1
|
Sniezek Carney O, Harris KW, Wohlfarter Y, Lee K, Butschek G, Anzmann AF, Hamacher-Brady A, Keller MA, Vernon HJ. Stem cell models of TAFAZZIN deficiency reveal novel tissue-specific pathologies in Barth syndrome. Hum Mol Genet 2024:ddae152. [PMID: 39535077 DOI: 10.1093/hmg/ddae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Barth syndrome (BTHS) is a rare mitochondrial disease caused by pathogenic variants in the gene TAFAZZIN, which leads to abnormal cardiolipin (CL) metabolism on the inner mitochondrial membrane. Although TAFAZZIN is ubiquitously expressed, BTHS involves a complex combination of tissue specific phenotypes including cardiomyopathy, neutropenia, skeletal myopathy, and growth delays, with a relatively minimal neurological burden. To understand both the developmental and functional effects of TAZ-deficiency in different tissues, we generated isogenic TAZ knockout (TAZ-KO) and WT cardiomyocytes (CMs) and neural progenitor cells (NPCs) from CRISPR-edited induced pluripotent stem cells (iPSCs). In TAZ-KO CMs we discovered evidence of dysregulated mitophagy including dysmorphic mitochondria and mitochondrial cristae, differential expression of key autophagy-associated genes, and an inability of TAZ-deficient CMs to properly initiate stress-induced mitophagy. In TAZ-deficient NPCs we identified novel phenotypes including a reduction in CIV abundance and CIV activity in the CIII2&CIV2 intermediate complex. Interestingly, while CL acyl chain manipulation was unable to alter mitophagy defects in TAZ-KO CMs, we found that linoleic acid or oleic acid supplementation was able to partially restore CIV abundance in TAZ-deficient NPCs. Taken together, our results have implications for understanding the tissue-specific pathology of BTHS and potential for tissue-specific therapeutic targeting. Moreover, our results highlight an emerging role for mitophagy in the cardiac pathophysiology of BTHS and reveal a potential neuron-specific bioenergetic phenotype.
Collapse
Affiliation(s)
- Olivia Sniezek Carney
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Kodi W Harris
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Yvonne Wohlfarter
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, Innsbruck 6020, Austria
| | - Kyuna Lee
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Grant Butschek
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Arianna F Anzmann
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Anne Hamacher-Brady
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, Innsbruck 6020, Austria
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| |
Collapse
|
2
|
Tran P, Lau C, Joshi M, Kuehl M, Maddock H, Banerjee P. Exploring Changes in Myocyte Structure, Contractility, and Energetics From Mechanical Unloading in Patients With Heart Failure Undergoing Ventricular Assist Device Implantation: A Systematic Review and Meta-Analysis. Heart Lung Circ 2024; 33:1097-1116. [PMID: 38704332 DOI: 10.1016/j.hlc.2024.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 05/06/2024]
Abstract
AIMS Recent reports of myocardial recovery after mechanical unloading with left ventricular assist devices (LVADs) have challenged the prevailing notion that end-stage heart failure (HF) is irreversible. To improve our understanding of this phenomenon, we comprehensively analysed the structural, functional, and energetic changes in failing human cardiomyocytes after LVAD implantation. METHODS Based on a prospectively registered protocol (PROSPERO-CRD42022380214), 30 eligible studies were identified from 940 records with a pooled population of 648 patients predominantly with non-ischaemic cardiomyopathy. RESULTS LVAD led to a substantial regression in myocyte size similar to that of donor hearts (standardised mean difference, -1.29; p<0.001). The meta-regression analysis revealed that HF duration was a significant modifier on the changes in myocyte size. There were some suggestions of fibrosis reversal (-5.17%; p=0.009); however, this was insignificant after sensitivity analysis. Developed force did not improve in cardiac trabeculae (n=5 studies); however, non-physiological isometric contractions were tested. At the myocyte level (n=4 studies), contractile kinetics improved where the time-to-peak force reduced by 41.7%-50.7% and time to 50% relaxation fell by 47.4%-62.1% (p<0.05). Qualitatively, LVAD enhanced substrate utilisation and mitochondrial function (n=6 studies). Most studies were at a high risk of bias. CONCLUSION The regression of maladaptive hypertrophy, partial fibrosis reversal, and normalisation in metabolic pathways after LVAD may be a testament to the heart's remarkable plasticity, even in the advanced stages of HF. However, inconsistencies exist in force-generating capabilities. Using more physiological force-length work-loop assays, addressing the high risks of bias and clinical heterogeneity are crucial to better understand the phenomenon of reverse remodelling.
Collapse
Affiliation(s)
- Patrick Tran
- Centre for Health & Life Sciences, Coventry University, Coventry, UK; Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK.
| | - Clement Lau
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Mithilesh Joshi
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK; Warwick Medical School, University of Warwick, Coventry, UK
| | - Michael Kuehl
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK; Warwick Medical School, University of Warwick, Coventry, UK
| | - Helen Maddock
- Centre for Health & Life Sciences, Coventry University, Coventry, UK
| | - Prithwish Banerjee
- Centre for Health & Life Sciences, Coventry University, Coventry, UK; Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK; Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
3
|
Piotin A, Oulehri W, Charles AL, Tacquard C, Collange O, Mertes PM, Geny B. Oxidative Stress and Mitochondria Are Involved in Anaphylaxis and Mast Cell Degranulation: A Systematic Review. Antioxidants (Basel) 2024; 13:920. [PMID: 39199166 PMCID: PMC11352116 DOI: 10.3390/antiox13080920] [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: 06/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Anaphylaxis, an allergic reaction caused by the massive release of active mediators, can lead to anaphylactic shock (AS), the most severe and potentially life-threatening form of anaphylactic reaction. Nevertheless, understanding of its pathophysiology to support new therapies still needs to be improved. We performed a systematic review, assessing the role and the complex cellular interplay of mitochondria and oxidative stress during anaphylaxis, mast cell metabolism and degranulation. After presenting the main characteristics of anaphylaxis, the oxidant/antioxidant balance and mitochondrial functions, we focused this review on the involvement of mitochondria and oxidative stress in anaphylaxis. Then, we discussed the role of oxidative stress and mitochondria following mast cell stimulation by allergens, leading to degranulation, in order to further elucidate mechanistic pathways. Finally, we considered potential therapeutic interventions implementing these findings for the treatment of anaphylaxis. Experimental studies evaluated mainly cardiomyocyte metabolism during AS. Cardiac dysfunction was associated with left ventricle mitochondrial impairment and lipid peroxidation. Studies evaluating in vitro mast cell degranulation, following Immunoglobulin E (IgE) or non-IgE stimulation, revealed that mitochondrial respiratory complex integrity and membrane potential are crucial for mast cell degranulation. Antigen stimulation raises reactive oxygen species (ROS) production from nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and mitochondria, leading to mast cell degranulation. Moreover, mast cell activation involved mitochondrial morphological changes and mitochondrial translocation to the cell surface near exocytosis sites. Interestingly, antioxidant administration reduced degranulation by lowering ROS levels. Altogether, these results highlight the crucial role of oxidative stress and mitochondria during anaphylaxis and mast cell degranulation. New therapeutics against anaphylaxis should probably target oxidative stress and mitochondria, in order to decrease anaphylaxis-induced systemic and major organ deleterious effects.
Collapse
Affiliation(s)
- Anays Piotin
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, 67000 Strasbourg, France
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Walid Oulehri
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Anne-Laure Charles
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Charles Tacquard
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
- Établissement Français du Sang (EFS) Grand Est, French National Institute of Health and Medical Research), (INSERM) BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, 67000 Strasbourg, France
| | - Olivier Collange
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Paul-Michel Mertes
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Bernard Geny
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| |
Collapse
|
4
|
Lahiri SK, Jin F, Zhou Y, Quick AP, Kramm CF, Wang MC, Wehrens XH. Altered myocardial lipid regulation in junctophilin-2-associated familial cardiomyopathies. Life Sci Alliance 2024; 7:e202302330. [PMID: 38438248 PMCID: PMC10912815 DOI: 10.26508/lsa.202302330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
Myocardial lipid metabolism is critical to normal heart function, whereas altered lipid regulation has been linked to cardiac diseases including cardiomyopathies. Genetic variants in the JPH2 gene can cause hypertrophic cardiomyopathy (HCM) and, in some cases, dilated cardiomyopathy (DCM). In this study, we tested the hypothesis that JPH2 variants identified in patients with HCM and DCM, respectively, cause distinct alterations in myocardial lipid profiles. Echocardiography revealed clinically significant cardiac dysfunction in both knock-in mouse models of cardiomyopathy. Unbiased myocardial lipidomic analysis demonstrated significantly reduced levels of total unsaturated fatty acids, ceramides, and various phospholipids in both mice with HCM and DCM, suggesting a common metabolic alteration in both models. On the contrary, significantly increased di- and triglycerides, and decreased co-enzyme were only found in mice with HCM. Moreover, mice with DCM uniquely exhibited elevated levels of cholesterol ester. Further in-depth analysis revealed significantly altered metabolites from all the lipid classes with either similar or opposing trends in JPH2 mutant mice with HCM or DCM. Together, these studies revealed, for the first time, unique alterations in the cardiac lipid composition-including distinct increases in neutral lipids and decreases in polar membrane lipids-in mice with HCM and DCM were caused by distinct JPH2 variants. These studies may aid the development of novel biomarkers or therapeutics for these inherited disorders.
Collapse
Affiliation(s)
- Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Feng Jin
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yue Zhou
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Ann P Quick
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Carlos F Kramm
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Meng C Wang
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Xander Ht Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Sniezek Carney O, Harris KW, Wohlfarter Y, Lee K, Butschek G, Anzmann A, Claypool SM, Hamacher-Brady A, Keller M, Vernon HJ. Stem cell models of TAFAZZIN deficiency reveal novel tissue-specific pathologies in Barth Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591534. [PMID: 38746168 PMCID: PMC11092433 DOI: 10.1101/2024.04.28.591534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Barth syndrome (BTHS) is a rare mitochondrial disease caused by pathogenic variants in the gene TAFAZZIN, which leads to abnormal cardiolipin (CL) metabolism on the inner mitochondrial membrane. Although TAFAZZIN is ubiquitously expressed, BTHS involves a complex combination of tissue specific phenotypes including cardiomyopathy, neutropenia, skeletal myopathy, and growth delays, with a relatively minimal neurological burden. To understand both the developmental and functional effects of TAZ-deficiency in different tissues, we generated isogenic TAZ knockout (TAZ- KO) and WT cardiomyocytes (CMs) and neural progenitor cells (NPCs) from CRISPR-edited induced pluripotent stem cells (iPSCs). In TAZ-KO CMs we discovered evidence of dysregulated mitophagy including dysmorphic mitochondria and mitochondrial cristae, differential expression of key autophagy-associated genes, and an inability of TAZ-deficient CMs to properly initiate stress-induced mitophagy. In TAZ-deficient NPCs we identified novel phenotypes including a reduction in CIV abundance and CIV activity in the CIII2&CIV2 intermediate complex. Interestingly, while CL acyl chain manipulation was unable to alter mitophagy defects in TAZ-KO CMs, we found that linoleic acid or oleic acid supplementation was able to partially restore CIV abundance in TAZ-deficient NPCs. Taken together, our results have implications for understanding the tissue-specific pathology of BTHS and potential for tissue-specific therapeutic targeting. Moreover, our results highlight an emerging role for mitophagy in the cardiac pathophysiology of BTHS and reveal a potential neuron-specific bioenergetic phenotype.
Collapse
|
6
|
Swain L, Bhave S, Qiao X, Reyelt L, Everett K, Awata J, Raghav R, Powers SN, Sunagawa G, Natov PS, Mahmoudi E, Warner M, Couper G, Kawabori M, Miyashita S, Aryaputra T, Huggins GS, Chin MT, Kapur NK. Novel Role for Cardiolipin as a Target of Therapy to Mitigate Myocardial Injury Caused by Venoarterial Extracorporeal Membrane Oxygenation. Circulation 2024; 149:1341-1353. [PMID: 38235580 PMCID: PMC11039383 DOI: 10.1161/circulationaha.123.065298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Cardiolipin is a mitochondrial-specific phospholipid that maintains integrity of the electron transport chain (ETC) and plays a central role in myocardial ischemia/reperfusion injury. Tafazzin is an enzyme that is required for cardiolipin maturation. Venoarterial extracorporeal membrane oxygenation (VA-ECMO) use to provide hemodynamic support for acute myocardial infarction has grown exponentially, is associated with poor outcomes, and is under active clinical investigation, yet the mechanistic effect of VA-ECMO on myocardial damage in acute myocardial infarction remains poorly understood. We hypothesized that VA-ECMO acutely depletes myocardial cardiolipin and exacerbates myocardial injury in acute myocardial infarction. METHODS We examined cardiolipin and tafazzin levels in human subjects with heart failure and healthy swine exposed to VA-ECMO and used a swine model of closed-chest myocardial ischemia/reperfusion injury to evaluate the effect of VA-ECMO on cardiolipin expression, myocardial injury, and mitochondrial function. RESULTS Cardiolipin and tafazzin levels are significantly reduced in the left ventricles of individuals requiring VA-ECMO compared with individuals without VA-ECMO before heart transplantation. Six hours of exposure to VA-ECMO also decreased left ventricular levels of cardiolipin and tafazzin in healthy swine compared with sham controls. To explore whether cardiolipin depletion by VA-ECMO increases infarct size, we performed left anterior descending artery occlusion for a total of 120 minutes followed by 180 minutes of reperfusion in adult swine in the presence and absence of MTP-131, an amphipathic molecule that interacts with cardiolipin to stabilize the inner mitochondrial membrane. Compared with reperfusion alone, VA-ECMO activation beginning after 90 minutes of left anterior descending artery occlusion increased infarct size (36±8% versus 48±7%; P<0.001). VA-ECMO also decreased cardiolipin and tafazzin levels, disrupted mitochondrial integrity, reduced electron transport chain function, and promoted oxidative stress. Compared with reperfusion alone or VA-ECMO before reperfusion, delivery of MTP-131 before VA-ECMO activation reduced infarct size (22±8%; P=0.03 versus reperfusion alone and P<0.001 versus VA-ECMO alone). MTP-131 restored cardiolipin and tafazzin levels, stabilized mitochondrial function, and reduced oxidative stress in the left ventricle. CONCLUSIONS We identified a novel mechanism by which VA-ECMO promotes myocardial injury and further identify cardiolipin as an important target of therapy to reduce infarct size and to preserve mitochondrial function in the setting of VA-ECMO for acute myocardial infarction.
Collapse
Affiliation(s)
- Lija Swain
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Shreyas Bhave
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Xiaoying Qiao
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Lara Reyelt
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Kay Everett
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Junya Awata
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Rahul Raghav
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Sarah N Powers
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Genya Sunagawa
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Peter S Natov
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Elena Mahmoudi
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Mary Warner
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Greg Couper
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Masa Kawabori
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Satoshi Miyashita
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Tejasvi Aryaputra
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Gordon S. Huggins
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| | - Navin K. Kapur
- Molecular Cardiology Research Institute, Interventional Research Laboratories, and The Cardiovascular Center, Tufts Medical Center
| |
Collapse
|
7
|
Zhang S, Huang Y, Zheng C, Wang L, Zhou Y, Chen W, Duan Y, Shan T. Leucine improves the growth performance, carcass traits, and lipid nutritional quality of pork in Shaziling pigs. Meat Sci 2024; 210:109435. [PMID: 38246121 DOI: 10.1016/j.meatsci.2024.109435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/03/2024] [Accepted: 01/14/2024] [Indexed: 01/23/2024]
Abstract
Leucine is involved in promoting fatty acid oxidation and lipolysis, mediating lipid metabolism and energy homeostasis, thus it has been widely used in livestock production. However, the effects of leucine on fat deposition and nutrition in Shaziling pigs remain unclear. A total of 72 Shaziling pigs (150 days old, weight 35.00 ± 1.00 kg) were randomly divided into 2 groups and fed with basal diet (control group) or basal diet containing 1% leucine (leucine group) for 60 days. The results showed that leucine significantly increased the average daily feed intake but decreased the ratio of feed to gain (P < 0.05), increased the loin muscle area and serum glucose content (P < 0.05) of Shaziling pigs. Besides, leucine regulated the re-distribution of fatty acids from adipose tissue to muscle as it significantly increased the contents of C18:1n-9 and C22:6n-3 (DHA) in the longissimus thoracis while decreased the contents of C22:5n-3 (DPA), C20:5n-3 (EPA), and DHA in the adipose tissue of Shaziling pigs (P < 0.05). Lipidomic analysis showed that the contents of phosphatidylethanolamines (PEs), cardiolipins (CLs), and phosphatidylglycerols (PGs) in the longissimus thoracis and the contents of lysophosphatidylethanolamines (LPEs), ceramides (Cers), phosphatidylinositols (PIs) in adipose tissue of Shaziling pigs were decreased in leucine group (P < 0.05). Collectively, this study clarified that dietary addition of 1% leucine have a better effect on growth performance and the deposition of beneficial fatty acids in the muscle of Shaziling pigs, which is conductive to the production of high quality and healthy pork. In addition, leucine altered the lipid composition of muscle and fat in Shaziling pigs. The related results provide a theoretical basis and application guidance for regulating fat deposition in Shaziling pigs, which is important for the healthy breeding of Shaziling pigs.
Collapse
Affiliation(s)
- Shu Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China
| | - Yuqin Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, PR China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China
| | - Yanbing Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China
| | - Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China
| | - Yehui Duan
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, PR China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, PR China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
8
|
Hill EB, Reisdorph RM, Rasolofomanana-Rajery S, Michel C, Khajeh-Sharafabadi M, Doenges KA, Weaver N, Quinn K, Sutliff AK, Tang M, Borengasser SJ, Frank DN, O'Connor LE, Campbell WW, Krebs NF, Hendricks AE, Reisdorph NA. Salmon Food-Specific Compounds and Their Metabolites Increase in Human Plasma and Are Associated with Cardiometabolic Health Indicators Following a Mediterranean-Style Diet Intervention. J Nutr 2024; 154:26-40. [PMID: 37918675 PMCID: PMC10808825 DOI: 10.1016/j.tjnut.2023.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Nutrimetabolomics allows for the comprehensive analysis of foods and human biospecimens to identify biomarkers of intake and begin to probe their associations with health. Salmon contains hundreds of compounds that may provide cardiometabolic benefits. OBJECTIVES We used untargeted metabolomics to identify salmon food-specific compounds (FSCs) and their predicted metabolites that were found in plasma after a salmon-containing Mediterranean-style (MED) diet intervention. Associations between changes in salmon FSCs and changes in cardiometabolic health indicators (CHIs) were also explored. METHODS For this secondary analysis of a randomized, crossover, controlled feeding trial, 41 participants consumed MED diets with 2 servings of salmon per week for 2 5-wk periods. CHIs were assessed, and fasting plasma was collected pre- and postintervention. Plasma, salmon, and 99 MED foods were analyzed using liquid chromatography-mass spectrometry-based metabolomics. Compounds were characterized as salmon FSCs if detected in all salmon replicates but none of the other foods. Metabolites of salmon FSCs were predicted using machine learning. For salmon FSCs and metabolites found in plasma, linear mixed-effect models were used to assess change from pre- to postintervention and associations with changes in CHIs. RESULTS Relative to the other 99 MED foods, there were 508 salmon FSCs with 237 unique metabolites. A total of 143 salmon FSCs and 106 metabolites were detected in plasma. Forty-eight salmon FSCs and 30 metabolites increased after the intervention (false discovery rate <0.05). Increases in 2 annotated salmon FSCs and 2 metabolites were associated with improvements in CHIs, including total cholesterol, low-density lipoprotein cholesterol, triglycerides, and apolipoprotein B. CONCLUSIONS A data-driven nutrimetabolomics strategy identified salmon FSCs and their predicted metabolites that were detectable in plasma and changed after consumption of a salmon-containing MED diet. Findings support this approach for the discovery of compounds in foods that may serve, upon further validation, as biomarkers or act as bioactive components influential to health. The trials supporting this work were registered at NCT02573129 (Mediterranean-style diet intervention) and NCT05500976 (ongoing clinical trial).
Collapse
Affiliation(s)
- Emily B Hill
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Richard M Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sakaiza Rasolofomanana-Rajery
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States; Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cole Michel
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mobin Khajeh-Sharafabadi
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States
| | - Katrina A Doenges
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nicholas Weaver
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Aimee K Sutliff
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Minghua Tang
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sarah J Borengasser
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel N Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren E O'Connor
- USDA, Agricultural Research Service, Beltsville Human Nutrition Research Center, Food Components and Health Laboratory, Beltsville, MD, United States
| | - Wayne W Campbell
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Nancy F Krebs
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Audrey E Hendricks
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, United States; Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | - Nichole A Reisdorph
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
9
|
Bhullar SK, Dhalla NS. Status of Mitochondrial Oxidative Phosphorylation during the Development of Heart Failure. Antioxidants (Basel) 2023; 12:1941. [PMID: 38001794 PMCID: PMC10669359 DOI: 10.3390/antiox12111941] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Mitochondria are specialized organelles, which serve as the "Power House" to generate energy for maintaining heart function. These organelles contain various enzymes for the oxidation of different substrates as well as the electron transport chain in the form of Complexes I to V for producing ATP through the process of oxidative phosphorylation (OXPHOS). Several studies have shown depressed OXPHOS activity due to defects in one or more components of the substrate oxidation and electron transport systems which leads to the depletion of myocardial high-energy phosphates (both creatine phosphate and ATP). Such changes in the mitochondria appear to be due to the development of oxidative stress, inflammation, and Ca2+-handling abnormalities in the failing heart. Although some investigations have failed to detect any changes in the OXPHOS activity in the failing heart, such results appear to be due to a loss of Ca2+ during the mitochondrial isolation procedure. There is ample evidence to suggest that mitochondrial Ca2+-overload occurs, which is associated with impaired mitochondrial OXPHOS activity in the failing heart. The depression in mitochondrial OXPHOS activity may also be due to the increased level of reactive oxygen species, which are formed as a consequence of defects in the electron transport complexes in the failing heart. Various metabolic interventions which promote the generation of ATP have been reported to be beneficial for the therapy of heart failure. Accordingly, it is suggested that depression in mitochondrial OXPHOS activity plays an important role in the development of heart failure.
Collapse
Affiliation(s)
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
10
|
Chowdhury A, Boshnakovska A, Aich A, Methi A, Vergel Leon AM, Silbern I, Lüchtenborg C, Cyganek L, Prochazka J, Sedlacek R, Lindovsky J, Wachs D, Nichtova Z, Zudova D, Koubkova G, Fischer A, Urlaub H, Brügger B, Katschinski DM, Dudek J, Rehling P. Metabolic switch from fatty acid oxidation to glycolysis in knock-in mouse model of Barth syndrome. EMBO Mol Med 2023; 15:e17399. [PMID: 37533404 PMCID: PMC10493589 DOI: 10.15252/emmm.202317399] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Mitochondria are central for cellular metabolism and energy supply. Barth syndrome (BTHS) is a severe disorder, due to dysfunction of the mitochondrial cardiolipin acyl transferase tafazzin. Altered cardiolipin remodeling affects mitochondrial inner membrane organization and function of membrane proteins such as transporters and the oxidative phosphorylation (OXPHOS) system. Here, we describe a mouse model that carries a G197V exchange in tafazzin, corresponding to BTHS patients. TAZG197V mice recapitulate disease-specific pathology including cardiac dysfunction and reduced oxidative phosphorylation. We show that mutant mitochondria display defective fatty acid-driven oxidative phosphorylation due to reduced levels of carnitine palmitoyl transferases. A metabolic switch in ATP production from OXPHOS to glycolysis is apparent in mouse heart and patient iPSC cell-derived cardiomyocytes. An increase in glycolytic ATP production inactivates AMPK causing altered metabolic signaling in TAZG197V . Treatment of mutant cells with AMPK activator reestablishes fatty acid-driven OXPHOS and protects mice against cardiac dysfunction.
Collapse
Affiliation(s)
- Arpita Chowdhury
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
- Present address:
Dewpoint Therapeutics GmbHDresdenGermany
| | - Angela Boshnakovska
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| | - Abhishek Aich
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
| | - Aditi Methi
- Department of Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Ana Maria Vergel Leon
- Department of Cardiovascular PhysiologyUniversity Medical Center GöttingenGöttingenGermany
| | - Ivan Silbern
- The Bioanalytical Mass Spectrometry GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Institute for Clinical Chemistry, University Medical Center GöttingenGöttingenGermany
| | | | - Lukas Cyganek
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research) partner site GöttingenGöttingenGermany
- Stem Cell Unit, Clinic for Cardiology and PneumologyUniversity Medical Center Göttingen, Georg‐August University GöttingenGöttingenGermany
| | - Jan Prochazka
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - Radislav Sedlacek
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - Jiri Lindovsky
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - Dominic Wachs
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| | - Zuzana Nichtova
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - Dagmar Zudova
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - Gizela Koubkova
- Czech Centre for PhenogenomicsInstitute of Molecular Genetics of the CASPragueCzech Republic
| | - André Fischer
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
- Department of Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Henning Urlaub
- The Bioanalytical Mass Spectrometry GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Institute for Clinical Chemistry, University Medical Center GöttingenGöttingenGermany
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH)HeidelbergGermany
| | - Dörthe M Katschinski
- Department of Cardiovascular PhysiologyUniversity Medical Center GöttingenGöttingenGermany
| | - Jan Dudek
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
- Max Planck Institute for Multidisciplinary ScienceGöttingenGermany
| |
Collapse
|
11
|
Jang S, Javadov S. Unraveling the mechanisms of cardiolipin function: The role of oxidative polymerization of unsaturated acyl chains. Redox Biol 2023; 64:102774. [PMID: 37300954 PMCID: PMC10363451 DOI: 10.1016/j.redox.2023.102774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Cardiolipin is a unique phospholipid of the inner mitochondrial membrane (IMM) as well as in bacteria. It performs several vital functions such as resisting osmotic rupture and stabilizing the supramolecular structure of large membrane proteins, like ATP synthases and respirasomes. The process of cardiolipin biosynthesis results in the production of immature cardiolipin. A subsequent step is required for its maturation when its acyl groups are replaced with unsaturated acyl chains, primarily linoleic acid. Linoleic acid is the major fatty acid of cardiolipin across all organs and tissues, except for the brain. Linoleic acid is not synthesized by mammalian cells. It has the unique ability to undergo oxidative polymerization at a moderately accelerated rate compared to other unsaturated fatty acids. This property can enable cardiolipin to form covalently bonded net-like structures essential for maintaining the complex geometry of the IMM and gluing the quaternary structure of large IMM protein complexes. Unlike triglycerides, phospholipids possess only two covalently linked acyl chains, which constrain their capacity to develop robust and complicated structures through oxidative polymerization of unsaturated acyl chains. Cardiolipin, on the other hand, has four fatty acids at its disposal to form covalently bonded polymer structures. Despite its significance, the oxidative polymerization of cardiolipin has been overlooked due to the negative perception surrounding biological oxidation and methodological difficulties. Here, we discuss an intriguing hypothesis that oxidative polymerization of cardiolipin is essential for the structure and function of cardiolipin in the IMM in physiological conditions. In addition, we highlight current challenges associated with the identification and characterization of oxidative polymerization of cardiolipin in vivo. Altogether, the study provides a better understanding of the structural and functional role of cardiolipin in mitochondria.
Collapse
Affiliation(s)
- Sehwan Jang
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA.
| |
Collapse
|
12
|
Sartre C, Peurois F, Ley M, Kryszke MH, Zhang W, Courilleau D, Fischmeister R, Ambroise Y, Zeghouf M, Cianferani S, Ferrandez Y, Cherfils J. Membranes prime the RapGEF EPAC1 to transduce cAMP signaling. Nat Commun 2023; 14:4157. [PMID: 37438343 DOI: 10.1038/s41467-023-39894-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023] Open
Abstract
EPAC1, a cAMP-activated GEF for Rap GTPases, is a major transducer of cAMP signaling and a therapeutic target in cardiac diseases. The recent discovery that cAMP is compartmentalized in membrane-proximal nanodomains challenged the current model of EPAC1 activation in the cytosol. Here, we discover that anionic membranes are a major component of EPAC1 activation. We find that anionic membranes activate EPAC1 independently of cAMP, increase its affinity for cAMP by two orders of magnitude, and synergize with cAMP to yield maximal GEF activity. In the cell cytosol, where cAMP concentration is low, EPAC1 must thus be primed by membranes to bind cAMP. Examination of the cell-active chemical CE3F4 in this framework further reveals that it targets only fully activated EPAC1. Together, our findings reformulate previous concepts of cAMP signaling through EPAC proteins, with important implications for drug discovery.
Collapse
Affiliation(s)
- Candice Sartre
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - François Peurois
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Marie Ley
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, IPHC, CNRS UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Marie-Hélène Kryszke
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Wenhua Zhang
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Delphine Courilleau
- Université Paris-Saclay, IPSIT-CIBLOT, Inserm US31, CNRS UAR3679, 91400, Orsay, France
| | | | - Yves Ambroise
- Université Paris-Saclay, CEA, Service de Chimie Bioorganique et de Marquage, 91191, Gif-sur-Yvette, France
| | - Mahel Zeghouf
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, IPHC, CNRS UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Yann Ferrandez
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Jacqueline Cherfils
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France.
| |
Collapse
|
13
|
Chen Y, Wu L, Liu J, Ma L, Zhang W. Adenine nucleotide translocase: Current knowledge in post-translational modifications, regulations and pathological implications for human diseases. FASEB J 2023; 37:e22953. [PMID: 37224026 DOI: 10.1096/fj.202201855rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/01/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Adenine nucleotide translocases (ANTs) are central to mitochondrial integrity and bioenergetic metabolism. This review aims to integrate the progresses and knowledge on ANTs over the last few years, contributing to a potential implication of ANTs for various diseases. Structures, functions, modifications, regulators and pathological implications of ANTs for human diseases are intensively demonstrated here. ANTs have four isoforms (ANT1-4), responsible for exchanging ATP/ADP, possibly composing of pro-apoptotic mPTP as a major component, and mediating FA-dependent uncoupling of proton efflux. ANT can be modified by methylation, nitrosylation and nitroalkylation, acetylation, glutathionylation, phosphorylation, carbonylation and hydroxynonenal-induced modifications. Compounds, including bongkrekic acid, atractyloside calcium, carbon monoxide, minocycline, 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid, cardiolipin, free long-chain fatty acids, agaric acid, long chain acyl-coenzyme A esters, all have an ability to regulate ANT activities. ANT impairment leads to bioenergetic failure and mitochondrial dysfunction, contributing to pathogenesis of diseases, such as diabetes (deficiency), heart disease (deficiency), Parkinson's disease (reduction), Sengers Syndrome (decrease), cancer (isoform shifting), Alzheimer's Disease (coaggregation with Tau), Progressive External Opthalmoplegia (mutation), and Fascioscapulohumeral muscular dystrophy (overexpression). This review improves the understanding of the mechanism of ANT in pathogenesis of human diseases, and opens a window for novel therapeutic strategies targeted on ANT in diseases.
Collapse
Affiliation(s)
- Yingfei Chen
- Grade 2020, Capital Medical University, Beijing, China
| | - Leshuang Wu
- Grade 2019, Dalian Medical University, Dalian, China
| | - Jun Liu
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Li J, Xin Y, Li J, Chen H, Li H. Phosphatidylethanolamine N-methyltransferase: from Functions to Diseases. Aging Dis 2023; 14:879-891. [PMID: 37191416 PMCID: PMC10187709 DOI: 10.14336/ad.2022.1025] [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: 08/02/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Locating on endoplasmic reticulum and mitochondria associated membrane, Phosphatidylethanolamine N-methyltransferase (PEMT), catalyzes phosphatidylethanolamine methylation to phosphatidylcholine. As the only endogenous pathway for choline biosynthesis in mammals, the dysregulation of PEMT can lead to imbalance of phospholipid metabolism. Dysregulation of phospholipid metabolism in the liver or heart can lead to deposition of toxic lipid species that adversely result in dysfunction of hepatocyte/cardiomyocyte. Studies have shown that PEMT-/- mice increased susceptibility of diet-induced fatty liver and steatohepatitis. However, knockout of PEMT protects against diet-induced atherosclerosis, diet-induced obesity, and insulin resistance. Thus, novel insights to the function of PEMT in various organs should be summarized. Here, we reviewed the structural and functional properties of PEMT, highlighting its role in the pathogenesis of obesity, liver diseases, cardiovascular diseases, and other conditions.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hui Chen
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, China.
| |
Collapse
|
15
|
Singh M, Kadhim MM, Turki Jalil A, Oudah SK, Aminov Z, Alsaikhan F, Jawhar ZH, Ramírez-Coronel AA, Farhood B. A systematic review of the protective effects of silymarin/silibinin against doxorubicin-induced cardiotoxicity. Cancer Cell Int 2023; 23:88. [PMID: 37165384 PMCID: PMC10173635 DOI: 10.1186/s12935-023-02936-4] [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: 01/17/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Although doxorubicin chemotherapy is commonly applied for treating different malignant tumors, cardiotoxicity induced by this chemotherapeutic agent restricts its clinical use. The use of silymarin/silibinin may mitigate the doxorubicin-induced cardiac adverse effects. For this aim, the potential cardioprotective effects of silymarin/silibinin against the doxorubicin-induced cardiotoxicity were systematically reviewed. METHODS In this study, we performed a systematic search in accordance with PRISMA guideline for identifying all relevant studies on "the role of silymarin/silibinin against doxorubicin-induced cardiotoxicity" in different electronic databases up to June 2022. Sixty-one articles were obtained and screened based on the predefined inclusion and exclusion criteria. Thirteen eligible papers were finally included in this review. RESULTS According to the echocardiographic and electrocardiographic findings, the doxorubicin-treated groups presented a significant reduction in ejection fraction, tissue Doppler peak mitral annulus systolic velocity, and fractional shortening as well as bradycardia, prolongation of QT and QRS interval. However, these echocardiographic abnormalities were obviously improved in the silymarin plus doxorubicin groups. As well, the doxorubicin administration led to induce histopathological and biochemical changes in the cardiac cells/tissue; in contrast, the silymarin/silibinin co-administration could mitigate these induced alterations (for most of the cases). CONCLUSION According to the findings, it was found that the co-administration of silymarin/silibinin alleviates the doxorubicin-induced cardiac adverse effects. Silymarin/silibinin exerts its cardioprotective effects via antioxidant, anti-inflammatory, anti-apoptotic activities, and other mechanisms.
Collapse
Affiliation(s)
- Mandeep Singh
- Department of Physical Education, University of Jammu, Srinagar, Jammu, India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
16
|
Pistritu DV, Vasiliniuc AC, Vasiliu A, Visinescu EF, Visoiu IE, Vizdei S, Martínez Anghel P, Tanca A, Bucur O, Liehn EA. Phospholipids, the Masters in the Shadows during Healing after Acute Myocardial Infarction. Int J Mol Sci 2023; 24:8360. [PMID: 37176067 PMCID: PMC10178977 DOI: 10.3390/ijms24098360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Phospholipids are major components of cell membranes with complex structures, high heterogeneity and critical biological functions and have been used since ancient times to treat cardiovascular disease. Their importance and role were shadowed by the difficulty or incomplete available research methodology to study their biological presence and functionality. This review focuses on the current knowledge about the roles of phospholipids in the pathophysiology and therapy of cardiovascular diseases, which have been increasingly recognized. Used in singular formulation or in inclusive combinations with current drugs, phospholipids proved their positive and valuable effects not only in the protection of myocardial tissue, inflammation and fibrosis but also in angiogenesis, coagulation or cardiac regeneration more frequently in animal models as well as in human pathology. Thus, while mainly neglected by the scientific community, phospholipids present negligible side effects and could represent an ideal target for future therapeutic strategies in healing myocardial infarction. Acknowledging and understanding their mechanisms of action could offer a new perspective into novel therapeutic strategies for patients suffering an acute myocardial infarction, reducing the burden and improving the general social and economic outcome.
Collapse
Affiliation(s)
- Dan-Valentin Pistritu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | | | - Anda Vasiliu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | - Elena-Florentina Visinescu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Ioana-Elena Visoiu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Smaranda Vizdei
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Paula Martínez Anghel
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Business Academy Aarhus, 30 Sønderhøj, 8260 Viby J, Denmark
| | - Antoanela Tanca
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Octavian Bucur
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Viron Molecular Medicine Institute, 201 Washington Street, Boston, MA 02108, USA
| | - Elisa Anamaria Liehn
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Institute for Molecular Medicine, University of Southern Denmark, 25 J.B Winsløws Vej, 5230 Odense, Denmark
- National Heart Center Singapore, 5 Hospital Dr., Singapore 169609, Singapore
| |
Collapse
|
17
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
18
|
Reynolds MB, Hong HS, Michmerhuizen BC, Lawrence ALE, Zhang L, Knight JS, Lyssiotis CA, Abuaita BH, O’Riordan MX. Cardiolipin coordinates inflammatory metabolic reprogramming through regulation of Complex II disassembly and degradation. SCIENCE ADVANCES 2023; 9:eade8701. [PMID: 36735777 PMCID: PMC9897665 DOI: 10.1126/sciadv.ade8701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/30/2022] [Indexed: 05/15/2023]
Abstract
Macrophage metabolic plasticity enables repurposing of electron transport from energy generation to inflammation and host defense. Altered respiratory complex II function has been implicated in cancer, diabetes, and inflammation, but regulatory mechanisms are incompletely understood. Here, we show that macrophage inflammatory activation triggers Complex II disassembly and succinate dehydrogenase subunit B loss through sequestration and selective mitophagy. Mitochondrial fission supported lipopolysaccharide-stimulated succinate dehydrogenase subunit B degradation but not sequestration. We hypothesized that this Complex II regulatory mechanism might be coordinated by the mitochondrial phospholipid cardiolipin. Cardiolipin synthase knockdown prevented lipopolysaccharide-induced metabolic remodeling and Complex II disassembly, sequestration, and degradation. Cardiolipin-depleted macrophages were defective in lipopolysaccharide-induced pro-inflammatory cytokine production, a phenotype partially rescued by Complex II inhibition. Thus, cardiolipin acts as a critical organizer of inflammatory metabolic remodeling.
Collapse
Affiliation(s)
- Mack B. Reynolds
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hanna S. Hong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Britton C Michmerhuizen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anna-Lisa E. Lawrence
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jason S. Knight
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Basel H. Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mary X. O’Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Myocardial metabolism is intricately linked to cardiac function. Perturbations of cardiac energy metabolism result in an energy-starved heart and the development of contractile dysfunction. In this review, we discuss alterations in myocardial energy supply, transcriptional changes in response to different energy demands, and mitochondrial function in the development of heart failure. RECENT FINDINGS Recent studies on substrate modulation through modifying energy substrate supply have shown cardioprotective properties. In addition, large cardiovascular outcome trials of anti-diabetic agents have demonstrated prognostic benefit, suggesting the importance of myocardial metabolism in cardiac function. Understanding molecular and transcriptional controls of cardiac metabolism promises new research avenues for metabolic treatment targets. Future studies assessing the impact of substrate modulation on cardiac energetic status and function will better inform development of metabolic therapies.
Collapse
Affiliation(s)
- Sher May Ng
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Stefan Neubauer
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Oliver J Rider
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK.
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
20
|
Bautista JS, Falabella M, Flannery PJ, Hanna MG, Heales SJ, Pope SA, Pitceathly RD. Advances in methods to analyse cardiolipin and their clinical applications. Trends Analyt Chem 2022; 157:116808. [PMID: 36751553 PMCID: PMC7614147 DOI: 10.1016/j.trac.2022.116808] [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] [Indexed: 11/06/2022]
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid, primarily localised within the inner mitochondrial membrane, that plays an essential role in mitochondrial architecture and function. Aberrant CL content, structure, and localisation have all been linked to impaired mitochondrial activity and are observed in the pathophysiology of cancer and neurological, cardiovascular, and metabolic disorders. The detection, quantification, and localisation of CL species is a valuable tool to investigate mitochondrial dysfunction and the pathophysiological mechanisms underpinning several human disorders. CL is measured using liquid chromatography, usually combined with mass spectrometry, mass spectrometry imaging, shotgun lipidomics, ion mobility spectrometry, fluorometry, and radiolabelling. This review summarises available methods to analyse CL, with a particular focus on modern mass spectrometry, and evaluates their advantages and limitations. We provide guidance aimed at selecting the most appropriate technique, or combination of techniques, when analysing CL in different model systems, and highlight the clinical contexts in which measuring CL is relevant.
Collapse
Affiliation(s)
- Javier S. Bautista
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Padraig J. Flannery
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Michael G. Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon J.R. Heales
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Simon A.S. Pope
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robert D.S. Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK, Corresponding author. Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK. (R.D.S. Pitceathly)
| |
Collapse
|
21
|
Russo S, De Rasmo D, Signorile A, Corcelli A, Lobasso S. Beneficial effects of SS-31 peptide on cardiac mitochondrial dysfunction in tafazzin knockdown mice. Sci Rep 2022; 12:19847. [PMID: 36400945 PMCID: PMC9674582 DOI: 10.1038/s41598-022-24231-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Barth Syndrome (BTHS), a genetic disease associated with early-onset cardioskeletal myopathy, is caused by loss-of-function mutations of the TAFAZZIN gene, which is responsible for remodeling the mitochondrial phospholipid cardiolipin (CL). Deregulation of CL biosynthesis and maturation in BTHS mitochondria result in a dramatically increased monolysocardiolipin (MLCL)/CL ratio associated with bioenergetic dysfunction. One of the most promising therapeutic approaches for BTHS includes the mitochondria-targeted tetrapeptide SS-31, which interacts with CL. Here, we used TAFAZZIN knockdown (TazKD) mice to investigate for the first time whether in vivo administration of SS-31 could affect phospholipid profiles and mitochondrial dysfunction. The CL fingerprinting of TazKD cardiac mitochondria obtained by MALDI-TOF/MS revealed the typical lipid changes associated with BTHS. TazKD mitochondria showed lower respiratory rates in state 3 and 4 together with a decreased in maximal respiratory rates. Treatment of TazKD mice with SS-31 improved mitochondrial respiratory capacity and promoted supercomplex organization, without affecting the MLCL/CL ratio. We hypothesize that SS-31 exerts its effect by influencing the function of the respiratory chain rather than affecting CL directly. In conclusion, our results indicate that SS-31 have beneficial effects on improving cardiac mitochondrial dysfunction in a BTHS animal model, suggesting the peptide as future pharmacologic agent for therapy.
Collapse
Affiliation(s)
- Silvia Russo
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Domenico De Rasmo
- grid.503043.1CNR-Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| | - Anna Signorile
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Angela Corcelli
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Simona Lobasso
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
22
|
Wohlfarter Y, Eidelpes R, Yu RD, Sailer S, Koch J, Karall D, Scholl-Bürgi S, Amberger A, Hillen HS, Zschocke J, Keller MA. ost in promiscuity? An evolutionary and biochemical evaluation of HSD10 function in cardiolipin metabolism. Cell Mol Life Sci 2022; 79:562. [PMID: 36271951 PMCID: PMC9587951 DOI: 10.1007/s00018-022-04579-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Multifunctional proteins are challenging as it can be difficult to confirm pathomechanisms associated with disease-causing genetic variants. The human 17β-hydroxysteroid dehydrogenase 10 (HSD10) is a moonlighting enzyme with at least two structurally and catalytically unrelated functions. HSD10 disease was originally described as a disorder of isoleucine metabolism, but the clinical manifestations were subsequently shown to be linked to impaired mtDNA transcript processing due to deficient function of HSD10 in the mtRNase P complex. A surprisingly large number of other, mostly enzymatic and potentially clinically relevant functions have been attributed to HSD10. Recently, HSD10 was reported to exhibit phospholipase C-like activity towards cardiolipins (CL), important mitochondrial phospholipids. To assess the physiological role of the proposed CL-cleaving function, we studied CL architectures in living cells and patient fibroblasts in different genetic backgrounds and lipid environments using our well-established LC-MS/MS cardiolipidomic pipeline. These experiments revealed no measurable effect on CLs, indicating that HSD10 does not have a physiologically relevant function towards CL metabolism. Evolutionary constraints could explain the broad range of reported substrates for HSD10 in vitro. The combination of an essential structural with a non-essential enzymatic function in the same protein could direct the evolutionary trajectory towards improvement of the former, thereby increasing the flexibility of the binding pocket, which is consistent with the results presented here.
Collapse
Affiliation(s)
- Yvonne Wohlfarter
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria
| | - Reiner Eidelpes
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Ryan D Yu
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sabrina Sailer
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria
| | - Jakob Koch
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria
| | - Daniela Karall
- Department of Paediatrics I (Inherited Metabolic Disorders), Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Scholl-Bürgi
- Department of Paediatrics I (Inherited Metabolic Disorders), Medical University of Innsbruck, Innsbruck, Austria
| | - Albert Amberger
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria
| | - Hauke S Hillen
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, 6020, Innsbruck, Austria.
| |
Collapse
|
23
|
Bikomeye JC, Terwoord JD, Santos JH, Beyer AM. Emerging mitochondrial signaling mechanisms in cardio-oncology: beyond oxidative stress. Am J Physiol Heart Circ Physiol 2022; 323:H702-H720. [PMID: 35930448 PMCID: PMC9529263 DOI: 10.1152/ajpheart.00231.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 12/27/2022]
Abstract
Many anticancer therapies (CTx) have cardiotoxic side effects that limit their therapeutic potential and cause long-term cardiovascular complications in cancer survivors. This has given rise to the field of cardio-oncology, which recognizes the need for basic, translational, and clinical research focused on understanding the complex signaling events that drive CTx-induced cardiovascular toxicity. Several CTx agents cause mitochondrial damage in the form of mitochondrial DNA deletions, mutations, and suppression of respiratory function and ATP production. In this review, we provide a brief overview of the cardiovascular complications of clinically used CTx agents and discuss current knowledge of local and systemic secondary signaling events that arise in response to mitochondrial stress/damage. Mitochondrial oxidative stress has long been recognized as a contributor to CTx-induced cardiotoxicity; thus, we focus on emerging roles for mitochondria in epigenetic regulation, innate immunity, and signaling via noncoding RNAs and mitochondrial hormones. Because data exploring mitochondrial secondary signaling in the context of cardio-oncology are limited, we also draw upon clinical and preclinical studies, which have examined these pathways in other relevant pathologies.
Collapse
Affiliation(s)
- Jean C Bikomeye
- Doctorate Program in Public and Community Health, Division of Epidemiology and Social Sciences, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Janée D Terwoord
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Biomedical Sciences Department, Rocky Vista University, Ivins, Utah
| | - Janine H Santos
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Andreas M Beyer
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
24
|
Doxorubicin induced cardio toxicity through sirtuins mediated mitochondrial disruption. Chem Biol Interact 2022; 365:110028. [DOI: 10.1016/j.cbi.2022.110028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 12/06/2022]
|
25
|
Su Y, Zhang W, Zhang R, Yuan Q, Wu R, Liu X, Wuri J, Li R, Yan T. Activation of Cholinergic Anti-Inflammatory Pathway Ameliorates Cerebral and Cardiac Dysfunction After Intracerebral Hemorrhage Through Autophagy. Front Immunol 2022; 13:870174. [PMID: 35812436 PMCID: PMC9260497 DOI: 10.3389/fimmu.2022.870174] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is the devastating subtype of stroke with cardiovascular complications, resulting in high rates of mortality and morbidity with the release of inflammatory factors. Previous studies have demonstrated that activation of α7nAChR can reduce immune and inflammation-related diseases by triggering the cholinergic anti-inflammatory pathway (CAIP). α7nAChR mediates protection from nervous system inflammation through AMPK-mTOR-p70S6K-associated autophagy. Therefore, the purpose of this study is to explore whether the activation of α7nAChR improves cerebral and cardiac dysfunction after ICH through autophagy. Methods Male C57BL/6 mice were randomly divided into five groups (1): Control + saline (2), ICH+ saline (3), ICH + PNU-282987 (4), ICH+ PNU-282987 + MLA (5), ICH + PNU-282987 + 3-MA. The neurological function was evaluated at multiple time points. Brain water content was measured at 3 days after ICH to assess the severity of brain edema. PCR, immunofluorescence staining, and Western Blot were performed at 7 days after ICH to detect inflammation and autophagy. Picro-Sirius Red staining was measured at 30 days after ICH to evaluate myocardial fibrosis, echocardiography was performed at 3 and 30 days to measure cardiac function. Results Our results indicated that the PNU-282987 reduced inflammatory factors (MCP-1, IL-1β, MMP-9, TNF-α, HMGB1, TLR2), promoted the polarization of macrophage/microglia into anti-inflammatory subtypes(CD206), repaired blood-brain barrier injury (ZO-1, Claudin-5, Occludin), alleviated acute brain edema and then recovered neurological dysfunction. Echocardiography and PSR indicated that activation of α7nAChR ameliorated cardiac dysfunction. Western Blot showed that activation of α7nAChR increased autophagy protein (LC3, Beclin) and decreased P62. It demonstrated that the activation of α7nAChR promotes autophagy and then recovers brain and heart function after ICH. Conclusions In conclusion, PNU-282987 promoted the cerebral and cardiac functional outcomes after ICH in mice through activated α7nAChR, which may be attributable to promoting autophagy and then reducing inflammatory reactions after ICH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tao Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
26
|
Yang J, Guo Q, Feng X, Liu Y, Zhou Y. Mitochondrial Dysfunction in Cardiovascular Diseases: Potential Targets for Treatment. Front Cell Dev Biol 2022; 10:841523. [PMID: 35646910 PMCID: PMC9140220 DOI: 10.3389/fcell.2022.841523] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are serious public health issues and are responsible for nearly one-third of global deaths. Mitochondrial dysfunction is accountable for the development of most CVDs. Mitochondria produce adenosine triphosphate through oxidative phosphorylation and inevitably generate reactive oxygen species (ROS). Excessive ROS causes mitochondrial dysfunction and cell death. Mitochondria can protect against these damages via the regulation of mitochondrial homeostasis. In recent years, mitochondria-targeted therapy for CVDs has attracted increasing attention. Various studies have confirmed that clinical drugs (β-blockers, angiotensin-converting enzyme inhibitors/angiotensin receptor-II blockers) against CVDs have mitochondrial protective functions. An increasing number of cardiac mitochondrial targets have shown their cardioprotective effects in experimental and clinical studies. Here, we briefly introduce the mechanisms of mitochondrial dysfunction and summarize the progression of mitochondrial targets against CVDs, which may provide ideas for experimental studies and clinical trials.
Collapse
|
27
|
Hoffmann JJ, Becker T. Crosstalk between Mitochondrial Protein Import and Lipids. Int J Mol Sci 2022; 23:ijms23095274. [PMID: 35563660 PMCID: PMC9101885 DOI: 10.3390/ijms23095274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 12/10/2022] Open
Abstract
Mitochondria import about 1000 precursor proteins from the cytosol. The translocase of the outer membrane (TOM complex) forms the major entry site for precursor proteins. Subsequently, membrane-bound protein translocases sort the precursor proteins into the outer and inner membrane, the intermembrane space, and the matrix. The phospholipid composition of mitochondrial membranes is critical for protein import. Structural and biochemical data revealed that phospholipids affect the stability and activity of mitochondrial protein translocases. Integration of proteins into the target membrane involves rearrangement of phospholipids and distortion of the lipid bilayer. Phospholipids are present in the interface between subunits of protein translocases and affect the dynamic coupling of partner proteins. Phospholipids are required for full activity of the respiratory chain to generate membrane potential, which in turn drives protein import across and into the inner membrane. Finally, outer membrane protein translocases are closely linked to organellar contact sites that mediate lipid trafficking. Altogether, intensive crosstalk between mitochondrial protein import and lipid biogenesis controls mitochondrial biogenesis.
Collapse
|
28
|
Dungan JR, Qin X, Gregory SG, Cooper-Dehoff R, Duarte JD, Qin H, Gulati M, Taylor JY, Pepine CJ, Hauser ER, Kraus WE. Sex-dimorphic gene effects on survival outcomes in people with coronary artery disease. AMERICAN HEART JOURNAL PLUS: CARDIOLOGY RESEARCH AND PRACTICE 2022; 17. [PMID: 35959094 PMCID: PMC9365120 DOI: 10.1016/j.ahjo.2022.100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: Ischemic coronary heart disease (IHD) is the leading cause of death worldwide. Genetic variation is presumed to be a major factor underlying sex differences for IHD events, including mortality. The purpose of this study was to identify sex-specific candidate genes associated with all-cause mortality among people diagnosed with coronary artery disease (CAD). Methods: We performed a sex-stratified, exploratory genome-wide association (GWAS) screen using existing data from CAD-diagnosed males (n = 510) and females (n = 174) who reported European ancestry from the Duke Catheterization Genetics biorepository. Extant genotype data for 785,945 autosomal SNPs generated with the Human Omni1-Quad BeadChip (Illumina, CA, USA) were analyzed using an additive inheritance model. We estimated instantaneous risk of all-cause mortality by genotype groups across the 11-year follow-up using Cox multivariate regression, covarying for age and genomic ancestry. Results: The top GWAS hits associated with all-cause mortality among people with CAD included 8 SNPs among males and 15 among females (p = 1 × 10−6 or 10−7), adjusted for covariates. Cross-sex comparisons revealed distinct candidate genes. Biologically relevant candidates included rs9932462 (EMP2/TEKT5) and rs2835913 (KCNJ6) among males and rs7217169 (RAP1GAP2), rs8021816 (PRKD1), rs8133010 (PDE9A), and rs12145981 (LPGAT1) among females. Conclusions: We report 20 sex-specific candidate genes having suggestive association with all-cause mortality among CAD-diagnosed subjects. Findings demonstrate proof of principle for identifying sex-associated genetic factors that may help explain differential mortality risk in people with CAD. Replication and meta-analyses in larger studies with more diverse samples will strengthen future work in this area.
Collapse
|
29
|
Macias LA, Brodbelt JS. Enhanced Characterization of Cardiolipins via Hybrid 193 nm Ultraviolet Photodissociation Mass Spectrometry. Anal Chem 2022; 94:3268-3277. [PMID: 35135194 PMCID: PMC9284920 DOI: 10.1021/acs.analchem.1c05071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiolipins (CLs) constitute a structurally complex class of glycerophospholipids with a unique tetraacylated structure accompanied by distinctive functional roles. Aberrations in the composition of this lipid class have been associated with disease states, spurring interest in the development of new approaches to differentiate the structures of diverse CLs in complex mixtures. The structural characterization of these complex lipids using conventional methods, however, suffers from limited resolution and frequently proves unable to discern subtle yet biologically significant features such as unsaturation sites or acyl chain position assignments. Here, we describe the synergistic use of chemical derivatization and hybrid dissociation techniques to characterize CL from complex biological mixtures with both double bond and sn positional isomer resolution in a shotgun mass spectrometry strategy. Utilizing (trimethylsilyl)diazomethane (TMSD), CL phosphate groups were methylated to promote positive-mode ionization by the production of metal-cationized lipids, enabling structural interrogation via hybrid higher-energy collisional activation/ultraviolet photodissociation (HCD/UVPD). This combination of TMSD derivatization and HCD/UVPD fragmentation results in diagnostic product ions that permit distinction and relative quantitation of sn-stereoisomers and the localization of double bonds. Applying this strategy to a total lipid extract from a thyroid carcinoma revealed a previously unreported 18:2/18:1 motif, elucidating a structural feature unique to the lipid class.
Collapse
Affiliation(s)
- Luis A Macias
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer S Brodbelt
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
30
|
Syahputra RA, Harahap U, Dalimunthe A, Nasution MP, Satria D. The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041320. [PMID: 35209107 PMCID: PMC8878416 DOI: 10.3390/molecules27041320] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Doxorubicin is a widely used and promising anticancer drug; however, a severe dose-dependent cardiotoxicity hampers its therapeutic value. Doxorubicin may cause acute and chronic issues, depending on the duration of toxicity. In clinical practice, the accumulative toxic dose is up to 400 mg/m2 and increasing the dose will increase the probability of cardiac toxicity. Several molecular mechanisms underlying the pathogenesis of doxorubicin cardiotoxicity have been proposed, including oxidative stress, topoisomerase beta II inhibition, mitochondrial dysfunction, Ca2+ homeostasis dysregulation, intracellular iron accumulation, ensuing cell death (apoptosis and necrosis), autophagy, and myofibrillar disarray and loss. Natural products including flavonoids have been widely studied both in cell, animal, and human models which proves that flavonoids alleviate cardiac toxicity caused by doxorubicin. This review comprehensively summarizes cardioprotective activity flavonoids including quercetin, luteolin, rutin, apigenin, naringenin, and hesperidin against doxorubicin, both in in vitro and in vivo models.
Collapse
Affiliation(s)
- Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
- Correspondence: (R.A.S.); (U.H.)
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
- Correspondence: (R.A.S.); (U.H.)
| | - Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - M. Pandapotan Nasution
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.P.N.); (D.S.)
| | - Denny Satria
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.P.N.); (D.S.)
| |
Collapse
|
31
|
Egea PF. Mechanisms of Non-Vesicular Exchange of Lipids at Membrane Contact Sites: Of Shuttles, Tunnels and, Funnels. Front Cell Dev Biol 2021; 9:784367. [PMID: 34912813 PMCID: PMC8667587 DOI: 10.3389/fcell.2021.784367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Eukaryotic cells are characterized by their exquisite compartmentalization resulting from a cornucopia of membrane-bound organelles. Each of these compartments hosts a flurry of biochemical reactions and supports biological functions such as genome storage, membrane protein and lipid biosynthesis/degradation and ATP synthesis, all essential to cellular life. Acting as hubs for the transfer of matter and signals between organelles and throughout the cell, membrane contacts sites (MCSs), sites of close apposition between membranes from different organelles, are essential to cellular homeostasis. One of the now well-acknowledged function of MCSs involves the non-vesicular trafficking of lipids; its characterization answered one long-standing question of eukaryotic cell biology revealing how some organelles receive and distribute their membrane lipids in absence of vesicular trafficking. The endoplasmic reticulum (ER) in synergy with the mitochondria, stands as the nexus for the biosynthesis and distribution of phospholipids (PLs) throughout the cell by contacting nearly all other organelle types. MCSs create and maintain lipid fluxes and gradients essential to the functional asymmetry and polarity of biological membranes throughout the cell. Membrane apposition is mediated by proteinaceous tethers some of which function as lipid transfer proteins (LTPs). We summarize here the current state of mechanistic knowledge of some of the major classes of LTPs and tethers based on the available atomic to near-atomic resolution structures of several "model" MCSs from yeast but also in Metazoans; we describe different models of lipid transfer at MCSs and analyze the determinants of their specificity and directionality. Each of these systems illustrate fundamental principles and mechanisms for the non-vesicular exchange of lipids between eukaryotic membrane-bound organelles essential to a wide range of cellular processes such as at PL biosynthesis and distribution, lipid storage, autophagy and organelle biogenesis.
Collapse
Affiliation(s)
- Pascal F Egea
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
32
|
Wu C, Zhang Z, Zhang W, Liu X. Mitochondrial dysfunction and mitochondrial therapies in heart failure. Pharmacol Res 2021; 175:106038. [PMID: 34929300 DOI: 10.1016/j.phrs.2021.106038] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide in the last decade, accompanied by immense health and economic burdens. Heart failure (HF), as the terminal stage of many cardiovascular diseases, is a common, intractable, and costly medical condition. Despite significant improvements in pharmacologic and device therapies over the years, life expectancy for this disease remains poor. Current therapies have not reversed the trends in morbidity and mortality as expected. Thus, there is an urgent need for novel potential therapeutic agents. Although the pathophysiology of the failing heart is extraordinarily complex, targeting mitochondrial dysfunction can be an effective approach for potential treatment. Increasing evidence has shown that mitochondrial abnormalities, including altered metabolic substrate utilization, impaired mitochondrial oxidative phosphorylation (OXPHOS), increased reactive oxygen species (ROS) formation, and aberrant mitochondrial dynamics, are closely related to HF. Here, we reviewed the findings on the role of mitochondrial dysfunction in HF, along with novel mitochondrial therapeutics and their pharmacological effects.
Collapse
Affiliation(s)
- Chennan Wu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhen Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
33
|
Smeir E, Leberer S, Blumrich A, Vogler G, Vasiliades A, Dresen S, Jaeger C, Gloaguen Y, Klose C, Beule D, Schulze PC, Bodmer R, Foryst-Ludwig A, Kintscher U. Depletion of cardiac cardiolipin synthase alters systolic and diastolic function. iScience 2021; 24:103314. [PMID: 34805785 PMCID: PMC8581512 DOI: 10.1016/j.isci.2021.103314] [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: 04/14/2021] [Revised: 09/13/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiolipin (CL) is a major cardiac mitochondrial phospholipid maintaining regular mitochondrial morphology and function in cardiomyocytes. Cardiac CL production includes its biosynthesis and a CL remodeling process. Here we studied the impact of CL biosynthesis and the enzyme cardiolipin synthase (CLS) on cardiac function. CLS and cardiac CL species were significantly downregulated in cardiomyocytes following catecholamine-induced cardiac damage in mice, accompanied by increased oxygen consumption rates, signs of oxidative stress, and mitochondrial uncoupling. RNAi-mediated cardiomyocyte-specific knockdown of CLS in Drosophila melanogaster resulted in marked cardiac dilatation, severe impairment of systolic performance, and slower diastolic filling velocity assessed by fluorescence-based heart imaging. Finally, we showed that CL72:8 is significantly decreased in cardiac samples from patients with heart failure with reduced ejection fraction (HFrEF). In summary, we identified CLS as a regulator of cardiac function. Considering the cardiac depletion of CL species in HFrEF, pharmacological targeting of CLS may be a promising therapeutic approach. Cardiolipin synthase (CLS) is reduced in isoproterenol (ISO)-induced cardiac damage This is accompanied by alterations of cardiolipins and mitochondrial function CLS mutant Drosophila melanogaster exhibit mild cardiac changes Cardiomyocyte-CLS knockdown in Drosophila results in severe cardiac dysfunction
Collapse
Affiliation(s)
- Elia Smeir
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Sarah Leberer
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Annelie Blumrich
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Anastasia Vasiliades
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Sandra Dresen
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Carsten Jaeger
- Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Yoann Gloaguen
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany.,Berlin Institute of Health, BIH, Metabolomics Platform, Berlin, Germany
| | | | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Anna Foryst-Ludwig
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Ulrich Kintscher
- Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health/ Institute of Pharmacology, Center for Cardiovascular Research, Hessische Street 3-4, 10115 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| |
Collapse
|
34
|
Fat of the Gut: Epithelial Phospholipids in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:ijms222111682. [PMID: 34769112 PMCID: PMC8584226 DOI: 10.3390/ijms222111682] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic inflammation within the gastrointestinal (GI) tract. Despite the significant progress in understanding the etiology and development of treatment strategies, IBD remain incurable for thousands of patients. Metabolic deregulation is indicative of IBD, including substantial shifts in lipid metabolism. Recent data showed that changes in some phospholipids are very common in IBD patients. For instance, phosphatidylcholine (PC)/phosphatidylethanolamine (PE) and lysophosphatidylcholine (LPC)/PC ratios are associated with the severity of the inflammatory process. Composition of phospholipids also changes upon IBD towards an increase in arachidonic acid and a decrease in linoleic and a-linolenic acid levels. Moreover, an increase in certain phospholipid metabolites, such as lysophosphatidylcholine, sphingosine-1-phosphate and ceramide, can result in enhanced intestinal inflammation, malignancy, apoptosis or necroptosis. Because some phospholipids are associated with pathogenesis of IBD, they may provide a basis for new strategies to treat IBD. Current attempts are aimed at controlling phospholipid and fatty acid levels through the diet or via pharmacological manipulation of lipid metabolism.
Collapse
|
35
|
Sapian S, Taib IS, Latip J, Katas H, Chin KY, Mohd Nor NA, Jubaidi FF, Budin SB. Therapeutic Approach of Flavonoid in Ameliorating Diabetic Cardiomyopathy by Targeting Mitochondrial-Induced Oxidative Stress. Int J Mol Sci 2021; 22:11616. [PMID: 34769045 PMCID: PMC8583796 DOI: 10.3390/ijms222111616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/26/2022] Open
Abstract
Diabetes cardiomyopathy is one of the key factors of mortality among diabetic patients around the globe. One of the prior contributors to the progression of diabetic cardiomyopathy is cardiac mitochondrial dysfunction. The cardiac mitochondrial dysfunction can induce oxidative stress in cardiomyocytes and was found to be the cause of majority of the heart morphological and dynamical changes in diabetic cardiomyopathy. To slow down the occurrence of diabetic cardiomyopathy, it is crucial to discover therapeutic agents that target mitochondrial-induced oxidative stress. Flavonoid is a plentiful phytochemical in plants that shows a wide range of biological actions against human diseases. Flavonoids have been extensively documented for their ability to protect the heart from diabetic cardiomyopathy. Flavonoids' ability to alleviate diabetic cardiomyopathy is primarily attributed to their antioxidant properties. In this review, we present the mechanisms involved in flavonoid therapies in ameliorating mitochondrial-induced oxidative stress in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Syaifuzah Sapian
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Izatus Shima Taib
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Jalifah Latip
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 46300, Malaysia;
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Kok-Yong Chin
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia;
| | - Nor Anizah Mohd Nor
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Fatin Farhana Jubaidi
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Siti Balkis Budin
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| |
Collapse
|
36
|
Trypanosoma brucei Tim50 Possesses PAP Activity and Plays a Critical Role in Cell Cycle Regulation and Parasite Infectivity. mBio 2021; 12:e0159221. [PMID: 34517757 PMCID: PMC8546626 DOI: 10.1128/mbio.01592-21] [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] [Indexed: 12/14/2022] Open
Abstract
Trypanosoma brucei, the infective agent for African trypanosomiasis, possesses a homologue of the translocase of the mitochondrial inner membrane 50 (TbTim50). It has a pair of characteristic phosphatase signature motifs, DXDX(T/V). Here, we demonstrated that, besides its protein phosphatase activity, the recombinant TbTim50 binds and hydrolyzes phosphatidic acid in a concentration-dependent manner. Mutations of D242 and D244, but not of D345and D347, to alanine abolished these activities. In silico structural homology models identified the putative binding interfaces that may accommodate different phosphosubstrates. Interestingly, TbTim50 depletion in the bloodstream form (BF) of T. brucei reduced cardiolipin (CL) levels and decreased mitochondrial membrane potential (ΔΨ). TbTim50 knockdown (KD) also reduced the population of G2/M phase and increased that of G1 phase cells; inhibited segregation and caused overreplication of kinetoplast DNA (kDNA), and reduced BF cell growth. Depletion of TbTim50 increased the levels of AMPK phosphorylation, and parasite morphology was changed with upregulation of expression of a few stumpy marker genes. Importantly, we observed that TbTim50-depleted parasites were unable to establish infection in mice. Proteomics analysis showed reductions in levels of the translation factors, flagellar transport proteins, and many proteasomal subunits, including those of the mitochondrial heat shock locus ATPase (HslVU), which is known to play a role in regulation of kinetoplast DNA (kDNA) replication. Reduction of the level of HslV in TbTim50 KD cells was further validated by immunoblot analysis. Together, our results showed that TbTim50 is essential for mitochondrial function, regulation of kDNA replication, and the cell cycle in the BF. Therefore, TbTim50 is an important target for structure-based drug design to combat African trypanosomiasis.
Collapse
|
37
|
Chen Z, Zhu S, Wang H, Wang L, Zhang J, Gu Y, Tan C, Dhanani M, Wever E, Wang X, Xie B, Wang S, Huang L, van Kampen AHC, Liu J, Han Z, Patel HH, Vaz FM, Fang X, Chen J, Ouyang K. PTPMT1 Is Required for Embryonic Cardiac Cardiolipin Biosynthesis to Regulate Mitochondrial Morphogenesis and Heart Development. Circulation 2021; 144:403-406. [PMID: 34339306 DOI: 10.1161/circulationaha.121.054768] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.).,Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Siting Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.).,Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Hong Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Li Wang
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Jianlin Zhang
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Yusu Gu
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Changming Tan
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.).,Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (C.T.)
| | - Mehul Dhanani
- Anesthesiology (M.D., H.H.P.), School of Medicine, University of California San Diego, La Jolla.,Veterans Administration San Diego Healthcare System, CA (M.D., H.H.P.)
| | - Eric Wever
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism (E.W., F.M.V.).,Core Facility Metabolomics (E.W., F.M.V.), Amsterdam Universitair Medische Centra.,Laboratory Genetic Metabolic Diseases; Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute (E.W., A.H.C.v.K.), Amsterdam Universitair Medische Centra
| | - Xinru Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Boyu Xie
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Shijia Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Antoine H C van Kampen
- Laboratory Genetic Metabolic Diseases; Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute (E.W., A.H.C.v.K.), Amsterdam Universitair Medische Centra.,Biosystems Data Analysis, Swammerdam Institute for Life Sciences (A.H.C.v.K.), University of Amsterdam, The Netherlands
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China (J.L.)
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| | - Hemal H Patel
- Anesthesiology (M.D., H.H.P.), School of Medicine, University of California San Diego, La Jolla.,Veterans Administration San Diego Healthcare System, CA (M.D., H.H.P.)
| | - Frédéric M Vaz
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism (E.W., F.M.V.).,Core Facility Metabolomics (E.W., F.M.V.), Amsterdam Universitair Medische Centra
| | - Xi Fang
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Ju Chen
- Departments of Medicine (Z.C., S.Z., L.W., J.Z., Y.G., C.T., X.F., J.C.)
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China (Z.C., S.Z., H.W., X.W., B.X., S.W., L.H., Z.H., K.O.)
| |
Collapse
|
38
|
Chaudhuri M, Tripathi A, Gonzalez FS. Diverse Functions of Tim50, a Component of the Mitochondrial Inner Membrane Protein Translocase. Int J Mol Sci 2021; 22:7779. [PMID: 34360547 PMCID: PMC8346121 DOI: 10.3390/ijms22157779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are essential in eukaryotes. Besides producing 80% of total cellular ATP, mitochondria are involved in various cellular functions such as apoptosis, inflammation, innate immunity, stress tolerance, and Ca2+ homeostasis. Mitochondria are also the site for many critical metabolic pathways and are integrated into the signaling network to maintain cellular homeostasis under stress. Mitochondria require hundreds of proteins to perform all these functions. Since the mitochondrial genome only encodes a handful of proteins, most mitochondrial proteins are imported from the cytosol via receptor/translocase complexes on the mitochondrial outer and inner membranes known as TOMs and TIMs. Many of the subunits of these protein complexes are essential for cell survival in model yeast and other unicellular eukaryotes. Defects in the mitochondrial import machineries are also associated with various metabolic, developmental, and neurodegenerative disorders in multicellular organisms. In addition to their canonical functions, these protein translocases also help maintain mitochondrial structure and dynamics, lipid metabolism, and stress response. This review focuses on the role of Tim50, the receptor component of one of the TIM complexes, in different cellular functions, with an emphasis on the Tim50 homologue in parasitic protozoan Trypanosoma brucei.
Collapse
Affiliation(s)
- Minu Chaudhuri
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.T.); (F.S.G.)
| | | | | |
Collapse
|
39
|
Cardiolipin, Non-Bilayer Structures and Mitochondrial Bioenergetics: Relevance to Cardiovascular Disease. Cells 2021; 10:cells10071721. [PMID: 34359891 PMCID: PMC8304834 DOI: 10.3390/cells10071721] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
The present review is an attempt to conceptualize a contemporary understanding about the roles that cardiolipin, a mitochondrial specific conical phospholipid, and non-bilayer structures, predominantly found in the inner mitochondrial membrane (IMM), play in mitochondrial bioenergetics. This review outlines the link between changes in mitochondrial cardiolipin concentration and changes in mitochondrial bioenergetics, including changes in the IMM curvature and surface area, cristae density and architecture, efficiency of electron transport chain (ETC), interaction of ETC proteins, oligomerization of respiratory complexes, and mitochondrial ATP production. A relationship between cardiolipin decline in IMM and mitochondrial dysfunction leading to various diseases, including cardiovascular diseases, is thoroughly presented. Particular attention is paid to the targeting of cardiolipin by Szeto–Schiller tetrapeptides, which leads to rejuvenation of important mitochondrial activities in dysfunctional and aging mitochondria. The role of cardiolipin in triggering non-bilayer structures and the functional roles of non-bilayer structures in energy-converting membranes are reviewed. The latest studies on non-bilayer structures induced by cobra venom peptides are examined in model and mitochondrial membranes, including studies on how non-bilayer structures modulate mitochondrial activities. A mechanism by which non-bilayer compartments are formed in the apex of cristae and by which non-bilayer compartments facilitate ATP synthase dimerization and ATP production is also presented.
Collapse
|
40
|
Cole LK, Agarwal P, Doucette CA, Fonseca M, Xiang B, Sparagna GC, Seshadri N, Vandel M, Dolinsky VW, Hatch GM. Tafazzin Deficiency Reduces Basal Insulin Secretion and Mitochondrial Function in Pancreatic Islets From Male Mice. Endocrinology 2021; 162:bqab102. [PMID: 34019639 PMCID: PMC8197286 DOI: 10.1210/endocr/bqab102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 12/13/2022]
Abstract
Tafazzin (TAZ) is a cardiolipin (CL) biosynthetic enzyme important for maintaining mitochondrial function. TAZ affects both the species and content of CL in the inner mitochondrial membrane, which are essential for normal cellular respiration. In pancreatic β cells, mitochondrial function is closely associated with insulin secretion. However, the role of TAZ and CL in the secretion of insulin from pancreatic islets remains unknown. Male 4-month-old doxycycline-inducible TAZ knock-down (KD) mice and wild-type littermate controls were used. Immunohistochemistry was used to assess β-cell morphology in whole pancreas sections, whereas ex vivo insulin secretion, CL content, RNA-sequencing analysis, and mitochondrial oxygen consumption were measured from isolated islet preparations. Ex vivo insulin secretion under nonstimulatory low-glucose concentrations was reduced ~52% from islets isolated from TAZ KD mice. Mitochondrial oxygen consumption under low-glucose conditions was also reduced ~58% in islets from TAZ KD animals. TAZ deficiency in pancreatic islets was associated with significant alteration in CL molecular species and elevated polyunsaturated fatty acid CL content. In addition, RNA-sequencing of isolated islets showed that TAZ KD increased expression of extracellular matrix genes, which are linked to pancreatic fibrosis, activated stellate cells, and impaired β-cell function. These data indicate a novel role for TAZ in regulating pancreatic islet function, particularly under low-glucose conditions.
Collapse
Affiliation(s)
- Laura K Cole
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Prasoon Agarwal
- KTH Royal Institute of Technology, School of Electrical Engineering and Computer Science, 10044 Stockholm, Sweden
- Science for Life Laboratory, 16939 Solna, Sweden
| | - Christine A Doucette
- Physiology and Pathophysiology, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Mario Fonseca
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Bo Xiang
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Nivedita Seshadri
- Physiology and Pathophysiology, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Marilyne Vandel
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Vernon W Dolinsky
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Grant M Hatch
- Department of Pharmacology, Winnipeg, R3E3P4, Canada
- Department of Therapeutics, Winnipeg, R3E3P4, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children’s Hospital Research Institute of Manitoba, Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E3P4, Canada
| |
Collapse
|
41
|
Del Campo A, Perez G, Castro PF, Parra V, Verdejo HE. Mitochondrial function, dynamics and quality control in the pathophysiology of HFpEF. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166208. [PMID: 34214606 DOI: 10.1016/j.bbadis.2021.166208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022]
Abstract
Heart failure (HF) is one of the leading causes of hospitalization for the adult population and a major cause of mortality worldwide. The HF syndrome is characterized by the heart's inability to supply the cardiac output required to meet the body's metabolic requirements or only at the expense of elevated filling pressures. HF without overt impairment of left ventricular ejection fraction (LVEF) was initially labeled as "diastolic HF" until recognizing the coexistence of both systolic and diastolic abnormalities in most cases. Acknowledging these findings, the preferred nomenclature is HF with preserved EF (HFpEF). This syndrome primarily affects the elderly population and is associated with a heterogeneous overlapping of comorbidities that makes its diagnosis challenging. Despite extensive research, there is still no evidence-based therapy for HFpEF, reinforcing the need for a thorough understanding of the pathophysiology underlying its onset and progression. The role of mitochondrial dysfunction in developing the pathophysiological changes that accompany HFpEF onset and progression (low-grade systemic inflammation, oxidative stress, endothelial dysfunction, and myocardial remodeling) has just begun to be acknowledged. This review summarizes our current understanding of the participation of the mitochondrial network in the pathogenesis of HFpEF, with particular emphasis on the signaling pathways involved, which may provide future therapeutic targets.
Collapse
Affiliation(s)
- Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo Perez
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Castro
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Chile
| | - Valentina Parra
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Autophagy Research Center, Universidad de Chile, Santiago, Chile; Network for the Study of High-lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Chile.
| | - Hugo E Verdejo
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Chile.
| |
Collapse
|
42
|
Zhao Y, Jia WW, Ren S, Xiao W, Li GW, Jin L, Lin Y. Difluoromethylornithine attenuates isoproterenol-induced cardiac hypertrophy by regulating apoptosis, autophagy and the mitochondria-associated membranes pathway. Exp Ther Med 2021; 22:870. [PMID: 34194548 PMCID: PMC8237397 DOI: 10.3892/etm.2021.10302] [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: 10/09/2020] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial hypertrophy is an independent risk factor of cardiovascular diseases and is closely associated with the incidence of heart failure. In the present study, we hypothesized that difluoromethylornithine (DFMO) could attenuate cardiac hypertrophy through mitochondria-associated membranes (MAM) and autophagy. Cardiac hypertrophy was induced in male rats by intravenous administration of isoproterenol (ISO; 5 mg/kg/day) for 1, 3,7 and 14 days. For DFMO treatment group, rats were given ISO (5 mg/kg/day) for 14 days and 2% DFMO in their water for 4 weeks. The expression of atrial natriuretic peptide (ANP) mRNA,heart parameters, apoptosis rate, fibrotic area and protein expressions of cleaved caspase3/9, GRP75, Mfn2, CypD and VDAC1 were measured to confirm the development of cardiac hypertrophy, apoptosis and autophagy induced by ISO. ANP mRNA and MAM protein expression levels were assessed by reverse transcription-quantitative PCR and western blotting to evaluate hypertrophy and the effects of DFMO oral administration. The results demonstrated that heart parameters, ANP mRNA levels, fibrotic area and apoptosis rate were significantly increased in the heart tissue for ISO 7 and 14 day groups compared with the control group. Furthermore, treatment with DFMO significantly inhibited these indicators, and DFMO downregulated the MAM signaling pathway and upregulated the autophagy pathway in heart tissue compared with the ISO 14 day group. Overall, all ISO-induced changes analyzed in the present study were attenuated following treatment with DFMO. The findings form this study suggested that DFMO treatment may be considered as a potential strategy for preventing ISO-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Wei-Wei Jia
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - San Ren
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Wei Xiao
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Guang-Wei Li
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Li Jin
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yan Lin
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
43
|
Randolph CE, Shenault DM, Blanksby SJ, McLuckey SA. Localization of Carbon-Carbon Double Bond and Cyclopropane Sites in Cardiolipins via Gas-Phase Charge Inversion Reactions. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:455-464. [PMID: 33370110 PMCID: PMC8557092 DOI: 10.1021/jasms.0c00348] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Cardiolipins (CLs) are comprised of two phosphatic acid moieties bound to a central glycerol backbone and are substituted with four acyl chains. Consequently, a vast number of distinct CL structures are possible in different biological contexts, representing a significant analytical challenge. Electrospray ionization tandem mass spectrometry (ESI-MS/MS) has become a widely used approach for the detection, characterization, and quantitation of complex lipids, including CLs. Central to this approach is fragmentation of the [CLs - H]- or [CL - 2H]2- anions by collision-induced dissociation (CID). Product ions in the resulting tandem mass spectra confirm the CL subclass assignment and reveal the numbers of carbons and degrees of unsaturation in each of the acyl chains. Conventional CID, however, affords limited structural elucidation of the fatty acyl chains, failing to discriminate isomers arising from different site(s) of unsaturation or cyclopropanation and potentially obscuring their metabolic origins. Here, we report the application of charge inversion ion/ion chemistry in the gas phase to enhance the structural elucidation of CLs. Briefly, CID of [CL - H]2- anions generated via negative ion ESI allowed for the assignment of individual fatty acyl substituents and phosphatidic acid moieties. Next, gas-phase derivatization of the resulting CL product ions, including fatty acyl carboxylate anions, was effected with gas-phase ion/ion charge inversion reactions with tris-phenanthroline magnesium reagent dications. Subsequent isolation and activation of the charge-inverted fatty acyl complex cations permitted the localization of both carbon-carbon double bond and cyclopropane motifs within each of the four acyl chains of CLs. This approach was applied to the de novo elucidation of unknown CLs in a biological extract revealing distinct isomeric populations and regiochemical relationships between double bonds and carbocyles.
Collapse
Affiliation(s)
- Caitlin E. Randolph
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907-2084, USA
| | | | - Stephen J. Blanksby
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Scott A. McLuckey
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907-2084, USA
| |
Collapse
|
44
|
Bisaccia G, Ricci F, Gallina S, Di Baldassarre A, Ghinassi B. Mitochondrial Dysfunction and Heart Disease: Critical Appraisal of an Overlooked Association. Int J Mol Sci 2021; 22:ijms22020614. [PMID: 33435429 PMCID: PMC7827742 DOI: 10.3390/ijms22020614] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/03/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
The myocardium is among the most energy-consuming tissues in the body, burning from 6 to 30 kg of ATP per day within the mitochondria, the so-called powerhouse of the cardiomyocyte. Although mitochondrial genetic disorders account for a small portion of cardiomyopathies, mitochondrial dysfunction is commonly involved in a broad spectrum of heart diseases, and it has been implicated in the development of heart failure via maladaptive circuits producing and perpetuating mitochondrial stress and energy starvation. In this bench-to-bedside review, we aimed to (i) describe the key functions of the mitochondria within the myocardium, including their role in ischemia/reperfusion injury and intracellular calcium homeostasis; (ii) examine the contribution of mitochondrial dysfunction to multiple cardiac disease phenotypes and their transition to heart failure; and (iii) discuss the rationale and current evidence for targeting mitochondrial function for the treatment of heart failure, including via sodium-glucose cotransporter 2 inhibitors.
Collapse
Affiliation(s)
- Giandomenico Bisaccia
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
| | - Fabrizio Ricci
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
- Department of Clinical Sciences, Lund University, E-205 02 Malmö, Sweden
- Casa di Cura Villa Serena, Città Sant’Angelo, 65013 Pescara, Italy
- Correspondence: ; Tel./Fax: +39-871-355-6922
| | - Sabina Gallina
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.B.); (B.G.)
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.B.); (B.G.)
| |
Collapse
|
45
|
Barth syndrome: cardiolipin, cellular pathophysiology, management, and novel therapeutic targets. Mol Cell Biochem 2021; 476:1605-1629. [PMID: 33415565 DOI: 10.1007/s11010-020-04021-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Barth syndrome is a rare X-linked genetic disease classically characterized by cardiomyopathy, skeletal myopathy, growth retardation, neutropenia, and 3-methylglutaconic aciduria. It is caused by mutations in the tafazzin gene localized to chromosome Xq28.12. Mutations in tafazzin may result in alterations in the level and molecular composition of the mitochondrial phospholipid cardiolipin and result in large elevations in the lysophospholipid monolysocardiolipin. The increased monolysocardiolipin:cardiolipin ratio in blood is diagnostic for the disease, and it leads to disruption in mitochondrial bioenergetics. In this review, we discuss cardiolipin structure, synthesis, and function and provide an overview of the clinical and cellular pathophysiology of Barth Syndrome. We highlight known pharmacological management for treatment of the major pathological features associated with the disease. In addition, we discuss non-pharmacological management. Finally, we highlight the most recent promising therapeutic options for this rare mitochondrial disease including lipid replacement therapy, peroxisome proliferator-activated receptor agonists, tafazzin gene replacement therapy, induced pluripotent stem cells, mitochondria-targeted antioxidants and peptides, and the polyphenolic compound resveratrol.
Collapse
|
46
|
Kücükköse C, Taskin AA, Marada A, Brummer T, Dennerlein S, Vögtle FN. Functional coupling of presequence processing and degradation in human mitochondria. FEBS J 2021; 288:600-613. [PMID: 32491259 DOI: 10.1111/febs.15358] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/31/2020] [Accepted: 05/04/2020] [Indexed: 12/31/2022]
Abstract
The mitochondrial proteome is built and maintained mainly by import of nuclear-encoded precursor proteins. Most of these precursors use N-terminal presequences as targeting signals that are removed by mitochondrial matrix proteases. The essential mitochondrial processing protease MPP cleaves presequences after import into the organelle thereby enabling protein folding and functionality. The cleaved presequences are subsequently degraded by peptidases. While most of these processes have been discovered in yeast, characterization of the human enzymes is still scarce. As the matrix presequence peptidase PreP has been reported to play a role in Alzheimer's disease, analysis of impaired peptide turnover in human cells is of huge interest. Here, we report the characterization of HEK293T PreP knockout cells. Loss of PreP causes severe defects in oxidative phosphorylation and changes in nuclear expression of stress response marker genes. The mitochondrial defects upon lack of PreP result from the accumulation of presequence peptides that trigger feedback inhibition of MPP and accumulation of nonprocessed precursor proteins. Also, the mitochondrial intermediate peptidase MIP that cleaves eight residues from a subset of precursors after MPP processing is compromised upon loss of PreP suggesting that PreP also degrades MIP generated octapeptides. Investigation of the PrePR183Q patient mutation associated with neurological disorders revealed that the mutation destabilizes the protein making it susceptible to enhanced degradation and aggregation upon heat shock. Taken together, our data reveal a functional coupling between precursor processing by MPP and MIP and presequence degradation by PreP in human mitochondria that is crucial to maintain a functional organellar proteome.
Collapse
Affiliation(s)
- Cansu Kücükköse
- Faculty of Medicine, Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, Germany
- Faculty of Biology, University of Freiburg, Germany
| | - Asli Aras Taskin
- Faculty of Medicine, Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany
| | - Adinarayana Marada
- Faculty of Medicine, Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, Germany
| | - Tilman Brummer
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany
- Centre for Biological Signalling Studies BIOSS, University of Freiburg, Germany
- Comprehensive Cancer Centre Freiburg, University of Freiburg, Germany
- DKTK Partner Site Freiburg and DKFZ, Heidelberg, Germany
| | - Sven Dennerlein
- Department of Cellular Biochemistry, University Medical Center Göttingen, Germany
| | - Friederike-Nora Vögtle
- Faculty of Medicine, Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany
| |
Collapse
|
47
|
Corrado M, Edwards-Hicks J, Villa M, Flachsmann LJ, Sanin DE, Jacobs M, Baixauli F, Stanczak M, Anderson E, Azuma M, Quintana A, Curtis JD, Clapes T, Grzes KM, Kabat AM, Kyle R, Patterson AE, Geltink RK, Amulic B, Steward CG, Strathdee D, Trompouki E, O'Sullivan D, Pearce EJ, Pearce EL. Dynamic Cardiolipin Synthesis Is Required for CD8 + T Cell Immunity. Cell Metab 2020; 32:981-995.e7. [PMID: 33264603 PMCID: PMC7721104 DOI: 10.1016/j.cmet.2020.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/25/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022]
Abstract
Mitochondria constantly adapt to the metabolic needs of a cell. This mitochondrial plasticity is critical to T cells, which modulate metabolism depending on antigen-driven signals and environment. We show here that de novo synthesis of the mitochondrial membrane-specific lipid cardiolipin maintains CD8+ T cell function. T cells deficient for the cardiolipin-synthesizing enzyme PTPMT1 had reduced cardiolipin and responded poorly to antigen because basal cardiolipin levels were required for activation. However, neither de novo cardiolipin synthesis, nor its Tafazzin-dependent remodeling, was needed for T cell activation. In contrast, PTPMT1-dependent cardiolipin synthesis was vital when mitochondrial fitness was required, most notably during memory T cell differentiation or nutrient stress. We also found CD8+ T cell defects in a small cohort of patients with Barth syndrome, where TAFAZZIN is mutated, and in a Tafazzin-deficient mouse model. Thus, the dynamic regulation of a single mitochondrial lipid is crucial for CD8+ T cell immunity.
Collapse
Affiliation(s)
- Mauro Corrado
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Joy Edwards-Hicks
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Matteo Villa
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Lea J Flachsmann
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - David E Sanin
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Maaike Jacobs
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Francesc Baixauli
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Michal Stanczak
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Eve Anderson
- Cancer Research UK Beatson Institute, Glasgow G61 1 BD, UK
| | - Mai Azuma
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Andrea Quintana
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jonathan D Curtis
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Thomas Clapes
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Katarzyna M Grzes
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Agnieszka M Kabat
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Ryan Kyle
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Annette E Patterson
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Ramon Klein Geltink
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Borko Amulic
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Colin G Steward
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TH, UK
| | | | - Eirini Trompouki
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - David O'Sullivan
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Edward J Pearce
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79098 Freiburg im Breisgau, Germany
| | - Erika L Pearce
- Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| |
Collapse
|
48
|
Excessive ROS production and enhanced autophagy contribute to myocardial injury induced by branched-chain amino acids: Roles for the AMPK-ULK1 signaling pathway and α7nAChR. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165980. [PMID: 32980459 DOI: 10.1016/j.bbadis.2020.165980] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 08/24/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUNDS AND AIMS Leucine, isoleucine, and valine are diet derived and essential amino acids that are termed branched-chain amino acids (BCAA). BCAA are widely used as dietary supplements to boost muscle growth and enhance exercise performance. However, the effects of BCAA on myocardial function are largely unknown. This study was designed to investigate whether BCAA affect heart function and, if so, to further explore the underlying molecular basis for the observed effects. METHODS AND RESULTS C57BL/6J mice were randomly divided into two groups, the control group received solvent (water) and the BCAA group received 2% BCAA dissolved in water, for a successive period of 12 weeks. Compared with control, BCAA treatment significantly increased water consumption without changing body weight or diet consumption; heart tissue BCAA levels were increased, markers representative of myocardial injury in heart tissue including c-reactive protein and cardiac muscle troponin were increased ; and creatine kinase, creatine kinase-MB, and lactate dehydrogenase were increased in serum; severe myocardial fibrosis was observed by Masson staining, which was accompanied by increased reactive oxygen species (ROS) production and decreased superoxide dismutase activity in heart tissue; both p-AMPK and p-ULK1 were significantly increased as was autophagy, judged by the presence of LC3 by western blotting and immunofluorescence, increased numbers of autophagosomes were found by transmission electron microscopy in the BCAA group. In vitro, 20 mmol/L BCAA significantly decreased cell viability and increased the production of ROS, as well as the expression of p-AMPK/AMPK and p-ULK1/ULK1 in cultured H9C2 cells. Treatment with the ROS scavenger N-acetyl-L-cysteine (NAC) improved cell viability and reversed ROS changes. Decreased H9C2 cell viability induced with 20 mmol/L BCAA was reversed by either blocking AMPK or inhibition of ULK1. Furthermore, blocking AMPK significantly decreased p-ULK1/ULK1, while inhibition of ULK1 reversed the enhanced expression of LC3-II/LC3-I induced by BCAA. Excessive ROS production and decreased cell viability induced by BCAA were further confirmed in primary cultured murine cardiomyocytes. Pharmacological activation of α7nAChR with PNU-282987 attenuated BCAA-induced injury in primary murine cardiomyocytes. However, this compound failed to suppress BCAA activation of AMPK and autophagy (LC3-II/I ratio). CONCLUSION These results provide the first evidence that treatment of mice with BCAA induced myocardial injury by triggering excessive ROS production and by enhancing AMPK-ULK1 pathway-dependent autophagy. These findings suggested that inhibition of either ROS production or autophagy may alleviate myocardial injury induced by BCAA.
Collapse
|
49
|
Greenwell AA, Gopal K, Ussher JR. Myocardial Energy Metabolism in Non-ischemic Cardiomyopathy. Front Physiol 2020; 11:570421. [PMID: 33041869 PMCID: PMC7526697 DOI: 10.3389/fphys.2020.570421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
As the most metabolically demanding organ in the body, the heart must generate massive amounts of energy adenosine triphosphate (ATP) from the oxidation of fatty acids, carbohydrates and other fuels (e.g., amino acids, ketone bodies), in order to sustain constant contractile function. While the healthy mature heart acts omnivorously and is highly flexible in its ability to utilize the numerous fuel sources delivered to it through its coronary circulation, the heart’s ability to produce ATP from these fuel sources becomes perturbed in numerous cardiovascular disorders. This includes ischemic heart disease and myocardial infarction, as well as in various cardiomyopathies that often precede the development of overt heart failure. We herein will provide an overview of myocardial energy metabolism in the healthy heart, while describing the numerous perturbations that take place in various non-ischemic cardiomyopathies such as hypertrophic cardiomyopathy, diabetic cardiomyopathy, arrhythmogenic cardiomyopathy, and the cardiomyopathy associated with the rare genetic disease, Barth Syndrome. Based on preclinical evidence where optimizing myocardial energy metabolism has been shown to attenuate cardiac dysfunction, we will discuss the feasibility of myocardial energetics optimization as an approach to treat the cardiac pathology associated with these various non-ischemic cardiomyopathies.
Collapse
Affiliation(s)
- Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
50
|
Tomášová P, Čermáková M, Pelantová H, Vecka M, Kratochvílová H, Lipš M, Lindner J, Ivák P, Netuka I, Šedivá B, Haluzík M, Kuzma M. Lipid Profiling in Epicardial and Subcutaneous Adipose Tissue of Patients with Coronary Artery Disease. J Proteome Res 2020; 19:3993-4003. [DOI: 10.1021/acs.jproteome.0c00269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Petra Tomášová
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
- 4th Medical Department, First Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U Nemocnice 2, 128 08 Praha 2, Czech Republic
| | - Martina Čermáková
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Marek Vecka
- 4th Medical Department, First Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U Nemocnice 2, 128 08 Praha 2, Czech Republic
| | - Helena Kratochvílová
- Institute of Medical Biochemistry and Laboratory Diagnostics; First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Michal Lipš
- Department of Anaesthesiology, Resuscitation and Intensive Care, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Jaroslav Lindner
- 2nd Department of Surgery - Department of Cardiovascular Surgery, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | | | | | - Blanka Šedivá
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
- Faculty of Applied Sciences, University of West Bohemia, Univerzitní 8, 306 14 Plzeň, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics; First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| |
Collapse
|