1
|
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
|
2
|
Hou H, Ji Y, Pan Y, Wang L, Liang Y. Persistent organic pollutants and metabolic diseases: From the perspective of lipid droplets. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124980. [PMID: 39293651 DOI: 10.1016/j.envpol.2024.124980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/12/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024]
Abstract
The characteristic of semi-volatility enables persistent organic pollutants (POPs) almost ubiquitous in the environment. There is increasing concern about the potential risks of exposure to POPs due to their lipophilicity and readily bioaccumulation. Lipid droplets (LDs) are highly dynamic lipid storage organelles, alterations of intracellular LDs play a vital role in the progression of many prevalent metabolic diseases, such as type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD). This article systematically reviewed the biological processes involved in LDs metabolism, the role of LDs proteins and LDs in metabolic diseases, and summarized updating researches on involvement of POPs in the progression of LDs-related metabolic diseases and potential mechanisms. POPs might change the physiological functions of LDs, also interfere the processes of adipogenesis and lipolysis by altering LDs synthesis, decomposition and function. However, further studies are still needed to explore the underlying mechanism of POPs-induced metabolic diseases, which can offer scientific evidences for metabolic disease prevention.
Collapse
Affiliation(s)
- Huixin Hou
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Yaoting Ji
- Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Pan
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Ling Wang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China.
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| |
Collapse
|
3
|
Caliskan B, Inanir M. SPECT myocardial perfusion imaging for the evaluation of left ventricular mechanical dyssynchrony in obese patients. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:1037-1043. [PMID: 39031559 DOI: 10.1002/jcu.23752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 07/22/2024]
Abstract
OBJECTIVE Left ventricular dyssynchrony (LVD), the loss of coordinated contraction in the left ventricle, is an early sign of heart failure. LVD can be assessed using phase analysis techniques with gated single-photon emission computed tomography (SPECT) myocardial perfusion imaging (MPI). This study aimed to investigate the impact of obesity on LVD through phase analysis. METHODS We retrospectively enrolled 152 obese patients and 80 age- and sex-matched nonobese patients who showed normal myocardial perfusion and normal left ventricular ejection fraction (LVEF) on MPI. Phase standard deviation (PSD) and phase histogram bandwidth (PBW), as phase analysis parameters, were compared between patients with and without obesity. RESULTS Although PSD values were within the normal range (cut-off value >23) for both groups, the PSD values of obese patients were higher than those of the nonobese (20.49 ± 8.66 vs. 14.81 ± 4.93; p < 0.05). PBW values of obese patients were statistically significantly higher than those of the nonobese (57.03 ± 23.17 vs. 41.40 ± 9.96; p < 0.05). The PBW values of obese patients exceeded the normal limits (cut-off value >49). A weak positive correlation was observed between body mass index (BMI) and PBW values in obese patients (r = 0.181, p < 0.05). In patients of normal weight, no correlations were found between BMI and phase analysis parameters. CONCLUSION LVD may develop in obese patients, even when myocardial perfusion and ejection fraction are preserved. The use of phase analysis with gated SPECT could be an additional finding improving the early detection of left ventricular dyssynchrony in obese patients.
Collapse
Affiliation(s)
- Billur Caliskan
- Department of Nuclear Medicine, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| | - Mehmet Inanir
- Department of Cardiology, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| |
Collapse
|
4
|
Feldman JM, Frishman WH, Aronow WS. Ninerafaxstat in the Treatment of Diabetic Cardiomyopathy and Nonobstructive Hypertrophic Cardiomyopathy. Cardiol Rev 2024:00045415-990000000-00318. [PMID: 39194232 DOI: 10.1097/crd.0000000000000776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Ninerafaxstat is a novel mitotrope under investigation in 2 large clinical trials: IMPROVE-DiCE (a phase IIa trial investigating ninerafaxstat) and IMPROVE-hypertrophic cardiomyopathy (HCM). IMPROVE-DiCE is a single-center, open-label, phase 2a trial investigating the effectiveness of ninerafaxstat in diabetic cardiomyopathy. Ninerafaxstat significantly improved phosphocreatine/adenosine triphosphate median by 32% (P < 0.01) and reduced myocardial triglyceride content by 34% (P = 0.026). Magnetic resonance imaging (MRI) analysis showed improved left ventricular peak circumferential diastolic strain rate by 15% (P < 0.047) and peak left ventricular filling rate by 11% (P < 0.05). Pyruvate dehydrogenase flux was increased in 7 of 9 patients (P = 0.08), consistent with improved glucose utilization. IMPROVE-HCM (ninerafaxstat safe, effective for nonobstructive hypertrophic cardiomyopathy patients) is a phase 2, multicenter, randomized controlled and double-blinded study. From baseline to 12 weeks, ninerafaxstat was associated with a significantly improved ventilatory efficiency slope compared with placebo (P = 0.006). In a post hoc analysis with 35 patients with baseline Kansas City Cardiomyopathy Questionnaire score ≤80, changes in ventilatory efficiency slope favored ninerafaxstat versus placebo (P = 0.02). Left atrial size, a surrogate marker of diastolic dysfunction, was significantly decreased in patients on ninerafaxstat versus placebo (P = 0.01). These findings support a larger phase 3 study in symptomatic nonobstructive HCM patients to further investigate ninerafaxstat. Several drugs that also improve glucose utilization including fatty acid oxidation inhibitors, carnitine palmitoyltransferase I inhibitors, and glucagon-like peptide-1 receptor agonists are presently under investigation in clinical trials.
Collapse
Affiliation(s)
- Jared M Feldman
- From the Department of Medicine, Long Island Jewish Medical Center and Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY
| | | | - Wilbert S Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
5
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
6
|
Guo D, Zhang M, Qi B, Peng T, Liu M, Li Z, Fu F, Guo Y, Li C, Wang Y, Hu L, Li Y. Lipid overload-induced RTN3 activation leads to cardiac dysfunction by promoting lipid droplet biogenesis. Cell Death Differ 2024; 31:292-308. [PMID: 38017147 PMCID: PMC10923887 DOI: 10.1038/s41418-023-01241-x] [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: 03/07/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Lipid droplet (LD) accumulation is a notable feature of obesity-induced cardiomyopathy, while underlying mechanism remains poorly understood. Here we show that mice fed with high-fat diet (HFD) exhibited significantly increase in cardiac LD and RTN3 expression, accompanied by cardiac function impairment. Multiple loss- and gain-of function experiments indicate that RTN3 is critical to HFD-induced cardiac LD accumulation. Mechanistically, RTN3 directly bonds with fatty acid binding protein 5 (FABP5) to facilitate the directed transport of fatty acids to endoplasmic reticulum, thereby promoting LD biogenesis in a diacylglycerol acyltransferase 2 dependent way. Moreover, lipid overload-induced RTN3 upregulation is due to increased expression of CCAAT/enhancer binding protein α (C/EBPα), which positively regulates RTN3 transcription by binding to its promoter region. Notably, above findings were verified in the myocardium of obese patients. Our findings suggest that manipulating LD biogenesis by modulating RTN3 may be a potential strategy for treating cardiac dysfunction in obese patients.
Collapse
Affiliation(s)
- Dong Guo
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Tingwei Peng
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Mingchuan Liu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Zhelong Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, Airforce Medical University, Xi'an, 710032, China
| | - Yanjie Guo
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, 710100, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Airforce Medical University, 710032, Xi'an, China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China.
| |
Collapse
|
7
|
Wang H, Shen M, Shu X, Guo B, Jia T, Feng J, Lu Z, Chen Y, Lin J, Liu Y, Zhang J, Zhang X, Sun D. Cardiac Metabolism, Reprogramming, and Diseases. J Cardiovasc Transl Res 2024; 17:71-84. [PMID: 37668897 DOI: 10.1007/s12265-023-10432-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular diseases (CVD) account for the largest bulk of deaths worldwide, posing a massive burden on societies and the global healthcare system. Besides, the incidence and prevalence of these diseases are on the rise, demanding imminent action to revert this trend. Cardiovascular pathogenesis harbors a variety of molecular and cellular mechanisms among which dysregulated metabolism is of significant importance and may even proceed other mechanisms. The healthy heart metabolism primarily relies on fatty acids for the ultimate production of energy through oxidative phosphorylation in mitochondria. Other metabolites such as glucose, amino acids, and ketone bodies come next. Under pathological conditions, there is a shift in metabolic pathways and the preference of metabolites, termed metabolic remodeling or reprogramming. In this review, we aim to summarize cardiovascular metabolism and remodeling in different subsets of CVD to come up with a new paradigm for understanding and treatment of these diseases.
Collapse
Affiliation(s)
- Haichang Wang
- Heart Hospital, Xi'an International Medical Center, Xi'an, China
| | - Min Shen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Baolin Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Tengfei Jia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiaxu Feng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zuocheng Lu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yanyan Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
8
|
Qian X, Jia H, Wang J, He S, Yu M, Feng X, Gong Q, An Y, Wang X, Shi N, Li H, Zou Z, Li G, Chen Y. Circulating palmitoyl sphingomyelin levels predict the 10-year increased risk of cardiovascular disease death in Chinese adults: findings from the Da Qing Diabetes Study. Cardiovasc Diabetol 2024; 23:37. [PMID: 38245731 PMCID: PMC10800040 DOI: 10.1186/s12933-023-02116-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/30/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Higher levels of palmitoyl sphingomyelin (PSM, synonymous with sphingomyelin 16:0) are associated with an increased risk of cardiovascular disease (CVD) in people with diabetes. Whether circulating PSM levels can practically predict the long-term risk of CVD and all-cause death remains unclear. This study aimed to investigate whether circulating PSM is a real predictor of CVD death in Chinese adults with or without diabetes. METHODS A total of 286 and 219 individuals with and without diabetes, respectively, from the original Da Qing Diabetes Study were enrolled. Blood samples collected in 2009 were used as a baseline to assess circulating PSM levels. The outcomes of CVD and all-cause death were followed up from 2009 to 2020, and 178 participants died, including 87 deaths due to CVD. Cox proportional hazards regression was used to estimate HRs and their 95% CIs for the outcomes. RESULTS Fractional polynomial regression analysis showed a linear association between baseline circulating PSM concentration (log-2 transformed) and the risk of all-cause and CVD death (p < 0.001), but not non-CVD death (p > 0.05), in all participants after adjustment for confounders. When the participants were stratified by PSM-tertile, the highest tertile, regardless of diabetes, had a higher incidence of CVD death (41.5 vs. 14.7 and 22.2 vs. 2.9 per 1000 person-years in patients with and without diabetes, respectively, all log-rank p < 0.01). Individuals with diabetes in the highest tertile group had a higher risk of CVD death than those in the lowest tertile (HR = 2.73; 95%CI, 1.20-6.22). CONCLUSIONS Elevated PSM levels are significantly associated with a higher 10-year risk of CVD death, but not non-CVD death, in Chinese adults with diabetes. These findings suggest that PSM is a potentially useful long-term predictor of CVD death in individuals with diabetes.
Collapse
Affiliation(s)
- Xin Qian
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongmei Jia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinping Wang
- Department of Cardiology, Da Qing First Hospital, Da Qing, China
| | - Siyao He
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinxing Feng
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiuhong Gong
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yali An
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuan Wang
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Shi
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Li
- Department of Cardiology, Da Qing First Hospital, Da Qing, China
| | - Zhongmei Zou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Guangwei Li
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China.
| | - Yanyan Chen
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
9
|
Shreya S, Alam MJ, Anupriya, Jaiswal S, Rani V, Jain BP. Lipotoxicity, ER Stress, and Cardiovascular Disease: Current Understanding and Future Directions. Cardiovasc Hematol Agents Med Chem 2024; 22:319-335. [PMID: 37859305 DOI: 10.2174/0118715257262366230928051902] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
The endoplasmic reticulum (ER) is a sub-cellular organelle that is responsible for the correct folding of proteins, lipid biosynthesis, calcium storage, and various post-translational modifications. In the disturbance of ER functioning, unfolded or misfolded proteins accumulate inside the ER lumen and initiate downstream signaling called unfolded protein response (UPR). The UPR signaling pathway is involved in lipolysis, triacylglycerol synthesis, lipogenesis, the mevalonate pathway, and low-density lipoprotein receptor recycling. ER stress also affects lipid metabolism by changing the levels of enzymes that are involved in the synthesis or modifications of lipids and causing lipotoxicity. Lipid metabolism and cardiac diseases are in close association as the deregulation of lipid metabolism leads to the development of various cardiovascular diseases (CVDs). Several studies have suggested that lipotoxicity is one of the important factors for cardiovascular disorders. In this review, we will discuss how ER stress affects lipid metabolism and their interplay in the development of cardiovascular disorders. Further, the current therapeutics available to target ER stress and lipid metabolism in various CVDs will be summarized.
Collapse
Affiliation(s)
- Smriti Shreya
- Gene Expression and Signaling lab, Department of Zoology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Md Jahangir Alam
- Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Anupriya
- Gene Expression and Signaling lab, Department of Zoology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Saumya Jaiswal
- Gene Expression and Signaling lab, Department of Zoology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Vibha Rani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Buddhi Prakash Jain
- Gene Expression and Signaling lab, Department of Zoology, Mahatma Gandhi Central University, Motihari, Bihar, India
| |
Collapse
|
10
|
Pedersen LN, Valenzuela Ripoll C, Ozcan M, Guo Z, Lotfinaghsh A, Zhang S, Ng S, Weinheimer C, Nigro J, Kovacs A, Diab A, Klaas A, Grogan F, Cho Y, Ataran A, Luehmann H, Heck A, Kolb K, Strong L, Navara R, Walls GM, Hugo G, Samson P, Cooper D, Reynoso FJ, Schwarz JK, Moore K, Lavine K, Rentschler SL, Liu Y, Woodard PK, Robinson C, Cuculich PS, Bergom C, Javaheri A. Cardiac radiation improves ventricular function in mice and humans with cardiomyopathy. MED 2023; 4:928-943.e5. [PMID: 38029754 PMCID: PMC10994563 DOI: 10.1016/j.medj.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Rapidly dividing cells are more sensitive to radiation therapy (RT) than quiescent cells. In the failing myocardium, macrophages and fibroblasts mediate collateral tissue injury, leading to progressive myocardial remodeling, fibrosis, and pump failure. Because these cells divide more rapidly than cardiomyocytes, we hypothesized that macrophages and fibroblasts would be more susceptible to lower doses of radiation and that cardiac radiation could therefore attenuate myocardial remodeling. METHODS In three independent murine heart failure models, including models of metabolic stress, ischemia, and pressure overload, mice underwent 5 Gy cardiac radiation or sham treatment followed by echocardiography. Immunofluorescence, flow cytometry, and non-invasive PET imaging were employed to evaluate cardiac macrophages and fibroblasts. Serial cardiac magnetic resonance imaging (cMRI) from patients with cardiomyopathy treated with 25 Gy cardiac RT for ventricular tachycardia (VT) was evaluated to determine changes in cardiac function. FINDINGS In murine heart failure models, cardiac radiation significantly increased LV ejection fraction and reduced end-diastolic volume vs. sham. Radiation resulted in reduced mRNA abundance of B-type natriuretic peptide and fibrotic genes, and histological assessment of the LV showed reduced fibrosis. PET and flow cytometry demonstrated reductions in pro-inflammatory macrophages, and immunofluorescence demonstrated reduced proliferation of macrophages and fibroblasts with RT. In patients who were treated with RT for VT, cMRI demonstrated decreases in LV end-diastolic volume and improvements in LV ejection fraction early after treatment. CONCLUSIONS These results suggest that 5 Gy cardiac radiation attenuates cardiac remodeling in mice and humans with heart failure. FUNDING NIH, ASTRO, AHA, Longer Life Foundation.
Collapse
Affiliation(s)
- Lauren N Pedersen
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | | | - Mualla Ozcan
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Zhen Guo
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Aynaz Lotfinaghsh
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Shiyang Zhang
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sherwin Ng
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Carla Weinheimer
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jessica Nigro
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ahmed Diab
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Amanda Klaas
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Felicia Grogan
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yoonje Cho
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Anahita Ataran
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Hannah Luehmann
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Abigail Heck
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kollin Kolb
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Lori Strong
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Rachita Navara
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Gerard M Walls
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT97AE, Northern Ireland
| | - Geoff Hugo
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Pamela Samson
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Cooper
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Francisco J Reynoso
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kaitlin Moore
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kory Lavine
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Stacey L Rentschler
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yongjian Liu
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Pamela K Woodard
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Clifford Robinson
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Phillip S Cuculich
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Carmen Bergom
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Ali Javaheri
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; John J. Cochran Veterans Affairs Medical Center, St. Louis, MO 63106, USA.
| |
Collapse
|
11
|
Ozkan B, Ndumele CE. Exploring the Mechanistic Link Between Obesity and Heart Failure. Curr Diab Rep 2023; 23:347-360. [PMID: 38100052 DOI: 10.1007/s11892-023-01526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 01/14/2024]
Abstract
PURPOSE OF REVIEW Among subtypes of cardiovascular disease, obesity has a potent and unique association with heart failure (HF) that is unexplained by traditional cardiovascular risk mediators. The concomitant rise in the prevalence of obesity and HF necessitates better understanding of their relationship to develop effective prevention and treatment strategies. The purpose of this review is to provide mechanistic insight regarding the link between obesity and HF by elucidating the direct and indirect pathways linking the two conditions. RECENT FINDINGS Several direct pathophysiologic mechanisms contribute to HF risk in individuals with excess weight, including hemodynamic alterations, neurohormonal activation, hormonal effects of dysfunctional adipose tissue, ectopic fat deposition with resulting lipotoxicity and microvascular dysfunction. Obesity further predisposes to HF indirectly through causal associations with hypertension, dyslipidemia, and most importantly, diabetes via insulin resistance. Low levels of physical activity and fitness further influence HF risk in the context of obesity. These various processes lead to myocardial injury and cardiac remodeling that are reflected by abnormalities in cardiac biomarkers and cardiac function on myocardial imaging. Understanding and addressing obesity-associated HF is a pressing clinical and public health challenge which can be informed by a deeper understanding of the complex pathways linking these two conditions together.
Collapse
Affiliation(s)
- Bige Ozkan
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie, Baltimore, MD, 568, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi E Ndumele
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie, Baltimore, MD, 568, USA.
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
12
|
Dajani AHJ, Liu MB, Olaopa MA, Cao L, Valenzuela-Ripoll C, Davis TJ, Poston MD, Smith EH, Contreras J, Pennino M, Waldmann CM, Hoover DB, Lee JT, Jay PY, Javaheri A, Slavik R, Qu Z, Ajijola OA. Heterogeneous cardiac sympathetic innervation gradients promote arrhythmogenesis in murine dilated cardiomyopathy. JCI Insight 2023; 8:e157956. [PMID: 37815863 PMCID: PMC10721311 DOI: 10.1172/jci.insight.157956] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
Ventricular arrhythmias (VAs) in heart failure are enhanced by sympathoexcitation. However, radiotracer studies of catecholamine uptake in failing human hearts demonstrate a proclivity for VAs in patients with reduced cardiac sympathetic innervation. We hypothesized that this counterintuitive finding is explained by heterogeneous loss of sympathetic nerves in the failing heart. In a murine model of dilated cardiomyopathy (DCM), delayed PET imaging of sympathetic nerve density using the catecholamine analog [11C]meta-Hydroxyephedrine demonstrated global hypoinnervation in ventricular myocardium. Although reduced, sympathetic innervation in 2 distinct DCM models invariably exhibited transmural (epicardial to endocardial) gradients, with the endocardium being devoid of sympathetic nerve fibers versus controls. Further, the severity of transmural innervation gradients was correlated with VAs. Transmural innervation gradients were also identified in human left ventricular free wall samples from DCM versus controls. We investigated mechanisms underlying this relationship by in silico studies in 1D, 2D, and 3D models of failing and normal human hearts, finding that arrhythmogenesis increased as heterogeneity in sympathetic innervation worsened. Specifically, both DCM-induced myocyte electrical remodeling and spatially inhomogeneous innervation gradients synergistically worsened arrhythmogenesis. Thus, heterogeneous innervation gradients in DCM promoted arrhythmogenesis. Restoration of homogeneous sympathetic innervation in the failing heart may reduce VAs.
Collapse
Affiliation(s)
- Al-Hassan J. Dajani
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Michael B. Liu
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Michael A. Olaopa
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Lucian Cao
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | | | - Timothy J. Davis
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Megan D. Poston
- Department of Biomedical Sciences, Quillen College of Medicine, and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Elizabeth H. Smith
- Department of Biomedical Sciences, Quillen College of Medicine, and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Jaime Contreras
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Marissa Pennino
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Christopher M. Waldmann
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Nuclear Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Donald B. Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Jason T. Lee
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | | | - Ali Javaheri
- Washington University School of Medicine, St. Louis, Missouri, USA
- John J. Cochran Veterans Hospital, St. Louis, Missouri, USA
| | - Roger Slavik
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Zhilin Qu
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Program of Excellence, and Department of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
13
|
Monga S, Valkovič L, Myerson SG, Neubauer S, Mahmod M, Rider OJ. Role of Cardiac Energetics in Aortic Stenosis Disease Progression: Identifying the High-risk Metabolic Phenotype. Circ Cardiovasc Imaging 2023; 16:e014863. [PMID: 37847766 PMCID: PMC10581424 DOI: 10.1161/circimaging.122.014863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/01/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Severe aortic stenosis (AS) is associated with left ventricular (LV) hypertrophy and cardiac metabolic alterations with evidence of steatosis and impaired myocardial energetics. Despite this common phenotype, there is an unexplained and wide individual heterogeneity in the degree of hypertrophy and progression to myocardial fibrosis and heart failure. We sought to determine whether the cardiac metabolic state may underpin this variability. METHODS We recruited 74 asymptomatic participants with AS and 13 healthy volunteers. Cardiac energetics were measured using phosphorus spectroscopy to define the myocardial phosphocreatine to adenosine triphosphate ratio. Myocardial lipid content was determined using proton spectroscopy. Cardiac function was assessed by cardiovascular magnetic resonance cine imaging. RESULTS Phosphocreatine/adenosine triphosphate was reduced early and significantly across the LV wall thickness quartiles (Q2, 1.50 [1.21-1.71] versus Q1, 1.64 [1.53-1.94]) with a progressive decline with increasing disease severity (Q4, 1.48 [1.18-1.70]; P=0.02). Myocardial triglyceride content levels were overall higher in all the quartiles with a significant increase seen across the AV pressure gradient quartiles (Q2, 1.36 [0.86-1.98] versus Q1, 1.03 [0.81-1.56]; P=0.034). While all AS groups had evidence of subclinical LV dysfunction with impaired strain parameters, impaired systolic longitudinal strain was related to the degree of energetic impairment (r=0.219; P=0.03). Phosphocreatine/adenosine triphosphate was not only an independent predictor of LV wall thickness (r=-0.20; P=0.04) but also strongly associated with myocardial fibrosis (r=-0.24; P=0.03), suggesting that metabolic changes play a role in disease progression. The metabolic and functional parameters showed comparable results when graded by clinical severity of AS. CONCLUSIONS A gradient of myocardial energetic deficit and steatosis exists across the spectrum of hypertrophied AS hearts, and these metabolic changes precede irreversible LV remodeling and subclinical dysfunction. As such, cardiac metabolism may play an important and potentially causal role in disease progression.
Collapse
Affiliation(s)
- Shveta Monga
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Ladislav Valkovič
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia (L.V.)
| | - Saul G. Myerson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Oliver J. Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| |
Collapse
|
14
|
Lewandowski ED. Metabolic flux in the driver's seat during cardiac health and disease. J Mol Cell Cardiol 2023; 182:15-24. [PMID: 37451081 PMCID: PMC10529670 DOI: 10.1016/j.yjmcc.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Cardiac function is a dynamic process that must adjust efficiently to the immediate demands of physical state and activity. So too, the metabolic support of cardiac function is a dynamic process that must respond, in time, to the demands of cardiac function and viability. Flux through metabolic pathways provides chemical energy and generates signaling molecules that regulate activity among intracellular compartments to meet these demands. Thus, flux through metabolic pathways provides a dynamic mode of support of cardiomyocytes during physiological and pathophysiological challenges. Any inability of metabolic flux to keep pace with the demands of the cardiomyocyte results in progressive dysfunction that contributes to cardiac disease. Thus, the priority in maintaining and regulating flux through metabolic pathways in the cardiomyocyte cannot be understated. Great potential exists in current efforts to elucidate metabolic mechanisms as therapeutic targets for the diseased heart. As a consequence, detecting metabolic flux in the functioning myocardium of the heart, under normal and diseased conditions, is essential in elucidating the metabolic basis of contractile dysfunction. As a companion to the 2022 ISHR Research Achievement Award lecture, this review examines the use and applications of stable isotope kinetics to quantify metabolic flux through intermediary pathways and the exchange and transport of intermediates across the mitochondrial membrane and sarcolemma of intact functioning hearts in determining how these intracellular events are coordinated to support cardiac function and health. Finally, this work reviews recently demonstrated metabolic defects in diseased hearts and the potential for metabolic alleviation of heart disease.
Collapse
Affiliation(s)
- E Douglas Lewandowski
- Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
15
|
Watson WD, Green PG, Lewis AJ, Arvidsson P, De Maria GL, Arheden H, Heiberg E, Clarke WT, Rodgers CT, Valkovič L, Neubauer S, Herring N, Rider OJ. Retained Metabolic Flexibility of the Failing Human Heart. Circulation 2023; 148:109-123. [PMID: 37199155 PMCID: PMC10417210 DOI: 10.1161/circulationaha.122.062166] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/01/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND The failing heart is traditionally described as metabolically inflexible and oxygen starved, causing energetic deficit and contractile dysfunction. Current metabolic modulator therapies aim to increase glucose oxidation to increase oxygen efficiency of adenosine triphosphate production, with mixed results. METHODS To investigate metabolic flexibility and oxygen delivery in the failing heart, 20 patients with nonischemic heart failure with reduced ejection fraction (left ventricular ejection fraction 34.9±9.1) underwent separate infusions of insulin+glucose infusion (I+G) or Intralipid infusion. We used cardiovascular magnetic resonance to assess cardiac function and measured energetics using phosphorus-31 magnetic resonance spectroscopy. To investigate the effects of these infusions on cardiac substrate use, function, and myocardial oxygen uptake (MVo2), invasive arteriovenous sampling and pressure-volume loops were performed (n=9). RESULTS At rest, we found that the heart had considerable metabolic flexibility. During I+G, cardiac glucose uptake and oxidation were predominant (70±14% total energy substrate for adenosine triphosphate production versus 17±16% for Intralipid; P=0.002); however, no change in cardiac function was seen relative to basal conditions. In contrast, during Intralipid infusion, cardiac long-chain fatty acid (LCFA) delivery, uptake, LCFA acylcarnitine production, and fatty acid oxidation were all increased (LCFA 73±17% of total substrate versus 19±26% total during I+G; P=0.009). Myocardial energetics were better with Intralipid compared with I+G (phosphocreatine/adenosine triphosphate 1.86±0.25 versus 2.01±0.33; P=0.02), and systolic and diastolic function were improved (LVEF 34.9±9.1 baseline, 33.7±8.2 I+G, 39.9±9.3 Intralipid; P<0.001). During increased cardiac workload, LCFA uptake and oxidation were again increased during both infusions. There was no evidence of systolic dysfunction or lactate efflux at 65% maximal heart rate, suggesting that a metabolic switch to fat did not cause clinically meaningful ischemic metabolism. CONCLUSIONS Our findings show that even in nonischemic heart failure with reduced ejection fraction with severely impaired systolic function, significant cardiac metabolic flexibility is retained, including the ability to alter substrate use to match both arterial supply and changes in workload. Increasing LCFA uptake and oxidation is associated with improved myocardial energetics and contractility. Together, these findings challenge aspects of the rationale underlying existing metabolic therapies for heart failure and suggest that strategies promoting fatty acid oxidation may form the basis for future therapies.
Collapse
Affiliation(s)
- William D. Watson
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Department of Cardiovascular Medicine (W.D.W.), University of Cambridge, UK
| | - Peregrine G. Green
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Department for Physiology, Anatomy and Genetics (P.G.G., N.H.), University of Oxford, UK
| | - Andrew J.M. Lewis
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| | - Per Arvidsson
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | | | - Håkan Arheden
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | - Einar Heiberg
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | - William T. Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences (W.T.C.), University of Oxford, UK
| | | | - Ladislav Valkovič
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Institute of Measurement Science, Slovak Academy of Sciences, Slovakia (L.V.)
| | - Stefan Neubauer
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics (P.G.G., N.H.), University of Oxford, UK
| | - Oliver J. Rider
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| |
Collapse
|
16
|
Guo P, Yao X, Jin X, Xv Y, Zhang J, Li Q, Yan C, Li X, Kim N. Interference with DGAT Gene Inhibited TAG Accumulation and Lipid Droplet Synthesis in Bovine Preadipocytes. Animals (Basel) 2023; 13:2223. [PMID: 37444021 DOI: 10.3390/ani13132223] [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: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Triacylglycerol (TGA) is the primary component of intramuscular fat. Expression of diacylglyceryl transferase (DGAT) determines the polyester differentiation ability of precursor adipocytes. The two DGAT isoforms (DGAT1 and DGAT2) play different roles in TAG metabolism. This study investigates the roles of DGAT1 and DGAT2 in signaling pathways related to differentiation and lipid metabolism in Yanbian bovine preadipocytes. sh-DGAT1 (sh-1), sh-DGAT2 (sh-2), and sh-DGAT1 + sh-DGAT2 (sh-1 + 2) were prepared using short interfering RNA (siRNA) interference technique targeting DGAT1 and DGAT2 genes and infected bovine preadipocytes. Molecular and transcriptomic techniques, including differentially expressed genes (DEGs) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis, were used to investigate the effects on the differentiation of Yanbian bovine preadipocytes. After interference with DGAT1 and DGAT2 genes, the contents of TAG and adiponectin were decreased. The TAG content in the sh-2 and sh-1 + 2 groups was significantly lower than that in the sh-NC group. RNA sequencing (RNA-seq) results showed 2070, 2242, and 2446 DEGs in the sh-1, sh-2, and sh-1 + 2 groups, respectively. The DEGs of the sh-2 group were mainly concentrated in the PPAR, AMPK, and Wnt signaling pathways associated with adipocyte proliferation and differentiation. These results demonstrated that at the mRNA level, DGAT2 plays a more important role in lipid metabolism than DGAT1.
Collapse
Affiliation(s)
- Panpan Guo
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- International Healthcare Innovation Institute, Jiangmen 529020, China
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Xuerui Yao
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- International Healthcare Innovation Institute, Jiangmen 529020, China
| | - Xin Jin
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, China
- Laboratory Animal Center, Yanbian University, Yanji 133002, China
| | - Yongnan Xv
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Junfang Zhang
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Qiang Li
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Changguo Yan
- Yanbian Hongchao Wisdom Animal Husbandry Co., Ltd., Yanji 133002, China
| | - Xiangzi Li
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Namhyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
17
|
Han L, Huang D, Wu S, Liu S, Wang C, Sheng Y, Lu X, Broxmeyer HE, Wan J, Yang L. Lipid droplet-associated lncRNA LIPTER preserves cardiac lipid metabolism. Nat Cell Biol 2023; 25:1033-1046. [PMID: 37264180 PMCID: PMC10344779 DOI: 10.1038/s41556-023-01162-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Lipid droplets (LDs) are cellular organelles critical for lipid homeostasis, with intramyocyte LD accumulation implicated in metabolic disorder-associated heart diseases. Here we identify a human long non-coding RNA, Lipid-Droplet Transporter (LIPTER), essential for LD transport in human cardiomyocytes. LIPTER binds phosphatidic acid and phosphatidylinositol 4-phosphate on LD surface membranes and the MYH10 protein, connecting LDs to the MYH10-ACTIN cytoskeleton and facilitating LD transport. LIPTER and MYH10 deficiencies impair LD trafficking, mitochondrial function and survival of human induced pluripotent stem cell-derived cardiomyocytes. Conditional Myh10 deletion in mouse cardiomyocytes leads to LD accumulation, reduced fatty acid oxidation and compromised cardiac function. We identify NKX2.5 as the primary regulator of cardiomyocyte-specific LIPTER transcription. Notably, LIPTER transgenic expression mitigates cardiac lipotoxicity, preserves cardiac function and alleviates cardiomyopathies in high-fat-diet-fed and Leprdb/db mice. Our findings unveil a molecular connector role of LIPTER in intramyocyte LD transport, crucial for lipid metabolism of the human heart, and hold significant clinical implications for treating metabolic syndrome-associated heart diseases.
Collapse
Affiliation(s)
- Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dayang Huang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shiyong Wu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheng Wang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
18
|
Yang X, Zhang X, Yang Z, Zhang Q, Hao W, Pang Y, Zhang D, Liu D. Transcriptional Regulation Associated with Subcutaneous Adipogenesis in Porcine ACSL1 Gene. Biomolecules 2023; 13:1057. [PMID: 37509093 PMCID: PMC10377008 DOI: 10.3390/biom13071057] [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: 04/24/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) plays an important role in fatty acid metabolism and fat deposition. The transcription of the ACSL1 gene is regulated specifically among cells and physiological processes, and transcriptional regulation of ACSL1 in adipogenesis remains elusive. Here, we characterize transcription factors (TFs) associated with adipogenesis in the porcine ACSL1 gene. CCAAT-enhancer binding protein (C/EBP)α, a well-known adipogenic marker, was found to enhance the expression of the ACSL1 gene via binding two tandem motifs in the promoter. Further, we demonstrate that ACSL1 mediates C/EBPα effects on adipogenesis in preadipocytes cultured from subcutaneous fat tissue of pigs via gain- and loss-of-function analyses. The cAMP-response element binding protein, another TF involved in adipogenesis, was also identified in the regulation of ACSL1 gene expression. Additionally, single nucleotide polymorphisms (SNPs) were screened in the promoter of ACSL1 among four breeds including the Chinese indigenous Min, and Duroc, Berkshire, and Yorkshire pigs through sequencing of PCR products. Two tightly linked SNPs, -517G>T and -311T>G, were found exclusively in Min pigs. The haplotype mutation decreases promoter activity in PK-15 and ST cells, and in vivo the expression of ACSL1, illustrating a possible role in adipogenesis regulated by C/EBPα/ACSL1 axis. Additionally, a total of 24 alternative splicing transcripts were identified, indicating the complexity of alternative splicing in the ACSL1 gene. The results will contribute to further revealing the regulatory mechanisms of ACSL1 during adipogenesis and to the characterization of molecular markers for selection of fat deposition in pigs.
Collapse
Affiliation(s)
- Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiaohan Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Zewei Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Wanjun Hao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yu Pang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Dongjie Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| |
Collapse
|
19
|
Kolleritsch S, Pajed L, Tilp A, Hois V, Pototschnig I, Kien B, Diwoky C, Hoefler G, Schoiswohl G, Haemmerle G. Adverse cardiac remodeling augments adipose tissue ß-adrenergic signaling and lipolysis counteracting diet-induced obesity. J Biol Chem 2023; 299:104788. [PMID: 37150323 PMCID: PMC10318461 DOI: 10.1016/j.jbc.2023.104788] [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: 10/03/2022] [Revised: 03/06/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023] Open
Abstract
Cardiac triacylglycerol accumulation is a common characteristic of obesity and type 2 diabetes and strongly correlates with heart morbidity and mortality. We have previously shown that cardiomyocyte-specific perilipin 5 overexpression (Plin5-Tg) provokes significant cardiac steatosis via lowering cardiac lipolysis and fatty acid (FA) oxidation. In strong contrast to cardiac steatosis and lethal heart dysfunction in adipose triglyceride lipase deficiency, Plin5-Tg mice do not develop heart dysfunction and show a normal life span on chow diet. This finding prompted us to study heart function and energy metabolism in Plin5-Tg mice fed high-fat diet (HFD). Plin5-Tg mice showed adverse cardiac remodeling on HFD with heart function only being compromised in one-year-old mice, likely due to reduced cardiac FA uptake, thereby delaying deleterious cardiac lipotoxicity. Notably, Plin5-Tg mice were less obese and protected from glucose intolerance on HFD. Changes in cardiac energy catabolism in Plin5-Tg mice increased ß-adrenergic signaling, lipolytic, and thermogenic protein expression in adipose tissue ultimately counteracting HFD-induced obesity. Acute cold exposure further augmented ß-adrenergic signaling in Plin5-Tg mice, whereas housing at thermoneutrality did not protect Plin5-Tg mice from HFD-induced obesity albeit blood glucose and insulin levels remained low in transgenic mice. Overall, our data suggest that the limited capacity for myocardial FA oxidation on HFD increases cardiac stress in Plin5-Tg mice, thereby stimulating adipose tissue ß-adrenergic signaling, triacylglycerol catabolism, and thermogenesis. However, long-term HFD-mediated metabolic stress causes contractile dysfunction in Plin5-Tg mice, which emphasizes the importance of a carefully controlled dietary regime in patients with cardiac steatosis and hypertrophy.
Collapse
Affiliation(s)
| | - Laura Pajed
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anna Tilp
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Victoria Hois
- Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | | | - Benedikt Kien
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria
| | - Gabriele Schoiswohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria; Department of Pharmacology and Toxicology, University of Graz, Graz, Austria.
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria.
| |
Collapse
|
20
|
Kong W, Peng Y, Ji C, Liu Z, Gao S, Zhang Y, Chen J, Li X, Bao M, Zhang Y, Jiang Q, Wang F, Li Z, Bian X, Ye J. Akt2 deficiency alleviates oxidative stress in the heart and liver via up-regulating SIRT6 during high-fat diet-induced obesity. Clin Sci (Lond) 2023; 137:823-841. [PMID: 37184210 DOI: 10.1042/cs20230433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/16/2023]
Abstract
The present study aims to investigate the role of AKT2 in the pathogenesis of hepatic and cardiac lipotoxicity induced by lipid overload-induced obesity and identify its downstream targets. WT and Akt2 KO mice were fed either normal diet, or high-fat diet (HFD) to induce obesity model in vivo. Human hepatic cell line (L02 cells) and neonatal rat cardiomyocytes (NRCMs) were used as in vitro models. We observed that during HFD-induced obesity, Akt2 loss-of-function mitigated lipid accumulation and oxidative stress in the liver and heart tissue. Mechanistically, down-regulation of Akt2 promotes SIRT6 expression in L02 cells and NRCMs, the latter deacetylates SOD2, which promotes SOD2 activity and therefore alleviates oxidative stress-induced injury of hepatocytes and cardiomyocytes. Furthermore, we also proved that AKT2 inhibitor protects hepatocytes and cardiomyocytes from HFD-induced oxidative stress. Therefore, our work prove that AKT2 plays an important role in the regulation of obesity-induced lipid metabolic disorder in the liver and heart. Our study also indicates AKT2 inhibitor as a potential therapy for obesity-induced hepatic and cardiac injury.
Collapse
Affiliation(s)
- Weixian Kong
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yue Peng
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Caoyu Ji
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Zekun Liu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Shuya Gao
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Yuexin Zhang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Jiawen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Xie Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Mengmeng Bao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yubin Zhang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Qizhou Jiang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Fuqun Wang
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou 514031, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xiaohong Bian
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Junmei Ye
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| |
Collapse
|
21
|
Cavaliere G, Cimmino F, Trinchese G, Catapano A, Petrella L, D'Angelo M, Lucchin L, Mollica MP. From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs. Antioxidants (Basel) 2023; 12:1172. [PMID: 37371902 DOI: 10.3390/antiox12061172] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Obesity is a major risk factor for several metabolic diseases, including type 2 diabetes, hyperlipidemia, cardiovascular diseases, and brain disorders. Growing evidence suggests the importance of inter-organ metabolic communication for the progression of obesity and the subsequent onset of related disorders. This review provides a broad overview of the pathophysiological processes that from adipose tissue dysfunction leading to altered multi-tissue crosstalk relevant to regulating energy homeostasis and the etiology of obesity. First, a comprehensive description of the role of adipose tissue was reported. Then, attention was turned toward the unhealthy expansion of adipose tissue, low-grade inflammatory state, metabolic inflexibility, and mitochondrial dysfunction as root causes of systemic metabolic alterations. In addition, a short spot was devoted to iron deficiency in obese conditions and the role of the hepcidin-ferroportin relationship in the management of this issue. Finally, different classes of bioactive food components were described with a perspective to enhance their potential preventive and therapeutic use against obesity-related diseases.
Collapse
Affiliation(s)
- Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
| | - Fabiano Cimmino
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Angela Catapano
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Margherita D'Angelo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, 39100 Bolzano, Italy
| | - Maria Pina Mollica
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80138 Naples, Italy
| |
Collapse
|
22
|
Gaebel R, Lang C, Vasudevan P, Lührs L, de Carvalho KAT, Abdelwahid E, David R. New Approaches in Heart Research: Prevention Instead of Cardiomyoplasty? Int J Mol Sci 2023; 24:ijms24109017. [PMID: 37240361 DOI: 10.3390/ijms24109017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death in industrialized nations. Due to the high number of patients and expensive treatments, according to the Federal Statistical Office (2017) in Germany, cardiovascular diseases account for around 15% of total health costs. Advanced coronary artery disease is mainly the result of chronic disorders such as high blood pressure, diabetes, and dyslipidemia. In the modern obesogenic environment, many people are at greater risk of being overweight or obese. The hemodynamic load on the heart is influenced by extreme obesity, which often leads to myocardial infarction (MI), cardiac arrhythmias, and heart failure. In addition, obesity leads to a chronic inflammatory state and negatively affects the wound-healing process. It has been known for many years that lifestyle interventions such as exercise, healthy nutrition, and smoking cessation drastically reduce cardiovascular risk and have a preventive effect against disorders in the healing process. However, little is known about the underlying mechanisms, and there is significantly less high-quality evidence compared to pharmacological intervention studies. Due to the immense potential of prevention in heart research, the cardiologic societies are calling for research work to be intensified, from basic understanding to clinical application. The topicality and high relevance of this research area are also evident from the fact that in March 2018, a one-week conference on this topic with contributions from top international scientists took place as part of the renowned "Keystone Symposia" ("New Insights into the Biology of Exercise"). Consistent with the link between obesity, exercise, and cardiovascular disease, this review attempts to draw lessons from stem-cell transplantation and preventive exercise. The application of state-of-the-art techniques for transcriptome analysis has opened new avenues for tailoring targeted interventions to very individual risk factors.
Collapse
Affiliation(s)
- Ralf Gaebel
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Cajetan Lang
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Praveen Vasudevan
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Larissa Lührs
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Prίncipe Research Institute & Pequeno Prίncipe Faculties, Ave. Silva Jardim, P.O. Box 80240-020, Curitiba 1632, Brazil
| | - Katherine Athayde Teixeira de Carvalho
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Prίncipe Research Institute & Pequeno Prίncipe Faculties, Ave. Silva Jardim, P.O. Box 80240-020, Curitiba 1632, Brazil
| | - Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL 60611, USA
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| |
Collapse
|
23
|
Kovilakath A, Wohlford G, Cowart LA. Circulating sphingolipids in heart failure. Front Cardiovasc Med 2023; 10:1154447. [PMID: 37229233 PMCID: PMC10203217 DOI: 10.3389/fcvm.2023.1154447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
Lack of significant advancements in early detection and treatment of heart failure have precipitated the need for discovery of novel biomarkers and therapeutic targets. Over the past decade, circulating sphingolipids have elicited promising results as biomarkers that premonish adverse cardiac events. Additionally, compelling evidence directly ties sphingolipids to these events in patients with incident heart failure. This review aims to summarize the current literature on circulating sphingolipids in both human cohorts and animal models of heart failure. The goal is to provide direction and focus for future mechanistic studies in heart failure, as well as pave the way for the development of new sphingolipid biomarkers.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - George Wohlford
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Richmond Veteran's Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
24
|
Zhang J, Tian C, Zhu K, Liu Y, Zhao C, Jiang M, Zhu C, Li G. Effects of Natural and Synthetic Astaxanthin on Growth, Body Color, and Transcriptome and Metabolome Profiles in the Leopard Coral Grouper (Plectropomus leopardus). Animals (Basel) 2023; 13:ani13071252. [PMID: 37048508 PMCID: PMC10093260 DOI: 10.3390/ani13071252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Natural and synthetic astaxanthin can promote pigmentation in fish. In this study, the effects of dietary astaxanthin on growth and pigmentation were evaluated in leopard coral grouper (Plectropomus leopardus). Fish were assigned to three groups: 0% astaxanthin (C), 0.02% natural astaxanthin (HP), and 0.02% synthetic astaxanthin (AS). Brightness (L*) was not influenced by astaxanthin. However, redness (a*) and yellowness (b*) were significantly higher for fish fed astaxanthin-containing diets than fish fed control diets and were significantly higher in the HP group than in the AS group. In a transcriptome analysis, 466, 33, and 32 differentially expressed genes (DEGs) were identified between C and HP, C and AS, and AS and HP, including various pigmentation-related genes. DEGs were enriched for carotenoid deposition and other pathways related to skin color. A metabolome analysis revealed 377, 249, and 179 differential metabolites (DMs) between C and HP, C and AS, and AS and HP, respectively. In conclusion, natural astaxanthin has a better coloration effect on P. leopardus, which is more suitable as a red colorant in aquaculture. These results improve our understanding of the effects of natural and synthetic astaxanthin on red color formation in fish.
Collapse
Affiliation(s)
- Junpeng Zhang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Changxu Tian
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Kecheng Zhu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Yong Liu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Can Zhao
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Mouyan Jiang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Chunhua Zhu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Guangli Li
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| |
Collapse
|
25
|
Jung J, Park J, Kim M, Ha J, Cho H, Park SB. SB2301-mediated perturbation of membrane composition in lipid droplets induces lipophagy and lipid droplets ubiquitination. Commun Biol 2023; 6:300. [PMID: 36944894 PMCID: PMC10030462 DOI: 10.1038/s42003-023-04682-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
Lipid droplets (LDs) are involved in various biological events in cells along with their primary role as a storage center for neutral lipids. Excessive accumulation of LDs is highly correlated with various diseases, including metabolic diseases. Therefore, a basic understanding of the molecular mechanism of LD degradation would be beneficial in both academic and industrial research. Lipophagy, a selective autophagy mechanism/LD degradation process, has gained increased attention in the research community. Herein, we sought to elucidate a novel lipophagy mechanism by utilizing the LD-degrading small molecule, SB2301, which activates ubiquitin-mediated lipophagy. Using a label-free target identification method, we revealed that ethanolamine-phosphate cytidylyltransferase 2 (PCYT2) is a potential target protein of SB2301. We also demonstrated that although SB2301 does not modulate PCYT2 function, it induces the cellular translocation of PCYT2 to the LD surface and spatially increases the phosphatidylethanolamine (PE)/phosphatidylcholine (PC) ratio of the LD membrane, causing LD coalescence, leading to the activation of lipophagy process to maintain energy homeostasis.
Collapse
Affiliation(s)
- Jinjoo Jung
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Jongbeom Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Mingi Kim
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Jaeyoung Ha
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea
| | - Hana Cho
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea
| | - Seung Bum Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea.
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
26
|
Oneglia AP, Szczepaniak LS, Jaffery MF, Cipher DJ, McDonald JG, Haykowsky MJ, Moreau KL, Clegg DJ, Zaha V, Nelson MD. Myocardial steatosis impairs left ventricular diastolic-systolic coupling in healthy humans. J Physiol 2023; 601:1371-1382. [PMID: 36891609 DOI: 10.1113/jp284272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Mounting evidence suggests that myocardial steatosis contributes to left ventricular diastolic dysfunction, but definitive evidence in humans is lacking due to confounding comorbidities. As such, we utilized a 48-h food restriction model to acutely increase myocardial triglyceride (mTG) content - measured by 1 H magnetic resonance spectroscopy - in 27 young healthy volunteers (13 men/14 women). Forty-eight hours of fasting caused a more than 3-fold increase in mTG content (P < 0.001). Diastolic function - defined as early diastolic circumferential strain rate (CSRd) - was unchanged following the 48-h fasting intervention, but systolic circumferential strain rate was elevated (P < 0.001), indicative of systolic-diastolic uncoupling. Indeed, in a separate control experiment in 10 individuals, administration of low-dose dobutamine (2 μg/kg/min) caused a similar change in systolic circumferential strain rate as was found during 48 h of food restriction, along with a proportionate increase in CSRd, such that the two metrics remained coupled. Taken together, these data indicate that myocardial steatosis contributes to diastolic dysfunction by impairing diastolic-systolic coupling in healthy adults, and suggest that steatosis may contribute to the progression of heart disease. KEY POINTS: Preclinical evidence strongly suggests that myocardial lipid accumulation (termed steatosis) is an important mechanism driving heart disease. Definitive evidence in humans is limited due to the confounding influence of multiple underlying comorbidities. Using a 48-h food restriction model to acutely increase myocardial triglyceride content in young healthy volunteers, we demonstrate an association between myocardial steatosis and left ventricular diastolic dysfunction. These data advance the hypothesis that myocardial steatosis may contribute to diastolic dysfunction and suggest myocardial steatosis as a putative therapeutic target.
Collapse
Affiliation(s)
- Andrew P Oneglia
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | | | - Manall F Jaffery
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Daisha J Cipher
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mark J Haykowsky
- College of Health Sciences, Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Kerrie L Moreau
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Vlad Zaha
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D Nelson
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
27
|
James AS, Ugbaja RN, Ugwor EI, Thomas FC, Akamo AJ, Akinloye DI, Eteng OE, Salami SK, Emmanuel EA, Ugbaja VC. Lycopene abolishes palmitate-mediated myocardial inflammation in female Wistar rats via modulation of lipid metabolism, NF-κB signalling pathway, and augmenting the antioxidant systems. Nutr Metab Cardiovasc Dis 2023; 33:671-681. [PMID: 36646601 DOI: 10.1016/j.numecd.2022.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Obesity-related heart failure is exacerbated by excessive intake of saturated fats such as palmitate (PA). Lycopene (LYC) possesses anti-lipidemic, antioxidant, cytoprotective, and anti-inflammatory effects. This study, therefore, evaluated the impact of LYC against PA-invoked cardiotoxicity. METHODS AND RESULTS Thirty-six female rats were equally divided into six groups: control; PA (5 mM); PA + LYC (24 mg/kg); PA + LYC (48 mg/kg); LYC (24 mg/kg); and LYC (48 mg/kg). The PA was administered five times weekly for seven weeks, while the LYC was given for the last two weeks. Lipids in the blood and the heart were estimated, as were oxidative stress and antioxidant indices, cardiac function, inflammation, and histology. Palmitate overload occasioned a significant (p < 0.05) increase in cardiac cholesterol (50%), phospholipids (19%), and non-esterified fatty acids (40%). However, triglyceride levels decreased (38%). Furthermore, malondialdehyde (45%), hydrogen peroxide (33%) levels and myeloperoxidase activity increased (79%). Also, cardiac gamma-glutamyl transferase (50%), serum creatine kinase activities (1.34 folds), NF-kB, interleukin1β, and interleukin-6 mRNA expression increased in the PA group relative to the control. In contrast, reduced glutathione (13%) and nitric oxide levels (22%), interleukin-10 mRNA expression, cardiac creatine kinase (35%), lactate dehydrogenase (33%), aspartate, and alanine transaminase activities decreased markedly (15- and 10%, respectively). Also, PA caused hyperemia, congestion of the cardiac interstitium, and infiltration of inflammatory cells. However, treatment with LYC reversed the features of cardiotoxicity and histological complications caused by PA. These observations are likely because LYC has anti-inflammatory, antioxidant, and cytoprotective properties. CONCLUSION Thus, LYC might be an appropriate remedy to manage PA-induced cardiotoxicity in female rats.
Collapse
Affiliation(s)
- Adewale S James
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria; Department of Chemical Sciences (Biochemistry Program), Augustine University Ilara-Epe, P.M.B 1010, Lagos State Nigeria.
| | - Regina N Ugbaja
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Emmanuel I Ugwor
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Funmilola C Thomas
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria, PMB 2240
| | - Adio J Akamo
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Dorcas I Akinloye
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Ofem E Eteng
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Shukurat K Salami
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Esther A Emmanuel
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| | - Victory C Ugbaja
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State, PMB 2240, Nigeria
| |
Collapse
|
28
|
Liu ZY, Liu F, Cao Y, Peng SL, Pan HW, Hong XQ, Zheng PF. ACSL1, CH25H, GPCPD1, and PLA2G12A as the potential lipid-related diagnostic biomarkers of acute myocardial infarction. Aging (Albany NY) 2023; 15:1394-1411. [PMID: 36863716 PMCID: PMC10042701 DOI: 10.18632/aging.204542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Lipid metabolism plays an essential role in the genesis and progress of acute myocardial infarction (AMI). Herein, we identified and verified latent lipid-related genes involved in AMI by bioinformatic analysis. Lipid-related differentially expressed genes (DEGs) involved in AMI were identified using the GSE66360 dataset from the Gene Expression Omnibus (GEO) database and R software packages. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted to analyze lipid-related DEGs. Lipid-related genes were identified by two machine learning techniques: least absolute shrinkage and selection operator (LASSO) regression and support vector machine recursive feature elimination (SVM-RFE). The receiver operating characteristic (ROC) curves were used to descript diagnostic accuracy. Furthermore, blood samples were collected from AMI patients and healthy individuals, and real-time quantitative polymerase chain reaction (RT-qPCR) was used to determine the RNA levels of four lipid-related DEGs. Fifty lipid-related DEGs were identified, 28 upregulated and 22 downregulated. Several enrichment terms related to lipid metabolism were found by GO and KEGG enrichment analyses. After LASSO and SVM-RFE screening, four genes (ACSL1, CH25H, GPCPD1, and PLA2G12A) were identified as potential diagnostic biomarkers for AMI. Moreover, the RT-qPCR analysis indicated that the expression levels of four DEGs in AMI patients and healthy individuals were consistent with bioinformatics analysis results. The validation of clinical samples suggested that 4 lipid-related DEGs are expected to be diagnostic markers for AMI and provide new targets for lipid therapy of AMI.
Collapse
Affiliation(s)
- Zheng-Yu Liu
- Department of Cardiology, Hunan Provincial People's Hospital, Changsha 410000, China
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
| | - Fen Liu
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410000, China
| | - Yan Cao
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
- Department of Emergency, Hunan Provincial People's Hospital, Changsha 410000, China
| | - Shao-Liang Peng
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Data Center, Hunan Provincial People's Hospital, Changsha 410000, China
| | - Hong-Wei Pan
- Department of Cardiology, Hunan Provincial People's Hospital, Changsha 410000, China
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
| | - Xiu-Qin Hong
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410000, China
| | - Peng-Fei Zheng
- Department of Cardiology, Hunan Provincial People's Hospital, Changsha 410000, China
- Department of Epidemiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410000, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha 410000, China
| |
Collapse
|
29
|
Borlaug BA, Jensen MD, Kitzman DW, Lam CSP, Obokata M, Rider OJ. Obesity and heart failure with preserved ejection fraction: new insights and pathophysiological targets. Cardiovasc Res 2023; 118:3434-3450. [PMID: 35880317 PMCID: PMC10202444 DOI: 10.1093/cvr/cvac120] [Citation(s) in RCA: 139] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity and heart failure with preserved ejection fraction (HFpEF) represent two intermingling epidemics driving perhaps the greatest unmet health problem in cardiovascular medicine in the 21st century. Many patients with HFpEF are either overweight or obese, and recent data have shown that increased body fat and its attendant metabolic sequelae have widespread, protean effects systemically and on the cardiovascular system leading to symptomatic HFpEF. The paucity of effective therapies in HFpEF underscores the importance of understanding the distinct pathophysiological mechanisms of obese HFpEF to develop novel therapies. In this review, we summarize the current understanding of the cardiovascular and non-cardiovascular features of the obese phenotype of HFpEF, how increased adiposity might pathophysiologically contribute to the phenotype, and how these processes might be targeted therapeutically.
Collapse
Affiliation(s)
- Barry A Borlaug
- Department of Cardiovascular Diseases, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Dalane W Kitzman
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Masaru Obokata
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Shama S, Jang H, Wang X, Zhang Y, Shahin NN, Motawi TK, Kim S, Gawrieh S, Liu W. Phosphatidylethanolamines Are Associated with Nonalcoholic Fatty Liver Disease (NAFLD) in Obese Adults and Induce Liver Cell Metabolic Perturbations and Hepatic Stellate Cell Activation. Int J Mol Sci 2023; 24:ijms24021034. [PMID: 36674549 PMCID: PMC9861886 DOI: 10.3390/ijms24021034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Pathogenesis roles of phospholipids (PLs) in nonalcoholic fatty liver disease (NAFLD) remain incompletely understood. This study investigated the role of PLs in the progression of NAFLD among obese individuals via studying the alterations in serum PL composition throughout the spectrum of disease progression and evaluating the effects of specific phosphatidylethanolamines (PEs) on FLD development in vitro. A total of 203 obese subjects, who were undergoing bariatric surgery, were included in this study. They were histologically classified into 80 controls (C) with normal liver histology, 93 patients with simple hepatic steatosis (SS), 16 with borderline nonalcoholic steatohepatitis (B-NASH) and 14 with progressive NASH (NASH). Serum PLs were profiled by automated electrospray ionization tandem mass spectrometry (ESI-MS/MS). HepG2 (hepatoma cells) and LX2 (immortalized hepatic stellate cells or HSCs) were used to explore the roles of PL in NAFLD/NASH development. Several PLs and their relative ratios were significantly associated with NAFLD progression, especially those involving PE. Incubation of HepG2 cells with two phosphatidylethanolamines (PEs), PE (34:1) and PE (36:2), resulted in significant inhibition of cell proliferation, reduction of mitochondrial mass and membrane potential, induction of lipid accumulation and mitochondrial ROS production. Meanwhile, treatment of LX2 cells with both PEs markedly increased cell activation and migration. These effects were associated with a significant change in the expression levels of genes involved in lipogenesis, lipid oxidation, autophagy, apoptosis, inflammation, and fibrosis. Thus, our study demonstrated that elevated level of PEs increases susceptibility to the disease progression of obesity associated NAFLD, likely through a causal cascade of impacts on the function of different liver cells.
Collapse
Affiliation(s)
- Samaa Shama
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Cell-Based Analysis Unit, Reference Laboratory, Egyptian Drug Authority, Cairo 12618, Egypt
| | - Hyejeong Jang
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Yang Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Tarek Kamal Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| | - Seongho Kim
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Samer Gawrieh
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| |
Collapse
|
31
|
Li H, Sun X, Li Z, Zhao R, Li M, Hu T. Machine learning-based integration develops biomarkers initial the crosstalk between inflammation and immune in acute myocardial infarction patients. Front Cardiovasc Med 2023; 9:1059543. [PMID: 36684609 PMCID: PMC9846646 DOI: 10.3389/fcvm.2022.1059543] [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: 10/01/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Great strides have been made in past years toward revealing the pathogenesis of acute myocardial infarction (AMI). However, the prognosis did not meet satisfactory expectations. Considering the importance of early diagnosis in AMI, biomarkers with high sensitivity and accuracy are urgently needed. On the other hand, the prevalence of AMI worldwide has rapidly increased over the last few years, especially after the outbreak of COVID-19. Thus, in addition to the classical risk factors for AMI, such as overwork, agitation, overeating, cold irritation, constipation, smoking, and alcohol addiction, viral infections triggers have been considered. Immune cells play pivotal roles in the innate immunosurveillance of viral infections. So, immunotherapies might serve as a potential preventive or therapeutic approach, sparking new hope for patients with AMI. An era of artificial intelligence has led to the development of numerous machine learning algorithms. In this study, we integrated multiple machine learning algorithms for the identification of novel diagnostic biomarkers for AMI. Then, the possible association between critical genes and immune cell infiltration status was characterized for improving the diagnosis and treatment of AMI patients.
Collapse
Affiliation(s)
- Hongyu Li
- Medical College of Soochow University, The People’s Liberation Army of China (PLA) Rocket Force Characteristic Medical Center, Beijing, China,Department of Cardiovascular Medicine, Baotou Central Hospital, Institute of Cardiovascular Diseases, Translational Medicine Center, Baotou, China
| | - Xinti Sun
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zesheng Li
- Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruiping Zhao
- Department of Cardiovascular Medicine, Baotou Central Hospital, Institute of Cardiovascular Diseases, Translational Medicine Center, Baotou, China
| | - Meng Li
- Department of Cardiovascular Medicine, Baotou Central Hospital, Institute of Cardiovascular Diseases, Translational Medicine Center, Baotou, China,*Correspondence: Meng Li,
| | - Taohong Hu
- Medical College of Soochow University, The People’s Liberation Army of China (PLA) Rocket Force Characteristic Medical Center, Beijing, China,Taohong Hu,
| |
Collapse
|
32
|
Ushakov A, Ivanchenko V, Gagarina A. Heart Failure And Type 2 Diabetes Mellitus: Neurohumoral, Histological And Molecular Interconnections. Curr Cardiol Rev 2023; 19:e170622206132. [PMID: 35718961 PMCID: PMC10201898 DOI: 10.2174/1573403x18666220617121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) is a global healthcare burden and a leading cause of morbidity and mortality worldwide. Type 2 diabetes mellitus (T2DM) appears to be one of the major risk factors that significantly worsen HF prognosis and increase the risk of fatal cardiovascular outcomes. Despite a great knowledge of pathophysiological mechanisms involved in HF development and progression, hospitalization rates in patients with HF and concomitant T2DM remain elevated. In this review, we discuss the complex interplay between systemic neurohumoral regulation and local cardiac mechanisms participating in myocardial remodeling and HF development in T2DM with special attention to cardiomyocyte energy metabolism, mitochondrial function and calcium metabolism, cardiomyocyte hypertrophy and death, extracellular matrix remodeling.
Collapse
Affiliation(s)
- A. Ushakov
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - V. Ivanchenko
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - A. Gagarina
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
33
|
Werbner B, Tavakoli-Rouzbehani OM, Fatahian AN, Boudina S. The dynamic interplay between cardiac mitochondrial health and myocardial structural remodeling in metabolic heart disease, aging, and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:9. [PMID: 36742465 PMCID: PMC9894375 DOI: 10.20517/jca.2022.42] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review provides a holistic perspective on the bi-directional relationship between cardiac mitochondrial dysfunction and myocardial structural remodeling in the context of metabolic heart disease, natural cardiac aging, and heart failure. First, a review of the physiologic and molecular drivers of cardiac mitochondrial dysfunction across a range of increasingly prevalent conditions such as metabolic syndrome and cardiac aging is presented, followed by a general review of the mechanisms of mitochondrial quality control (QC) in the heart. Several important mechanisms by which cardiac mitochondrial dysfunction triggers or contributes to structural remodeling of the heart are discussed: accumulated metabolic byproducts, oxidative damage, impaired mitochondrial QC, and mitochondrial-mediated cell death identified as substantial mechanistic contributors to cardiac structural remodeling such as hypertrophy and myocardial fibrosis. Subsequently, the less studied but nevertheless important reverse relationship is explored: the mechanisms by which cardiac structural remodeling feeds back to further alter mitochondrial bioenergetic function. We then provide a condensed pathogenesis of several increasingly important clinical conditions in which these relationships are central: diabetic cardiomyopathy, age-associated declines in cardiac function, and the progression to heart failure, with or without preserved ejection fraction. Finally, we identify promising therapeutic opportunities targeting mitochondrial function in these conditions.
Collapse
Affiliation(s)
- Benjamin Werbner
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Amir Nima Fatahian
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
34
|
Therapeutic Potential of VEGF-B in Coronary Heart Disease and Heart Failure: Dream or Vision? Cells 2022; 11:cells11244134. [PMID: 36552897 PMCID: PMC9776740 DOI: 10.3390/cells11244134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary heart disease (CHD) is the leading cause of death around the world. Based on the roles of vascular endothelial growth factor (VEGF) family members to regulate blood and lymphatic vessels and metabolic functions, several therapeutic approaches have been attempted during the last decade. However proangiogenic therapies based on classical VEGF-A have been disappointing. Therefore, it has become important to focus on other VEGFs such as VEGF-B, which is a novel member of the VEGF family. Recent studies have shown the very promising potential of the VEGF-B to treat CHD and heart failure. The aim of this review article is to present the role of VEGF-B in endothelial biology and as a potential therapeutic agent for CHD and heart failure. In addition, key differences between the VEGF-A and VEGF-B effects on endothelial functions are demonstrated.
Collapse
|
35
|
Cao Y, Yu Y, Zhang L, Liu Y, Zheng K, Wang S, Jin H, Liu L, Cao Y. Transcript variants of long-chain acyl-CoA synthase 1 have distinct roles in sheep lipid metabolism. Front Genet 2022; 13:1021103. [PMID: 36482895 PMCID: PMC9723241 DOI: 10.3389/fgene.2022.1021103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/16/2022] [Indexed: 07/30/2023] Open
Abstract
Mutton has recently been identified to be a consumer favorite, and intermuscular fat is the key factor in determining meat tenderness. Long-chain acyl-CoA synthetase 1 (ACSL1) is a vital subtype of the ACSL family that is involved in the synthesis of lipids from acyl-CoA and the oxidation of fatty acids. The amplification of the ACSL1 gene using rapid amplification of cDNA ends revealed that the alternative polyadenylation (APA) results in two transcripts of the ACSL1 gene. Exon 18 had premature termination, resulting in a shorter CDS region. In this study, the existence of two transcripts of varying lengths translated normally and designated ACSL1-a and ACSL1-b was confirmed. Overexpression of ACSL1-a can promote the synthesis of an intracellular diglyceride, while ACSL1-b can promote triglyceride synthesis. The transfection of ACSL1 shRNA knocks down both the transcripts, the triglyceride content was significantly reduced after differentiation and induction; and lipidome sequencing results exhibited a significant decrease in 14-22 carbon triglyceride metabolites. The results of the present study indicated that the ACSL1 gene played a crucial role in the synthesis of triglycerides. Furthermore, the two transcripts involved in various interactions in the triglyceride synthesis process may be the topic of interest for future research and provide a more theoretical basis for sheep breeding.
Collapse
Affiliation(s)
- Yang Cao
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
- Institute of Animal Husbandry and Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yongsheng Yu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lichun Zhang
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Yu Liu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Kaizhi Zheng
- Institute of Animal Husbandry and Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Sutian Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Haiguo Jin
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lixiang Liu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Yang Cao
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Gongzhuling, China
| |
Collapse
|
36
|
Ke J, Pan J, Lin H, Gu J. Diabetic cardiomyopathy: a brief summary on lipid toxicity. ESC Heart Fail 2022; 10:776-790. [PMID: 36369594 PMCID: PMC10053269 DOI: 10.1002/ehf2.14224] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/30/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes mellitus (DM) is a serious epidemic around the globe, and cardiovascular diseases account for the majority of deaths in patients with DM. Diabetic cardiomyopathy (DCM) is defined as a cardiac dysfunction derived from DM without the presence of coronary artery diseases and hypertension. Patients with either type 1 or type 2 DM are at high risk of developing DCM and even heart failure. Metabolic disorders of obesity and insulin resistance in type 2 diabetic environments result in dyslipidaemia and subsequent lipid-induced toxicity (lipotoxicity) in organs including the heart. Although various mechanisms have been proposed underlying DCM, it remains incompletely understood how lipotoxicity alters cardiac function and how DM induces clinical heart syndrome. With recent progress, we here summarize the latest discoveries on lipid-induced cardiac toxicity in diabetic hearts and discuss the underlying therapies and controversies in clinical DCM.
Collapse
Affiliation(s)
- Jiahan Ke
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jianan Pan
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Hao Lin
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jun Gu
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
37
|
Lim SY, Lim FLS, Criado-Navarro I, Yeo XH, Dayal H, Vemulapalli SD, Seah SJ, Laserna AKC, Yang X, Tan SH, Chan MY, Li SFY. Multi-Omics Investigation into Acute Myocardial Infarction: An Integrative Method Revealing Interconnections amongst the Metabolome, Lipidome, Glycome, and Metallome. Metabolites 2022; 12:1080. [PMID: 36355163 PMCID: PMC9693522 DOI: 10.3390/metabo12111080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Acute myocardial infarction (AMI) is a leading cause of mortality and morbidity worldwide. This work aims to investigate the translational potential of a multi-omics study (comprising metabolomics, lipidomics, glycomics, and metallomics) in revealing biomechanistic insights into AMI. Following the N-glycomics and metallomics studies performed by our group previously, untargeted metabolomic and lipidomic profiles were generated and analysed in this work via the use of a simultaneous metabolite/lipid extraction and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis workflow. The workflow was applied to blood plasma samples from AMI cases (n = 101) and age-matched healthy controls (n = 66). The annotated metabolomic (number of features, n = 27) and lipidomic (n = 48) profiles, along with the glycomic (n = 37) and metallomic (n = 30) profiles of the same set of AMI and healthy samples were integrated and analysed. The integration method used here works by identifying a linear combination of maximally correlated features across the four omics datasets, via utilising both block-partial least squares-discriminant analysis (block-PLS-DA) based on sparse generalised canonical correlation analysis. Based on the multi-omics mapping of biomolecular interconnections, several postulations were derived. These include the potential roles of glycerophospholipids in N-glycan-modulated immunoregulatory effects, as well as the augmentation of the importance of Ca-ATPases in cardiovascular conditions, while also suggesting contributions of phosphatidylethanolamine in their functions. Moreover, it was shown that combining the four omics datasets synergistically enhanced the classifier performance in discriminating between AMI and healthy subjects. Fresh and intriguing insights into AMI, otherwise undetected via single-omics analysis, were revealed in this multi-omics study. Taken together, we provide evidence that a multi-omics strategy may synergistically reinforce and enhance our understanding of diseases.
Collapse
Affiliation(s)
- Si Ying Lim
- NUS Graduate School’s Integrative Sciences & Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Felicia Li Shea Lim
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | | | - Xin Hao Yeo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Hiranya Dayal
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | | | - Song Jie Seah
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Anna Karen Carrasco Laserna
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Central Instrumentation Facility (Laguna Campus), Office of the Vice President for Research and Innovation, De La Salle University, Manila 1004, Philippines
| | - Xiaoxun Yang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Sock Hwee Tan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Mark Y. Chan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Sam Fong Yau Li
- NUS Graduate School’s Integrative Sciences & Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
38
|
Furukawa E, Chen Z, Kubo T, Wu Y, Ueda K, Chelenga M, Chiba H, Yanagawa Y, Katagiri S, Nagano M, Hui SP. Simultaneous free fatty acid elevations and accelerated desaturation in plasma and oocytes in early postpartum dairy cows under intensive feeding management. Theriogenology 2022; 193:20-29. [PMID: 36122530 DOI: 10.1016/j.theriogenology.2022.09.006] [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: 04/05/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
A severe negative energy balance and high circulating free fatty acids (FFA) in postpartum cows impair fertility. The lipotoxicity of FFA has been shown to decrease the quality of bovine oocytes in vitro. Therefore, excess FFA in cells is converted to triacylglycerol (TAG), a non-toxic form, to avoid lipotoxicity. We recently reported that the TAG content in oocytes was higher in postpartum lactating cows subjected to grazing management than in heifers (Theriogenology 176: 174-182, 2021). The present study investigated the compositions of the energy metabolism-related lipids, FFA and TAG, in the plasma and oocytes of cows at different lactation stages under indoor intensive feeding management in order to obtain insights into lipotoxicity in oocytes, particularly those in early postpartum cows. Blood and oocytes were collected from 20 lactating cows categorized into the following lactation groups: 20-30 days in milk (DIM) (n = 5), 40-50 DIM (n = 5), 60-80 DIM (n = 5), and 130-160 DIM (n = 5). Daily energy balance data were obtained for 3 weeks prior to oocyte collection using the ovum pick up (OPU) method. The contents and compositions of FFA and TAG in plasma and oocytes were analyzed using liquid chromatography-mass spectrometry. As expected, plasma FFA was high at 20-30 DIM, decreased by 50 DIM, and was maintained at a low level for the remainder of the experimental period. Similar changes were observed in oocyte FFA and TAG with DIM as plasma FFA. Oocyte FFA positively correlated with plasma FFA (P < 0.05), but negatively correlated with the mean energy balance 1 and 21 days before OPU (P < 0.05). Relationships were noted between the composition and content of FFA in plasma and oocytes, with the FFA 16:1/16:0 and 18:1/18:0 ratios positively correlating with the total amount of FFA (P < 0.05). Elevated oocyte FFA in cows in the early postpartum period under intensive feeding management suggested that oocytes were at a high risk of FFA lipotoxicity. Furthermore, the present results implied that the severe negative energy balance in the previous few weeks was closely related to increases in oocyte FFA, which supports the importance of long-term cow feeding management for preserving the quality of oocytes in the early postpartum period. The present results provide insights into the effects of high circulating FFA on the fertility of postpartum cows.
Collapse
Affiliation(s)
- Eri Furukawa
- Laboratory of Theriogenology, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Zhen Chen
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Tomoaki Kubo
- Dairy Cattle Group, Dairy Research Center, Hokkaido Research Organization, 7, Asahigaoka, Nakashibetsu, Hokkaido, 086-1135, Japan
| | - Yue Wu
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Koichiro Ueda
- Laboratory of Animal Production System, Research Faculty of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-8589, Japan
| | - Madalitso Chelenga
- Laboratory of Theriogenology, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Nakanuma Nishi-4-2-1-15, Higashi-ku, Sapporo, Hokkaido, 007-0894, Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Seiji Katagiri
- Laboratory of Theriogenology, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Masashi Nagano
- Laboratory of Theriogenology, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan; Laboratory of Animal Reproduction, Department of Animal Science, School of Veterinary Medicine, Kitasato University, 35-1, Higashi-23 Bancho, Towada, 034-8628, Japan.
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
39
|
Flam E, Jang C, Murashige D, Yang Y, Morley MP, Jung S, Kantner DS, Pepper H, Bedi KC, Brandimarto J, Prosser BL, Cappola T, Snyder NW, Rabinowitz JD, Margulies KB, Arany Z. Integrated landscape of cardiac metabolism in end-stage human nonischemic dilated cardiomyopathy. NATURE CARDIOVASCULAR RESEARCH 2022; 1:817-829. [PMID: 36776621 PMCID: PMC9910091 DOI: 10.1038/s44161-022-00117-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/08/2022] [Indexed: 01/03/2023]
Abstract
Heart failure (HF) is a leading cause of mortality. Failing hearts undergo profound metabolic changes, but a comprehensive evaluation in humans is lacking. We integrate plasma and cardiac tissue metabolomics of 678 metabolites, genome-wide RNA-sequencing, and proteomic studies to examine metabolic status in 87 explanted human hearts from 39 patients with end-stage HF compared with 48 nonfailing donors. We confirm bioenergetic defects in human HF and reveal selective depletion of adenylate purines required for maintaining ATP levels. We observe substantial reductions in fatty acids and acylcarnitines in failing tissue, despite plasma elevations, suggesting defective import of fatty acids into cardiomyocytes. Glucose levels, in contrast, are elevated. Pyruvate dehydrogenase, which gates carbohydrate oxidation, is de-repressed, allowing increased lactate and pyruvate burning. Tricarboxylic acid cycle intermediates are significantly reduced. Finally, bioactive lipids are profoundly reprogrammed, with marked reductions in ceramides and elevations in lysoglycerophospholipids. These data unveil profound metabolic abnormalities in human failing hearts.
Collapse
Affiliation(s)
- Emily Flam
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Danielle Murashige
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Yifan Yang
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Morley
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Daniel S. Kantner
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hannah Pepper
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kenneth C. Bedi
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeff Brandimarto
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin L. Prosser
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas Cappola
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel W. Snyder
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Joshua D. Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kenneth B. Margulies
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Zolt Arany
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Fiserova I, Trinh MD, Elkalaf M, Vacek L, Heide M, Martinkova S, Bechynska K, Kosek V, Hajslova J, Fiser O, Tousek P, Polak J. Isoprenaline modified the lipidomic profile and reduced β-oxidation in HL-1 cardiomyocytes: In vitro model of takotsubo syndrome. Front Cardiovasc Med 2022; 9:917989. [PMID: 36072861 PMCID: PMC9441769 DOI: 10.3389/fcvm.2022.917989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies have suggested a pathogenetic link between impaired mitochondria and Takotsubo syndrome (TTS), which is closely connected with catecholamine overstimulation, poor outcomes, and changes in lipid metabolism. We investigated the changes in lipid metabolism at the level of fatty acid β-oxidation and changes in the intracellular lipidomic spectrum. The immortalized cell line of HL-1 cardiomyocytes was used in this study as an established in vitro model of TTS. The cells were exposed to the non-selective β-agonist isoprenaline (ISO) for acute (2 h) and prolonged (24 h) periods. We investigated the impact on mitochondrial adenosine 5’-triphosphate (ATP) production and β-oxidation using real-time cell metabolic analysis, total lipid content, and changes in the lipidomic spectrum using high-performance liquid chromatography (HPLC) and mass spectrometry. Furthermore, modifications of selected lipid transporters were determined using real-time – polymerase chain reaction (RT-PCR) and/or Western blot techniques. By choosing this wide range of targets, we provide a detailed overview of molecular changes in lipid metabolism during catecholamine overstimulation. The present study demonstrates that acute exposure to ISO decreased ATP production by up to 42.2%, and prolonged exposure to ISO decreased β-oxidation by 86.4%. Prolonged exposure to ISO also increased lipid accumulation by 4%. Lipid spectrum analysis of prolonged exposure to ISO showed a reduced concentration of cardioprotective and an increased concentration of lipotoxic lipid molecules during long-term exposure. Decreased lipid utilization can lead to higher intracellular lipid accumulation and the formation of lipotoxic molecules. Changes in the lipid spectrum can induce pathophysiological signaling pathways leading to cardiomyocyte remodeling or apoptosis. Thus, changes in lipid metabolism induced by excessive doses of catecholamines may cause TTS and contribute to a progression of heart failure, which is at increased risk after a TTS episode.
Collapse
Affiliation(s)
- Ivana Fiserova
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cardiology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czechia
| | - Minh Duc Trinh
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cardiology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czechia
| | - Moustafa Elkalaf
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Lukas Vacek
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Marek Heide
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Stanislava Martinkova
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Kamila Bechynska
- Department of Food Analysis and Nutrition, University of Chemistry and Technology Prague, Prague, Czechia
| | - Vit Kosek
- Department of Food Analysis and Nutrition, University of Chemistry and Technology Prague, Prague, Czechia
| | - Jana Hajslova
- Department of Food Analysis and Nutrition, University of Chemistry and Technology Prague, Prague, Czechia
| | - Ondrej Fiser
- Department of Biomedical Technology, Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czechia
| | - Petr Tousek
- Department of Cardiology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czechia
| | - Jan Polak
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czechia
- *Correspondence: Jan Polak,
| |
Collapse
|
41
|
Contribution of specific ceramides to obesity-associated metabolic diseases. Cell Mol Life Sci 2022; 79:395. [PMID: 35789435 PMCID: PMC9252958 DOI: 10.1007/s00018-022-04401-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022]
Abstract
Ceramides are a heterogeneous group of bioactive membrane sphingolipids that play specialized regulatory roles in cellular metabolism depending on their characteristic fatty acyl chain lengths and subcellular distribution. As obesity progresses, certain ceramide molecular species accumulate in metabolic tissues and cause cell-type-specific lipotoxic reactions that disrupt metabolic homeostasis and lead to the development of cardiometabolic diseases. Several mechanisms for ceramide action have been inferred from studies in vitro, but only recently have we begun to better understand the acyl chain length specificity of ceramide-mediated signaling in the context of physiology and disease in vivo. New discoveries show that specific ceramides affect various metabolic pathways and that global or tissue-specific reduction in selected ceramide pools in obese rodents is sufficient to improve metabolic health. Here, we review the tissue-specific regulation and functions of ceramides in obesity, thus highlighting the emerging concept of selectively inhibiting production or action of ceramides with specific acyl chain lengths as novel therapeutic strategies to ameliorate obesity-associated diseases.
Collapse
|
42
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
43
|
Pei Y, Song Y, Wang B, Lin C, Yang Y, Li H, Feng Z. Integrated lipidomics and RNA sequencing analysis reveal novel changes during 3T3-L1 cell adipogenesis. PeerJ 2022; 10:e13417. [PMID: 35529487 PMCID: PMC9074861 DOI: 10.7717/peerj.13417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/19/2022] [Indexed: 01/14/2023] Open
Abstract
After adipogenic differentiation, key regulators of adipogenesis are stimulated and cells begin to accumulate lipids. To identify specific changes in lipid composition and gene expression patterns during 3T3-L1 cell adipogenesis, we carried out lipidomics and RNA sequencing analysis of undifferentiated and differentiated 3T3-L1 cells. The analysis revealed significant changes in lipid content and gene expression patterns during adipogenesis. Slc2a4 was up-regulated, which may enhance glucose transport; Gpat3, Agpat2, Lipin1 and Dgat were also up-regulated, potentially to enrich intracellular triacylglycerol (TG). Increased expression levels of Pnpla2, Lipe, Acsl1 and Lpl likely increase intracellular free fatty acids, which can then be used for subsequent synthesis of other lipids, such as sphingomyelin (SM) and ceramide (Cer). Enriched intracellular diacylglycerol (DG) can also provide more raw materials for the synthesis of phosphatidylinositol (PI), phosphatidylcholine (PC), phosphatidylethanolamine (PE), ether-PE, and ether-PC, whereas high expression of Pla3 may enhance the formation of lysophophatidylcholine (LPC) and lysophosphatidylethanolamine (LPE). Therefore, in the process of adipogenesis of 3T3-L1 cells, a series of genes are activated, resulting in large changes in the contents of various lipid metabolites in the cells, especially TG, DG, SM, Cer, PI, PC, PE, etherPE, etherPC, LPC and LPE. These findings provide a theoretical basis for our understanding the pathophysiology of obesity.
Collapse
Affiliation(s)
- Yangli Pei
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yuxin Song
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Bingyuan Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chenghong Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Ying Yang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Feng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
44
|
Zhao M, Wei H, Li C, Zhan R, Liu C, Gao J, Yi Y, Cui X, Shan W, Ji L, Pan B, Cheng S, Song M, Sun H, Jiang H, Cai J, Garcia-Barrio MT, Chen YE, Meng X, Dong E, Wang DW, Zheng L. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat Commun 2022; 13:1757. [PMID: 35365608 PMCID: PMC8976029 DOI: 10.1038/s41467-022-29060-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/14/2022] [Indexed: 12/31/2022] Open
Abstract
Numerous studies found intestinal microbiota alterations which are thought to affect the development of various diseases through the production of gut-derived metabolites. However, the specific metabolites and their pathophysiological contribution to cardiac hypertrophy or heart failure progression still remain unclear. N,N,N-trimethyl-5-aminovaleric acid (TMAVA), derived from trimethyllysine through the gut microbiota, was elevated with gradually increased risk of cardiac mortality and transplantation in a prospective heart failure cohort (n = 1647). TMAVA treatment aggravated cardiac hypertrophy and dysfunction in high-fat diet-fed mice. Decreased fatty acid oxidation (FAO) is a hallmark of metabolic reprogramming in the diseased heart and contributes to impaired myocardial energetics and contractile dysfunction. Proteomics uncovered that TMAVA disturbed cardiac energy metabolism, leading to inhibition of FAO and myocardial lipid accumulation. TMAVA treatment altered mitochondrial ultrastructure, respiration and FAO and inhibited carnitine metabolism. Mice with γ-butyrobetaine hydroxylase (BBOX) deficiency displayed a similar cardiac hypertrophy phenotype, indicating that TMAVA functions through BBOX. Finally, exogenous carnitine supplementation reversed TMAVA induced cardiac hypertrophy. These data suggest that the gut microbiota-derived TMAVA is a key determinant for the development of cardiac hypertrophy through inhibition of carnitine synthesis and subsequent FAO. Intestinal microbiota alterations may affect heart function through the production of gut-derived metabolites. Here the authors found that gut microbiota-derived TMAVA is a key determinant for the development of cardiac hypertrophy through inhibition of carnitine synthesis and subsequent fatty acid oxidation.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenze Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rui Zhan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Changjie Liu
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jianing Gao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Yaodong Yi
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao Cui
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenxin Shan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Si Cheng
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, 100050, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Cai
- Fuwai Hospital, State Key Laboratory of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minerva T Garcia-Barrio
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China. .,Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
45
|
Marwick TH, Gimelli A, Plein S, Bax JJ, Charron P, Delgado V, Donal E, Lancellotti P, Levelt E, Maurovich-Horvat P, Neubauer S, Pontone G, Saraste A, Cosyns B, Edvardsen T, Popescu BA, Galderisi M, Derumeaux G, Bäck M, Bertrand PB, Dweck M, Keenan N, Magne J, Neglia D, Stankovic I. Multimodality imaging approach to left ventricular dysfunction in diabetes: an expert consensus document from the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 2022; 23:e62-e84. [PMID: 34739054 DOI: 10.1093/ehjci/jeab220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 01/14/2023] Open
Abstract
Heart failure (HF) is among the most important and frequent complications of diabetes mellitus (DM). The detection of subclinical dysfunction is a marker of HF risk and presents a potential target for reducing incident HF in DM. Left ventricular (LV) dysfunction secondary to DM is heterogeneous, with phenotypes including predominantly systolic, predominantly diastolic, and mixed dysfunction. Indeed, the pathogenesis of HF in this setting is heterogeneous. Effective management of this problem will require detailed phenotyping of the contributions of fibrosis, microcirculatory disturbance, abnormal metabolism, and sympathetic innervation, among other mechanisms. For this reason, an imaging strategy for the detection of HF risk needs to not only detect subclinical LV dysfunction (LVD) but also characterize its pathogenesis. At present, it is possible to identify individuals with DM at increased risk HF, and there is evidence that cardioprotection may be of benefit. However, there is insufficient justification for HF screening, because we need stronger evidence of the links between the detection of LVD, treatment, and improved outcome. This review discusses the options for screening for LVD, the potential means of identifying the underlying mechanisms, and the pathways to treatment.
Collapse
Affiliation(s)
- Thomas H Marwick
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Alessia Gimelli
- Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, 56124 Pisa, Italy
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Center & Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Phillippe Charron
- Sorbonne Université, INSERM UMRS 1166 and ICAN Institute, Paris, France
- APHP, Centre de référence pour les maladies cardiaques héréditaires ou rares, Hôpital Pitié-Salpêtrière, Paris, France
| | - Victoria Delgado
- Department of Cardiology, Leiden University Medical Centre, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Erwan Donal
- Service de Cardiologie Et Maladies Vasculaires Et CIC-IT 1414, CHU Rennes, 35000 Rennes, France
- Université de Rennes 1, LTSI, 35000 Rennes, France
| | - Patrizio Lancellotti
- Department of Cardiology, University of Liège Hospital, GIGA Cardiovascular Sciences, CHU SartTilman, Liège, Belgium
- Gruppo Villa Maria Care and Research, Maria Cecilia Hospital, Cotignola, and Anthea Hospital, Bari, Italy
| | - Eylem Levelt
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital , Groby Road, Leicester LE3 9QF, UK
| | - Pal Maurovich-Horvat
- MTA-SE Cardiovascular Imaging Research Group, Medical Imaging Centre, Semmelweis University, 2 Koranyi u., 1083 Budapest, Hungary
| | - Stefan Neubauer
- Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Gianluca Pontone
- Centro Cardiologico Monzino IRCCS, University of Milan, Cardiovascular Imaging, Milan, Italy
| | - Antti Saraste
- Turku PET Centre, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
| | - Bernard Cosyns
- Cardiology, CHVZ (Centrum voor Hart en Vaatziekten), ICMI (In Vivo Cellular and Molecular Imaging) Laboratory, Universitair ziekenhuis Brussel, 109 Laarbeeklaan, Brussels 1090, Belgium
| | - Thor Edvardsen
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Postbox 4950 Nydalen, Sognsvannsveien 20, NO-0424 Oslo, Norway
- Institute for clinical medicine, University of Oslo, Sognsvannsveien 20, NO-0424 Oslo, Norway
| | - Bogdan A Popescu
- Department of Cardiology, University of Medicine and Pharmacy "Carol Davila", Euroecolab, Emergency Institute for Cardiovascular Diseases "Prof. Dr. C. C. Iliescu", Bucharest, Romania
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Genevieve Derumeaux
- IMRB - Inserm U955 Senescence, metabolism and cardiovascular diseases 8, rue du Général Sarrail, 94010 Créteil, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Shalaby YM, Al Aidaros A, Valappil A, Ali BR, Akawi N. Role of Ceramides in the Molecular Pathogenesis and Potential Therapeutic Strategies of Cardiometabolic Diseases: What we Know so Far. Front Cell Dev Biol 2022; 9:816301. [PMID: 35127726 PMCID: PMC8808480 DOI: 10.3389/fcell.2021.816301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Ceramides represent a class of biologically active lipids that are involved in orchestrating vital signal transduction pathways responsible for regulating cellular differentiation and proliferation. However, accumulating clinical evidence have shown that ceramides are playing a detrimental role in the pathogenesis of several diseases including cardiovascular disease, type II diabetes and obesity, collectively referred to as cardiometabolic disease. Therefore, it has become necessary to study in depth the role of ceramides in the pathophysiology of such diseases, aiming to tailor more efficient treatment regimens. Furthermore, understanding the contribution of ceramides to the pathological molecular mechanisms of those interrelated conditions may improve not only the therapeutic but also the diagnostic and preventive approaches of the preceding hazardous events. Hence, the purpose of this article is to review currently available evidence on the role of ceramides as a common factor in the pathological mechanisms of cardiometabolic diseases as well as the mechanism of action of the latest ceramides-targeted therapies.
Collapse
Affiliation(s)
- Youssef M Shalaby
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Egypt
| | - Anas Al Aidaros
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Anjana Valappil
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Nadia Akawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
47
|
Tarkhnishvili A, Koentges C, Pfeil K, Gollmer J, Byrne NJ, Vosko I, Lueg J, Vogelbacher L, Birkle S, Tang S, Bon-Nawul Mwinyella T, Hoffmann MM, Odening KE, Michel NA, Wolf D, Stachon P, Hilgendorf I, Wallner M, Ljubojevic-Holzer S, von Lewinski D, Rainer P, Sedej S, Sourij H, Bode C, Zirlik A, Bugger H. Effects of Short Term Adiponectin Receptor Agonism on Cardiac Function and Energetics in Diabetic db/db Mice. J Lipid Atheroscler 2022; 11:161-177. [PMID: 35656151 PMCID: PMC9133777 DOI: 10.12997/jla.2022.11.2.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/01/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Impaired cardiac efficiency is a hallmark of diabetic cardiomyopathy in models of type 2 diabetes. Adiponectin receptor 1 (AdipoR1) deficiency impairs cardiac efficiency in non-diabetic mice, suggesting that hypoadiponectinemia in type 2 diabetes may contribute to impaired cardiac efficiency due to compromised AdipoR1 signaling. Thus, we investigated whether targeting cardiac adiponectin receptors may improve cardiac function and energetics, and attenuate diabetic cardiomyopathy in type 2 diabetic mice. Methods A non-selective adiponectin receptor agonist, AdipoRon, and vehicle were injected intraperitoneally into Eight-week-old db/db or C57BLKS/J mice for 10 days. Cardiac morphology and function were evaluated by echocardiography and working heart perfusions. Results Based on echocardiography, AdipoRon treatment did not alter ejection fraction, left ventricular diameters or left ventricular wall thickness in db/db mice compared to vehicle-treated mice. In isolated working hearts, an impairment in cardiac output and efficiency in db/db mice was not improved by AdipoRon. Mitochondrial respiratory capacity, respiration in the presence of oligomycin, and 4-hydroxynonenal levels were similar among all groups. However, AdipoRon induced a marked shift in the substrate oxidation pattern in db/db mice towards increased reliance on glucose utilization. In parallel, the diabetes-associated increase in serum triglyceride levels in vehicle-treated db/db mice was blunted by AdipoRon treatment, while an increase in myocardial triglycerides in vehicle-treated db/db mice was not altered by AdipoRon treatment. Conclusion AdipoRon treatment shifts myocardial substrate preference towards increased glucose utilization, likely by decreasing fatty acid delivery to the heart, but was not sufficient to improve cardiac output and efficiency in db/db mice.
Collapse
Affiliation(s)
| | - Christoph Koentges
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Katharina Pfeil
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Johannes Gollmer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nikole J Byrne
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Ivan Vosko
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julia Lueg
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Laura Vogelbacher
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Stephan Birkle
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Sibai Tang
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | | | - Michael M Hoffmann
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Center – University of Freiburg, Germany
| | - Katja E Odening
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Nathaly Anto Michel
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dennis Wolf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus Wallner
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Dirk von Lewinski
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Simon Sedej
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Cardiovascular Diabetology Research Group, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Christoph Bode
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Bugger
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Huang W, Gao F, Zhang Y, Chen T, Xu C. Lipid Droplet-Associated Proteins in Cardiomyopathy. ANNALS OF NUTRITION AND METABOLISM 2021; 78:1-13. [PMID: 34856540 DOI: 10.1159/000520122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The heart requires a high rate of fatty-acid oxidation (FAO) to meet its energy needs. Neutral lipids are the main source of energy for the heart and are stored in lipid droplets (LDs), which are cytosolic organelles that primarily serve to store neutral lipids and regulate cellular lipid metabolism. LD-associated proteins (LDAPs) are proteins either located on the surface of the LDs or reside in the cytosol and contribute to lipid metabolism. Therefore, abnormal cardiac lipid accumulation or FAO can alter the redox state of the heart, resulting in cardiomyopathy, a group of diseases that negatively affect the myocardial function, thereby leading to heart failure and even cardiac death. SUMMARY LDs, along with LDAPs, are pivotal for modulating heart lipid homeostasis. The proper cardiac development and the maintenance of its normal function depend largely on lipid homeostasis regulated by LDs and LDAPs. Overexpression or deletion of specific LDAPs can trigger myocardial dysfunction and may contribute to the development of cardiomyopathy. Extensive connections and interactions may also exist between LDAPs. Key Message: In this review, the various mechanisms involved in LDAP-mediated regulation of lipid metabolism, the association between cardiac development and lipid metabolism, as well as the role of LDAPs in cardiomyopathy progression are discussed.
Collapse
Affiliation(s)
- Weiwei Huang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuting Zhang
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tianhui Chen
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Chen Xu
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Li DK, Smith LE, Rookyard AW, Lingam SJ, Koay YC, McEwen HP, Twigg SM, Don AS, O'Sullivan JF, Cordwell SJ, White MY. Multi-omics of a pre-clinical model of diabetic cardiomyopathy reveals increased fatty acid supply impacts mitochondrial metabolic selectivity. J Mol Cell Cardiol 2021; 164:92-109. [PMID: 34826416 DOI: 10.1016/j.yjmcc.2021.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023]
Abstract
The incidence of type 2 diabetes (T2D) is increasing globally, with long-term implications for human health and longevity. Heart disease is the leading cause of death in T2D patients, who display an elevated risk of an acute cardiovascular event and worse outcomes following such an insult. The underlying mechanisms that predispose the diabetic heart to this poor prognosis remain to be defined. This study developed a pre-clinical model (Rattus norvegicus) that complemented caloric excess from a high-fat diet (HFD) and pancreatic β-cell dysfunction from streptozotocin (STZ) to produce hyperglycaemia, peripheral insulin resistance, hyperlipidaemia and elevated fat mass to mimic the clinical features of T2D. Ex vivo cardiac function was assessed using Langendorff perfusion with systolic and diastolic contractile depression observed in T2D hearts. Cohorts representing untreated, individual HFD- or STZ-treatments and the combined HFD + STZ approach were used to generate ventricular samples (n = 9 per cohort) for sequential and integrated analysis of the proteome, lipidome and metabolome by liquid chromatography-tandem mass spectrometry. This study found that in T2D hearts, HFD treatment primed the metabolome, while STZ treatment was the major driver for changes in the proteome. Both treatments equally impacted the lipidome. Our data suggest that increases in β-oxidation and early TCA cycle intermediates promoted rerouting via 2-oxaloacetate to glutamate, γ-aminobutyric acid and glutathione. Furthermore, we suggest that the T2D heart activates networks to redistribute excess acetyl-CoA towards ketogenesis and incomplete β-oxidation through the formation of short-chain acylcarnitine species. Multi-omics provided a global and comprehensive molecular view of the diabetic heart, which distributes substrates and products from excess β-oxidation, reduces metabolic flexibility and impairs capacity to restore high energy reservoirs needed to respond to and prevent subsequent acute cardiovascular events.
Collapse
Affiliation(s)
- Desmond K Li
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia
| | - Lauren E Smith
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia
| | - Alexander W Rookyard
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Life and Environmental Sciences, Camperdown, The University of Sydney, Australia
| | - Shivanjali J Lingam
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia
| | - Yen C Koay
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; Sydney Medical School, The University of Sydney, Camperdown, Australia; Heart Research Institute, Newtown, Australia
| | - Holly P McEwen
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; Centenary Institute, The University of Sydney, Camperdown, Australia
| | - Stephen M Twigg
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; Sydney Medical School, The University of Sydney, Camperdown, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Anthony S Don
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia; Centenary Institute, The University of Sydney, Camperdown, Australia
| | - John F O'Sullivan
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; Sydney Medical School, The University of Sydney, Camperdown, Australia; Heart Research Institute, Newtown, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Stuart J Cordwell
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia; School of Life and Environmental Sciences, Camperdown, The University of Sydney, Australia; Sydney Mass Spectrometry, The University of Sydney, Camperdown, Australia
| | - Melanie Y White
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia; School of Medical Sciences, The University of Sydney, Camperdown, Australia.
| |
Collapse
|
50
|
Relevance of mitochondrial dysfunction in heart disease associated with insulin resistance conditions. Pflugers Arch 2021; 474:21-31. [PMID: 34807312 DOI: 10.1007/s00424-021-02638-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/27/2022]
Abstract
Insulin resistance plays a key role in the development and progression of obesity, diabetes, and their complications. Moreover, insulin resistance is considered the principal link between metabolic diseases and cardiovascular diseases. Heart disease associated with insulin resistance is one of the most important consequences of both obesity and diabetes, and it is characterized by impaired cardiac energetics, diastolic dysfunction, and finally heart failure. Mitochondrion plays a key role in cell energy homeostasis and is the main source of reactive oxygen species. Obesity and diabetes are associated with alterations in mitochondrial function and dynamics. Mitochondrial dysfunction is characterized by changes in mitochondrial respiratory chain with reduced ATP production and elevated reactive oxygen species production. These mitochondrial alterations together with inflammation contribute to the development and progression of heart disease under insulin resistance conditions. Finally, numerous miRNAs participate in the regulation of energy substrate metabolism, reactive oxygen species production, and apoptotic pathways within the mitochondria. This notion supports the relevance of interactions between miRNAs and mitochondrial dysfunction in the pathophysiology of metabolic heart disease.
Collapse
|