
- Health Management Center, Guizhou International General Hospital, Guizhou Province, China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Fawang Du
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Shui Tian
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Yang Zhang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Xinghui Liu
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Baolin Chen
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Junhua Ge
- Department of Cardiology, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), The Affiliated Hospital of Qingdao University, Shandong Province, China.
| | - Zhi Jiang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China.
- Guizhou University Medical College, Guizhou Province, China.
| |
Collapse
|
15
|
Lv T, Liu C, Guo S, Wu M, Wang X, Zhang Z, Zhou J, Yao Y, Shen Z, Yang J, Sun S, Liu Z, Chi J. Targeting Ketone Body Metabolism Improves Cardiac Function and Hemodynamics in Patients With Heart Failure: A Systematic Review and Meta-Analysis. Nutr Rev 2025:nuae179. [PMID: 39873669 DOI: 10.1093/nutrit/nuae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
CONTEXT The impacts of elevated ketone body levels on cardiac function and hemodynamics in patients with heart failure (HF) remain unclear. OBJECTIVE The effects of ketone intervention on these parameters in patients with HF were evaluated quantitatively in this meta-analysis. DATA SOURCES We searched the PubMed, Cochrane Library, and Embase databases for relevant studies published from inception to April 13, 2024. Ketone therapy included ketone ester and β-hydroxybutyrate intervention. DATA EXTRACTION Seven human studies were included for the quantitative analysis. DATA ANALYSIS Our results showed that ketone therapy significantly improved left ventricular ejection fraction (standardized mean difference, 0.52 [95% CI, 0.25-0.80]; I2 = 0%), cardiac output (0.84 [95% CI, 0.36-1.32]; I2 = 68%) and stroke volume (0.47 [95% CI, 0.10-0.84]; I2 = 39%), and significantly reduced systemic vascular resistance (-0.92 [95% CI, -1.52 to -0.33]; I2 = 74%) without influencing mean arterial pressure (-0.09 [95% CI: -0.40 to 0.22]; I2 = 0%) in patients with HF. Subgroup analysis revealed that the enhanced cardiac function and favorable hemodynamic effects of ketone therapy were also applicable to individuals without HF. CONCLUSIONS Ketone therapy may significantly improve cardiac systolic function and hemodynamics in patients with HF and in patients without HF, suggesting it may be a promising treatment for patients with HF and also a beneficial medical strategy for patients without HF or healthy individuals.
Collapse
Affiliation(s)
- Tingting Lv
- Department of General Practice, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Chunyan Liu
- Department of Infection Management, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
| | - Shitian Guo
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P. R. China
| | - Menglu Wu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Xiang Wang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ziyi Zhang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yiying Yao
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zeyu Shen
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Juntao Yang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Shijia Sun
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zheng Liu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jufang Chi
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Department of Cardiology, Zhuji People's Hospital (Zhuji Hospital, Wenzhou Medical University), Zhuji, Zhejiang 311800, P. R. China
| |
Collapse
|
16
|
Li L, Zhong S, Ye J, Hu S, Xiong X, Chen G, Hu Z. Shenmai injection revives cardiac function in rats with hypertensive heart failure: involvement of microbial-host co-metabolism. BMC Complement Med Ther 2025; 25:24. [PMID: 39856640 PMCID: PMC11761217 DOI: 10.1186/s12906-024-04737-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Heart failure (HF) is a complex syndrome marked by considerable expenditures and elevated mortality and morbidity rates globally. Shenmai injection (SMI), a form of Traditional Chinese Medicine-based therapy, has demonstrated effectiveness in treating HF. Recent research suggests that Traditional Chinese Medicine (TCM) may induce beneficial changes in microbial-host co-metabolism, potentially providing cardiovascular protection. This study used a rat model of hypertensive heart failure (H-HF) to explore the mechanism of SMI. The possible compounds and key targets of SMI against H-HF were investigated using network pharmacology. The pharmacodynamics of SMI were validated using the H-HF animal model, with analysis of fecal gut microbiota integrating metabolomics and 16S rRNA sequencing. Metorigin metabolite traceability analysis and the MetaboAnalyst platform were utilized to explore the action mechanism. To evaluate changes in serum TMAO levels, targeted metabolomics was performed. Finally, the study looked at the intrinsic relationships among modifications in the intestinal flora, metabolite profile changes, and the targets of SMI compounds to clarify how they might be used to treat H-HF. According to metabolomics and 16S rRNA sequencing, by reestablishing homeostasis in the gut microbiota, SMI affects vital metabolic pathways, such as energy metabolism, amino acid metabolism, and bile acid metabolism. Increased serum TMAO levels were identified to be a risk factor for H-HF, and SMI was able to downregulate the levels of TMAO-related metabolites. Network pharmacology analysis identified 13 active components of SMI targeting 46 proteins, resulting in differential expression changes in 8 metabolites and 24 gut microbes. In conclusion, this study highlights the effectiveness of SMI in alleviating H-HF and its potential to modulate microbial-host co-metabolism. Through a comprehensive discussion of the interconnected relationships among the components, targets, metabolites, and gut microbiota, it provided fresh light on the therapeutic mechanism of SMI on H-HF.
Collapse
Affiliation(s)
- Lin Li
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Senjie Zhong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiahao Ye
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyuan Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiajun Xiong
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guangyu Chen
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Engineering Technology Research Center For Medicinal and Functional Food, Changsha, Hunan, China
| | - Zhixi Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
17
|
Wang Z, Zhu H, Xiong W. Metabolism and metabolomics in senescence, aging, and age-related diseases: a multiscale perspective. Front Med 2025:10.1007/s11684-024-1116-0. [PMID: 39821730 DOI: 10.1007/s11684-024-1116-0] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
The pursuit of healthy aging has long rendered aging and senescence captivating. Age-related ailments, such as cardiovascular diseases, diabetes, and neurodegenerative disorders, pose significant threats to individuals. Recent studies have shed light on the intricate mechanisms encompassing genetics, epigenetics, transcriptomics, and metabolomics in the processes of senescence and aging, as well as the establishment of age-related pathologies. Amidst these underlying mechanisms governing aging and related pathology metabolism assumes a pivotal role that holds promise for intervention and therapeutics. The advancements in metabolomics techniques and analysis methods have significantly propelled the study of senescence and aging, particularly with the aid of multiscale metabolomics which has facilitated the discovery of metabolic markers and therapeutic potentials. This review provides an overview of senescence and aging, emphasizing the crucial role metabolism plays in the aging process as well as age-related diseases.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongying Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Wei Xiong
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| |
Collapse
|
18
|
Geng J, Zhang X, Guo Y, Wen H, Guo D, Liang Q, Pu S, Wang Y, Liu M, Li Z, Hu W, Yang X, Chang P, Hu L, Li Y. Moderate-intensity interval exercise exacerbates cardiac lipotoxicity in high-fat, high-calories diet-fed mice. Nat Commun 2025; 16:613. [PMID: 39800728 PMCID: PMC11725574 DOI: 10.1038/s41467-025-55917-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Physical exercise is a cornerstone for preventing diet-induced obesity, while it is unclear whether physical exercise could offset high-fat, high-calories diet (HFCD)-induced cardiac dysfunction. Here, mice were fed with HFCD and simultaneously subjected to physical exercise. As expected, physical exercise prevented HFCD-induced whole-body fat deposition. However, physical exercise exacerbated HFCD-induced cardiac damage. Further metabolomic analysis results showed that physical exercise induced circulating lipid redistribution, leading to excessive cardiac lipid uptake and lipotoxicity. Our study provides valuable insights into the cardiac effects of exercise in mice fed with HFCD, suggesting that counteracting the negative effect of HFCD by simultaneous physical exercise might be detrimental. Moreover, inappropriate physical exercise may damage certain organs even though it leads to weight loss and overall metabolic benefits. Of note, the current findings are based on animal experiments, the generalizability of these findings beyond this specific diet and mouse strain remains to be further explored.
Collapse
Affiliation(s)
- Jing Geng
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Xiaoliang Zhang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
- Department of Cardiology, No.901 Hospital of PLA, Hefei, China
| | - Yanjie Guo
- Xi'an International Medical Center Hospital, Xi'an, China
| | - He Wen
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Qi Liang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Siying Pu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Mingchuan Liu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Zhelong Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Wei Hu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Xue Yang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China
| | - Pan Chang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Medical College, Xi'an, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, China.
| |
Collapse
|
19
|
UMEZAWA M, AOKI T, NIIMI S, TAKANO H, MAMADA K, FUJII Y. A pilot study investigating serum carnitine profile of cats with preclinical hypertrophic cardiomyopathy. J Vet Med Sci 2025; 87:75-79. [PMID: 39567015 PMCID: PMC11735214 DOI: 10.1292/jvms.24-0171] [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: 05/07/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
This study aimed to prospectively investigate the hypothesis that myocardial energy metabolism changes in the early stages of feline hypertrophic cardiomyopathy (HCM) by evaluating the blood carnitine profiles. Fourteen client owned cats with HCM stage B1 and 22 clinically healthy cats were included in this study. Total-carnitine, free-carnitine, acylcarnitine, and acylcarnitine/free-carnitine ratio (AF ratio) in serum were measured in both groups. No significant differences were observed in total-carnitine, free-carnitine, and acylcarnitine concentrations between the groups. However, the AF ratio in the HCM group was significantly lower than that of the control group (P=0.02). The lower serum AF ratio in cats with early-stage HCM, may suggest an alteration in their myocardial energy metabolism.
Collapse
Affiliation(s)
- Mutsuki UMEZAWA
- Laboratory of Small Animal Surgery, Azabu University, Kanagawa, Japan
| | - Takuma AOKI
- Laboratory of Small Animal Surgery, Azabu University, Kanagawa, Japan
| | - Seiya NIIMI
- Laboratory of Small Animal Surgery, Azabu University, Kanagawa, Japan
| | | | - Kazuya MAMADA
- Veterinary Specialists Emergency Center, Saitama, Japan
| | - Yoko FUJII
- Laboratory of Small Animal Surgery, Azabu University, Kanagawa, Japan
| |
Collapse
|
20
|
Tudorancea I, Șerban IL, Șerban DN, Costache-Enache II, Cătălin C, Naum AG, Iliescu R. Sympathetic nervous system inhibition enhances cardiac metabolism and improves hemodynamics and glucose-insulin dynamics in obese and lean rat models. Sci Rep 2025; 15:503. [PMID: 39747975 PMCID: PMC11697016 DOI: 10.1038/s41598-024-84218-1] [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: 09/18/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
This study aimed to investigate the effects of chronic sympathoinhibition on glucose uptake by the myocardium and by the skeletal muscle in an animal model of obesity associated with leptin signaling deficiency. 6 obese Zucker rats (OZR) and 6 control Lean Zucker rats (LZR) were studied during basal conditions, chronic clonidine administration (30 days, 300 µg/kg), and washout recovery period. Glucose uptake in the myocardium and in the skeletal muscle was measured using positron emission tomography (PET) and 2-[18F] fluoro-2-deoxy-D-glucose ([18F]FDG). The standardized uptake value (SUV) corrected for blood glucose was used for the semi-quantitative analysis. Body weight, food and water intake, blood glucose concentration, blood pressure variability as an index of sympathetic activity and hemodynamic parameters such as mean arterial blood pressure (MAP), systolic blood pressure (SBP), diastolic blood pressure (DBP) and heart rate (HR) were analyzed. Myocardial glucose uptake was significantly lower during basal conditions in OZR versus LZR. In both OZR and LZR, chronic clonidine significantly reduced myocardial glucose uptake and hemodynamic variables (such as MAP, SBP, DBP, HR), and sympathetic activity (SA). [18F]FDG skeletal muscle uptake did not significantly differ in OZR versus LZR. Our findings indicate that cardiac glucose metabolism is reduced in obesity presumably in relation with the level of sympathetic activation.
Collapse
Affiliation(s)
- Ionuț Tudorancea
- Department of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 1 Independenței Blvd, Iași, Romania
| | - Ionela Lăcrămioara Șerban
- Department of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
| | - Dragomir N Șerban
- Department of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
| | - Irina-Iuliana Costache-Enache
- Cardiology Clinic, "St. Spiridon" County Clinical Emergency Hospital, 1 Independenței Blvd, Iași, Romania
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
| | - Caratașu Cătălin
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
| | - Alexandru Grațian Naum
- Department of Biophysics, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania.
| | - Radu Iliescu
- Department of Pharmacology, "Grigore T. Popa" University of Medicine and Pharmacy, 16 University St., Iași, Romania
| |
Collapse
|
21
|
Peng M, Fu Y, Qin C, Shi L, Zhang M, Zhou S. A stratified study of human blood metabolites and coronary artery diseases-A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103754. [PMID: 39448312 DOI: 10.1016/j.numecd.2024.09.024] [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: 02/21/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysregulation is closely associated with coronary artery diseases (CAD). Exploring the relationship between metabolites and CAD is helpful in identifying changes in energy metabolism during disease progression. METHODS AND RESULTS We use Mendelian Randomization (MR) analysis to assess the relationships between 275 serum metabolites and CAD such as angina pectoris, post-myocardial infarction complications, coronary atherosclerosis, myocardial infarction (MI), and unstable angina pectoris (UA). The inverse variance-weighted method (IVW) served as the primary approach for causal analysis, with MR-Egger and weighted median (WM) as supplementary methods. Sensitivity analyses were conducted to assess heterogeneity and multiple effects. We also analyzed potentially related metabolic pathways.We identified causal relationships between 42 known metabolites and CAD. Among them, the genetic susceptibility to elevated levels of amino acid Isobutyrylcarnitine is associated with an increased risk of coronary artery atherosclerosis; but it provides protection against the development of MI. Genetic susceptibility to elevated levels of fatty acids Stearate, Caprylate is associated with higher risk of angina pectoris, while Threonate has a protective effect in the development of angina; Stearate is associated with an increased risk of UA, whereas higher levels of the lipids Choline, 1-arachidonoylglycerophosphoinositol∗, Hexadecanedioate, Tetradecanedioate play a protective role in UA.Metabolic pathway analysis identified 6 pathways that may be associated with CAD. CONCLUSION We identified causal relationships between 42 serum metabolites and CAD. Specifically, changes in metabolites such as Isobutyrylcarnitine, Caprylate, and Stearate were associated with risks of CAD. These findings provide new insights into the metabolic mechanisms of CAD.
Collapse
Affiliation(s)
- Mengling Peng
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Yu Fu
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Cong Qin
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Lei Shi
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Meiwei Zhang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Shanshan Zhou
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China.
| |
Collapse
|
22
|
Lee JE, Kim BG, Won JC. Molecular Pathways in Diabetic Cardiomyopathy and the Role of Anti-hyperglycemic Drugs Beyond Their Glucose Lowering Effect. J Lipid Atheroscler 2025; 14:54-76. [PMID: 39911956 PMCID: PMC11791414 DOI: 10.12997/jla.2025.14.1.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/02/2024] [Accepted: 09/27/2024] [Indexed: 02/07/2025] Open
Abstract
Epidemiological evidence has shown that diabetes is associated with overt heart failure (HF) and worse clinical outcomes. However, the presence of a distinct primary diabetic cardiomyopathy (DCM) has not been easy to prove because the association between diabetes and HF is confounded by hypertension, obesity, microvascular dysfunction, and autonomic neuropathy. In addition, the molecular mechanisms underlying DCM are not yet fully understood, DCM usually remains asymptomatic in the early stage, and no specific biomarkers have been identified. Nonetheless, several mechanistic associations at the systemic, cardiac, and cellular/molecular levels explain different aspects of myocardial dysfunction, including impaired cardiac relaxation, compliance, and contractility. In this review, we focus on recent clinical and preclinical advances in our understanding of the molecular mechanisms of DCM and the role of anti-hyperglycemic agents in preventing DCM beyond their glucose lowering effect.
Collapse
Affiliation(s)
- Jie-Eun Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Byung Gyu Kim
- Division of Cardiology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Jong Chul Won
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Hunter B, Li M, Parker BL, Koay YC, Harney DJ, Pearson E, Cao J, Chen GT, Guneratne O, Smyth GK, Larance M, O'Sullivan JF, Lal S. Proteomic and metabolomic analyses of the human adult myocardium reveal ventricle-specific regulation in end-stage cardiomyopathies. Commun Biol 2024; 7:1666. [PMID: 39702518 DOI: 10.1038/s42003-024-07306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
The left and right ventricles of the human heart are functionally and developmentally distinct such that genetic or acquired insults can cause dysfunction in one or both ventricles resulting in heart failure. To better understand ventricle-specific molecular changes influencing heart failure development, we first performed unbiased quantitative mass spectrometry on pre-mortem non-diseased human myocardium to compare the metabolome and proteome between the normal left and right ventricles. Constituents of gluconeogenesis, glycolysis, lipogenesis, lipolysis, fatty acid catabolism, the citrate cycle and oxidative phosphorylation were down-regulated in the left ventricle, while glycogenesis, pyruvate and ketone metabolism were up-regulated. Inter-ventricular significance of these metabolic pathways was then found to be diminished within end-stage dilated cardiomyopathy and ischaemic cardiomyopathy, while heart failure-associated pathways were increased in the left ventricle relative to the right within ischaemic cardiomyopathy, such as fluid sheer-stress, increased glutamine-glutamate ratio, and down-regulation of contractile proteins, indicating a left ventricular pathological bias.
Collapse
Affiliation(s)
- Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Chin Koay
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
| | - Dylan J Harney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Evangeline Pearson
- Paediatric Oncology and Haematology, Oxford Children's Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, England
| | - Jacob Cao
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gavin T Chen
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Oneka Guneratne
- Kolling Institute, Royal North Shore Hospital, and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Larance
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Heart Research Institute, Newtown, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, NSW, Australia.
| |
Collapse
|
24
|
Goedeke L, Ma Y, Gaspar RC, Nasiri A, Lee J, Zhang D, Galsgaard KD, Hu X, Zhang J, Guerrera N, Li X, LaMoia T, Hubbard BT, Haedersdal S, Wu X, Stack J, Dufour S, Butrico GM, Kahn M, Perry RJ, Cline GW, Young LH, Shulman GI. SGLT2 inhibition alters substrate utilization and mitochondrial redox in healthy and failing rat hearts. J Clin Invest 2024; 134:e176708. [PMID: 39680452 PMCID: PMC11645152 DOI: 10.1172/jci176708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/17/2024] [Indexed: 12/18/2024] Open
Abstract
Previous studies highlight the potential for sodium-glucose cotransporter type 2 (SGLT2) inhibitors (SGLT2i) to exert cardioprotective effects in heart failure by increasing plasma ketones and shifting myocardial fuel utilization toward ketone oxidation. However, SGLT2i have multiple in vivo effects and the differential impact of SGLT2i treatment and ketone supplementation on cardiac metabolism remains unclear. Here, using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) methodology combined with infusions of [13C6]glucose or [13C4]βOHB, we demonstrate that acute SGLT2 inhibition with dapagliflozin shifts relative rates of myocardial mitochondrial metabolism toward ketone oxidation, decreasing pyruvate oxidation with little effect on fatty acid oxidation in awake rats. Shifts in myocardial ketone oxidation persisted when plasma glucose levels were maintained. In contrast, acute βOHB infusion similarly augmented ketone oxidation, but markedly reduced fatty acid oxidation and did not alter glucose uptake or pyruvate oxidation. After inducing heart failure, dapagliflozin increased relative rates of ketone and fatty acid oxidation, but decreased pyruvate oxidation. Dapagliflozin increased mitochondrial redox and reduced myocardial oxidative stress in heart failure, which was associated with improvements in left ventricular ejection fraction after 3 weeks of treatment. Thus, SGLT2i have pleiotropic effects on systemic and heart metabolism, which are distinct from ketone supplementation and may contribute to the long-term cardioprotective benefits of SGLT2i.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Medicine (Cardiology) and The Cardiovascular Research Institute and
- Department of Medicine (Endocrinology) and The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yina Ma
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
| | - Rafael C. Gaspar
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Ali Nasiri
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jieun Lee
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Dongyan Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Katrine Douglas Galsgaard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoyue Hu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jiasheng Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Nicole Guerrera
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Xiruo Li
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Traci LaMoia
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Brandon T. Hubbard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Sofie Haedersdal
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Clinical Research, Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Xiaohong Wu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - John Stack
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Sylvie Dufour
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Gina Marie Butrico
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Mario Kahn
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Rachel J. Perry
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gary W. Cline
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Lawrence H. Young
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gerald I. Shulman
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
25
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [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: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
26
|
Zoccarato A, Smyrnias I, Reumiller CM, Hafstad AD, Chong M, Richards DA, Santos CXC, Visnagri A, Verma S, Bromage DI, Zhang M, Zhang X, Sawyer G, Thompson R, Shah AM. NRF2 activation in the heart induces glucose metabolic reprogramming and reduces cardiac dysfunction via upregulation of the pentose phosphate pathway. Cardiovasc Res 2024:cvae250. [PMID: 39657243 DOI: 10.1093/cvr/cvae250] [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: 04/05/2024] [Revised: 08/02/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
AIMS The transcription factor NRF2 is well recognized as a master regulator of antioxidant responses and cytoprotective genes. Previous studies showed that NRF2 enhances resistance of mouse hearts to chronic hemodynamic overload at least in part by reducing oxidative stress. Evidence from other tissues suggests that NRF2 may modulate glucose intermediary metabolism but whether NRF2 has such effects in the heart is unclear. Here, we investigate the role of NRF2 in regulating glucose intermediary metabolism and cardiac function during disease stress. METHODS AND RESULTS Cardiomyocyte-specific Keap1 knockout (csKeap1KO) mice, deficient in the endogenous inhibitor of NRF2, were used as a novel model of constitutively active NRF2 signaling. Targeted metabolomics and isotopomer analysis were employed in studies with 13C6-glucose in csKeap1KO and wild-type (WT) mice. Pharmacological and genetic approaches were utilized in neonatal rat ventricular cardiomyocytes (NRVM) to explore molecular mechanisms. We found that cardiac-specific activation of NRF2 redirected glucose metabolism towards the pentose phosphate pathway (PPP), a branch pathway of glycolysis, and mitigated pressure overload-induced cardiomyocyte death and cardiac dysfunction. Activation of NRF2 also protected against myocardial infarction-induced DNA damage in remote myocardium and cardiac dysfunction. In vitro, knockdown of Keap1 upregulated PPP enzymes and reduced cell death in NRVM subjected to chronic neurohumoral stimulation. These pro-survival effects were abolished by pharmacological inhibition of the PPP or silencing of the PPP rate-limiting enzyme glucose-6-phosphate dehydrogenase (G6PD). Knockdown of NRF2 in NRVM increased stress-induced DNA damage which was rescued by supplementing the cells with either NADPH or nucleosides, the two main products of the PPP. CONCLUSIONS These results indicate that NRF2 regulates cardiac metabolic reprogramming by stimulating the diversion of glucose into the PPP, thereby generating NADPH and providing nucleotides to prevent stress-induced DNA damage and cardiac dysfunction.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ioannis Smyrnias
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
- Comparative Biomedical Sciences, University of Surrey, Guildford, UK
| | - Christina M Reumiller
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Anne D Hafstad
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
- Cardiovascular Research Group, Department of Medical Biology, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Mei Chong
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Daniel A Richards
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Asjad Visnagri
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Sharwari Verma
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Daniel I Bromage
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Min Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Xiaohong Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Greta Sawyer
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Richard Thompson
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
27
|
Liu C, Shen M, Liu Y, Manhas A, Zhao SR, Zhang M, Belbachir N, Ren L, Zhang JZ, Caudal A, Nishiga M, Thomas D, Zhang A, Yang H, Zhou Y, Ameen M, Sayed N, Rhee JW, Qi LS, Wu JC. CRISPRi/a screens in human iPSC-cardiomyocytes identify glycolytic activation as a druggable target for doxorubicin-induced cardiotoxicity. Cell Stem Cell 2024; 31:1760-1776.e9. [PMID: 39515331 PMCID: PMC11646563 DOI: 10.1016/j.stem.2024.10.007] [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: 12/08/2022] [Revised: 07/31/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Doxorubicin is limited in its therapeutic utility due to its life-threatening cardiovascular side effects. Here, we present an integrated drug discovery pipeline combining human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMs), CRISPR interference and activation (CRISPRi/a) bidirectional pooled screens, and a small-molecule screening to identify therapeutic targets mitigating doxorubicin-induced cardiotoxicity (DIC) without compromising its oncological effects. The screens revealed several previously unreported candidate genes contributing to DIC, including carbonic anhydrase 12 (CA12). Genetic inhibition of CA12 protected iCMs against DIC by improving cell survival, sarcomere structural integrity, contractile function, and calcium handling. Indisulam, a CA12 antagonist, can effectively attenuate DIC in iCMs, engineered heart tissue, and animal models. Mechanistically, doxorubicin-induced CA12 potentiated a glycolytic activation in cardiomyocytes, contributing to DIC by interfering with cellular metabolism and functions. Collectively, our study provides a roadmap for future drug discovery efforts, potentially leading to more targeted therapies with minimal off-target toxicity.
Collapse
Affiliation(s)
- Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Physiology and Cancer Center, Milwaukee, WI, USA; Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Mengcheng Shen
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Yanxia Liu
- Department of Bioengineering, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Amit Manhas
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Mao Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Nadjet Belbachir
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Lu Ren
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Joe Z Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Dilip Thomas
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Greentstone Biosciences, Palo Alto, CA, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Yang Zhou
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Mohamed Ameen
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA; Department of Surgery, Division of Vascular Surgery, Stanford University, Stanford, CA, USA
| | - June-Wha Rhee
- Department of Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine (Division of Cardiology), Stanford, CA, USA.
| |
Collapse
|
28
|
Staršíchová A. SR-B1-/-ApoE-R61h/h Mice Mimic Human Coronary Heart Disease. Cardiovasc Drugs Ther 2024; 38:1123-1137. [PMID: 37273155 PMCID: PMC10240136 DOI: 10.1007/s10557-023-07475-8] [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] [Accepted: 05/22/2023] [Indexed: 06/06/2023]
Abstract
Cardiovascular diseases are the leading cause of death in the modern world. Atherosclerosis underlies the majority of these pathologies and may result in sudden life-threatening events such as myocardial infarction or stroke. Current concepts consider a rupture (resp. erosion) of "unstable/vulnerable" atherosclerotic plaques as a primary cause leading to thrombus formation and subsequent occlusion of the artery lumen finally triggering an acute clinical event. We and others described SR-B1-/-ApoE-R61h/h mice mimicking clinical coronary heart disease in all major aspects: from coronary atherosclerosis through vulnerable plaque ruptures leading to thrombus formation/coronary artery occlusion, finally resulting in myocardial infarction/ischemia. SR-B1-/-ApoE-R61h/h mouse provides a valuable model to study vulnerable/occlusive plaques, to evaluate bioactive compounds as well as new anti-inflammatory and "anti-rupture" drugs, and to test new technologies in experimental cardiovascular medicine. This review summarizes and discuss our knowledge about SR-B1-/-ApoE-R61h/h mouse model based on recent publications and experimental observations from the lab.
Collapse
Affiliation(s)
- Andrea Staršíchová
- Graduate School Cell Dynamics and Disease, University of Muenster, Muenster, Germany.
- European Institute for Molecular Imaging, University of Muenster, Muenster, Germany.
- Novogenia Covid GmbH, Eugendorf, Austria.
| |
Collapse
|
29
|
Chen Z, Zhang M, Xu Q, Lu P, Liu M, Yin R, Liu X, Dai Y, Gao X, Gong J, Zhang S, Wang X. Huangqi-Danshen decoction improves heart failure by regulating pericardial adipose tissue derived extracellular vesicular miR-27a-3p to activate AMPKα2 mediated mitophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156187. [PMID: 39488874 DOI: 10.1016/j.phymed.2024.156187] [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: 08/08/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Huangqi-Danshen decoction (HDD) is a classic traditional Chinese medicine for treating heart failure. Pericardial adipose tissue (PAT) has recently gained increasing attention in cardiovascular diseases. PURPOSE This study aimed to investigate the effect of pericardial adipose tissue-derived extracellular vesicles on heart failure, the protective effect of HDD on myocardial remodel in heart failure rats, and identify the potential molecular mechanisms involved. METHODS UPLC-MS/MS identified active components of HDD. Extracellular vesicles (EVs) from pericardial adipose tissue of sham-operated and HF rats were identified through transmission electron microscopy, nanoparticle tracking analysis and western blot. EVs were co-cultured with H9c2 cardiomyocytes in order to examine their uptake and effects. MicroRNA sequencing, dual-luciferase reporter assay and PCR were conducted for exploring specific mechanisms of EVs on hypertrophic cardiomyocytes. In vivo, heart failure was modeled in rats via transverse aortic constriction (TAC). In vitro, the hypertrophic cardiomyocyte model were established using Ang II-induced H9c2 cardiomyocytes. RESULTS UPLC-MS/MS identified 11 active components in serum of HDD administrated rats. Echocardiography showed HDD improved cardiac function in TAC model rats. HE and Masson staining indicated HDD ameliorated myocardial hypertrophy and fibrosis. MicroRNA sequencing found that HDD treatment resulted in 37 differentially expressed miRNAs (DMEs) (p < 0.05 and |log2FC| ≥ 1). KEGG analysis revealed that DEMs were enriched in the AMPK signaling pathway. PCR identified miR-27a-3p with the greatest difference in AMPK-related DMEs. Dual-luciferase reporter assay and Targetscan website were utilized to identify the target relationship between miR-27a-3p and PRKAA2 (AMPKα2). The miR-27a-3p negatively regulated AMPKα2 to inhibit mitophagy mediated by PINK1/Parkin pathway. HDD inhibited miR-27a-3p secretion from failing heart pericardial adipose tissue-derived extracellular vesicles, thereby improving inflammation, cardiac function, and myocardial remodeling through above pathways. CONCLUSION HDD inhibited the PAT-derived extracellular vesicular miR-27a-3p in failing hearts to activate AMPK/PINK1/Parkin signaling-mediated mitophagy, which improved cardiomyocyte energy metabolism, myocardial remodeling and heart failure.
Collapse
Affiliation(s)
- Zhaoyang Chen
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Meng Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiyao Xu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Pengyu Lu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Min Liu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Rui Yin
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Xuan Liu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Yang Dai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China
| | - Xin Gao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Juexiao Gong
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Sujie Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
30
|
Watson WD, Arvidsson PM, Miller JJJ, Lewis AJ, Rider OJ. A Mitochondrial Basis for Heart Failure Progression. Cardiovasc Drugs Ther 2024; 38:1161-1171. [PMID: 38878138 PMCID: PMC11680631 DOI: 10.1007/s10557-024-07582-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 12/29/2024]
Abstract
In health, the human heart is able to match ATP supply and demand perfectly. It requires 6 kg of ATP per day to satisfy demands of external work (mechanical force generation) and internal work (ion movements and basal metabolism). The heart is able to link supply with demand via direct responses to ADP and AMP concentrations but calcium concentrations within myocytes play a key role, signalling both inotropy, chronotropy and matched increases in ATP production. Calcium/calmodulin-dependent protein kinase (CaMKII) is a key adapter to increased workload, facilitating a greater and more rapid calcium concentration change. In the failing heart, this is dysfunctional and ATP supply is impaired. This review aims to examine the mechanisms and pathologies that link increased energy demand to this disrupted situation. We examine the roles of calcium loading, oxidative stress, mitochondrial structural abnormalities and damage-associated molecular patterns.
Collapse
Affiliation(s)
- William D Watson
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK.
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK.
| | - Per M Arvidsson
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Lund, Sweden
| | - Jack J J Miller
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Andrew J Lewis
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Rizza V, Tondi L, Patti AM, Cecchi D, Lombardi M, Perone F, Ambrosetti M, Rizzo M, Cianflone D, Maranta F. Diabetic cardiomyopathy: pathophysiology, imaging assessment and therapeutical strategies. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 23:200338. [PMID: 39734497 PMCID: PMC11681223 DOI: 10.1016/j.ijcrp.2024.200338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetes mellitus (DM) is one of the most prevalent cardiovascular risk factors in the general population, being associated with high morbidity and socioeconomic burden. Diabetic cardiomyopathy (DCM) is a non-negligible complication of DM, whose pathophysiological fundaments are the altered cardiac metabolism, the hyperglycemia-triggered formation of advanced glycation end-products (AGEs) and the inflammatory milieu which are typical in diabetic patients. These metabolic abnormalities lead to cardiomyocytes apoptosis, interstitial fibrosis and mechanical cardiac dysfunction, which can be identified with non-invasive imaging techniques, like echocardiography and cardiac magnetic resonance. This review aims to: 1) describe the major imaging features of DCM; 2) highlight how early identification of DCM-related anatomical and functional remodeling might allow patients' therapy optimization and prognosis improvement.
Collapse
Affiliation(s)
| | - Lara Tondi
- Multimodality Cardiac Imaging Section, Policlinico San Donato, San Donato Milanese, Italy
- Postgraduate School of Radiology, University of Milan, Milan, Italy
| | - Angelo Maria Patti
- Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | | | - Massimo Lombardi
- Multimodality Cardiac Imaging Section, Policlinico San Donato, San Donato Milanese, Italy
| | - Francesco Perone
- Cardiac Rehabilitation Unit, Rehabilitation Clinic ‘Villa Delle Magnolie', Castel Morrone, Caserta, Italy
| | - Marco Ambrosetti
- Cardiovascular Rehabilitation Unit, ASST Crema, Santa Marta Hospital, Rivolta D'Adda, Italy
| | - Manfredi Rizzo
- Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Domenico Cianflone
- IRCCS Ospedale San Raffaele, Milan, Italy
- Cardiovascular Rehabilitation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Maranta
- IRCCS Ospedale San Raffaele, Milan, Italy
- Cardiovascular Rehabilitation Unit, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
32
|
Zhang X, Zhou X, Tu Z, Qiang L, Lu Z, Xie Y, Liu CH, Zhang L, Fu Y. Proteomic and ubiquitinome analysis reveal that microgravity affects glucose metabolism of mouse hearts by remodeling non-degradative ubiquitination. PLoS One 2024; 19:e0313519. [PMID: 39541295 PMCID: PMC11563481 DOI: 10.1371/journal.pone.0313519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Long-term exposure to a microgravity environment leads to structural and functional changes in hearts of astronauts. Although several studies have reported mechanisms of cardiac damage under microgravity conditions, comprehensive research on changes at the protein level in these hearts is still lacking. In this study, proteomic analysis of microgravity-exposed hearts identified 156 differentially expressed proteins, and ubiquitinomic analysis of these hearts identified 169 proteins with differential ubiquitination modifications. Integrated ubiquitinomic and proteomic analysis revealed that differential proteomic changes caused by transcription affect the immune response in microgravity-exposed hearts. Additionally, changes in ubiquitination modifications under microgravity conditions excessively activated certain kinases, such as hexokinase and phosphofructokinase, leading to cardiac metabolic disorders. These findings provide new insights into the mechanisms of cardiac damage under microgravity conditions.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xuemei Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhiwei Tu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lihua Qiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhe Lu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuping Xie
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Cui Hua Liu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yesheng Fu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
33
|
Łukasiewicz Mierzejewska M, Kotuszewska M, Puppel K, Madras Majewska B. Effects of In Ovo Taurine Injection on Embryo Development, Antioxidant Status, and Bioactive Peptide Content in Chicken Embryos ( Gallus gallus domesticus). Int J Mol Sci 2024; 25:11783. [PMID: 39519333 PMCID: PMC11546265 DOI: 10.3390/ijms252111783] [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: 09/20/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Stress in birds disrupts the homeostasis of the organism, leading to an inability to neutralize reactive oxygen species. Taurine, an effective antioxidant, affects various cellular mechanisms, including cation modulation, protein phosphorylation, and cell proliferation. The aim of the study was to evaluate the effect of colloid with taurine applied in ovo to Albumin on embryonic development, oxidative stress indicators and the content of bioactive peptides-carnosine and anserine-in the pectoral muscle. The research materials were eggs of the parent flock (Ross 308) divided into four groups (K (without injection), T50-concentration of taurine hydrocolloid 50 ppm (mg/L); T100-colloid concentration 100 ppm (mg/L) taurine; T500-colloid concentration of 500 ppm (mg/L) taurine). The experimental solutions were injected in an amount of 0.3 mL into egg white. Eggs were incubated under standard incubation conditions. The addition of 100 and 500 ppm taurine had a highly significant (p = 0.001) effect on the plasma antioxidant potential in chicks. The level of anserine increased with increasing concentrations of taurine. These changes were highly significant (p = 0.007). The level of anserine in the T2 and T3 groups was determined to be 2.5 times higher than in the pectoral muscles of embryos not treated with taurine colloid. An analysis of the results showed that the administration of an increased dose of hydrocolloid with taurine increased the content of carnosine and anserine in the pectoral muscle. Colloid with taurine applied in ovo to chicken white egg reduces oxidative stress and increases homeostasis of the organism.
Collapse
Affiliation(s)
- Monika Łukasiewicz Mierzejewska
- Institute of Animal Science, Warsaw University of Life Sciences, Ciszewskiego 8 Street, 02-786 Warsaw, Poland; (K.P.); (B.M.M.)
| | - Marta Kotuszewska
- Scientific Circle “Aves”, Warsaw University of Life Sciences, Ciszewskiego 8 Street, 02-786 Warsaw, Poland;
| | - Kamila Puppel
- Institute of Animal Science, Warsaw University of Life Sciences, Ciszewskiego 8 Street, 02-786 Warsaw, Poland; (K.P.); (B.M.M.)
| | - Beata Madras Majewska
- Institute of Animal Science, Warsaw University of Life Sciences, Ciszewskiego 8 Street, 02-786 Warsaw, Poland; (K.P.); (B.M.M.)
| |
Collapse
|
34
|
Dimasi CG, Darby JRT, Cho SKS, Saini BS, Holman SL, Meakin AS, Wiese MD, Macgowan CK, Seed M, Morrison JL. Reduced in utero substrate supply decreases mitochondrial abundance and alters the expression of metabolic signalling molecules in the fetal sheep heart. J Physiol 2024; 602:5901-5922. [PMID: 37996982 DOI: 10.1113/jp285572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
Babies born with fetal growth restriction (FGR) are at higher risk of developing cardiometabolic diseases across the life course. The reduction in substrate supply to the developing fetus that causes FGR not only alters cardiac growth and structure but may have deleterious effects on metabolism and function. Using a sheep model of placental restriction to induce FGR, we investigated key cardiac metabolic and functional markers that may be altered in FGR. We also employed phase-contrast magnetic resonance imaging MRI to assess left ventricular cardiac output (LVCO) as a measure of cardiac function. We hypothesized that signalling molecules involved in cardiac fatty acid utilisation and contractility would be impaired by FGR and that this would have a negative impact on LVCO in the late gestation fetus. Key glucose (GLUT4 protein) and fatty acid (FATP, CD36 gene expression) substrate transporters were significantly reduced in the hearts of FGR fetuses. We also found reduced mitochondrial numbers as well as abundance of electron transport chain complexes (complexes II and IV). These data suggest that FGR diminishes metabolic and mitochondrial capacity in the fetal heart; however, alterations were not correlated with fetal LVCO. Overall, these data show that FGR alters fetal cardiac metabolism in late gestation. If sustained ex utero, this altered metabolic profile may contribute to poor cardiac outcomes in FGR-born individuals after birth. KEY POINTS: Around the time of birth, substrate utilisation in the fetal heart switches from carbohydrates to fatty acids. However, the effect of fetal growth restriction (FGR) on this switch, and thus the ability of the fetal heart to effectively metabolise fatty acids, is not fully understood. Using a sheep model of early onset FGR, we observed significant downregulation in mRNA expression of fatty acid receptors CD36 and FABP in the fetal heart. FGR fetuses also had significantly lower cardiac mitochondrial abundance than controls. There was a reduction in abundance of complexes II and IV within the electron transport chain of the FGR fetal heart, suggesting altered ATP production. This indicates reduced fatty acid metabolism and mitochondrial function in the heart of the FGR fetus, which may have detrimental long-term implications and contribute to increased risk of cardiovascular disease later in life.
Collapse
Affiliation(s)
- Catherine G Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Steven K S Cho
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Brahmdeep S Saini
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ashley S Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Michael D Wiese
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Christopher K Macgowan
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mike Seed
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Birkedal R, Branovets J, Vendelin M. Compartmentalization in cardiomyocytes modulates creatine kinase and adenylate kinase activities. FEBS Lett 2024; 598:2623-2640. [PMID: 39112921 DOI: 10.1002/1873-3468.14994] [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: 03/14/2024] [Revised: 06/03/2024] [Accepted: 07/21/2024] [Indexed: 11/12/2024]
Abstract
Intracellular molecules are transported by motor proteins or move by diffusion resulting from random molecular motion. Cardiomyocytes are packed with structures that are crucial for function, but also confine the diffusional spaces, providing cells with a means to control diffusion. They form compartments in which local concentrations are different from the overall, average concentrations. For example, calcium and cyclic AMP are highly compartmentalized, allowing these versatile second messengers to send different signals depending on their location. In energetic compartmentalization, the ratios of AMP and ADP to ATP are different from the average ratios. This is important for the performance of ATPases fuelling cardiac excitation-contraction coupling and mechanical work. A recent study suggested that compartmentalization modulates the activity of creatine kinase and adenylate kinase in situ. This could have implications for energetic signaling through, for example, AMP-activated kinase. It highlights the importance of taking compartmentalization into account in our interpretation of cellular physiology and developing methods to assess local concentrations of AMP and ADP to enhance our understanding of compartmentalization in different cell types.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| |
Collapse
|
36
|
Geiser A, Currie S, Al-Hasani H, Chadt A, McConnell G, Gould GW. A novel 3D imaging approach for quantification of GLUT4 levels across the intact myocardium. J Cell Sci 2024; 137:jcs262146. [PMID: 38958032 DOI: 10.1242/jcs.262146] [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: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Cellular heterogeneity is a well-accepted feature of tissues, and both transcriptional and metabolic diversity have been revealed by numerous approaches, including optical imaging. However, the high magnification objective lenses needed for high-resolution imaging provides information from only small layers of tissue, which can result in poor cell statistics. There is therefore an unmet need for an imaging modality that can provide detailed molecular and cellular insight within intact tissue samples in 3D. Using GFP-tagged GLUT4 as proof of concept, we present here a novel optical mesoscopy approach that allows precise measurement of the spatial location of GLUT4 within specific anatomical structures across the myocardium in ultrathick sections (5 mm×5 mm×3 mm) of intact mouse heart. We reveal distinct GLUT4 distribution patterns across cardiac walls and highlight specific changes in GLUT4 expression levels in response to high fat diet-feeding, and we identify sex-dependent differences in expression patterns. This method is applicable to any target that can be labelled for light microscopy, and to other complex tissues when organ structure needs to be considered simultaneously with cellular detail.
Collapse
Affiliation(s)
- Angéline Geiser
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Currie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical faculty, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical faculty, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
37
|
Piperis C, Marathonitis A, Anastasiou A, Theofilis P, Mourouzis K, Giannakodimos A, Tryfou E, Oikonomou E, Siasos G, Tousoulis D. Multifaceted Impact of SGLT2 Inhibitors in Heart Failure Patients: Exploring Diverse Mechanisms of Action. Biomedicines 2024; 12:2314. [PMID: 39457625 PMCID: PMC11504660 DOI: 10.3390/biomedicines12102314] [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: 09/04/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Heart failure (HF) is a growing concern due to the aging population and increasing prevalence of comorbidities. Despite advances in treatment, HF remains a significant burden, necessitating novel therapeutic approaches. Sodium-glucose cotransporter 2 inhibitors (SGLT2is) have emerged as a promising treatment option, demonstrating benefits across the entire spectrum of HF, regardless of left ventricular ejection fraction (LVEF). This review explores the multifaceted mechanisms through which SGLT2is exert cardioprotective effects, including modulation of energy metabolism, reduction of oxidative stress, attenuation of inflammation, and promotion of autophagy. SGLT2is shift myocardial energy substrate utilization from carbohydrates to more efficient fatty acids and ketone bodies, enhancing mitochondrial function and reducing insulin resistance. These inhibitors also mitigate oxidative stress by improving mitochondrial biogenesis, reducing reactive oxygen species (ROS) production, and regulating calcium-signaling pathways. Inflammation, a key driver of HF progression, is alleviated through the suppression of proinflammatory cytokines and modulation of immune cell activity. Additionally, SGLT2is promote autophagy, facilitating the clearance of damaged cellular components and preserving myocardial structure and function. Beyond their glucose-lowering effects, SGLT2is provide significant benefits in patients with chronic kidney disease (CKD) and HF, reducing the progression of CKD and improving overall survival. The pleiotropic actions of SGLT2is highlight their potential as a cornerstone in HF management. Further research is needed to fully elucidate their mechanisms and optimize their use in clinical practice.
Collapse
Affiliation(s)
- Christos Piperis
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Anastasios Marathonitis
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Artemis Anastasiou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Panagiotis Theofilis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Konstantinos Mourouzis
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Alexios Giannakodimos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Elsi Tryfou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.M.); (A.A.); (K.M.); (A.G.); (E.T.); (E.O.); (G.S.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
38
|
Song Y, Spurlock B, Liu J, Qian L. Cardiac Aging in the Multi-Omics Era: High-Throughput Sequencing Insights. Cells 2024; 13:1683. [PMID: 39451201 PMCID: PMC11506570 DOI: 10.3390/cells13201683] [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: 09/11/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiovascular diseases are a leading cause of mortality worldwide, and the risks of both developing a disease and receiving a poor prognosis increase with age. With increasing life expectancy, understanding the mechanisms underlying heart aging has become critical. Traditional techniques have supported research into finding the physiological changes and hallmarks of cardiovascular aging, including oxidative stress, disabled macroautophagy, loss of proteostasis, and epigenetic alterations, among others. The advent of high-throughput multi-omics techniques offers new perspectives on the molecular mechanisms and cellular processes in the heart, guiding the development of therapeutic targets. This review explores the contributions and characteristics of these high-throughput techniques to unraveling heart aging. We discuss how different high-throughput omics approaches, both alone and in combination, produce robust and exciting new findings and outline future directions and prospects in studying heart aging in this new era.
Collapse
Affiliation(s)
- Yiran Song
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (Y.S.); (B.S.); (J.L.)
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian Spurlock
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (Y.S.); (B.S.); (J.L.)
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (Y.S.); (B.S.); (J.L.)
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (Y.S.); (B.S.); (J.L.)
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
39
|
Takeuchi A, Matsuoka S. A simulation study on the role of mitochondria-sarcoplasmic reticulum Ca 2+ interaction in cardiomyocyte energetics during exercise. J Physiol 2024. [PMID: 39387569 DOI: 10.1113/jp286054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/15/2024] [Indexed: 10/15/2024] Open
Abstract
Previous studies demonstrated that the mitochondrial Ca2+ uniporter MCU and the Na+-Ca2+ exchanger NCLX exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction. However, the physiological relevance of the mitochondria-SR Ca2+ interaction has remained unsolved. Furthermore, although mitochondrial Ca2+ has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases, the contribution of the Ca2+-dependent regulatory mechanisms to cellular functions under physiological conditions has been controversial. In this study, we constructed a new integrated model of human ventricular myocyte with excitation-contraction-energetics coupling and investigated systematically the contribution of mitochondria-SR Ca2+ interaction, especially focusing on cardiac energetics during dynamic workload transitions in exercise. Simulation analyses revealed that the spatial coupling of mitochondria and SR, particularly via mitochondrial Ca2+ uniport activity-RyR, was the primary determinant of mitochondrial Ca2+ concentration, and that the Ca2+-dependent regulatory mechanism facilitated mitochondrial NADH recovery during exercise and contributed to the stability of NADH in the workload transition by about 40%, while oxygen consumption rate and cytoplasmic ATP level were not influenced. We concluded that the mitochondria-SR Ca2+ interaction, created via the uneven distribution of Ca2+ handling proteins, optimizes the contribution of the mitochondrial Ca2+-dependent regulatory mechanism to stabilizing NADH during exercise. KEY POINTS: The mitochondrial Ca2+ uniporter protein MCU and the Na+-Ca2+ exchanger protein NCLX are reported to exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction in cardiomyocytes. Mitochondrial Ca2+ (Ca2+ mit) has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases. Here we constructed an integrated model of a human ventricular myocyte with excitation-contraction-energetics coupling and investigated the role of the mitochondria-SR Ca2+ interaction in cardiac energetics during exercise. Simulation analyses revealed that the spatial coupling particularly via mitochondrial Ca2+ uniport activity-RyR is the primary determinant of Ca2+ mit concentration, and that the activation of NADH-producing dehydrogenases by Ca2+ mit contributes to NADH stability during exercise. The mitochondria-SR Ca2+ interaction optimizes the contribution of Ca2+ mit to the activation of NADH-producing dehydrogenases.
Collapse
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| |
Collapse
|
40
|
Shi J, Jin Y, Lin S, Li X, Zhang D, Wu J, Qi Y, Li Y. Mitochondrial non-energetic function and embryonic cardiac development. Front Cell Dev Biol 2024; 12:1475603. [PMID: 39435335 PMCID: PMC11491369 DOI: 10.3389/fcell.2024.1475603] [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: 08/04/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
The initial contraction of the heart during the embryonic stage necessitates a substantial energy supply, predominantly derived from mitochondrial function. However, during embryonic heart development, mitochondria influence beyond energy supplementation. Increasing evidence suggests that mitochondrial permeability transition pore opening and closing, mitochondrial fusion and fission, mitophagy, reactive oxygen species production, apoptosis regulation, Ca2+ homeostasis, and cellular redox state also play critical roles in early cardiac development. Therefore, this review aims to describe the essential roles of mitochondrial non-energetic function embryonic cardiac development.
Collapse
Affiliation(s)
- Jingxian Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Patel S, Ahuja R, Vallejo JA, Siddiqui G, Colson J, Edegbe J, Salzman G, Hamidpour S, Monaghan-Nichols AP, Poisner A, Molteni A, Wacker MJ. Fat Embolism Does Not Alter Cardiac Structure or Induce Pathological Changes in a Rat Model. J Surg Res 2024; 302:628-640. [PMID: 39190973 DOI: 10.1016/j.jss.2024.07.104] [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: 10/01/2023] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Fat embolism (FE) encompasses conditions in which fatty substance becomes embedded in a tissue/organ. Fat emboli most commonly affect the lungs in a trauma setting. This can lead to both significant pathology locally and systemically including changes in structure, inflammatory response, activation of the renin-angiotensin system, and subsequent hypoxia. In fact, changes in skin, brain, lungs, and kidneys have been noted in FE syndrome. Because there is an extensive record of pathology reports on this condition without evidence of direct cardiac involvement, as well as our studies showing apparent complete recovery after the acute embolism, we hypothesized that structural changes similar to the lung and at the same time course would not be observed in the heart. METHODS We used a rat model of FE previously described by our group where we have documented significant lung pathology. In this study, we analyzed both pulmonary and cardiac structure, histology, and gene expression at 48 h and 10 wks post fat injection to mimic FE. RESULTS Despite severe inflammatory evidence and structural changes to the lung and vasculature up to 10 wks after FE, we found no significant alterations to cardiovascular morphometry including lumen patency ratio, adventitia/media ratio, fibrosis content, and heart chamber/wall dimensions in stained histological sections. Additionally, genetic markers of cardiac pathological hypertrophy were not significantly elevated 48 h or 10 wks after fat treatment. Oil Red O staining showed increased fat droplet content within lung and aorta tissue, but not in the myocardium. CONCLUSIONS Our study suggests that, in contrast to the lungs, the heart is more resistant to the inflammatory and remodeling responses that result from FE, possibly due to the organ-specific differences in fat retention.
Collapse
Affiliation(s)
- Shaan Patel
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Rohan Ahuja
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Julian A Vallejo
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Gulnaz Siddiqui
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Jordan Colson
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Joy Edegbe
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Gary Salzman
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Soheila Hamidpour
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | | | - Alan Poisner
- University of Kansas School of Medicine, Kansas City, Kansas
| | - Agostino Molteni
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Michael J Wacker
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri.
| |
Collapse
|
42
|
Xu W, Richmond M. Advances in understanding and managing pediatric heart failure and transplant. Curr Opin Pediatr 2024; 36:489-495. [PMID: 39254752 PMCID: PMC11408753 DOI: 10.1097/mop.0000000000001393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW This article highlights the most recent advances in a review of the current literature in the field of pediatric heart failure and transplantation. RECENT FINDINGS Diagnostically, the identification of new genetic factors has contributed to a deeper understanding of cardiomyopathy in children. Novel medications like sacubitril/valsartan and Sodium-Glucose cotransporter-2 (SGLT2) inhibitors, which are now standard in the adult population are being studied in pediatric population and offer new promise of pediatric heart failure treatment. Ventricular assist devices are more commonly used in cardiomyopathy patients and single ventricle patients as a bridge to transplant. Recent pediatric heart transplant society (PHTS) data demonstrated that waitlist survival improved significantly over the past decades (i) and new treatments such as daratumumab and eculizumab have been used in high-risk populations and demonstrate promising results. TEAMMATE trial is the first multicenter randomized clinical trial (RCT) in pediatric heart transplant (HT) to evaluate the safety and efficacy of everolimus (EVL) and low-dose tacrolimus (TAC) compared to standard-dose TAC and mycophenolate mofetil (MMF). It will provide valuable information about the safety and efficacy of EVL, TAC, and MMF (ii).Donor cell-free DNA has been used more in pediatric transplant recipients and has significantly decreased invasive EMB (iii). SUMMARY This past 5 years have witness dramatic progress in the field of pediatric heart failure and transplantation including more use of mechanical support in heart failure patients with various underlying etiology, especially use of mechanical support in single ventricle patients and the use of sacubitril/valsartan and SGLT2 inhibitors in the pediatric population. The problem of the highly sensitized transplant recipient remains, although novel therapeutics have been added to our toolbox of options to maintain healthy allograft function. Ongoing research aims to further enhance our understanding and management of pediatric heart failure, emphasizing the need for continued innovation in this complex field.
Collapse
Affiliation(s)
- Wenyuan Xu
- Pediatric Advanced Cardiac Care and Transplantation, Division of Pediatric Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
43
|
Jin F, Liu LJ. Mitochondrial abnormalities in septic cardiomyopathy. Minerva Anestesiol 2024; 90:922-930. [PMID: 39051884 DOI: 10.23736/s0375-9393.24.18045-5] [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: 07/27/2024]
Abstract
Septic cardiomyopathy is a common complication in patients with sepsis, and is one of the indicators of poor prognosis. Its pathogenesis is complex, involving calcium ion imbalance in cardiomyocytes, nitric oxide (NO) synthesis disorder, mitochondrial abnormalities and immune inflammatory reaction, especially mitochondrial abnormalities. In this paper, the mechanism of mitochondrial abnormalities causing septic cardiomyopathy was discussed from the aspects of mitochondrial structure change, mitochondrial energy metabolism disorder, redox imbalance, mitochondrial calcium overload, mitochondrial biosynthesis and autophagy abnormalities.
Collapse
Affiliation(s)
- Fang Jin
- Department of Critical Care Medicine, The First People's Hospital of Kunshan, Kunshan, Suzhou, China
| | - Li-Jun Liu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China -
| |
Collapse
|
44
|
Liao L, Wang T, Zhang L, Wei Y, Fan X. Protective Mechanisms of SGLTi in Ischemic Heart Disease. J Cardiovasc Transl Res 2024; 17:1018-1035. [PMID: 38767796 DOI: 10.1007/s12265-024-10513-x] [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: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 05/22/2024]
Abstract
Ischemic heart disease (IHD) is a common clinical cardiovascular disease with high morbidity and mortality. Sodium glucose cotransporter protein inhibitor (SGLTi) is a novel hypoglycemic drug. To date, both clinical trials and animal experiments have shown that SGLTi play a protective role in IHD, including myocardial infarction (MI) and ischemia/reperfusion (I/R). The protective effects may be involved in mechanisms of energy metabolic conversion, anti-inflammation, anti-fibrosis, ionic homeostasis improvement, immune cell development, angiogenesis and functional regulation, gut microbiota regulation, and epicardial lipids. Thus, this review summarizes the above mechanisms and aims to provide theoretical evidence for therapeutic strategies for IHD.
Collapse
Affiliation(s)
- Lei Liao
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tong Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lu Zhang
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Xinrong Fan
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
45
|
Shrestha UM, Chae HD, Fang Q, Lee RJ, Packiasamy J, Huynh L, Blecha J, Huynh TL, VanBrocklin HF, Levi J, Seo Y. A Feasibility Study of [ 18F]F-AraG Positron Emission Tomography (PET) for Cardiac Imaging-Myocardial Viability in Ischemia-Reperfusion Injury Model. Mol Imaging Biol 2024; 26:869-878. [PMID: 39060882 DOI: 10.1007/s11307-024-01932-y] [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: 04/11/2024] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE Myocardial infarction (MI) with subsequent inflammation is one of the most common heart conditions leading to progressive tissue damage. A reliable imaging marker to assess tissue viability after MI would help determine the risks and benefits of any intervention. In this study, we investigate whether a new mitochondria-targeted imaging agent, 18F-labeled 2'-deoxy-2'-18F-fluoro-9-β-d-arabinofuranosylguanine ([18F]F-AraG), a positron emission tomography (PET) agent developed for imaging activated T cells, is suitable for cardiac imaging and to test the myocardial viability after MI. PROCEDURE To test whether the myocardial [18F]-F-AraG signal is coming from cardiomyocytes or immune infiltrates, we compared cardiac signal in wild-type (WT) mice with that of T cell deficient Rag1 knockout (Rag1 KO) mice. We assessed the effect of dietary nucleotides on myocardial [18F]F-AraG uptake in normal heart by comparing [18F]F-AraG signals between mice fed with purified diet and those fed with purified diet supplemented with nucleotides. The myocardial viability was investigated in rodent model by imaging rat with [18F]F-AraG and 2-deoxy-2[18F]fluoro-D-glucose ([18F]FDG) before and after MI. All PET signals were quantified in terms of the percent injected dose per cc (%ID/cc). We also explored [18F]FDG signal variability and potential T cell infiltration into fibrotic area in the affected myocardium with H&E analysis. RESULTS The difference in %ID/cc for Rag1 KO and WT mice was not significant (p = ns) indicating that the [18F]F-AraG signal in the myocardium was primarily coming from cardiomyocytes. No difference in myocardial uptake was observed between [18F]F-AraG signals in mice fed with purified diet and with purified diet supplemented with nucleotides (p = ns). The [18F]FDG signals showed wider variability at different time points. Noticeable [18F]F-AraG signals were observed in the affected MI regions. There were T cells in the fibrotic area in the H&E analysis, but they did not constitute the predominant infiltrates. CONCLUSIONS Our preliminary preclinical data show that [18F]F-AraG accumulates in cardiomyocytes indicating that it may be suitable for cardiac imaging and to evaluate the myocardial viability after MI.
Collapse
Affiliation(s)
- Uttam M Shrestha
- Department of Radiology and Biomedical Imaging, UCSF Physics Research Laboratory, University of California, 185 Berry Street, STE 350, San Francisco, CA, 94143, USA.
| | - Hee-Don Chae
- CellSight Technologies, Inc., 185 Berry Street, STE 350, San Francisco, CA, 94107, USA
| | - Qizhi Fang
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Randall J Lee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Juliet Packiasamy
- CellSight Technologies, Inc., 185 Berry Street, STE 350, San Francisco, CA, 94107, USA
| | - Lyna Huynh
- CellSight Technologies, Inc., 185 Berry Street, STE 350, San Francisco, CA, 94107, USA
| | - Joseph Blecha
- Department of Radiology and Biomedical Imaging, UCSF Physics Research Laboratory, University of California, 185 Berry Street, STE 350, San Francisco, CA, 94143, USA
| | - Tony L Huynh
- Department of Radiology and Biomedical Imaging, UCSF Physics Research Laboratory, University of California, 185 Berry Street, STE 350, San Francisco, CA, 94143, USA
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, UCSF Physics Research Laboratory, University of California, 185 Berry Street, STE 350, San Francisco, CA, 94143, USA
| | - Jelena Levi
- CellSight Technologies, Inc., 185 Berry Street, STE 350, San Francisco, CA, 94107, USA.
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, UCSF Physics Research Laboratory, University of California, 185 Berry Street, STE 350, San Francisco, CA, 94143, USA
| |
Collapse
|
46
|
Sorge M, Savoré G, Gallo A, Acquarone D, Sbroggiò M, Velasco S, Zamporlini F, Femminò S, Moiso E, Morciano G, Balmas E, Raimondi A, Nattenberg G, Stefania R, Tacchetti C, Rizzo AM, Corsetto P, Ghigo A, Turco E, Altruda F, Silengo L, Pinton P, Raffaelli N, Sniadecki NJ, Penna C, Pagliaro P, Hirsch E, Riganti C, Tarone G, Bertero A, Brancaccio M. An intrinsic mechanism of metabolic tuning promotes cardiac resilience to stress. EMBO Mol Med 2024; 16:2450-2484. [PMID: 39271959 PMCID: PMC11473679 DOI: 10.1038/s44321-024-00132-z] [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] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Defining the molecular mechanisms underlying cardiac resilience is crucial to find effective approaches to protect the heart. A physiologic level of ROS is produced in the heart by fatty acid oxidation, but stressful events can boost ROS and cause mitochondrial dysfunction and cardiac functional impairment. Melusin is a muscle specific chaperone required for myocardial compensatory remodeling during stress. Here we report that Melusin localizes in mitochondria where it binds the mitochondrial trifunctional protein, a key enzyme in fatty acid oxidation, and decreases it activity. Studying both mice and human induced pluripotent stem cell-derived cardiomyocytes, we found that Melusin reduces lipid oxidation in the myocardium and limits ROS generation in steady state and during pressure overload and doxorubicin treatment, preventing mitochondrial dysfunction. Accordingly, the treatment with the lipid oxidation inhibitor Trimetazidine concomitantly with stressful stimuli limits ROS accumulation and prevents long-term heart dysfunction. These findings disclose a physiologic mechanism of metabolic regulation in the heart and demonstrate that a timely restriction of lipid metabolism represents a potential therapeutic strategy to improve cardiac resilience to stress.
Collapse
Affiliation(s)
- Matteo Sorge
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
| | - Giulia Savoré
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Andrea Gallo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Davide Acquarone
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Mauro Sbroggiò
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Silvia Velasco
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Enrico Moiso
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara, 44121, Italy
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, 48033, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Andrea Raimondi
- Experimental Imaging Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Gabrielle Nattenberg
- Departments of Mechanical Engineering, Bioengineering, and Laboratory Medicine and Pathology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Rachele Stefania
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Carlo Tacchetti
- Experimental Imaging Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, 20133, Italy
| | - Paola Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, 20133, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Emilia Turco
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Lorenzo Silengo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, 44121, Italy
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, 48033, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60121, Italy
| | - Nathan J Sniadecki
- Departments of Mechanical Engineering, Bioengineering, and Laboratory Medicine and Pathology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Torino, 10126, Italy
| | - Guido Tarone
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
| |
Collapse
|
47
|
Li Y, Zhang H, Yu C, Dong X, Yang F, Wang M, Wen Z, Su M, Li B, Yang L. New Insights into Mitochondria in Health and Diseases. Int J Mol Sci 2024; 25:9975. [PMID: 39337461 PMCID: PMC11432609 DOI: 10.3390/ijms25189975] [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: 08/28/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondria are a unique type of semi-autonomous organelle within the cell that carry out essential functions crucial for the cell's survival and well-being. They are the location where eukaryotic cells carry out energy metabolism. Aside from producing the majority of ATP through oxidative phosphorylation, which provides essential energy for cellular functions, mitochondria also participate in other metabolic processes within the cell, such as the electron transport chain, citric acid cycle, and β-oxidation of fatty acids. Furthermore, mitochondria regulate the production and elimination of ROS, the synthesis of nucleotides and amino acids, the balance of calcium ions, and the process of cell death. Therefore, it is widely accepted that mitochondrial dysfunction is a factor that causes or contributes to the development and advancement of various diseases. These include common systemic diseases, such as aging, diabetes, Parkinson's disease, and cancer, as well as rare metabolic disorders, like Kearns-Sayre syndrome, Leigh disease, and mitochondrial myopathy. This overview outlines the various mechanisms by which mitochondria are involved in numerous illnesses and cellular physiological activities. Additionally, it provides new discoveries regarding the involvement of mitochondria in both disorders and the maintenance of good health.
Collapse
Affiliation(s)
- Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Chunjuan Yu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Xiaolei Dong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Fanghao Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Mengjun Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Ziyuan Wen
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Mohan Su
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| |
Collapse
|
48
|
Zhao J, Han L, Zhang YR, Liu SM, Ji DR, Wang R, Yu YR, Jia MZ, Chai SB, Tang HF, Huang W, Qi YF. Intermedin Alleviates Diabetic Cardiomyopathy by Up-Regulating CPT-1β through Activation of the Phosphatidyl Inositol 3 Kinase/Protein Kinase B Signaling Pathway. Pharmaceuticals (Basel) 2024; 17:1204. [PMID: 39338366 PMCID: PMC11435185 DOI: 10.3390/ph17091204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), one of the most serious long-term consequences of diabetes, is closely associated with myocardial fatty acid metabolism. Carnitine palmitoyltransferase-1β (CPT-1β) is the rate-limiting enzyme responsible for β-oxidation of long-chain fatty acids. Intermedin (IMD) is a pivotal bioactive small molecule peptide, participating in the protection of various cardiovascular diseases. However, the role and underlying mechanisms of IMD in DCM are still unclear. In this study, we investigated whether IMD alleviates DCM via regulating CPT-1β. A rat DCM model was established by having rats to drink fructose water for 12 weeks. A mouse DCM model was induced by feeding mice a high-fat diet for 16 weeks. We showed that IMD and its receptor complexes levels were significantly down-regulated in the cardiac tissues of DCM rats and mice. Reduced expression of IMD was also observed in neonatal rat cardiomyocytes treated with palmitic acid (PA, 300 μM) in vitro. Exogenous and endogenous IMD mitigated cardiac hypertrophy, fibrosis, dysfunction, and lipid accumulation in DCM rats and IMD-transgenic DCM mice, whereas knockout of IMD worsened these pathological processes in IMD-knockout DCM mice. In vitro, IMD alleviated PA-induced cardiomyocyte hypertrophy and cardiac fibroblast activation. We found that CPT-1β enzyme activity, mRNA and protein levels, and acetyl-CoA content were increased in T2DM patients, rats and mice. IMD up-regulated the CPT-1β levels and acetyl-CoA content in T2DM rats and mice. Knockdown of CPT-1β blocked the effects of IMD on increasing acetyl-CoA content and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. IMD receptor antagonist IMD17-47 and the phosphatidyl inositol 3 kinase (PI3K)/protein kinase B (Akt) inhibitor LY294002 reversed the effects of IMD on up-regulating CPT-1β and acetyl-CoA expression and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. We revealed that IMD alleviates DCM by up-regulating CPT-1β via calcitonin receptor-like receptor/receptor activity-modifying protein (CRLR/RAMP) receptor complexes and PI3K/Akt signaling. IMD may serve as a potent therapeutic target for the treatment of DCM.
Collapse
Affiliation(s)
- Jie Zhao
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Ling Han
- Department of Cardiology, Fuxing Hospital, Capital Medical University, Beijing 100038, China
| | - Ya-Rong Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Shi-Meng Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Deng-Ren Ji
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Rui Wang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yan-Rong Yu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Mo-Zhi Jia
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - San-Bao Chai
- Department of Endocrinology and Metabolism, Peking University International Hospital, Beijing 102206, China
| | - Hui-Fang Tang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Wei Huang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| |
Collapse
|
49
|
Joergensen SH, Hansen ESS, Bøgh N, Bertelsen LB, Tougaard RS, Staehr PB, Laustsen C, Wiggers H. Hyperpolarized [1- 13C]pyruvate cardiovascular magnetic resonance imaging identifies metabolic phenotypes in patients with heart failure. J Cardiovasc Magn Reson 2024; 26:101095. [PMID: 39270801 PMCID: PMC11635003 DOI: 10.1016/j.jocmr.2024.101095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Hyperpolarized [1-13C]pyruvate cardiovascular magnetic resonance imaging (HP [1-13C]pyruvate CMR) visualizes key steps in myocardial metabolism. The present study aimed to examine patients with heart failure (HF) using HP [1-13C]pyruvate CMR. METHODS A cross-sectional study of patients with HF and healthy controls using HP [1-13C]pyruvate CMR. Metabolic imaging was obtained using a cardiac-gated spectral-spatial excitation with spiral read-out acquisition. The metabolite signal was analyzed for lactate, bicarbonate, and the alanine signal. Metabolite signal was normalized to the total carbon signal (TC). At the 1-year follow-up, echocardiography was performed in all patients and HP [1-13C]pyruvate MRI in two patients. RESULTS We included six patients with ischemic heart disease (IHD), six with dilated cardiomyopathy, and six healthy controls. In patients, left ventricular ejection fraction (LVEF) correlated with lactate/bicarbonate (r = -0.6, p = 0.03) and lactate/TC (r = -0.7, p = 0.01). In patients with LVEF <30%, lactate/TC was increased (p = 0.01) and bicarbonate/TC reduced (p = 0.03). Circumferential strain correlated with metabolite ratios: lactate/bicarbonate, r = 0.87 (p = 0.0002); lactate/TC, r = 0.85 (p = 0.0005); bicarbonate/TC, r = -0.82 (p = 0.001). In patients with IHD, a strong correlation was found between baseline metabolite ratios and the change in LVEF at follow-up: lactate/bicarbonate (p = 0.001), lactate/TC (p = 0.011), and bicarbonate/TC (p = 0.012). CONCLUSIONS This study highlighted the ability of HP [1-13C]pyruvate CMR to detect changes in metabolism in HF. HP [1-13C]pyruvate CMR has the potential for metabolic phenotyping of patients with HF and for predicting treatment response. TRIAL REGISTRATION EUDRACT, 2018-003533-15. Registered December 4, 2018, https://www.clinicaltrialsregister.eu/ctr-search/search?query=2018-003533-15.
Collapse
Affiliation(s)
- Steen Hylgaard Joergensen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Esben Soevsoe S Hansen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nikolaj Bøgh
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lotte Bonde Bertelsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rasmus Stilling Tougaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Christoffer Laustsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Henrik Wiggers
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
50
|
Tang D, Gu Y, Chen S, Niu T, Zhu J, Liu P, Ding M, Guo Y. Alpha-ketoglutarate is required for chronic hypoxia-induced cardiac remodeling. Am J Physiol Cell Physiol 2024; 327:C728-C736. [PMID: 39069824 DOI: 10.1152/ajpcell.00257.2024] [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: 04/18/2024] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Chronic hypoxia (CH) is commonly associated with various cardiovascular diseases, with cardiac hypertrophy being the most frequently observed alteration. Metabolic remodeling is another consequence seen in the hypoxic heart. However, the mechanistic linkage between metabolic remodeling and cardiac hypertrophy in the hypoxic heart remains unclear. In this study, wild-type C57BL/6J mice were subjected to CH for 4 wk. Echocardiography and morphological analysis were used to assess the cardiac effects. We found that 4 wk of CH led to significant cardiac hypertrophy in the mice, whereas cardiac function remained unchanged compared with normoxic mice. In addition, CH induced an elevation in cardiac alpha-ketoglutarate (α-KG) content. Promoting α-KG degradation in the CH hearts prevented CH-induced cardiac hypertrophy but led to noticeable cardiac dysfunction. Mechanistically, α-KG promoted the transcription of hypertrophy-related genes by regulating histone methylation. Silencing lysine-specific demethylase 5 (KDM5), a histone demethylation enzyme, blunted α-KG-induced transcription of hypertrophy-related genes. These data suggest that α-KG is required for CH-induced cardiac remodeling, thus establishing a connection between metabolic changes and cardiac remodeling in hypoxic hearts.NEW & NOTEWORTHY We reported that alpha-ketoglutarate (α-KG) is indispensable for chronic hypoxia (CH)-induced cardiac remodeling, which builds the bridge between metabolic intermediates and cardiac remodeling.
Collapse
Affiliation(s)
- Daishi Tang
- Digestive System Department, Shaanxi Provincial Crops Hospital of Chinese People's Armed Police Force, Xi'an, People's Republic of China
| | - Yong Gu
- Digestive System Department, Shaanxi Provincial Crops Hospital of Chinese People's Armed Police Force, Xi'an, People's Republic of China
| | - Shasha Chen
- The Second Clinical School of Medicine, Shaanxi University of Chinese Medicine, Xianyang, People's Republic of China
| | - Tong Niu
- Department of Hematology, Xi'an Daxing Hospital, Xi'an, People's Republic of China
| | - Jin'ao Zhu
- The Second Clinical School of Medicine, Shaanxi University of Chinese Medicine, Xianyang, People's Republic of China
| | - Panpan Liu
- Department of Cardiology, Xi'an International Medical Center Hospital, Northwest University, Xi'an, People's Republic of China
| | - Mingge Ding
- Department of Geriatrics Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanjie Guo
- Department of Cardiology, Xi'an International Medical Center Hospital, Northwest University, Xi'an, People's Republic of China
| |
Collapse
|