1
|
Brothwell MJ, Cao (曹国燊) G, Maschek JA, Poss AM, Peterlin AD, Wang (汪立平) L, Baker TB, Shahtout JL, Siripoksup P, Pearce QJ, Johnson JM, Finger FM, Prola A, Pellizzari SA, Hale GL, Manuel AM, Watanabe (渡邉真也) S, Miranda ER, Affolter KE, Tippetts TS, Nikolova LS, Choi (崔蘭煕) RH, Decker ST, Patil M, Catrow JL, Holland WL, Nowinski SM, Lark DS, Fisher-Wellman KH, Mimche PN, Evason KJ, Cox JE, Summers SA, Gerhart-Hines Z, Funai (船井勝彦) K. Cardiolipin deficiency disrupts CoQ redox state and induces steatohepatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617517. [PMID: 39416056 PMCID: PMC11482932 DOI: 10.1101/2024.10.10.617517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disorder marked by lipid accumulation, leading to steatohepatitis (MASH). A key feature of the transition to MASH involves oxidative stress resulting from defects in mitochondrial oxidative phosphorylation (OXPHOS). Here, we show that pathological alterations in the lipid composition of the inner mitochondrial membrane (IMM) directly instigate electron transfer inefficiency to promote oxidative stress. Specifically, cardiolipin (CL) was downregulated across four mouse models of MASLD. Hepatocyte-specific CL synthase knockout (CLS-LKO) led to spontaneous MASH with elevated mitochondrial electron leak. Loss of CL interfered with the ability of coenzyme Q (CoQ) to transfer electrons, promoting leak primarily at sites IIF and IIIQ0. Data from human liver biopsies revealed a highly robust correlation between mitochondrial CL and CoQ, co-downregulated with MASH. Thus, reduction in mitochondrial CL promotes oxidative stress and contributes to pathogenesis of MASH.
Collapse
Affiliation(s)
- Marisa J. Brothwell
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Guoshen Cao (曹国燊)
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
| | - J. Alan Maschek
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Annelise M. Poss
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Alek D. Peterlin
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Liping Wang (汪立平)
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Talia B. Baker
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Division of Transplantation and Advanced Hepatobiliary Surgery, Department of Surgery; University of Utah; Salt Lake City, UT; USA
| | - Justin L. Shahtout
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
| | - Piyarat Siripoksup
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
| | - Quentinn J. Pearce
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Jordan M. Johnson
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Fabian M. Finger
- Novo Nordisk Foundation Center for Basic Metabolic Research; University of Copenhagen; Copenhagen; DK
- Center for Adipocyte Signaling (ADIPOSIGN); University of Southern Denmark; Odense; DK
| | - Alexandre Prola
- Laboratory of Fundamental and Applied Bioenergetics; University of Grenoble Alpes, Inserm U1055; Grenoble; FR
| | - Sarah A. Pellizzari
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - Gillian L. Hale
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - Allison M. Manuel
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Shinya Watanabe (渡邉真也)
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Edwin R. Miranda
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Kajsa E. Affolter
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Laboratory of Fundamental and Applied Bioenergetics; University of Grenoble Alpes, Inserm U1055; Grenoble; FR
| | - Trevor S. Tippetts
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Linda S. Nikolova
- Electron Microscopy Core Facility; University of Utah; Salt Lake City, UT; USA
| | - Ran Hee Choi (崔蘭煕)
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Stephen T. Decker
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Mallikarjun Patil
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - J. Leon Catrow
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - William L. Holland
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Sara M. Nowinski
- Department of Metabolism and Nutritional Programming; Van Andel Institute; Grand Rapids, MI; USA
| | - Daniel S. Lark
- College of Health and Human Sciences; Colorado State University; Fort Collins, CO; USA
- Columbine Health Systems Center for Healthy Aging; Colorado State University; Fort Collins, CO; USA
| | - Kelsey H. Fisher-Wellman
- Department of Cancer Biology, Wake Forest University School of Medicine; Atrium Health Wake Forest Baptist Comprehensive Cancer Center; Winston-Salem, NC; USA
| | - Patrice N. Mimche
- Departments of Dermatology and Medicine; Division of Gastroenterology and Hepatology, Indiana University School of Medicine; Indianapolis, IN; USA
| | - Kimberley J. Evason
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - James E. Cox
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Scott A. Summers
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research; University of Copenhagen; Copenhagen; DK
- Center for Adipocyte Signaling (ADIPOSIGN); University of Southern Denmark; Odense; DK
| | - Katsuhiko Funai (船井勝彦)
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| |
Collapse
|
2
|
Fuentes JM, Morcillo P. The Role of Cardiolipin in Mitochondrial Function and Neurodegenerative Diseases. Cells 2024; 13:609. [PMID: 38607048 PMCID: PMC11012098 DOI: 10.3390/cells13070609] [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: 02/18/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid synthesized in the inner mitochondrial membrane. CL plays a key role in mitochondrial membranes, impacting a plethora of functions this organelle performs. Consequently, it is conceivable that abnormalities in the CL content, composition, and level of oxidation may negatively impact mitochondrial function and dynamics, with important implications in a variety of diseases. This review concentrates on papers published in recent years, combined with basic and underexplored research in CL. We capture new findings on its biological functions in the mitochondria, as well as its association with neurodegenerative diseases such as Alzheimer's disease or Parkinson's disease. Lastly, we explore the potential applications of CL as a biomarker and pharmacological target to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
| | - Patricia Morcillo
- Departmentof Neurology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
3
|
Ikari N, Honjo K, Sagami Y, Nakamura Y, Arakawa H. Mieap forms membrane-less organelles involved in cardiolipin metabolism. iScience 2024; 27:108916. [PMID: 38322995 PMCID: PMC10845071 DOI: 10.1016/j.isci.2024.108916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 11/16/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Biomolecular condensates (BCs) are formed by proteins with intrinsically disordered regions (IDRs) via liquid-liquid phase separation. Mieap/Spata18, a p53-inducible protein, participates in suppression of colorectal tumors by promoting mitochondrial quality control. However, the regulatory mechanism involved remains unclear. Here, we report that Mieap is an IDR-containing protein that drives formation of BCs involved in cardiolipin metabolism. Mieap BCs specifically phase separate the mitochondrial phospholipid, cardiolipin. Mieap directly binds to cardiolipin in vitro. Lipidomic analysis of cardiolipin suggests that Mieap promotes enzymatic reactions in cardiolipin biosynthesis and remodeling. Accordingly, four cardiolipin biosynthetic enzymes, TAMM41, PGS1, PTPMT1, and CRLS1 and two remodeling enzymes, PLA2G6 and TAZ, are phase-separated by Mieap BCs. Mieap-deficient cells exhibit altered crista structure, leading to decreased respiration activity and ATP production in mitochondria. These results suggest that Mieap may form membrane-less organelles to compartmentalize and facilitate cardiolipin metabolism, thus potentially contributing to mitochondrial quality control.
Collapse
Affiliation(s)
- Naoki Ikari
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Katsuko Honjo
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yoko Sagami
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yasuyuki Nakamura
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hirofumi Arakawa
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
4
|
Deir S, Mozhdehbakhsh Mofrad Y, Mashayekhan S, Shamloo A, Mansoori-Kermani A. Step-by-step fabrication of heart-on-chip systems as models for cardiac disease modeling and drug screening. Talanta 2024; 266:124901. [PMID: 37459786 DOI: 10.1016/j.talanta.2023.124901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases are caused by hereditary factors, environmental conditions, and medication-related issues. On the other hand, the cardiotoxicity of drugs should be thoroughly examined before entering the market. In this regard, heart-on-chip (HOC) systems have been developed as a more efficient and cost-effective solution than traditional methods, such as 2D cell culture and animal models. HOCs must replicate the biology, physiology, and pathology of human heart tissue to be considered a reliable platform for heart disease modeling and drug testing. Therefore, many efforts have been made to find the best methods to fabricate different parts of HOCs and to improve the bio-mimicry of the systems in the last decade. Beating HOCs with different platforms have been developed and techniques, such as fabricating pumpless HOCs, have been used to make HOCs more user-friendly systems. Recent HOC platforms have the ability to simultaneously induce and record electrophysiological stimuli. Additionally, systems including both heart and cancer tissue have been developed to investigate tissue-tissue interactions' effect on cardiac tissue response to cancer drugs. In this review, all steps needed to be considered to fabricate a HOC were introduced, including the choice of cellular resources, biomaterials, fabrication techniques, biomarkers, and corresponding biosensors. Moreover, the current HOCs used for modeling cardiac diseases and testing the drugs are discussed. We finally introduced some suggestions for fabricating relatively more user-friendly HOCs and facilitating the commercialization process.
Collapse
Affiliation(s)
- Sara Deir
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran.
| | | |
Collapse
|
5
|
Verkerk AO, Wilders R. Injection of I K1 through dynamic clamp can make all the difference in patch-clamp studies on hiPSC-derived cardiomyocytes. Front Physiol 2023; 14:1326160. [PMID: 38152247 PMCID: PMC10751953 DOI: 10.3389/fphys.2023.1326160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Human-induced stem cell-derived cardiomyocytes (hiPSC-CMs) are a valuable tool for studying development, pharmacology, and (inherited) arrhythmias. Unfortunately, hiPSC-CMs are depolarized and spontaneously active, even the working cardiomyocyte subtypes such as atrial- and ventricular-like hiPSC-CMs, in contrast to the situation in the atria and ventricles of adult human hearts. Great efforts have been made, using many different strategies, to generate more mature, quiescent hiPSC-CMs with more close-to-physiological resting membrane potentials, but despite promising results, it is still difficult to obtain hiPSC-CMs with such properties. The dynamic clamp technique allows to inject a current with characteristics of the inward rectifier potassium current (IK1), computed in real time according to the actual membrane potential, into patch-clamped hiPSC-CMs during action potential measurements. This results in quiescent hiPSC-CMs with a close-to-physiological resting membrane potential. As a result, action potential measurements can be performed with normal ion channel availability, which is particularly important for the physiological functioning of the cardiac SCN5A-encoded fast sodium current (INa). We performed in vitro and in silico experiments to assess the beneficial effects of the dynamic clamp technique in dissecting the functional consequences of the SCN5A-1795insD+/- mutation. In two separate sets of patch-clamp experiments on control hiPSC-CMs and on hiPSC-CMs with mutations in ACADVL and GNB5, we assessed the value of dynamic clamp in detecting delayed afterdepolarizations and in investigating factors that modulate the resting membrane potential. We conclude that the dynamic clamp technique has highly beneficial effects in all of the aforementioned settings and should be widely used in patch-clamp studies on hiPSC-CMs while waiting for the ultimate fully mature hiPSC-CMs.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
6
|
Joshi A, Gohil VM. Cardiolipin deficiency leads to the destabilization of mitochondrial magnesium channel MRS2 in Barth syndrome. Hum Mol Genet 2023; 32:3353-3360. [PMID: 37721533 DOI: 10.1093/hmg/ddad153] [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: 07/17/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
Barth syndrome (BTHS) is a debilitating X-linked cardio-skeletal myopathy caused by loss-of-function mutations in TAFAZZIN, a cardiolipin (CL)-remodeling enzyme required for the maintenance of normal levels of CL species in mitochondrial membranes. At present, how perturbations in CL abundance and composition lead to many debilitating clinical presentations in BTHS patients have not been fully elucidated. Inspired by our recent findings that CL is essential for optimal mitochondrial calcium uptake, we measured the levels of other biologically important metal ions in BTHS mitochondria and found that in addition to calcium, magnesium levels are significantly reduced. Consistent with this observation, we report a decreased abundance of the mitochondrial magnesium influx channel MRS2 in multiple models of BTHS including yeast, murine myoblast, and BTHS patient cells and cardiac tissue. Mechanistically, we attribute reduced steady-state levels of MRS2 to its increased turnover in CL-deficient BTHS models. By expressing Mrs2 in well-characterized yeast mutants of the phospholipid biosynthetic pathways, we demonstrate a specific requirement of CL for Mrs2 abundance and assembly. Finally, we provide in vitro evidence for the direct binding of CL with human MRS2. Together, our study has identified a critical requirement of CL for MRS2 stability and suggests perturbation of mitochondrial magnesium homeostasis as a novel contributing factor to BTHS pathology.
Collapse
Affiliation(s)
- Alaumy Joshi
- Department of Biochemistry and Biophysics, Texas A&M University, 301 Old Main Drive, TAMU 3474, College Station, TX 77843, United States
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, 301 Old Main Drive, TAMU 3474, College Station, TX 77843, United States
| |
Collapse
|
7
|
Kutschka I, Bertero E, Wasmus C, Xiao K, Yang L, Chen X, Oshima Y, Fischer M, Erk M, Arslan B, Alhasan L, Grosser D, Ermer KJ, Nickel A, Kohlhaas M, Eberl H, Rebs S, Streckfuss-Bömeke K, Schmitz W, Rehling P, Thum T, Higuchi T, Rabinowitz J, Maack C, Dudek J. Activation of the integrated stress response rewires cardiac metabolism in Barth syndrome. Basic Res Cardiol 2023; 118:47. [PMID: 37930434 PMCID: PMC10628049 DOI: 10.1007/s00395-023-01017-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 09/29/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
Barth Syndrome (BTHS) is an inherited cardiomyopathy caused by defects in the mitochondrial transacylase TAFAZZIN (Taz), required for the synthesis of the phospholipid cardiolipin. BTHS is characterized by heart failure, increased propensity for arrhythmias and a blunted inotropic reserve. Defects in Ca2+-induced Krebs cycle activation contribute to these functional defects, but despite oxidation of pyridine nucleotides, no oxidative stress developed in the heart. Here, we investigated how retrograde signaling pathways orchestrate metabolic rewiring to compensate for mitochondrial defects. In mice with an inducible knockdown (KD) of TAFAZZIN, and in induced pluripotent stem cell-derived cardiac myocytes, mitochondrial uptake and oxidation of fatty acids was strongly decreased, while glucose uptake was increased. Unbiased transcriptomic analyses revealed that the activation of the eIF2α/ATF4 axis of the integrated stress response upregulates one-carbon metabolism, which diverts glycolytic intermediates towards the biosynthesis of serine and fuels the biosynthesis of glutathione. In addition, strong upregulation of the glutamate/cystine antiporter xCT increases cardiac cystine import required for glutathione synthesis. Increased glutamate uptake facilitates anaplerotic replenishment of the Krebs cycle, sustaining energy production and antioxidative pathways. These data indicate that ATF4-driven rewiring of metabolism compensates for defects in mitochondrial uptake of fatty acids to sustain energy production and antioxidation.
Collapse
Affiliation(s)
- Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - Italian IRCCS Cardiology Network, Genoa, Italy
| | - Christina Wasmus
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Lifeng Yang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai, 200031, China
| | - Xinyu Chen
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Yasuhiro Oshima
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Marcus Fischer
- Division of Pediatric Cardiology and Intensive Care, University Hospital LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Manuela Erk
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Berkan Arslan
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Lin Alhasan
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Daria Grosser
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Katharina J Ermer
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Hanna Eberl
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
| | - Sabine Rebs
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
| | - Katrin Streckfuss-Bömeke
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
- Clinic for Cardiology and Pneumology, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany
| | - Werner Schmitz
- Department of Biochemistry and Molecular Biology, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Peter Rehling
- University Göttingen, Institute of Biochemistry and Molecular Cell Biology, Humboldtallee 23, 37072, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
- Rebirth Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Joshua Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- Medical Clinic I, University Clinic Würzburg, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
8
|
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: 2.0] [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
|
9
|
Rebs S, Streckfuss-Bömeke K. How can we use stem cell-derived cardiomyocytes to understand the involvement of energetic metabolism in alterations of cardiac function? FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1222986. [PMID: 39086669 PMCID: PMC11285589 DOI: 10.3389/fmmed.2023.1222986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/15/2023] [Indexed: 08/02/2024]
Abstract
Mutations in the mitochondrial-DNA or mitochondria related nuclear-encoded-DNA lead to various multisystemic disorders collectively termed mitochondrial diseases. One in three cases of mitochondrial disease affects the heart muscle, which is called mitochondrial cardiomyopathy (MCM) and is associated with hypertrophic, dilated, and noncompact cardiomyopathy. The heart is an organ with high energy demand, and mitochondria occupy 30%-40% of its cardiomyocyte-cell volume. Mitochondrial dysfunction leads to energy depletion and has detrimental effects on cardiac performance. However, disease development and progression in the context of mitochondrial and nuclear DNA mutations, remains incompletely understood. The system of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) is an excellent platform to study MCM since the unique genetic identity to their donors enables a robust recapitulation of the predicted phenotypes in a dish on a patient-specific level. Here, we focus on recent insights into MCM studied by patient-specific iPSC-CM and further discuss research gaps and advances in metabolic maturation of iPSC-CM, which is crucial for the study of mitochondrial dysfunction and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Sabine Rebs
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Magistrati M, Gilea AI, Gerra MC, Baruffini E, Dallabona C. Drug Drop Test: How to Quickly Identify Potential Therapeutic Compounds for Mitochondrial Diseases Using Yeast Saccharomyces cerevisiae. Int J Mol Sci 2023; 24:10696. [PMID: 37445873 DOI: 10.3390/ijms241310696] [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: 05/30/2023] [Revised: 06/22/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Mitochondrial diseases (MDs) refer to a group of clinically and genetically heterogeneous pathologies characterized by defective mitochondrial function and energy production. Unfortunately, there is no effective treatment for most MDs, and current therapeutic management is limited to relieving symptoms. The yeast Saccharomyces cerevisiae has been efficiently used as a model organism to study mitochondria-related disorders thanks to its easy manipulation and well-known mitochondrial biogenesis and metabolism. It has been successfully exploited both to validate alleged pathogenic variants identified in patients and to discover potential beneficial molecules for their treatment. The so-called "drug drop test", a phenotype-based high-throughput screening, especially if coupled with a drug repurposing approach, allows the identification of molecules with high translational potential in a cost-effective and time-saving manner. In addition to drug identification, S. cerevisiae can be used to point out the drug's target or pathway. To date, drug drop tests have been successfully carried out for a variety of disease models, leading to very promising results. The most relevant aspect is that studies on more complex model organisms confirmed the effectiveness of the drugs, strengthening the results obtained in yeast and demonstrating the usefulness of this screening as a novel approach to revealing new therapeutic molecules for MDs.
Collapse
Affiliation(s)
- Martina Magistrati
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Alexandru Ionut Gilea
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Maria Carla Gerra
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Enrico Baruffini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| |
Collapse
|
11
|
Moon SH, Dilthey BG, Guan S, Sims HF, Pittman SK, Keith AL, Jenkins CM, Weihl CC, Gross RW. Genetic deletion of skeletal muscle iPLA 2γ results in mitochondrial dysfunction, muscle atrophy and alterations in whole-body energy metabolism. iScience 2023; 26:106895. [PMID: 37275531 PMCID: PMC10239068 DOI: 10.1016/j.isci.2023.106895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/28/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023] Open
Abstract
Skeletal muscle is the major site of glucose utilization in mammals integrating serum glucose clearance with mitochondrial respiration. To mechanistically elucidate the roles of iPLA2γ in skeletal muscle mitochondria, we generated a skeletal muscle-specific calcium-independent phospholipase A2γ knockout (SKMiPLA2γKO) mouse. Genetic ablation of skeletal muscle iPLA2γ resulted in pronounced muscle weakness, muscle atrophy, and increased blood lactate resulting from defects in mitochondrial function impairing metabolic processing of pyruvate and resultant bioenergetic inefficiency. Mitochondria from SKMiPLA2γKO mice were dysmorphic displaying marked changes in size, shape, and interfibrillar juxtaposition. Mitochondrial respirometry demonstrated a marked impairment in respiratory efficiency with decreases in the mass and function of oxidative phosphorylation complexes and cytochrome c. Further, a pronounced decrease in mitochondrial membrane potential and remodeling of cardiolipin molecular species were prominent. Collectively, these alterations prevented body weight gain during high-fat feeding through enhanced glucose disposal without efficient capture of chemical energy thereby altering whole-body bioenergetics.
Collapse
Affiliation(s)
- Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Beverly Gibson Dilthey
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shaoping Guan
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Harold F. Sims
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Sara K. Pittman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Amy L. Keith
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Christopher M. Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Conrad C. Weihl
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Richard W. Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Chemistry, Washington University, Saint Louis, MO 63130, USA
| |
Collapse
|
12
|
Wang S, Yazawa E, Keating EM, Mazumdar N, Hauschild A, Ma Q, Wu H, Xu Y, Shi X, Strathdee D, Gerszten RE, Schlame M, Pu WT. Genetic modifiers modulate phenotypic expression of tafazzin deficiency in a mouse model of Barth syndrome. Hum Mol Genet 2023; 32:2055-2067. [PMID: 36917259 PMCID: PMC10244222 DOI: 10.1093/hmg/ddad041] [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: 01/31/2023] [Revised: 02/27/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Barth syndrome is an X-linked disorder caused by loss-of-function mutations in Tafazzin (TAZ), an acyltransferase that catalyzes remodeling of cardiolipin, a signature phospholipid of the inner mitochondrial membrane. Patients develop cardiac and skeletal muscle weakness, growth delay and neutropenia, although phenotypic expression varies considerably between patients. Taz knockout mice recapitulate many of the hallmark features of the disease. We used mouse genetics to test the hypothesis that genetic modifiers alter the phenotypic manifestations of Taz inactivation. We crossed TazKO/X females in the C57BL6/J inbred strain to males from eight inbred strains and evaluated the phenotypes of first-generation (F1) TazKO/Y progeny, compared to TazWT/Y littermates. We observed that genetic background strongly impacted phenotypic expression. C57BL6/J and CAST/EiJ[F1] TazKO/Y mice developed severe cardiomyopathy, whereas A/J[F1] TazKO/Y mice had normal heart function. C57BL6/J and WSB/EiJ[F1] TazKO/Y mice had severely reduced treadmill endurance, whereas endurance was normal in A/J[F1] and CAST/EiJ[F1] TazKO/Y mice. In all genetic backgrounds, cardiolipin showed similar abnormalities in knockout mice, and transcriptomic and metabolomic investigations identified signatures of mitochondrial uncoupling and activation of the integrated stress response. TazKO/Y cardiac mitochondria were small, clustered and had reduced cristae density in knockouts in severely affected genetic backgrounds but were relatively preserved in the permissive A/J[F1] strain. Gene expression and mitophagy measurements were consistent with reduced mitophagy in knockout mice in genetic backgrounds intolerant of Taz mutation. Our data demonstrate that genetic modifiers powerfully modulate phenotypic expression of Taz loss-of-function and act downstream of cardiolipin, possibly by altering mitochondrial quality control.
Collapse
Affiliation(s)
- Suya Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Erika Yazawa
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Erin M Keating
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Neil Mazumdar
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Alexander Hauschild
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
| | - Haiyan Wu
- Department of Pharmacology, Sichuan University West China School of Basic Sciences and Forensic Medicine, Chengdu, Sichuan, China
| | - Yang Xu
- Department of Anesthesiology, New York University School of Medicine, New York, NY, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Douglas Strathdee
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Glasgow, UK
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Michael Schlame
- Department of Anesthesiology, New York University School of Medicine, New York, NY, USA
| | - William T Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02215NYU 10016, USA
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Glasgow, UK
- Harvard Stem Cell Institute, Harvard University, 02138 Beatson, Cambridge, MA G61 1BD, USA
| |
Collapse
|
13
|
Yang D, Li J, Li Z, Zhao M, Wang D, Sun Z, Wen P, Gou F, Dai Y, Ji Y, Li W, Zhao D, Yang L. Cardiolipin externalization mediates prion protein (PrP) peptide 106-126-associated mitophagy and mitochondrial dysfunction. Front Mol Neurosci 2023; 16:1163981. [PMID: 37333615 PMCID: PMC10272765 DOI: 10.3389/fnmol.2023.1163981] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/02/2023] [Indexed: 06/20/2023] Open
Abstract
Proper mitochondrial performance is imperative for the maintenance of normal neuronal function to prevent the development of neurodegenerative diseases. Persistent accumulation of damaged mitochondria plays a role in prion disease pathogenesis, which involves a chain of events that culminate in the generation of reactive oxygen species and neuronal death. Our previous studies have demonstrated that PINK1/Parkin-mediated mitophagy induced by PrP106-126 is defective and leads to an accumulation of damaged mitochondria after PrP106-126 treatment. Externalized cardiolipin (CL), a mitochondria-specific phospholipid, has been reported to play a role in mitophagy by directly interacting with LC3II at the outer mitochondrial membrane. The involvement of CL externalization in PrP106-126-induced mitophagy and its significance in other physiological processes of N2a cells treated with PrP106-126 remain unknown. We demonstrate that the PrP106-126 peptide caused a temporal course of mitophagy in N2a cells, which gradually increased and subsequently decreased. A similar trend in CL externalization to the mitochondrial surface was seen, resulting in a gradual decrease in CL content at the cellular level. Inhibition of CL externalization by knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3 and NDPK-D, responsible for CL translocation to the mitochondrial surface, significantly decreased PrP106-126-induced mitophagy in N2a cells. Meanwhile, the inhibition of CL redistribution significantly decreased PINK1 and DRP1 recruitment in PrP106-126 treatment but had no significant decrease in Parkin recruitment. Furthermore, the inhibition of CL externalization resulted in impaired oxidative phosphorylation and severe oxidative stress, which led to mitochondrial dysfunction. Our results indicate that CL externalization induced by PrP106-126 on N2a cells plays a positive role in the initiation of mitophagy, leading to the stabilization of mitochondrial function.
Collapse
|
14
|
Olivar-Villanueva M, Ren M, Schlame M, Phoon CK. The critical role of cardiolipin in metazoan differentiation, development, and maturation. Dev Dyn 2023; 252:691-712. [PMID: 36692477 PMCID: PMC10238668 DOI: 10.1002/dvdy.567] [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: 08/11/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Cardiolipins are phospholipids that are central to proper mitochondrial functioning. Because mitochondria play crucial roles in differentiation, development, and maturation, we would also expect cardiolipin to play major roles in these processes. Indeed, cardiolipin has been implicated in the mechanism of three human diseases that affect young infants, implying developmental abnormalities. In this review, we will: (1) Review the biology of cardiolipin; (2) Outline the evidence for essential roles of cardiolipin during organismal development, including embryogenesis and cell maturation in vertebrate organisms; (3) Place the role(s) of cardiolipin during embryogenesis within the larger context of the roles of mitochondria in development; and (4) Suggest avenues for future research.
Collapse
Affiliation(s)
| | - Mindong Ren
- Department of Anesthesiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Michael Schlame
- Department of Anesthesiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Colin K.L. Phoon
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
15
|
Quintana-Cabrera R, Scorrano L. Determinants and outcomes of mitochondrial dynamics. Mol Cell 2023; 83:857-876. [PMID: 36889315 DOI: 10.1016/j.molcel.2023.02.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/13/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023]
Abstract
Mitochondria are not only central organelles in metabolism and energy conversion but are also platforms for cellular signaling cascades. Classically, the shape and ultrastructure of mitochondria were depicted as static. The discovery of morphological transitions during cell death and of conserved genes controlling mitochondrial fusion and fission contributed to establishing the concept that mitochondrial morphology and ultrastructure are dynamically regulated by mitochondria-shaping proteins. These finely tuned, dynamic changes in mitochondrial shape can in turn control mitochondrial function, and their alterations in human diseases suggest that this space can be explored for drug discovery. Here, we review the basic tenets and molecular mechanisms of mitochondrial morphology and ultrastructure, describing how they can coordinately define mitochondrial function.
Collapse
Affiliation(s)
| | - Luca Scorrano
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy; Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy.
| |
Collapse
|
16
|
Baker MJ, Crameri JJ, Thorburn DR, Frazier AE, Stojanovski D. Mitochondrial biology and dysfunction in secondary mitochondrial disease. Open Biol 2022; 12:220274. [PMID: 36475414 PMCID: PMC9727669 DOI: 10.1098/rsob.220274] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial diseases are a broad, genetically heterogeneous class of metabolic disorders characterized by deficits in oxidative phosphorylation (OXPHOS). Primary mitochondrial disease (PMD) defines pathologies resulting from mutation of mitochondrial DNA (mtDNA) or nuclear genes affecting either mtDNA expression or the biogenesis and function of the respiratory chain. Secondary mitochondrial disease (SMD) arises due to mutation of nuclear-encoded genes independent of, or indirectly influencing OXPHOS assembly and operation. Despite instances of novel SMD increasing year-on-year, PMD is much more widely discussed in the literature. Indeed, since the implementation of next generation sequencing (NGS) techniques in 2010, many novel mitochondrial disease genes have been identified, approximately half of which are linked to SMD. This review will consolidate existing knowledge of SMDs and outline discrete categories within which to better understand the diversity of SMD phenotypes. By providing context to the biochemical and molecular pathways perturbed in SMD, we hope to further demonstrate the intricacies of SMD pathologies outside of their indirect contribution to mitochondrial energy generation.
Collapse
Affiliation(s)
- Megan J. Baker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jordan J. Crameri
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - David R. Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia,Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Ann E. Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
17
|
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
|
18
|
Corey RA, Harrison N, Stansfeld PJ, Sansom MSP, Duncan AL. Cardiolipin, and not monolysocardiolipin, preferentially binds to the interface of complexes III and IV. Chem Sci 2022; 13:13489-13498. [PMID: 36507170 PMCID: PMC9682889 DOI: 10.1039/d2sc04072g] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/25/2022] [Indexed: 12/15/2022] Open
Abstract
The mitochondrial electron transport chain comprises a series of protein complexes embedded in the inner mitochondrial membrane that generate a proton motive force via oxidative phosphorylation, ultimately generating ATP. These protein complexes can oligomerize to form larger structures called supercomplexes. Cardiolipin (CL), a conical lipid, unique within eukaryotes to the inner mitochondrial membrane, has proven essential in maintaining the stability and function of supercomplexes. Monolysocardiolipin (MLCL) is a CL variant that accumulates in people with Barth syndrome (BTHS). BTHS is caused by defects in CL biosynthesis and characterised by abnormal mitochondrial bioenergetics and destabilised supercomplexes. However, the mechanisms by which MLCL causes pathogenesis remain unclear. Here, multiscale molecular dynamics characterise the interactions of CL and MLCL with yeast and mammalian mitochondrial supercomplexes containing complex III (CIII) and complex IV (CIV). Coarse-grained simulations reveal that both CL and MLCL bind to sites at the interface between CIII and CIV of the supercomplex. Free energy perturbation calculations show that MLCL interaction is weaker than that of CL and suggest that interaction with CIV drives this difference. Atomistic contact analyses show that, although interaction with CIII is similar for CL and MLCL, CIV makes more contacts with CL than MLCL, demonstrating that CL is a more successful "glue" between the two complexes. Simulations of the human CIII2CIV supercomplex show that this interface site is maintained between species. Our study suggests that MLCL accumulation in people with BTHS disrupts supercomplex stability by formation of relatively weak interactions at the interface lipid binding site.
Collapse
Affiliation(s)
- Robin A Corey
- Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| | - Noah Harrison
- Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| | - Philllp J Stansfeld
- School of Life Sciences & Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| | - Anna L Duncan
- Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| |
Collapse
|
19
|
Greenwell AA, Tabatabaei Dakhili SA, Gopal K, Saed CT, Chan JSF, Kazungu Mugabo N, Zhabyeyev P, Eaton F, Kruger J, Oudit GY, Ussher JR. Stimulating myocardial pyruvate dehydrogenase activity fails to alleviate cardiac abnormalities in a mouse model of human Barth syndrome. Front Cardiovasc Med 2022; 9:997352. [PMID: 36211560 PMCID: PMC9537754 DOI: 10.3389/fcvm.2022.997352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Barth syndrome (BTHS) is a rare genetic disorder due to mutations in the TAFAZZIN gene, leading to impaired maturation of cardiolipin and thereby adversely affecting mitochondrial function and energy metabolism, often resulting in cardiomyopathy. In a murine model of BTHS involving short-hairpin RNA mediated knockdown of Tafazzin (TazKD mice), myocardial glucose oxidation rates were markedly reduced, likely secondary to an impairment in the activity of pyruvate dehydrogenase (PDH), the rate-limiting enzyme of glucose oxidation. Furthermore, TazKD mice exhibited cardiac hypertrophy with minimal cardiac dysfunction. Because the stimulation of myocardial glucose oxidation has been shown to alleviate diabetic cardiomyopathy and heart failure, we hypothesized that stimulating PDH activity would alleviate the cardiac hypertrophy present in TazKD mice. In order to address our hypothesis, 6-week-old male TazKD mice and their wild-type (WT) littermates were treated with dichloroacetate (DCA; 70 mM in the drinking water), which stimulates PDH activity via inhibiting PDH kinase to prevent inhibitory phosphorylation of PDH. We utilized ultrasound echocardiography to assess cardiac function and left ventricular wall structure in all mice prior to and following 6-weeks of treatment. Consistent with systemic activation of PDH and glucose oxidation, DCA treatment improved glycemia in both TazKD mice and their WT littermates, and decreased PDH phosphorylation equivalently at all 3 of its inhibitory sites (serine 293/300/232). However, DCA treatment had no impact on left ventricular structure, or systolic and diastolic function in TazKD mice. Therefore, it is unlikely that stimulating glucose oxidation is a viable target to improve BTHS-related cardiomyopathy.
Collapse
Affiliation(s)
- Amanda A. Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Christina T. Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jordan S. F. Chan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Nick Kazungu Mugabo
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Pavel Zhabyeyev
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jennifer Kruger
- Health Sciences Laboratory Animal Services, University of Alberta, Edmonton, AB, Canada
| | - Gavin Y. Oudit
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Division of Cardiology, Department of Medicine, 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
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
- *Correspondence: John R. Ussher
| |
Collapse
|
20
|
Zegallai HM, Abu-El-Rub E, Olayinka-Adefemi F, Cole LK, Sparagna GC, Marshall AJ, Hatch GM. Tafazzin deficiency in mouse mesenchymal stem cells promote reprogramming of activated B lymphocytes toward immunosuppressive phenotypes. FASEB J 2022; 36:e22443. [PMID: 35816277 DOI: 10.1096/fj.202200145r] [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: 01/25/2022] [Revised: 05/20/2022] [Accepted: 06/27/2022] [Indexed: 11/11/2022]
Abstract
Barth Syndrome (BTHS) is a rare X-linked genetic disorder caused by mutation in the TAFAZZIN gene. Tafazzin (Taz) deficiency in BTHS patients results in an increased risk of infections. Mesenchymal stem cells (MSCs) are well known for their immune-inhibitory function. We examined how Taz-deficiency in murine MSCs impact their ability to modulate the function of lipopolysaccharide (LPS)-activated wild type (WT) B lymphocytes. MSCs from tafazzin knockdown (TazKD) mice exhibited a reduction in mitochondrial cardiolipin compared to wild type (WT) MSCs. However, mitochondrial bioenergetics and membrane potential were unaltered. In contrast, TazKD MSCs exhibited increased reactive oxygen species generation and increased glycolysis. The increased glycolysis was associated with an elevated proliferation, phosphatidylinositol-3-kinase expression and expression of the immunosuppressive markers indoleamine-2,3-dioxygenase, cytotoxic T-lymphocyte-associated protein 4, interleukin-10, and cluster of differentiation 59 compared to controls. Inhibition of glycolysis with 2-deoxyglucose attenuated the TazKD-mediated increased expression of cytotoxic T-lymphocyte-associated protein 4 and interleukin-10. When co-cultured with LPS-activated WT B cells, TazKD MSCs inhibited B cell proliferation and growth rate and reduced B cell secretion of immunoglobulin M compared to controls. In addition, co-culture of LPS-activated WT B cells with TazKD MSCs promoted B cell differentiation toward interleukin-10 secreting plasma cells and B regulatory cells compared to controls. The results indicate that Taz deficiency in MSCs promote reprogramming of activated B lymphocytes toward immunosuppressive phenotypes.
Collapse
Affiliation(s)
- Hana M Zegallai
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ejlal Abu-El-Rub
- Physiology and Pathophysiology, Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan.,Physiology and Pathophysiology, Regenerative Medicine, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Folayemi Olayinka-Adefemi
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Laura K Cole
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Aaron J Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Grant M Hatch
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
21
|
Chowdhury S, Jackson L, Byrne BJ, Bryant RM, Cade WT, Churchill TL, Buchanan J, Taylor C. Longitudinal Observational Study of Cardiac Outcome Risk Factor Prediction in Children, Adolescents, and Adults with Barth Syndrome. Pediatr Cardiol 2022; 43:1251-1263. [PMID: 35238957 PMCID: PMC9462389 DOI: 10.1007/s00246-022-02846-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/04/2022] [Indexed: 12/01/2022]
Abstract
Barth Syndrome (BTHS) is an X-linked mitochondrial cardioskeletal myopathy caused by defects in TAFAZZIN, a gene responsible for cardiolipin remodeling. Altered mitochondrial levels of cardiolipin lead to cardiomyopathy (CM), muscle weakness, exercise intolerance, and mortality. Cardiac risk factors predicting outcome are unknown. Therefore, we conducted a longitudinal observational study to determine risk factors for outcome in BTHS. Subjects with minimum two evaluations (or one followed by death or transplant) were included. Cardiac size, function, and QTc data were measured by echocardiography and electrocardiography at 7 time points from 2002 to 2018. Analysis included baseline, continuous, and categorical variables. Categorical risk factors included prolonged QTc, abnormal right ventricle fractional area change (RV FAC), left ventricle (LV) or RV non-compaction, and restrictive CM phenotype. The association between variables and cardiac death or transplant (CD/TX) was assessed. Median enrollment age was 7 years (range 0.5-22; n = 44). Transplant-free survival (TFS) was 74.4% at 15 years from first evaluation. The cohort demonstrated longitudinal declines in LV size and stroke volume z-scores (end-diastolic volume, p = 0.0002; stroke volume p < 0.0001), worsening RV FAC (p = 0.0405), and global longitudinal strain (GLS) (p = 0.0001) with stable ejection (EF) and shortening (FS) fraction. CD/TX subjects (n = 9) displayed worsening LV dilation (p = 0.0066), EF (p ≤ 0.0001), FS (p = 0.0028), and RV FAC (p = .0032) versus stability in TFS. Having ≥ 2 categorical risk factors predicted CD/TX (p = 0.0073). Over 15 years, 25% of BTHS subjects progressed to CD/TX. Those with progressive LV enlargement, dysfunction, and multiple cardiac risk factors warrant increased surveillance and intense therapy.
Collapse
Affiliation(s)
| | - Lanier Jackson
- Medical University of South Carolina, Charleston, SC, USA
| | - Barry J. Byrne
- School of Medicine, University of Florida, Gainesville, FL, USA
| | | | - W. Todd Cade
- Duke University School of Medicine, Durham, NC, USA
| | | | - Julia Buchanan
- Medical University of South Carolina, Charleston, SC, USA
| | - Carolyn Taylor
- MUSC Children's Heart Program, Pediatric Echocardiography Lab, Pediatric Cardiology, Shawn Jenkins Children's Hospital, MSC 915, 10 McClennan Banks Dr., Charleston, SC, 29425-8905, USA.
| |
Collapse
|
22
|
Sohn J, Milosevic J, Brouse T, Aziz N, Elkhoury J, Wang S, Hauschild A, van Gastel N, Cetinbas M, Tufa SF, Keene DR, Sadreyev RI, Pu WT, Sykes DB. A new murine model of Barth syndrome neutropenia links TAFAZZIN deficiency to increased ER stress-induced apoptosis. Blood Adv 2022; 6:2557-2577. [PMID: 34979560 PMCID: PMC9043941 DOI: 10.1182/bloodadvances.2021005720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Barth syndrome is an inherited X-linked disorder that leads to cardiomyopathy, skeletal myopathy, and neutropenia. These symptoms result from the loss of function of the enzyme TAFAZZIN, a transacylase located in the inner mitochondrial membrane that is responsible for the final steps of cardiolipin production. The link between defective cardiolipin maturation and neutropenia remains unclear. To address potential mechanisms of neutropenia, we examined myeloid progenitor development within the fetal liver of TAFAZZIN knockout (KO) animals as well as within the adult bone marrow of wild-type recipients transplanted with TAFAZZIN-KO hematopoietic stem cells. We also used the ER-Hoxb8 system (estrogen receptor fused to Hoxb8) of conditional immortalization to establish a new murine model system for the ex vivo study of TAFAZZIN-deficient neutrophils. The TAFAZZIN-KO cells demonstrated the expected dramatic differences in cardiolipin maturation that result from a lack of TAFAZZIN enzyme activity. Contrary to our hypothesis, we did not identify any significant differences in neutrophil development or neutrophil function across a variety of assays including phagocytosis and the production of cytokines or reactive oxygen species. However, transcriptomic analysis of the TAFAZZIN-deficient neutrophil progenitors demonstrated an upregulation of markers of endoplasmic reticulum stress and confirmatory testing demonstrated that the TAFAZZIN-deficient cells had increased sensitivity to certain ER stress-mediated and non-ER stress-mediated triggers of apoptosis. Although the link between increased sensitivity to apoptosis and the variably penetrant neutropenia phenotype seen in some patients with Barth syndrome remains to be clarified, our studies and new model system set a foundation for further investigation.
Collapse
Affiliation(s)
- Jihee Sohn
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Jelena Milosevic
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Thomas Brouse
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Najihah Aziz
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Jenna Elkhoury
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Suya Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | | | - Nick van Gastel
- de Duve Institute, Brussels, Belgium
- Harvard Stem Cell Institute, Cambridge, MA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
- Department of Genetics, Harvard Medical School, Boston, MA
| | - Sara F. Tufa
- Micro-Imaging Center, Shriners Hospitals for Children, Portland, OR
| | - Douglas R. Keene
- Micro-Imaging Center, Shriners Hospitals for Children, Portland, OR
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA; and
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Massachusetts General Hospital Cancer Center, Boston, MA
| |
Collapse
|
23
|
Pang J, Bao Y, Mitchell-Silbaugh K, Veevers J, Fang X. Barth Syndrome Cardiomyopathy: An Update. Genes (Basel) 2022; 13:genes13040656. [PMID: 35456462 PMCID: PMC9030331 DOI: 10.3390/genes13040656] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 04/02/2022] [Indexed: 12/28/2022] Open
Abstract
Barth syndrome (BTHS) is an X-linked mitochondrial lipid disorder caused by mutations in the TAFAZZIN (TAZ) gene, which encodes a mitochondrial acyltransferase/transacylase required for cardiolipin (CL) biosynthesis. Cardiomyopathy is a major clinical feature of BTHS. During the past four decades, we have witnessed many landmark discoveries that have led to a greater understanding of clinical features of BTHS cardiomyopathy and their molecular basis, as well as the therapeutic targets for this disease. Recently published Taz knockout mouse models provide useful experimental models for studying BTHS cardiomyopathy and testing potential therapeutic approaches. This review aims to summarize key findings of the clinical features, molecular mechanisms, and potential therapeutic approaches for BTHS cardiomyopathy, with particular emphasis on the most recent studies.
Collapse
Affiliation(s)
- Jing Pang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Department of Biological Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Yutong Bao
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Department of Biological Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Kalia Mitchell-Silbaugh
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
| | - Jennifer Veevers
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Correspondence: ; Tel.: +1-858-246-4637
| |
Collapse
|
24
|
Chu I, Chen YC, Lai RY, Chan JF, Lee YH, Balazova M, Hsu YHH. Phosphatidylglycerol Supplementation Alters Mitochondrial Morphology and Cardiolipin Composition. MEMBRANES 2022; 12:membranes12040383. [PMID: 35448353 PMCID: PMC9028734 DOI: 10.3390/membranes12040383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023]
Abstract
The pathogenic variant of the TAZ gene is directly associated with Barth syndrome. Because tafazzin in the mitochondria is responsible for cardiolipin (CL) remodeling, all molecules related to the metabolism of CL can affect or be affected by TAZ mutation. In this study, we intend to recover the distortion of the mitochondrial lipid composition, especially CL, for Barth syndrome treatment. The genetically edited TAZ knockout HAP1 cells were demonstrated to be a suitable cellular model, where CL desaturation occurred and monolyso-CL (MLCL) was accumulated. From the species analysis by mass spectrometry, phosphatidylethanolamine showed changed species content after TAZ knockout. TAZ knockout also caused genetic down-regulation of PGS gene and up-regulation of PNPLA8 gene, which may decrease the biosynthesis of CLs and increase the hydrolysis product MLCL. Supplemented phosphatidylglycerol(18:1)2 (PG(18:1)2) was successfully biosynthesized to mature symmetrical CL and drastically decrease the concentration of MLCL to recover the morphology of mitochondria and the cristae shape of inner mitochondria. Newly synthesized mature CL may induce the down-regulation of PLA2G6 and PNPLA8 genes to potentially decrease MLCL production. The excess supplemented PG was further metabolized into phosphatidylcholine and phosphatidylethanolamine.
Collapse
Affiliation(s)
- I Chu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
| | - Ying-Chih Chen
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
| | - Ruo-Yun Lai
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
| | - Jui-Fen Chan
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
| | - Ya-Hui Lee
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
| | - Maria Balazova
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia;
| | - Yuan-Hao Howard Hsu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan; (I.C.); (Y.-C.C.); (R.-Y.L.); (J.-F.C.); (Y.-H.L.)
- Correspondence: ; Tel.: +886-4-23590121 (ext. 32230); Fax: +886-4-23590426
| |
Collapse
|
25
|
Ji J, Damschroder D, Bessert D, Lazcano P, Wessells R, Reynolds CA, Greenberg ML. NAD supplementation improves mitochondrial performance of cardiolipin mutants. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159094. [PMID: 35051613 PMCID: PMC8883178 DOI: 10.1016/j.bbalip.2021.159094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 12/01/2022]
Abstract
Cardiolipin (CL) deficiency causes mitochondrial dysfunction and aberrant metabolism that are associated in humans with the severe disease Barth syndrome (BTHS). Several metabolic abnormalities are observed in BTHS patients and model systems, including decreased oxidative phosphorylation, reduced tricarboxylic acid (TCA) cycle flux, and accumulated lactate and D-β-hydroxybutyrate, which strongly suggests that nicotinamide adenine dinucleotide (NAD) redox metabolism may be altered in CL-deficient cells. In this study, we identified abnormal NAD+ metabolism in multiple BTHS model systems and demonstrate that supplementation of NAD+ precursors such as nicotinamide mononucleotide (NMN) improves mitochondrial function. Improved mitochondrial function in the Drosophila model was associated with restored exercise endurance, which suggests a potential therapeutic benefit of NAD+ precursor supplementation in the management of BTHS patients.
Collapse
Affiliation(s)
- Jiajia Ji
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States of America
| | - Deena Damschroder
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Denise Bessert
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Pablo Lazcano
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States of America
| | - Robert Wessells
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Christian A Reynolds
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, United States of America.
| | - Miriam L Greenberg
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States of America.
| |
Collapse
|
26
|
Dudek J, Maack C. Mechano-energetic aspects of Barth syndrome. J Inherit Metab Dis 2022; 45:82-98. [PMID: 34423473 DOI: 10.1002/jimd.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022]
Abstract
Energy-demanding organs like the heart are strongly dependent on oxidative phosphorylation in mitochondria. Oxidative phosphorylation is governed by the respiratory chain located in the inner mitochondrial membrane. The inner mitochondrial membrane is the only cellular membrane with significant amounts of the phospholipid cardiolipin, and cardiolipin was found to directly interact with a number of essential protein complexes, including respiratory chain complexes I to V. An inherited defect in the biogenesis of cardiolipin causes Barth syndrome, which is associated with cardiomyopathy, skeletal myopathy, neutropenia and growth retardation. Energy conversion is dependent on reducing equivalents, which are replenished by oxidative metabolism in the Krebs cycle. Cardiolipin deficiency in Barth syndrome also affects Krebs cycle activity, metabolite transport and mitochondrial morphology. During excitation-contraction coupling, calcium (Ca2+ ) released from the sarcoplasmic reticulum drives sarcomeric contraction. At the same time, Ca2+ influx into mitochondria drives the activation of Krebs cycle dehydrogenases and the regeneration of reducing equivalents. Reducing equivalents are essential not only for energy conversion, but also for maintaining a redox buffer, which is required to detoxify reactive oxygen species (ROS). Defects in CL may also affect Ca2+ uptake into mitochondria and thereby hamper energy supply and demand matching, but also detoxification of ROS. Here, we review the impact of cardiolipin deficiency on mitochondrial function in Barth syndrome and discuss potential therapeutic strategies.
Collapse
Affiliation(s)
- Jan Dudek
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
27
|
Pu WT. Experimental models of Barth syndrome. J Inherit Metab Dis 2022; 45:72-81. [PMID: 34370877 PMCID: PMC8814986 DOI: 10.1002/jimd.12423] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023]
Abstract
Mutation of the gene Tafazzin (TAZ) causes Barth syndrome, an X-linked disorder characterized by cardiomyopathy, skeletal muscle weakness, and neutropenia. TAZ is an acyltransferase that catalyzes the remodeling of cardiolipin, the signature phospholipid of the inner mitochondrial membrane. Here, we review the major model systems that have been established to study the role of cardiolipin remodeling in mitochondrial function and the pathogenesis of Barth syndrome. We summarize key features of each model and provide examples of how each has contributed to advance our understanding of TAZ function and Barth syndrome pathophysiology.
Collapse
Affiliation(s)
- William T. Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138
- correspondence:
| |
Collapse
|
28
|
Cogliati S, Cabrera-Alarcón JL, Enriquez JA. Regulation and functional role of the electron transport chain supercomplexes. Biochem Soc Trans 2021; 49:2655-2668. [PMID: 34747989 PMCID: PMC8786287 DOI: 10.1042/bst20210460] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Mitochondria are one of the most exhaustively investigated organelles in the cell and most attention has been paid to the components of the mitochondrial electron transport chain (ETC) in the last 100 years. The ETC collects electrons from NADH or FADH2 and transfers them through a series of electron carriers within multiprotein respiratory complexes (complex I to IV) to oxygen, therefore generating an electrochemical gradient that can be used by the F1-F0-ATP synthase (also named complex V) in the mitochondrial inner membrane to synthesize ATP. The organization and function of the ETC is a continuous source of surprises. One of the latest is the discovery that the respiratory complexes can assemble to form a variety of larger structures called super-complexes (SCs). This opened an unexpected level of complexity in this well-known and fundamental biological process. This review will focus on the current evidence for the formation of different SCs and will explore how they modulate the ETC organization according to the metabolic state. Since the field is rapidly growing, we also comment on the experimental techniques used to describe these SC and hope that this overview may inspire new technologies that will help to advance the field.
Collapse
Affiliation(s)
- Sara Cogliati
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | | | - Jose Antonio Enriquez
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| |
Collapse
|
29
|
Bertero E, Nickel A, Kohlhaas M, Hohl M, Sequeira V, Brune C, Schwemmlein J, Abeßer M, Schuh K, Kutschka I, Carlein C, Münker K, Atighetchi S, Müller A, Kazakov A, Kappl R, von der Malsburg K, van der Laan M, Schiuma AF, Böhm M, Laufs U, Hoth M, Rehling P, Kuhn M, Dudek J, von der Malsburg A, Prates Roma L, Maack C. Loss of Mitochondrial Ca 2+ Uniporter Limits Inotropic Reserve and Provides Trigger and Substrate for Arrhythmias in Barth Syndrome Cardiomyopathy. Circulation 2021; 144:1694-1713. [PMID: 34648376 DOI: 10.1161/circulationaha.121.053755] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Barth syndrome (BTHS) is caused by mutations of the gene encoding tafazzin, which catalyzes maturation of mitochondrial cardiolipin and often manifests with systolic dysfunction during early infancy. Beyond the first months of life, BTHS cardiomyopathy typically transitions to a phenotype of diastolic dysfunction with preserved ejection fraction, blunted contractile reserve during exercise, and arrhythmic vulnerability. Previous studies traced BTHS cardiomyopathy to mitochondrial formation of reactive oxygen species (ROS). Because mitochondrial function and ROS formation are regulated by excitation-contraction coupling, integrated analysis of mechano-energetic coupling is required to delineate the pathomechanisms of BTHS cardiomyopathy. METHODS We analyzed cardiac function and structure in a mouse model with global knockdown of tafazzin (Taz-KD) compared with wild-type littermates. Respiratory chain assembly and function, ROS emission, and Ca2+ uptake were determined in isolated mitochondria. Excitation-contraction coupling was integrated with mitochondrial redox state, ROS, and Ca2+ uptake in isolated, unloaded or preloaded cardiac myocytes, and cardiac hemodynamics analyzed in vivo. RESULTS Taz-KD mice develop heart failure with preserved ejection fraction (>50%) and age-dependent progression of diastolic dysfunction in the absence of fibrosis. Increased myofilament Ca2+ affinity and slowed cross-bridge cycling caused diastolic dysfunction, in part, compensated by accelerated diastolic Ca2+ decay through preactivated sarcoplasmic reticulum Ca2+-ATPase. Taz deficiency provoked heart-specific loss of mitochondrial Ca2+ uniporter protein that prevented Ca2+-induced activation of the Krebs cycle during β-adrenergic stimulation, oxidizing pyridine nucleotides and triggering arrhythmias in cardiac myocytes. In vivo, Taz-KD mice displayed prolonged QRS duration as a substrate for arrhythmias, and a lack of inotropic response to β-adrenergic stimulation. Cellular arrhythmias and QRS prolongation, but not the defective inotropic reserve, were restored by inhibiting Ca2+ export through the mitochondrial Na+/Ca2+ exchanger. All alterations occurred in the absence of excess mitochondrial ROS in vitro or in vivo. CONCLUSIONS Downregulation of mitochondrial Ca2+ uniporter, increased myofilament Ca2+ affinity, and preactivated sarcoplasmic reticulum Ca2+-ATPase provoke mechano-energetic uncoupling that explains diastolic dysfunction and the lack of inotropic reserve in BTHS cardiomyopathy. Furthermore, defective mitochondrial Ca2+ uptake provides a trigger and a substrate for ventricular arrhythmias. These insights can guide the ongoing search for a cure of this orphaned disease.
Collapse
Affiliation(s)
- Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Now with Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy (E.B.)
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Mathias Hohl
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Vasco Sequeira
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Carolin Brune
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Julia Schwemmlein
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Christopher Carlein
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Kai Münker
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Sarah Atighetchi
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Andreas Müller
- Clinic for Radiology (A.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Andrey Kazakov
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Reinhard Kappl
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Karina von der Malsburg
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Martin van der Laan
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Anna-Florentine Schiuma
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Michael Böhm
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Ulrich Laufs
- Now with Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (U.L.)
| | - Markus Hoth
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, Georg-August University, Göttingen, Germany (P.R., J.D.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (P.R.).,Max-Planck Institute for Biophysical Chemistry, Göttingen, Germany (P.R.)
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Department of Cellular Biochemistry, Georg-August University, Göttingen, Germany (P.R., J.D.)
| | - Alexander von der Malsburg
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Leticia Prates Roma
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany.,Department for Internal Medicine 1, University Clinic Würzburg, Germany (C.M.)
| |
Collapse
|
30
|
Ralph-Epps T, Onu CJ, Vo L, Schmidtke MW, Le A, Greenberg ML. Studying Lipid-Related Pathophysiology Using the Yeast Model. Front Physiol 2021; 12:768411. [PMID: 34777024 PMCID: PMC8581491 DOI: 10.3389/fphys.2021.768411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 01/01/2023] Open
Abstract
Saccharomyces cerevisiae, commonly known as baker's yeast, is one of the most comprehensively studied model organisms in science. Yeast has been used to study a wide variety of human diseases, and the yeast model system has proved to be an especially amenable tool for the study of lipids and lipid-related pathophysiologies, a topic that has gained considerable attention in recent years. This review focuses on how yeast has contributed to our understanding of the mitochondrial phospholipid cardiolipin (CL) and its role in Barth syndrome (BTHS), a genetic disorder characterized by partial or complete loss of function of the CL remodeling enzyme tafazzin. Defective tafazzin causes perturbation of CL metabolism, resulting in many downstream cellular consequences and clinical pathologies that are discussed herein. The influence of yeast research in the lipid-related pathophysiologies of Alzheimer's and Parkinson's diseases is also summarized.
Collapse
Affiliation(s)
- Tyler Ralph-Epps
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Chisom J Onu
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Linh Vo
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Michael W Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Anh Le
- Muskegon Catholic Central High School, Muskegon, MI, United States
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
31
|
Abstract
Barth syndrome is a rare and potentially fatal X-linked disease characterized by cardiomyopathy, skeletal muscle weakness, growth delays, and cyclic neutropenia. Patients with Barth syndrome are prone to high risk of mortality in infancy and the development of cardiomyopathy with severe weakening of the immune system. Elamipretide is a water-soluble, aromatic-cationic, mitochondria-targeting tetrapeptide that readily penetrates and transiently localizes to the inner mitochondrial membrane. Therapy with elamipretide facilitates cell health by improving energy production and inhibiting excessive formation of reactive oxygen species, thus alleviating oxidative stress. Elamipretide crosses the outer membrane of the mitochondrion and becomes associated with cardiolipin, a constituent phospholipid of the inner membrane. Elamipretide improves mitochondrial bioenergetics and morphology rapidly in induced pluripotent stem cells from patients with Barth syndrome and other genetically related diseases characterized by pediatric cardiomyopathy. Data with elamipretide across multiple models of disease are especially promising, with results from several studies supporting the use of elamipretide as potential therapy for patients with Barth syndrome, particularly where there is a confirmed diagnosis of cardiomyopathy. This review highlights the challenges and opportunities presented in treating Barth syndrome cardiomyopathy patients with elamipretide and addresses evidence supporting the durability of effect of elamipretide as a therapeutic agent for Barth syndrome, especially its likely durable effects on progression of cardiomyopathy following the cessation of drug treatment and the capability of elamipretide to structurally reverse remodel the failing left ventricle at the global, cellular, and molecular level in a gradual manner through specific targeting of the mitochondrial inner membrane.
Collapse
|
32
|
Pánek T, Eliáš M, Vancová M, Lukeš J, Hashimi H. Returning to the Fold for Lessons in Mitochondrial Crista Diversity and Evolution. Curr Biol 2021; 30:R575-R588. [PMID: 32428499 DOI: 10.1016/j.cub.2020.02.053] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cristae are infoldings of the mitochondrial inner membrane jutting into the organelle's innermost compartment from narrow stems at their base called crista junctions. They are emblematic of aerobic mitochondria, being the fabric for the molecular machinery driving cellular respiration. Electron microscopy revealed that diverse eukaryotes possess cristae of different shapes. Yet, crista diversity has not been systematically examined in light of our current knowledge about eukaryotic evolution. Since crista form and function are intricately linked, we take a holistic view of factors that may underlie both crista diversity and the adherence of cristae to a recognizable form. Based on electron micrographs of 226 species from all major lineages, we propose a rational crista classification system that postulates cristae as variations of two general morphotypes: flat and tubulo-vesicular. The latter is most prevalent and likely ancestral, but both morphotypes are found interspersed throughout the eukaryotic tree. In contrast, crista junctions are remarkably conserved, supporting their proposed role as diffusion barriers that sequester cristae contents. Since cardiolipin, ATP synthase dimers, the MICOS complex, and dynamin-like Opa1/Mgm1 are known to be involved in shaping cristae, we examined their variation in the context of crista diversity. Moreover, we have identified both commonalities and differences that may collectively be manifested as diverse variations of crista form and function.
Collapse
Affiliation(s)
- Tomáš Pánek
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava 710 00, Czech Republic
| | - Marek Eliáš
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava 710 00, Czech Republic
| | - Marie Vancová
- Institute of Parasitology, Biology Center, Czech Academy of Sciences and Faculty of Science, University of South Bohemia, České Budějovice 370 05, Czech Republic
| | - Julius Lukeš
- Institute of Parasitology, Biology Center, Czech Academy of Sciences and Faculty of Science, University of South Bohemia, České Budějovice 370 05, Czech Republic
| | - Hassan Hashimi
- Institute of Parasitology, Biology Center, Czech Academy of Sciences and Faculty of Science, University of South Bohemia, České Budějovice 370 05, Czech Republic.
| |
Collapse
|
33
|
Marchal GA, Jouni M, Chiang DY, Pérez-Hernández M, Podliesna S, Yu N, Casini S, Potet F, Veerman CC, Klerk M, Lodder EM, Mengarelli I, Guan K, Vanoye CG, Rothenberg E, Charpentier F, Redon R, George AL, Verkerk AO, Bezzina CR, MacRae CA, Burridge PW, Delmar M, Galjart N, Portero V, Remme CA. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na V1.5 at Intercalated Discs. Circ Res 2021; 129:349-365. [PMID: 34092082 PMCID: PMC8298292 DOI: 10.1161/circresaha.120.318643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mariam Jouni
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - David Y Chiang
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | | | - Svitlana Podliesna
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Nuo Yu
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Franck Potet
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Christiaan C Veerman
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany (K.G.)
| | - Carlos G Vanoye
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology (E.R.), NYU School of Medicine
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Richard Redon
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Alfred L George
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | - Paul W Burridge
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Mario Delmar
- Division of Cardiology (M.P.-H., M.D.), NYU School of Medicine
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| |
Collapse
|
34
|
McKnight CL, Low YC, Elliott DA, Thorburn DR, Frazier AE. Modelling Mitochondrial Disease in Human Pluripotent Stem Cells: What Have We Learned? Int J Mol Sci 2021; 22:7730. [PMID: 34299348 PMCID: PMC8306397 DOI: 10.3390/ijms22147730] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial diseases disrupt cellular energy production and are among the most complex group of inherited genetic disorders. Affecting approximately 1 in 5000 live births, they are both clinically and genetically heterogeneous, and can be highly tissue specific, but most often affect cell types with high energy demands in the brain, heart, and kidneys. There are currently no clinically validated treatment options available, despite several agents showing therapeutic promise. However, modelling these disorders is challenging as many non-human models of mitochondrial disease do not completely recapitulate human phenotypes for known disease genes. Additionally, access to disease-relevant cell or tissue types from patients is often limited. To overcome these difficulties, many groups have turned to human pluripotent stem cells (hPSCs) to model mitochondrial disease for both nuclear-DNA (nDNA) and mitochondrial-DNA (mtDNA) contexts. Leveraging the capacity of hPSCs to differentiate into clinically relevant cell types, these models permit both detailed investigation of cellular pathomechanisms and validation of promising treatment options. Here we catalogue hPSC models of mitochondrial disease that have been generated to date, summarise approaches and key outcomes of phenotypic profiling using these models, and discuss key criteria to guide future investigations using hPSC models of mitochondrial disease.
Collapse
Affiliation(s)
- Cameron L. McKnight
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Yau Chung Low
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - David A. Elliott
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - David R. Thorburn
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- Victorian Clinical Genetics Services, Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Ann E. Frazier
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
35
|
Dudek J, Kutschka I, Maack C. Metabolic and Redox Regulation of Cardiovascular Stem Cell Biology and Pathology. Antioxid Redox Signal 2021; 35:163-181. [PMID: 33121253 DOI: 10.1089/ars.2020.8201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Cardiovascular stem cells are important for regeneration and repair of damaged tissue. Recent Advances: Pluripotent stem cells have a unique metabolism, which is adopted for their energetic and biosynthetic demand as rapidly proliferating cells. Stem cell differentiation requires an exceptional metabolic flexibility allowing for metabolic remodeling between glycolysis and oxidative phosphorylation. Critical Issues: Respiration is associated with the generation of reactive oxygen species (ROS) by the mitochondrial respiratory chain. But also the membrane-bound protein nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, NOX) contributes to ROS levels. ROS not only play a significant role in stem cell differentiation and tissue renewal but also cause senescence and contribute to tissue aging. Future Directions: For utilization of stem cells in therapeutic approaches, a deep understanding of the molecular mechanisms how metabolism and the cellular redox state regulate stem cell differentiation is required. Modulating the redox state of stem cells using antioxidative agents may be suitable to enhance activity of endothelial progenitor cells. Antioxid. Redox Signal. 35, 163-181.
Collapse
Affiliation(s)
- Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
36
|
Azar J, Bahmad HF, Daher D, Moubarak MM, Hadadeh O, Monzer A, Al Bitar S, Jamal M, Al-Sayegh M, Abou-Kheir W. The Use of Stem Cell-Derived Organoids in Disease Modeling: An Update. Int J Mol Sci 2021; 22:7667. [PMID: 34299287 PMCID: PMC8303386 DOI: 10.3390/ijms22147667] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Organoids represent one of the most important advancements in the field of stem cells during the past decade. They are three-dimensional in vitro culturing models that originate from self-organizing stem cells and can mimic the in vivo structural and functional specificities of body organs. Organoids have been established from multiple adult tissues as well as pluripotent stem cells and have recently become a powerful tool for studying development and diseases in vitro, drug screening, and host-microbe interaction. The use of stem cells-that have self-renewal capacity to proliferate and differentiate into specialized cell types-for organoids culturing represents a major advancement in biomedical research. Indeed, this new technology has a great potential to be used in a multitude of fields, including cancer research, hereditary and infectious diseases. Nevertheless, organoid culturing is still rife with many challenges, not limited to being costly and time consuming, having variable rates of efficiency in generation and maintenance, genetic stability, and clinical applications. In this review, we aim to provide a synopsis of pluripotent stem cell-derived organoids and their use for disease modeling and other clinical applications.
Collapse
Affiliation(s)
- Joseph Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Darine Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Maya M. Moubarak
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Samar Al Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| | - Mohamed Jamal
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 66566, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2260, Lebanon; (J.A.); (H.F.B.); (D.D.); (M.M.M.); (O.H.); (A.M.); (S.A.B.)
| |
Collapse
|
37
|
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: 2.3] [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
|
38
|
Zhu S, Chen Z, Zhu M, Shen Y, Leon LJ, Chi L, Spinozzi S, Tan C, Gu Y, Nguyen A, Zhou Y, Feng W, Vaz FM, Wang X, Gustafsson AB, Evans SM, Kunfu O, Fang X. Cardiolipin Remodeling Defects Impair Mitochondrial Architecture and Function in a Murine Model of Barth Syndrome Cardiomyopathy. Circ Heart Fail 2021; 14:e008289. [PMID: 34129362 PMCID: PMC8210459 DOI: 10.1161/circheartfailure.121.008289] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiomyopathy is a major clinical feature in Barth syndrome (BTHS), an X-linked mitochondrial lipid disorder caused by mutations in Tafazzin (TAZ), encoding a mitochondrial acyltransferase required for cardiolipin remodeling. Despite recent description of a mouse model of BTHS cardiomyopathy, an in-depth analysis of specific lipid abnormalities and mitochondrial form and function in an in vivo BTHS cardiomyopathy model is lacking. METHODS We performed in-depth assessment of cardiac function, cardiolipin species profiles, and mitochondrial structure and function in our newly generated Taz cardiomyocyte-specific knockout mice and Cre-negative control mice (n≥3 per group). RESULTS Taz cardiomyocyte-specific knockout mice recapitulate typical features of BTHS and mitochondrial cardiomyopathy. Fewer than 5% of cardiomyocyte-specific knockout mice exhibited lethality before 2 months of age, with significantly enlarged hearts. More than 80% of cardiomyocyte-specific knockout displayed ventricular dilation at 16 weeks of age and survived until 50 weeks of age. Full parameter analysis of cardiac cardiolipin profiles demonstrated lower total cardiolipin concentration, abnormal cardiolipin fatty acyl composition, and elevated monolysocardiolipin to cardiolipin ratios in Taz cardiomyocyte-specific knockout, relative to controls. Mitochondrial contact site and cristae organizing system and F1F0-ATP synthase complexes, required for cristae morphogenesis, were abnormal, resulting in onion-shaped mitochondria. Organization of high molecular weight respiratory chain supercomplexes was also impaired. In keeping with observed mitochondrial abnormalities, seahorse experiments demonstrated impaired mitochondrial respiration capacity. CONCLUSIONS Our mouse model mirrors multiple physiological and biochemical aspects of BTHS cardiomyopathy. Our results give important insights into the underlying cause of BTHS cardiomyopathy and provide a framework for testing therapeutic approaches to BTHS cardiomyopathy, or other mitochondrial-related cardiomyopathies.
Collapse
Affiliation(s)
- Siting Zhu
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- 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
| | - Ze’e Chen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- 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
| | - Mason Zhu
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Ying Shen
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Leonardo J Leon
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Liguo Chi
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Simone Spinozzi
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Changming Tan
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yusu Gu
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Anh Nguyen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Yi Zhou
- Department of Molecular Biology, University of California, San Diego, La Jolla, California, USA
| | - Wei Feng
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Asa B Gustafsson
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Sylvia M Evans
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Ouyang Kunfu
- 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
| | - Xi Fang
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
39
|
Bennett CF, O’Malley KE, Perry EA, Balsa E, Latorre-Muro P, Riley CL, Luo C, Jedrychowski M, Gygi SP, Puigserver P. Peroxisomal-derived ether phospholipids link nucleotides to respirasome assembly. Nat Chem Biol 2021; 17:703-710. [PMID: 33723432 PMCID: PMC8159895 DOI: 10.1038/s41589-021-00772-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
The protein complexes of the mitochondrial electron transport chain exist in isolation and in higher order assemblies termed supercomplexes (SCs) or respirasomes (SC I+III2+IV). The association of complexes I, III and IV into the respirasome is regulated by unknown mechanisms. Here, we designed a nanoluciferase complementation reporter for complex III and IV proximity to determine in vivo respirasome levels. In a chemical screen, we found that inhibitors of the de novo pyrimidine synthesis enzyme dihydroorotate dehydrogenase (DHODH) potently increased respirasome assembly and activity. By-passing DHODH inhibition via uridine supplementation decreases SC assembly by altering mitochondrial phospholipid composition, specifically elevated peroxisomal-derived ether phospholipids. Cell growth rates upon DHODH inhibition depend on ether lipid synthesis and SC assembly. These data reveal that nucleotide pools signal to peroxisomes to modulate synthesis and transport of ether phospholipids to mitochondria for SC assembly, which are necessary for optimal cell growth in conditions of nucleotide limitation.
Collapse
Affiliation(s)
- Christopher F. Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine E. O’Malley
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A. Perry
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher L. Riley
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Correspondence:
| |
Collapse
|
40
|
Liu X, Wang S, Guo X, Li Y, Ogurlu R, Lu F, Prondzynski M, Buzon SDLS, Ma Q, Zhang D, Wang G, Cotton J, Guo Y, Xiao L, Milan DJ, Xu Y, Schlame M, Bezzerides VJ, Pu WT. Increased Reactive Oxygen Species-Mediated Ca 2+/Calmodulin-Dependent Protein Kinase II Activation Contributes to Calcium Handling Abnormalities and Impaired Contraction in Barth Syndrome. Circulation 2021; 143:1894-1911. [PMID: 33793303 PMCID: PMC8691127 DOI: 10.1161/circulationaha.120.048698] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mutations in tafazzin (TAZ), a gene required for biogenesis of cardiolipin, the signature phospholipid of the inner mitochondrial membrane, causes Barth syndrome (BTHS). Cardiomyopathy and risk of sudden cardiac death are prominent features of BTHS, but the mechanisms by which impaired cardiolipin biogenesis causes cardiac muscle weakness and arrhythmia are poorly understood. METHODS We performed in vivo electrophysiology to define arrhythmia vulnerability in cardiac-specific TAZ knockout mice. Using cardiomyocytes derived from human induced pluripotent stem cells and cardiac-specific TAZ knockout mice as model systems, we investigated the effect of TAZ inactivation on Ca2+ handling. Through genome editing and pharmacology, we defined a molecular link between TAZ mutation and abnormal Ca2+ handling and contractility. RESULTS A subset of mice with cardiac-specific TAZ inactivation developed arrhythmias, including bidirectional ventricular tachycardia, atrial tachycardia, and complete atrioventricular block. Compared with wild-type controls, BTHS-induced pluripotent stem cell-derived cardiomyocytes had increased diastolic Ca2+ and decreased Ca2+ transient amplitude. BTHS-induced pluripotent stem cell-derived cardiomyocytes had higher levels of mitochondrial and cellular reactive oxygen species than wild-type controls, which activated CaMKII (Ca2+/calmodulin-dependent protein kinase II). Activated CaMKII phosphorylated the RYR2 (ryanodine receptor 2) on serine 2814, increasing Ca2+ leak through RYR2. Inhibition of this reactive oxygen species-CaMKII-RYR2 pathway through pharmacological inhibitors or genome editing normalized aberrant Ca2+ handling in BTHS-induced pluripotent stem cell-derived cardiomyocytes and improved their contractile function. Murine Taz knockout cardiomyocytes also exhibited elevated diastolic Ca2+ and decreased Ca2+ transient amplitude. These abnormalities were ameliorated by Ca2+/calmodulin-dependent protein kinase II or reactive oxygen species inhibition. CONCLUSIONS This study identified a molecular pathway that links TAZ mutation with abnormal Ca2+ handling and decreased cardiomyocyte contractility. This pathway may offer therapeutic opportunities to treat BTHS and potentially other diseases with elevated mitochondrial reactive oxygen species production.
Collapse
Affiliation(s)
- Xujie Liu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Radiology, Basic Medical School, Chongqing Medical University, Chongqing, 400016, China
| | - Suya Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Xiaoling Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Center of Scientific Research, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Roza Ogurlu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | | | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Donghui Zhang
- State key laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China
| | - Gang Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Justin Cotton
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard College, Cambridge, MA 02138, USA
| | - Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ling Xiao
- Department of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David J. Milan
- Department of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yang Xu
- Department of Anesthesiology, New York University School of Medicine, New York, New York
| | - Michael Schlame
- Department of Anesthesiology, New York University School of Medicine, New York, New York
| | | | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
41
|
Hernansanz-Agustín P, Enríquez JA. Functional segmentation of CoQ and cyt c pools by respiratory complex superassembly. Free Radic Biol Med 2021; 167:232-242. [PMID: 33722627 DOI: 10.1016/j.freeradbiomed.2021.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 12/25/2022]
Abstract
Electron transfer between respiratory complexes is an essential step for the efficiency of the mitochondrial oxidative phosphorylation. Until recently, it was stablished that ubiquinone and cytochrome c formed homogenous single pools in the inner mitochondrial membrane which were not influenced by the presence of respiratory supercomplexes. However, this idea was challenged by the fact that bottlenecks in electron transfer appeared after disruption of supercomplexes into their individual complexes. The postulation of the plasticity model embraced all these observations and concluded that complexes and supercomplexes co-exist and are dedicated to a spectrum of metabolic requirements. Here, we review the involvement of superassembly in complex I stability, the role of supercomplexes in ROS production and the segmentation of the CoQ and cyt c pools, together with their involvement in signaling and disease. Taking apparently conflicting literature we have built up a comprehensive model for the segmentation of CoQ and cyt c mediated by supercomplexes, discuss the current limitations and provide a prospect of the current knowledge in the field.
Collapse
Affiliation(s)
- Pablo Hernansanz-Agustín
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III CNIC, Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| | - José Antonio Enríquez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III CNIC, Melchor Fernández Almagro 3, Madrid, 28029, Spain; Centro de Investigaciones Biomédicas en Red de Fragilidad y Envejecimiento Saludable-CIBERFES. Av. Monforte de Lemos, 3-5. Pabellón 11, Planta 0 28029, Madrid, Spain.
| |
Collapse
|
42
|
Ramaccini D, Montoya-Uribe V, Aan FJ, Modesti L, Potes Y, Wieckowski MR, Krga I, Glibetić M, Pinton P, Giorgi C, Matter ML. Mitochondrial Function and Dysfunction in Dilated Cardiomyopathy. Front Cell Dev Biol 2021; 8:624216. [PMID: 33511136 PMCID: PMC7835522 DOI: 10.3389/fcell.2020.624216] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiac tissue requires a persistent production of energy in order to exert its pumping function. Therefore, the maintenance of this function relies on mitochondria that represent the “powerhouse” of all cardiac activities. Mitochondria being one of the key players for the proper functioning of the mammalian heart suggests continual regulation and organization. Mitochondria adapt to cellular energy demands via fusion-fission events and, as a proof-reading ability, undergo mitophagy in cases of abnormalities. Ca2+ fluxes play a pivotal role in regulating all mitochondrial functions, including ATP production, metabolism, oxidative stress balance and apoptosis. Communication between mitochondria and others organelles, especially the sarcoplasmic reticulum is required for optimal function. Consequently, abnormal mitochondrial activity results in decreased energy production leading to pathological conditions. In this review, we will describe how mitochondrial function or dysfunction impacts cardiac activities and the development of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Daniela Ramaccini
- University of Hawaii Cancer Center, Honolulu, HI, United States.,Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | | - Femke J Aan
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Lorenzo Modesti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Irena Krga
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Marija Glibetić
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | |
Collapse
|
43
|
Verkerk AO, Knottnerus SJG, Portero V, Bleeker JC, Ferdinandusse S, Guan K, IJlst L, Visser G, Wanders RJA, Wijburg FA, Bezzina CR, Mengarelli I, Houtkooper RH. Electrophysiological Abnormalities in VLCAD Deficient hiPSC-Cardiomyocytes Do not Improve with Carnitine Supplementation. Front Pharmacol 2021; 11:616834. [PMID: 33597881 PMCID: PMC7883678 DOI: 10.3389/fphar.2020.616834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Patients with a deficiency in very long-chain acyl-CoA dehydrogenase (VLCAD), an enzyme that is involved in the mitochondrial beta-oxidation of long-chain fatty acids, are at risk for developing cardiac arrhythmias. In human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs), VLCAD deficiency (VLCADD) results in a series of abnormalities, including: 1) accumulation of long-chain acylcarnitines, 2) action potential shortening, 3) higher systolic and diastolic intracellular Ca2+ concentrations, and 4) development of delayed afterdepolarizations. In the fatty acid oxidation process, carnitine is required for bidirectional transport of acyl groups across the mitochondrial membrane. Supplementation has been suggested as potential therapeutic approach in VLCADD, but its benefits are debated. Here, we studied the effects of carnitine supplementation on the long-chain acylcarnitine levels and performed electrophysiological analyses in VLCADD patient-derived hiPSC-CMs with a ACADVL gene mutation (p.Val283Ala/p.Glu381del). Under standard culture conditions, VLCADD hiPSC-CMs showed high concentrations of long-chain acylcarnitines, short action potentials, and high delayed afterdepolarizations occurrence. Incubation of the hiPSC-CMs with 400 µM L-carnitine for 48 h led to increased long-chain acylcarnitine levels both in medium and cells. In addition, carnitine supplementation neither restored abnormal action potential parameters nor the increased occurrence of delayed afterdepolarizations in VLCADD hiPSC-CMs. We conclude that long-chain acylcarnitine accumulation and electrophysiological abnormalities in VLCADD hiPSC-CMs are not normalized by carnitine supplementation, indicating that this treatment is unlikely to be beneficial against cardiac arrhythmias in VLCADD patients.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Suzan J G Knottnerus
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Vincent Portero
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jeannette C Bleeker
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Gepke Visser
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Isabella Mengarelli
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|
44
|
Goncalves RLS, Schlame M, Bartelt A, Brand MD, Hotamışlıgil GS. Cardiolipin deficiency in Barth syndrome is not associated with increased superoxide/H 2 O 2 production in heart and skeletal muscle mitochondria. FEBS Lett 2020; 595:415-432. [PMID: 33112430 PMCID: PMC7894513 DOI: 10.1002/1873-3468.13973] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 12/11/2022]
Abstract
Barth syndrome (BTHS) is a rare X-linked genetic disorder caused by mutations in the gene encoding the transacylase tafazzin and characterized by loss of cardiolipin and severe cardiomyopathy. Mitochondrial oxidants have been implicated in the cardiomyopathy in BTHS. Eleven mitochondrial sites produce superoxide/hydrogen peroxide (H2 O2 ) at significant rates. Which of these sites generate oxidants at excessive rates in BTHS is unknown. Here, we measured the maximum capacity of superoxide/H2 O2 production from each site and the ex vivo rate of superoxide/H2 O2 production in the heart and skeletal muscle mitochondria of the tafazzin knockdown mice (tazkd) from 3 to 12 months of age. Despite reduced oxidative capacity, superoxide/H2 O2 production was indistinguishable between tazkd mice and wild-type littermates. These observations raise questions about the involvement of mitochondrial oxidants in BTHS pathology.
Collapse
Affiliation(s)
- Renata L S Goncalves
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael Schlame
- Departments of Anesthesiology and Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Alexander Bartelt
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
45
|
Metabolic Alterations Caused by Defective Cardiolipin Remodeling in Inherited Cardiomyopathies. Life (Basel) 2020; 10:life10110277. [PMID: 33187128 PMCID: PMC7697959 DOI: 10.3390/life10110277] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/21/2022] Open
Abstract
The heart is the most energy-consuming organ in the human body. In heart failure, the homeostasis of energy supply and demand is endangered by an increase in cardiomyocyte workload, or by an insufficiency in energy-providing processes. Energy metabolism is directly associated with mitochondrial redox homeostasis. The production of toxic reactive oxygen species (ROS) may overwhelm mitochondrial and cellular ROS defense mechanisms in case of heart failure. Mitochondria are essential cell organelles and provide 95% of the required energy in the heart. Metabolic remodeling, changes in mitochondrial structure or function, and alterations in mitochondrial calcium signaling diminish mitochondrial energy provision in many forms of cardiomyopathy. The mitochondrial respiratory chain creates a proton gradient across the inner mitochondrial membrane, which couples respiration with oxidative phosphorylation and the preservation of energy in the chemical bonds of ATP. Akin to other mitochondrial enzymes, the respiratory chain is integrated into the inner mitochondrial membrane. The tight association with the mitochondrial phospholipid cardiolipin (CL) ensures its structural integrity and coordinates enzymatic activity. This review focuses on how changes in mitochondrial CL may be associated with heart failure. Dysfunctional CL has been found in diabetic cardiomyopathy, ischemia reperfusion injury and the aging heart. Barth syndrome (BTHS) is caused by an inherited defect in the biosynthesis of cardiolipin. Moreover, a dysfunctional CL pool causes other types of rare inherited cardiomyopathies, such as Sengers syndrome and Dilated Cardiomyopathy with Ataxia (DCMA). Here we review the impact of cardiolipin deficiency on mitochondrial functions in cellular and animal models. We describe the molecular mechanisms concerning mitochondrial dysfunction as an incitement of cardiomyopathy and discuss potential therapeutic strategies.
Collapse
|
46
|
Le CH, Benage LG, Specht KS, Li Puma LC, Mulligan CM, Heuberger AL, Prenni JE, Claypool SM, Chatfield KC, Sparagna GC, Chicco AJ. Tafazzin deficiency impairs CoA-dependent oxidative metabolism in cardiac mitochondria. J Biol Chem 2020; 295:12485-12497. [PMID: 32665401 DOI: 10.1074/jbc.ra119.011229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 07/07/2020] [Indexed: 12/20/2022] Open
Abstract
Barth syndrome is a mitochondrial myopathy resulting from mutations in the tafazzin (TAZ) gene encoding a phospholipid transacylase required for cardiolipin remodeling. Cardiolipin is a phospholipid of the inner mitochondrial membrane essential for the function of numerous mitochondrial proteins and processes. However, it is unclear how tafazzin deficiency impacts cardiac mitochondrial metabolism. To address this question while avoiding confounding effects of cardiomyopathy on mitochondrial phenotype, we utilized Taz-shRNA knockdown (TazKD ) mice, which exhibit defective cardiolipin remodeling and respiratory supercomplex instability characteristic of human Barth syndrome but normal cardiac function into adulthood. Consistent with previous reports from other models, mitochondrial H2O2 emission and oxidative damage were greater in TazKD than in wild-type (WT) hearts, but there were no differences in oxidative phosphorylation coupling efficiency or membrane potential. Fatty acid and pyruvate oxidation capacities were 40-60% lower in TazKD mitochondria, but an up-regulation of glutamate oxidation supported respiration rates approximating those with pyruvate and palmitoylcarnitine in WT. Deficiencies in mitochondrial CoA and shifts in the cardiac acyl-CoA profile paralleled changes in fatty acid oxidation enzymes and acyl-CoA thioesterases, suggesting limitations of CoA availability or "trapping" in TazKD mitochondrial metabolism. Incubation of TazKD mitochondria with exogenous CoA partially rescued pyruvate and palmitoylcarnitine oxidation capacities, implicating dysregulation of CoA-dependent intermediary metabolism rather than respiratory chain defects in the bioenergetic impacts of tafazzin deficiency. These findings support links among cardiolipin abnormalities, respiratory supercomplex instability, and mitochondrial oxidant production and shed new light on the distinct metabolic consequences of tafazzin deficiency in the mammalian heart.
Collapse
Affiliation(s)
- Catherine H Le
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, Colorado, USA
| | - Lindsay G Benage
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Kalyn S Specht
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Lance C Li Puma
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Christopher M Mulligan
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado, USA
| | - Adam L Heuberger
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, Colorado, USA
| | - Jessica E Prenni
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, Colorado, USA
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathryn C Chatfield
- Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Genevieve C Sparagna
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adam J Chicco
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, Colorado, USA .,Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.,Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
47
|
Colina-Tenorio L, Horten P, Pfanner N, Rampelt H. Shaping the mitochondrial inner membrane in health and disease. J Intern Med 2020; 287:645-664. [PMID: 32012363 DOI: 10.1111/joim.13031] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/19/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Mitochondria play central roles in cellular energetics, metabolism and signalling. Efficient respiration, mitochondrial quality control, apoptosis and inheritance of mitochondrial DNA depend on the proper architecture of the mitochondrial membranes and a dynamic remodelling of inner membrane cristae. Defects in mitochondrial architecture can result in severe human diseases affecting predominantly the nervous system and the heart. Inner membrane morphology is generated and maintained in particular by the mitochondrial contact site and cristae organizing system (MICOS), the F1 Fo -ATP synthase, the fusion protein OPA1/Mgm1 and the nonbilayer-forming phospholipids cardiolipin and phosphatidylethanolamine. These protein complexes and phospholipids are embedded in a network of functional interactions. They communicate with each other and additional factors, enabling them to balance different aspects of cristae biogenesis and to dynamically remodel the inner mitochondrial membrane. Genetic alterations disturbing these membrane-shaping factors can lead to human pathologies including fatal encephalopathy, dominant optic atrophy, Leigh syndrome, Parkinson's disease and Barth syndrome.
Collapse
Affiliation(s)
- L Colina-Tenorio
- From the, Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - P Horten
- From the, Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - N Pfanner
- From the, Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - H Rampelt
- From the, Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Panuzzo C, Jovanovski A, Pergolizzi B, Pironi L, Stanga S, Fava C, Cilloni D. Mitochondria: A Galaxy in the Hematopoietic and Leukemic Stem Cell Universe. Int J Mol Sci 2020; 21:ijms21113928. [PMID: 32486249 PMCID: PMC7312164 DOI: 10.3390/ijms21113928] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the main fascinating energetic source into the cells. Their number, shape, and dynamism are controlled by the cell’s type and current behavior. The perturbation of the mitochondrial inward system via stress response and/or oncogenic insults could activate several trafficking molecular mechanisms with the intention to solve the problem. In this review, we aimed to clarify the crucial pathways in the mitochondrial system, dissecting the different metabolic defects, with a special emphasis on hematological malignancies. We investigated the pivotal role of mitochondria in the maintenance of hematopoietic stem cells (HSCs) and their main alterations that could induce malignant transformation, culminating in the generation of leukemic stem cells (LSCs). In addition, we presented an overview of LSCs mitochondrial dysregulated mechanisms in terms of (1) increasing in oxidative phosphorylation program (OXPHOS), as a crucial process for survival and self-renewal of LSCs,(2) low levels of reactive oxygen species (ROS), and (3) aberrant expression of B-cell lymphoma 2 (Bcl-2) with sustained mitophagy. Furthermore, these peculiarities may represent attractive new “hot spots” for mitochondrial-targeted therapy. Finally, we remark the potential of the LCS metabolic effectors to be exploited as novel therapeutic targets.
Collapse
Affiliation(s)
- Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
- Correspondence: (C.P.); (D.C.)
| | - Aleksandar Jovanovski
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Lucrezia Pironi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
| | - Carmen Fava
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
- Correspondence: (C.P.); (D.C.)
| |
Collapse
|
49
|
Guo J, Huebsch N. Modeling the Response of Heart Muscle to Mechanical Stimulation In Vitro. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43152-020-00007-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
50
|
Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RWJ, Krotenberg Garcia A, Mircea M, Kostidis S, Davis RP, van Meer BJ, Jost CR, Koster AJ, Mei H, Míguez DG, Mulder AA, Ledesma-Terrón M, Pompilio G, Sala L, Salvatori DCF, Slieker RC, Sommariva E, de Vries AAF, Giera M, Semrau S, Tertoolen LGJ, Orlova VV, Bellin M, Mummery CL. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell 2020; 26:862-879.e11. [PMID: 32459996 PMCID: PMC7284308 DOI: 10.1016/j.stem.2020.05.004] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/05/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022]
Abstract
Cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) are functionally immature, but this is improved by incorporation into engineered tissues or forced contraction. Here, we showed that tri-cellular combinations of hiPSC-derived CMs, cardiac fibroblasts (CFs), and cardiac endothelial cells also enhance maturation in easily constructed, scaffold-free, three-dimensional microtissues (MTs). hiPSC-CMs in MTs with CFs showed improved sarcomeric structures with T-tubules, enhanced contractility, and mitochondrial respiration and were electrophysiologically more mature than MTs without CFs. Interactions mediating maturation included coupling between hiPSC-CMs and CFs through connexin 43 (CX43) gap junctions and increased intracellular cyclic AMP (cAMP). Scaled production of thousands of hiPSC-MTs was highly reproducible across lines and differentiated cell batches. MTs containing healthy-control hiPSC-CMs but hiPSC-CFs from patients with arrhythmogenic cardiomyopathy strikingly recapitulated features of the disease. Our MT model is thus a simple and versatile platform for modeling multicellular cardiac diseases that will facilitate industry and academic engagement in high-throughput molecular screening. Cardiac fibroblasts and endothelial cells induce hiPSC-cardiomyocyte maturation CX43 gap junctions form between cardiac fibroblasts and cardiomyocytes cAMP-pathway activation contributes to hiPSC-cardiomyocyte maturation Patient-derived hiPSC-cardiac fibroblasts cause arrhythmia in microtissues
Collapse
Affiliation(s)
- Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Amy Cochrane
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Xu Cao
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ruben W J van Helden
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ana Krotenberg Garcia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Maria Mircea
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Carolina R Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Abraham J Koster
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - David G Míguez
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Mario Ledesma-Terrón
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Luca Sala
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Daniela C F Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Epidemiology and Biostatistics, Amsterdam Public Health Institute, VU University Medical Center, 1007 Amsterdam, the Netherlands
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Antoine A F de Vries
- Department of Cardiology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Stefan Semrau
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Leon G J Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands.
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Biology, University of Padua, 35121 Padua, Italy; Veneto Institute of Molecular Medicine, 35129 Padua, Italy.
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Applied Stem Cell Technologies, University of Twente, 7500 Enschede, the Netherlands.
| |
Collapse
|