1
|
Tanase DM, Valasciuc E, Costea CF, Scripcariu DV, Ouatu A, Hurjui LL, Tarniceriu CC, Floria DE, Ciocoiu M, Baroi LG, Floria M. Duality of Branched-Chain Amino Acids in Chronic Cardiovascular Disease: Potential Biomarkers versus Active Pathophysiological Promoters. Nutrients 2024; 16:1972. [PMID: 38931325 PMCID: PMC11206939 DOI: 10.3390/nu16121972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Branched-chain amino acids (BCAAs), comprising leucine (Leu), isoleucine (Ile), and valine (Val), are essential nutrients vital for protein synthesis and metabolic regulation via specialized signaling networks. Their association with cardiovascular diseases (CVDs) has become a focal point of scientific debate, with emerging evidence suggesting both beneficial and detrimental roles. This review aims to dissect the multifaceted relationship between BCAAs and cardiovascular health, exploring the molecular mechanisms and clinical implications. Elevated BCAA levels have also been linked to insulin resistance (IR), type 2 diabetes mellitus (T2DM), inflammation, and dyslipidemia, which are well-established risk factors for CVD. Central to these processes are key pathways such as mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-light-chain-enhancer of activate B cells (NF-κB)-mediated inflammation, and oxidative stress. Additionally, the interplay between BCAA metabolism and gut microbiota, particularly the production of metabolites like trimethylamine-N-oxide (TMAO), adds another layer of complexity. Contrarily, some studies propose that BCAAs may have cardioprotective effects under certain conditions, contributing to muscle maintenance and metabolic health. This review critically evaluates the evidence, addressing the biological basis and signal transduction mechanism, and also discusses the potential for BCAAs to act as biomarkers versus active mediators of cardiovascular pathology. By presenting a balanced analysis, this review seeks to clarify the contentious roles of BCAAs in CVD, providing a foundation for future research and therapeutic strategies required because of the rising prevalence, incidence, and total burden of CVDs.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iași, Romania
| | - Dragos Viorel Scripcariu
- Department of General Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences II, Physiology Discipline, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Laboratory, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Diana Elena Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Livia Genoveva Baroi
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Vascular Surgery, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
2
|
Zhou Y, Zhu Y, Wu Y, Xiang X, Ouyang X, Liu L, Li T. 4-phenylbutyric acid improves sepsis-induced cardiac dysfunction by modulating amino acid metabolism and lipid metabolism via Comt/Ptgs2/Ppara. Metabolomics 2024; 20:46. [PMID: 38641695 PMCID: PMC11031492 DOI: 10.1007/s11306-024-02112-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/22/2024] [Indexed: 04/21/2024]
Abstract
INTRODUCTION Cardiac dysfunction after sepsis the most common and severe sepsis-related organ failure. The severity of cardiac damage in sepsis patients was positively associated to mortality. It is important to look for drugs targeting sepsis-induced cardiac damage. Our previous studies found that 4-phenylbutyric acid (PBA) was beneficial to septic shock by improving cardiovascular function and survival, while the specific mechanism is unclear. OBJECTIVES We aimed to explore the specific mechanism and PBA for protecting cardiac function in sepsis. METHODS The cecal ligation and puncture-induced septic shock models were used to observe the therapeutic effects of PBA on myocardial contractility and the serum levels of cardiac troponin-T. The mechanisms of PBA against sepsis were explored by metabolomics and network pharmacology. RESULTS The results showed that PBA alleviated the sepsis-induced cardiac damage. The metabolomics results showed that there were 28 metabolites involving in the therapeutic effects of PBA against sepsis. According to network pharmacology, 11 hub genes were found that were involved in lipid metabolism and amino acid transport following PBA treatment. The further integrated analysis focused on 7 key targets, including Comt, Slc6a4, Maoa, Ppara, Pparg, Ptgs2 and Trpv1, as well as their core metabolites and pathways. In an in vitro assay, PBA effectively inhibited sepsis-induced reductions in Comt, Ptgs2 and Ppara after sepsis. CONCLUSIONS PBA protects sepsis-induced cardiac injury by targeting Comt/Ptgs2/Ppara, which regulates amino acid metabolism and lipid metabolism. The study reveals the complicated mechanisms of PBA against sepsis.
Collapse
Affiliation(s)
- Yuanqun Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xinming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xingnan Ouyang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
3
|
Lai P, Zhang L, Qiu Y, Ren J, Sun X, Zhang T, Wang L, Cheng S, Liu S, Zhuang H, Lu D, Zhang S, Liang H, Chen S. Heat stress reduces brown adipose tissue activity by exacerbating mitochondrial damage in type 2 diabetic mice. J Therm Biol 2024; 119:103799. [PMID: 38342042 DOI: 10.1016/j.jtherbio.2024.103799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/13/2024]
Abstract
Epidemiological evidence shows that diabetic patients are susceptible to high temperature weather, and brown adipose tissue (BAT) activity is closely related to type 2 diabetes (T2DM). Activation of BAT under cold stress helps improve T2DM. However, the impact of high temperature on the activity of BAT is still unclear. The study aimed to investigate the impact of heat stress on glucose and lipid metabolism in T2DM mice by influencing BAT activity. High-fat feeding and injecting streptozotocin (STZ) induced model of T2DM mice. All mice were randomly divided into three groups: a normal(N) group, a diabetes (DM) group and a heat stress diabetes (DMHS) group. The DMHS group received heat stress intervention for 3 days. Fasting blood glucose, fasting serum insulin and blood lipids were measured in all three groups. The activity of BAT was assessed by using quantitative real-time PCR (qRT-PCR), electron microscopy, and PET CT. Furthermore, the UHPLC-Q-TOF MS technique was employed to perform metabolomics analysis of BAT on both DM group and DMHS group. The results of this study indicated that heat stress aggravated the dysregulation of glucose and lipid metabolism, exacerbated mitochondrial dysfunction in BAT and reduced the activity of BAT in T2DM mice. This may be related to the abnormal accumulation of branched-chain amino acids (BCAAs) in the mitochondria of BAT.
Collapse
Affiliation(s)
- Penghua Lai
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China; School of Traditional Chinese Medicine, Xiamen University, Malaysia
| | - Linlin Zhang
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Yan Qiu
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Jie Ren
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Xue Sun
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Ting Zhang
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Liuyi Wang
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Sijie Cheng
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Sijia Liu
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Hongli Zhuang
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Xiamen University, China
| | - Daiwei Lu
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Shaoliang Zhang
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China
| | - Huiqing Liang
- Liver Disease Center, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361009, Fujian Province, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Shaodong Chen
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian Province, China; School of Traditional Chinese Medicine, Xiamen University, Malaysia.
| |
Collapse
|
4
|
Chi OZ, Liu X, Magsino J, Weiss HR. Leucine Reduced Blood-Brain Barrier Disruption and Infarct Size in Early Cerebral Ischemia-Reperfusion. Brain Sci 2023; 13:1372. [PMID: 37891741 PMCID: PMC10605042 DOI: 10.3390/brainsci13101372] [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: 08/22/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
A disruption of the blood-brain barrier (BBB) is a crucial pathophysiological change that can impact the outcome of a stroke. Ribosomal protein S6 (S6) and protein kinase B (Akt) play significant roles in early cerebral ischemia-reperfusion injury. Studies have suggested that branched-chain amino acids (BCAAs) may have neuroprotective properties for spinal cord or brain injuries. Therefore, we conducted research to investigate if leucine, one of the BCAAs, could offer neuroprotection and alter BBB disruption, along with its effects on the phosphorylation of S6 and Akt during the early phase of cerebral ischemia-reperfusion, specifically within the thrombolytic therapy time window. In rats, ten min after left middle cerebral artery occlusion (MCAO), 5 µL of 20 mM L-leucine or normal saline was injected into the left lateral ventricle. After two hours of reperfusion following one hour of MCAO, we determined the transfer coefficient (Ki) of 14C-α-aminoisobutyric acid to assess the BBB disruption, infarct size, and phosphorylation of S6 and Akt. Ischemia-reperfusion increased the Ki (+143%, p < 0.001) and the intra-cerebroventricular injection of leucine lowered the Ki in the ischemic-reperfused cortex (-34%, p < 0.001). Leucine reduced the percentage of cortical infarct (-42%, p < 0.0001) out of the total cortical area. Ischemia-reperfusion alone significantly increased the phosphorylation of both S6 and Akt (p < 0.05). However, the administration of leucine had no further effect on the phosphorylation of S6 or Akt in the ischemic-reperfused cortex. This study suggests that an acute increase in leucine levels in the brain during early ischemia-reperfusion within a few hours of stroke may offer neuroprotection, possibly due to reduced BBB disruption being one of the major contributing factors. Leucine did not further increase the already elevated phosphorylation of S6 or Akt by ischemia-reperfusion under the current experimental conditions. Our data warrant further studies on the effects of leucine on neuronal survival and its mechanisms in the later stages of cerebral ischemia-reperfusion.
Collapse
Affiliation(s)
- Oak Z. Chi
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, Suite 3100, New Brunswick, NJ 08901-1977, USA;
| | - Xia Liu
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, Suite 3100, New Brunswick, NJ 08901-1977, USA;
| | - Jedrick Magsino
- Department of Biochemistry and Molecular Biology, Rutgers Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854-8021, USA;
| | - Harvey R. Weiss
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854-8021, USA;
| |
Collapse
|
5
|
Fan H, Liu X, Ren Z, Fei X, Luo J, Yang X, Xue Y, Zhang F, Liang B. Gut microbiota and cardiac arrhythmia. Front Cell Infect Microbiol 2023; 13:1147687. [PMID: 37180433 PMCID: PMC10167053 DOI: 10.3389/fcimb.2023.1147687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
One of the most prevalent cardiac diseases is cardiac arrhythmia, however the underlying causes are not entirely understood. There is a lot of proof that gut microbiota (GM) and its metabolites have a significant impact on cardiovascular health. In recent decades, intricate impacts of GM on cardiac arrythmia have been identified as prospective approaches for its prevention, development, treatment, and prognosis. In this review, we discuss about how GM and its metabolites might impact cardiac arrhythmia through a variety of mechanisms. We proposed to explore the relationship between the metabolites produced by GM dysbiosis including short-chain fatty acids(SCFA), Indoxyl sulfate(IS), trimethylamine N-oxide(TMAO), lipopolysaccharides(LPS), phenylacetylglutamine(PAGln), bile acids(BA), and the currently recognized mechanisms of cardiac arrhythmias including structural remodeling, electrophysiological remodeling, abnormal nervous system regulation and other disease associated with cardiac arrythmia, detailing the processes involving immune regulation, inflammation, and different types of programmed cell death etc., which presents a key aspect of the microbial-host cross-talk. In addition, how GM and its metabolites differ and change in atrial arrhythmias and ventricular arrhythmias populations compared with healthy people are also summarized. Then we introduced potential therapeutic strategies including probiotics and prebiotics, fecal microbiota transplantation (FMT) and immunomodulator etc. In conclusion, the GM has a significant impact on cardiac arrhythmia through a variety of mechanisms, offering a wide range of possible treatment options. The discovery of therapeutic interventions that reduce the risk of cardiac arrhythmia by altering GM and metabolites is a real challenge that lies ahead.
Collapse
Affiliation(s)
- Hongxuan Fan
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xuchang Liu
- Department of Urology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhaoyu Ren
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoning Fei
- Clinical College, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Luo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinyu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaya Xue
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fenfang Zhang
- Department of Cardiology, Yangquan First People’s Hospital, Yangquan, Shanxi, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
The Effect of miR-505-5p on Inhibition of Serum Uromodulin Ameliorates Myocardial Inflammation and Apoptosis Induced by Ischemia-Reperfusion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3521971. [PMID: 36225178 PMCID: PMC9550459 DOI: 10.1155/2022/3521971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/05/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022]
Abstract
Background It has been found that miR-505-5p is closely related to cardiovascular metabolic risk factors. Nonetheless, there is little research analyzing miR-505-5p for its role as well as molecular mechanism in myocardial injury caused by ischemia-reperfusion (I/R). Methods This work utilized quantitative reverse transcriptase PCR (qRT-PCR) for detecting miR-505-5p and serum uromodulin (sUmod) levels. sUmod, interleukin-1beta (IL-1β), IL-6, IL-10, caspase7, caspase9, tumor necrosis factor-alpha (TNF-α), Bax, and Bcl-xL expression was detected by western blot. Bioinformatics database was used for target prediction and miR-505-5's target was determined by luciferase reporter gene assay. Results Relative to sham group, sUmod was highly expressed within myocardial I/R injury (MIRI), whereas sUmod silencing significantly decreased the heart weight/body weight ratio, reduced serum myocardial enzymes expression, ameliorated I/R-mediated myocardial apoptosis, and inflammation. TargetScan bioinformatics database and luciferase reporter genes confirmed that sUmod was miR-505-5p's direct target gene, besides, miR-505-5p overexpression significantly improved the myocardial injury score, increased IL-10, decreased TNF-α, IL-1β, IL-6 expression, decreased caspase7, caspase9, Bax expression, and increased Bcl-xL expression. More importantly, overexpression of sUmod abolished miR-505-5p overexpression's role in I/R-mediated myocardial apoptosis and inflammation. Conclusion miR-505-5p can improve I/R-mediated myocardial apoptosis and inflammation by targeting sUmod. In this study, miR-505-5p is related to MIRI pathogenesis, which provides the new possible targeted therapy in patients with MIRI.
Collapse
|
7
|
Portero V, Nicol T, Podliesna S, Marchal GA, Baartscheer A, Casini S, Tadros R, Treur JL, Tanck MWT, Cox IJ, Probert F, Hough TA, Falcone S, Beekman L, Müller-Nurasyid M, Kastenmüller G, Gieger C, Peters A, Kääb S, Sinner MF, Blease A, Verkerk AO, Bezzina CR, Potter PK, Remme CA. Chronically elevated branched chain amino acid levels are pro-arrhythmic. Cardiovasc Res 2022; 118:1742-1757. [PMID: 34142125 PMCID: PMC9215196 DOI: 10.1093/cvr/cvab207] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/16/2021] [Indexed: 01/03/2023] Open
Abstract
AIMS Cardiac arrhythmias comprise a major health and economic burden and are associated with significant morbidity and mortality, including cardiac failure, stroke, and sudden cardiac death (SCD). Development of efficient preventive and therapeutic strategies is hampered by incomplete knowledge of disease mechanisms and pathways. Our aim is to identify novel mechanisms underlying cardiac arrhythmia and SCD using an unbiased approach. METHODS AND RESULTS We employed a phenotype-driven N-ethyl-N-nitrosourea mutagenesis screen and identified a mouse line with a high incidence of sudden death at young age (6-9 weeks) in the absence of prior symptoms. Affected mice were found to be homozygous for the nonsense mutation Bcat2p.Q300*/p.Q300* in the Bcat2 gene encoding branched chain amino acid transaminase 2. At the age of 4-5 weeks, Bcat2p.Q300*/p.Q300* mice displayed drastic increase of plasma levels of branch chain amino acids (BCAAs-leucine, isoleucine, valine) due to the incomplete catabolism of BCAAs, in addition to inducible arrhythmias ex vivo as well as cardiac conduction and repolarization disturbances. In line with these findings, plasma BCAA levels were positively correlated to electrocardiogram indices of conduction and repolarization in the German community-based KORA F4 Study. Isolated cardiomyocytes from Bcat2p.Q300*/p.Q300* mice revealed action potential (AP) prolongation, pro-arrhythmic events (early and late afterdepolarizations, triggered APs), and dysregulated calcium homeostasis. Incubation of human pluripotent stem cell-derived cardiomyocytes with elevated concentration of BCAAs induced similar calcium dysregulation and pro-arrhythmic events which were prevented by rapamycin, demonstrating the crucial involvement of mTOR pathway activation. CONCLUSIONS Our findings identify for the first time a causative link between elevated BCAAs and arrhythmia, which has implications for arrhythmogenesis in conditions associated with BCAA metabolism dysregulation such as diabetes, metabolic syndrome, and heart failure.
Collapse
Affiliation(s)
- Vincent Portero
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Thomas Nicol
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Svitlana Podliesna
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Gerard A Marchal
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Antonius Baartscheer
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Simona Casini
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute and Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Jorien L Treur
- Department of Psychiatry, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Michael W T Tanck
- Amsterdam UMC, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health (APH), The Netherlands
| | - I Jane Cox
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, Kings College, London, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Tertius A Hough
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Sara Falcone
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Leander Beekman
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Martina Müller-Nurasyid
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, Ludwig Maximilian’s University (LMU) Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Stefan Kääb
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Moritz F Sinner
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Andrew Blease
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Arie O Verkerk
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Connie R Bezzina
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Paul K Potter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Carol Ann Remme
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| |
Collapse
|
8
|
Exercise Enhances Branched-Chain Amino Acid Catabolism and Decreases Cardiac Vulnerability to Myocardial Ischemic Injury. Cells 2022; 11:cells11101706. [PMID: 35626742 PMCID: PMC9139679 DOI: 10.3390/cells11101706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term exercise-induced metabolic adaptations occupy a central position in exercise-afforded cardiac benefits. Emerging evidence suggests that branched-chain amino acid (BCAA) catabolic defect contributes to cardiac dysfunction in multiple cardiometabolic diseases. However, the role of BCAA catabolism in exercise-afforded cardiac benefits remains unknown. Here, we show that exercise improves BCAA catabolism and thus reduce cardiac vulnerability to myocardial ischemic injury. Exercise increased circulating BCAA levels in both humans (male adolescent athletes) and mice (following an 8-week swimming intervention). It increased the expression of mitochondrial localized 2C-type serine-threonine protein phosphatase (PP2Cm), a key enzyme in regulating BCAA catabolism, and decreased BCAA accumulation in mouse hearts, indicating an increase in BCAA catabolism. Pharmacological promotion of BCAA catabolism protected the mouse heart against myocardial infarction (MI) induced by permanent ligation of the left descending coronary artery. Although cardiac-specific PP2Cm knockout showed no significant effects on cardiac structural and functional adaptations to exercise, it blunted the cardioprotective effects of exercise against MI. Mechanistically, exercise alleviated BCAA accumulation and subsequently inactivated the mammalian target of rapamycin in MI hearts. These results showed that exercise elevated BCAA catabolism and protected the heart against myocardial ischemic injury, reinforcing the role of exercise in the promotion of cardiac health.
Collapse
|
9
|
Morio A, Tsutsumi R, Satomi S, Kondo T, Miyoshi H, Kato T, Kuroda M, Kitamura T, Hara K, Saeki N, Sakaue H, Tsutsumi YM. Leucine imparts cardioprotective effects by enhancing mTOR activity and mitochondrial fusion in a myocardial ischemia/reperfusion injury murine model. Diabetol Metab Syndr 2021; 13:139. [PMID: 34801078 PMCID: PMC8606064 DOI: 10.1186/s13098-021-00755-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Coronary artery disease is a leading cause of morbidity and mortality among patients with diabetes. Previously, we demonstrated that branched-chain amino acids (BCAAs) showed cardioprotective effects against cardiac ischemia/reperfusion (I/R) injury. A recent study suggested that leucine (Leu), a BCAA, is a key amino acid involved in mammalian target of rapamycin (mTOR) activity and mitochondrial function. However, whether Leu has cardioprotective effects on diabetic hearts is unclear. In this study, we examined the preconditioning effect of Leu treatment on high-fat diet (HFD)-induced obese mouse which simulate prediabetic heart. METHODS In vivo mice models of I/R injury were divided into the following groups: control, mTOR+/-, and high-fat diet (HFD)-induced obese groups. Mice were randomly administered with Leu, the mTOR inhibitor rapamycin (Rap), or Leu with Rap. Isolated rat cardiomyocytes were subjected to simulated I/R injury. Biochemical and mitochondrial functional assays were performed to evaluate the changes in mTOR activity and mitochondrial dynamics caused by Leu treatment. RESULTS Leu-treated mice showed a significant reduction in infarct size when compared with the control group (34.8% ± 3.8% vs. 43.1% ± 2.4%, n = 7, p < 0.05), whereas Rap-treated mice did not show the protective effects of Leu. This preconditioning effect of Leu was attenuated in mTOR+/- mice. Additionally, Leu increased the percentage of fused mitochondria and the mitochondrial volume, and decreased the number of mitochondria per cell in isolated cardiomyocytes. In HFD-induced obese mice, Leu treatment significantly reduced infarct size (41.0% ± 1.1% vs. 51.0% ± 1.4%, n = 7, p < 0.05), which was not induced by ischemic preconditioning, and this effect was inhibited by Rap. Furthermore, we observed enhanced mTOR protein expression and mitochondrial fusion with decreased reactive oxygen species production with Leu treatment in HFD-induced obese mice, but not in mTOR+/- mice. CONCLUSIONS Leu treatment improved the damage caused by myocardial I/R injury by promoting mTOR activity and mitochondrial fusion on prediabetic hearts in mice.
Collapse
Affiliation(s)
- Atsushi Morio
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Shiho Satomi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Takashi Kondo
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Takahiro Kato
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Tadahiro Kitamura
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan
| | - Kenta Hara
- Kita Harima Medical Center, 926-250 Ichiba, Ono, Hyogo, 675-1392, Japan
| | - Noboru Saeki
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan.
| |
Collapse
|
10
|
Morio A, Tsutsumi R, Kondo T, Miyoshi H, Kato T, Narasaki S, Satomi S, Nakaya E, Kuroda M, Sakaue H, Kitamura T, Tsutsumi YM. Leucine induces cardioprotection in vitro by promoting mitochondrial function via mTOR and Opa-1 signaling. Nutr Metab Cardiovasc Dis 2021; 31:2979-2986. [PMID: 34362635 DOI: 10.1016/j.numecd.2021.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Coronary heart disease is a major global health concern. Further, severity of this condition is greatly influenced by myocardial ischemia/reperfusion (I/R) injury. Branched-chain amino acids (BCAAs) have cardioprotective effects against I/R via mammalian target of rapamycin (mTOR) activity, wherein Leu is considered to particularly regulate mTOR activation. However, the mechanism underlying cardioprotective effects of Leu via mTOR activity is not fully elucidated. Here, we aimed to study the signaling pathway of cardioprotection and mitochondrial function induced by Leu treatment. METHODS AND RESULTS Cardiac myocytes isolated from adult male Wistar rats were incubated and exposed to simulated I/R (SI/R) injury by replacing the air content. Cardiac myocytes were treated with Leu and subsequently, their survival rate was calculated. To elucidate the signaling pathway and mitochondrial function, immunoblots and mitochondrial permeability transition pore were examined. Cell survival rate was decreased with SI/R but improved by 160 μM Leu (38.5 ± 3.6% vs. 64.5 ± 4.2%, respectively, p < 0.001). Although rapamycin (mTOR inhibitor) prevented this cardioprotective effect induced by Leu, wortmannin (PI3K inhibitor) did not interfere with this effect. In addition, we indicated that overexpression of Opa-1 and mitochondrial function are ameliorated via Leu-induced mitochondrial biogenesis. In contrast, knockdown of Opa-1 suppressed Leu-induced cardioprotection. CONCLUSION Leu treatment is critical in rendering a cardioprotective effect exhibited by BCAAs via mTOR signaling. Furthermore, Leu improved mitochondrial function.
Collapse
Affiliation(s)
- Atsushi Morio
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takashi Kondo
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Takahiro Kato
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Soshi Narasaki
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Shiho Satomi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Erika Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tadahiro Kitamura
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
11
|
Supruniuk E, Żebrowska E, Chabowski A. Branched chain amino acids-friend or foe in the control of energy substrate turnover and insulin sensitivity? Crit Rev Food Sci Nutr 2021; 63:2559-2597. [PMID: 34542351 DOI: 10.1080/10408398.2021.1977910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Branched chain amino acids (BCAA) and their derivatives are bioactive molecules with pleiotropic functions in the human body. Elevated fasting blood BCAA concentrations are considered as a metabolic hallmark of obesity, insulin resistance, dyslipidaemia, nonalcoholic fatty liver disease, type 2 diabetes and cardiovascular disease. However, since increased BCAA amount is observed both in metabolically healthy and obese subjects, a question whether BCAA are mechanistic drivers of insulin resistance and its morbidities or only markers of metabolic dysregulation, still remains open. The beneficial effects of BCAA on body weight and composition, aerobic capacity, insulin secretion and sensitivity demand high catabolic potential toward amino acids and/or adequate BCAA intake. On the opposite, BCAA-related inhibition of lipogenesis and lipolysis enhancement may preclude impairment in insulin sensitivity. Thereby, the following review addresses various strategies pertaining to the modulation of BCAA catabolism and the possible roles of BCAA in energy homeostasis. We also aim to elucidate mechanisms behind the heterogeneity of ramifications associated with BCAA modulation.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
12
|
Liu K, Chen Y, Ai F, Li YQ, Zhang K, Zhang WT. PHLDA3 inhibition attenuates endoplasmic reticulum stress-induced apoptosis in myocardial hypoxia/reoxygenation injury by activating the PI3K/AKT signaling pathway. Exp Ther Med 2021; 21:613. [PMID: 33936270 PMCID: PMC8082641 DOI: 10.3892/etm.2021.10045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum stress (ERS)-induced apoptosis serves a crucial role in the pathogenesis of myocardial ischemia/reperfusion injury (MIRI). Previous studies have confirmed that pleckstrin homology-like domain family A member 3 (PHLDA3) is an important mediator in ERS-associated apoptosis. The aim of the current study focused on whether PHLDA3 served protective effects on hypoxia/reoxygenation (H/R)-injured cardiomyocytes by inhibiting ERS-induced apoptosis. Furthermore, the molecular mechanisms associated with the PI3K/AKT signaling pathway were investigated. Primary neonatal rat cardiomyocytes were isolated and randomized into four groups: i) Control + adenovirus encoding scrambled short hairpin RNA (AdshRNA); ii) control + adenoviral vectors encoding PHLDA3 shRNA (AdshPHLDA3); iii) H/R+ AdshRNA and iv) H/R+AdshPHLDA3. AdshPHLDA3 was used to knock down PHLDA3. An H/R injury model was constructed by treatment with hypoxia for 4 h followed by reoxygenation for 6 h. A PI3K/AKT inhibitor, LY294002, was supplemented in mechanistic studies. Cell viability and LDH/CK releases were detected to evaluate myocardial damage. Flow cytometry assays were used to assess apoptotic response. Western blotting assays were used to detect protein expression. The results demonstrated that H/R induced myocardial damage and increased PHLDA3 expression. ERS-induced apoptosis was significantly increased following H/R injury, as indicated by increased apoptotic rates and ERS-associated protein expression, including those of CHOP, 78 kDa glucose-regulated protein and caspase-12. However, PHLDA3 inhibition following AdshPHLDA3 transfection reversed cell damage and ERS-associated apoptosis on H/R injury. Studies for molecular mechanisms concluded that the apoptosis-inhibition effects and cardioprotective roles of PHLDA3 inhibition were induced partly by the activation of the PI3K/AKT pathway, which was verified by LY294002 treatment. In conclusion, in the process of H/R injury, PHLDA3 inhibition reduced ERS-induced apoptosis and H/R injury by activating the PI3K/AKT pathway. PHLDA3 may be a therapeutic target for the treatment of MIRI.
Collapse
Affiliation(s)
- Kai Liu
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Ying Chen
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yun-Qian Li
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Kun Zhang
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Wei-Tong Zhang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
13
|
Sciarretta S, Forte M, Frati G, Sadoshima J. The complex network of mTOR signaling in the heart. Cardiovasc Res 2021; 118:424-439. [PMID: 33512477 DOI: 10.1093/cvr/cvab033] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/13/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) integrates several intracellular and extracellular signals involved in the regulation of anabolic and catabolic processes. mTOR assembles into two macromolecular complexes, named mTORC1 and mTORC2, which have different regulators, substrates and functions. Studies of gain- and loss-of-function animal models of mTOR signaling revealed that mTORC1/2 elicit both adaptive and maladaptive functions in the cardiovascular system. Both mTORC1 and mTORC2 are indispensable for driving cardiac development and cardiac adaption to stress, such as pressure overload. However, persistent and deregulated mTORC1 activation in the heart is detrimental during stress and contributes to the development and progression of cardiac remodeling and genetic and metabolic cardiomyopathies. In this review, we discuss the latest findings regarding the role of mTOR in the cardiovascular system, both under basal conditions and during stress, such as pressure overload, ischemia and metabolic stress. Current data suggest that mTOR modulation may represent a potential therapeutic strategy for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | | | - Giacomo Frati
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
14
|
Watanabe T, Miyoshi H, Noda Y, Narasaki S, Morio A, Toyota Y, Kimura H, Mukaida K, Yasuda T, Tsutsumi YM. Effects of Remimazolam and Propofol on Ca 2+ Regulation by Ryanodine Receptor 1 with Malignant Hyperthermia Mutation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8845129. [PMID: 33490280 PMCID: PMC7801102 DOI: 10.1155/2021/8845129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/21/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND We investigated the potential safety of remimazolam and propofol in malignant hyperthermia- (HM-) susceptible patients using ryanodine receptor 1- (RYR1-) expressing human embryonic kidney- (HEK-) 293 cells. METHODS We compared the enhanced responsiveness of HEK-293 cells expressing wild-type RYR1 with that of mutant RYR1 to caffeine following perfusion with remimazolam or propofol. Furthermore, we investigated whether RYR1 enhanced the responsiveness of cells to remimazolam or propofol and compared the median effective concentration (EC50; i.e., the concentration required to reach half-maximal activation) using an unpaired two-tailed t-test while a P < 0.05 was considered significant. RESULTS Remimazolam and propofol did not promote the caffeine-induced increase in intracellular Ca2+ levels in HEK-293 cells expressing mutant RYR1 even with exposure to approximately 100-fold the clinically used concentration. In wild-type RYR1, EC50 values of remimazolam following refusion vs. nonperfusion were 2.86 mM vs. 2.75 mM (P = 0.76) while for propofol perfusion vs. nonperfusion, they were 2.76 mM vs. 2.75 mM, respectively (P = 0.83). In mutant RYR1, EC50 values of remimazolam refusion vs. nonperfusion were 1.58 mM vs. 1.71 mM, respectively (P = 0.63) while for propofol perfusion vs. nonperfusion, they were 1.65 mM vs. 1.71 mM, respectively (P = 0.73). Remimazolam and propofol increased intracellular Ca2+ levels in a concentration-dependent manner, but the effect was not enhanced by RYR1. EC50 values of remimazolam with non-RYR1 vs. wild-type RYR1 were 1.00 mM vs. 0.92 mM, respectively (P = 0.91) while those of propofol were 1.09 mM vs. 1.05 mM, respectively (P = 0.84). CONCLUSIONS The increase in intracellular Ca2+ concentration caused by remimazolam or propofol was not considered an RYR1-mediated reaction. We conclude that remimazolam and propofol can be safely used as an anesthetic in MH-susceptible patients with RYR1-mutation without causing MH and may be safely substituted for an MH-triggering anesthetic when RYR1-mediated MH occurs.
Collapse
Affiliation(s)
- Tomoyuki Watanabe
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yuko Noda
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Soshi Narasaki
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Atsushi Morio
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yukari Toyota
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hiroshi Kimura
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Keiko Mukaida
- Department of Anesthesiology, Hiroshima Prefectural Rehabilitation Center, Higashihiroshima 739-0036, Japan
| | - Toshimichi Yasuda
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yasuo M. Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
15
|
Functional analysis of newly identified RYR1 variants in patients susceptible to malignant hyperthermia. J Anesth 2020; 34:658-665. [PMID: 32535660 DOI: 10.1007/s00540-020-02803-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE This study aimed to evaluate whether the three ryanodine receptor type 1 (RYR1) variants (p.Ser2345Thr, p.Ser2345Arg, and p.Lys3367Arg) which we identified in Japanese malignant hyperthermia (MH) patients with a clinical grading scale rank of 6 were causative for MH. METHODS We prepared human embryonic kidney (HEK)-293 cells transfected with wild-type RYR1 or one of the RYR1 variants, along with myotubes cultured from muscle pieces. Calcium kinetics were examined by calculating the 340/380-nm ratio under various caffeine and 4-chloro-m-cresol (4CmC) concentrations with the ratiometric dye Fura-2 AM. Half-maximal effective concentration (EC50) values were calculated from dose-response curves. Statistical analysis was based on one-way analysis of variance with a Dunnett's multiple comparison test, using a P value < 0.05 as evidence of statistical significance. RESULTS In functional analysis using HEK-293 cells, we found significant reductions in the EC50 of p.Ser2345Thr and p.Ser2345Arg in comparison with wild-type RYR1 (P < 0.001), while the EC50 of p.Lys3367Arg was not significantly different (P = 0.062 for caffeine and P > 0.999 for 4CmC). On the other hand, functional analysis using myotubes showed significant differences in the EC50 values for all variants (P < 0.001 for all comparisons). CONCLUSIONS p.Ser2345Thr and p.Ser2345Arg appear capable of causing a calcium metabolism disorder that leads to the onset of MH, and p.Ser2345Arg can be considered as a diagnostic mutation, because it meets the European Malignant Hyperthermia Group criteria. However, patients with p.Lys3367Arg might have mutations in genes other than RYR1 that are capable of causing MH.
Collapse
|