1
|
Hutchinson A, Taylor CL, Chowdhury SM, Jackson L. ECG Findings Are Poor Predictors for Adverse Events and Cardiac Death in Barth Syndrome. PROGRESS IN PEDIATRIC CARDIOLOGY 2024; 75:101750. [PMID: 39281339 PMCID: PMC11392022 DOI: 10.1016/j.ppedcard.2024.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Patients with Barth syndrome (BTHS) can present with cardiomyopathy. BTHS subjects are at risk for cardiac adverse outcomes throughout life, including malignant arrhythmias and death. Electrocardiogram (ECG) parameters have never been assessed as a tool to predict adverse outcomes in individuals with BTHS. Objectives The purpose of this study was to identify any ECG parameters including QRS fragmentation, presence of arrhythmia, or abnormal intervals that could predict adverse outcomes and cardiac death among the BTHS population. Methods We performed a retrospective case referent study on subjects with BTHS (n=43), and compared them with our reference group, subjects with idiopathic dilated cardiomyopathy (DCM) from a single institution (n=53) from 2007-2021. BTHS data was obtained from subjects attending the biennial Barth Syndrome Foundation International Scientific, Medical, and Family Conferences (BSFISMFC) from 2002-2018. ECG data from first and last available ECG's prior to an adverse event or cardiac death was analyzed, and then multivariable regression was performed to determine odd ratios between ECG characteristics and adverse events/cardiac death. Results No ECG variables were statistically significant predictors of adverse events or cardiac death in the BTHS group. Last ECG QRS fragmentation trended to statistically significance (OR 13.3, p=0.12) in predicting adverse events in the DCM group. Conclusion No ECG parameters, including QRS fragmentation, presence of arrhythmia, or abnormal interval values predict adverse events or cardiac death among BTHS patients. QRS fragmentation may be a predictor of adverse events in the DCM population.
Collapse
Affiliation(s)
- Alexander Hutchinson
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425 USA
| | - Carolyn L Taylor
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425 USA
| | - Shahryar M Chowdhury
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425 USA
| | - Lanier Jackson
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425 USA
| |
Collapse
|
2
|
Horonyova P, Durisova I, Cermakova P, Babelova L, Buckova B, Sofrankova L, Valachovic M, Hsu YHH, Balazova M. The subtherapeutic dose of valproic acid induces the activity of cardiolipin-dependent proteins. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2024; 1865:149501. [PMID: 39079622 DOI: 10.1016/j.bbabio.2024.149501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/29/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
A mood-stabilizing anticonvulsant valproic acid (VPA) is a drug with a pleiotropic effect on cells. Here, we describe the impact of VPA on the metabolic function of human HAP1 cells. We show that VPA altered the biosynthetic pathway of cardiolipin (CL) and affected the activities of mitochondrial enzymes such as pyruvate dehydrogenase, α-ketoglutarate dehydrogenase and NADH dehydrogenase. We demonstrate that a therapeutic dose of VPA (0.6 mM) has a harmful effect on cell growth and increases the production of reactive oxygen species and superoxides. On the contrary, less concentrated VPA (0.06 mM) increased the activities of CL-dependent enzymes leading to an increased level of oxidative phosphorylation and ATP production. The effect of VPA was also tested on the Barth syndrome model, which is characterized by a reduced amount of CL and an increased level of monolyso-CL. In this model, VPA treatment slightly attenuated the mitochondrial defects by altering the activities of CL-dependent enzymes. However, the presence of CL was essential for the increase in ATP production by VPA. Our findings highlight the potential therapeutic role of VPA in normalizing mitochondrial function in BTHS and shed light on the intricate interplay between lipid metabolism and mitochondrial physiology in health and disease. SUMMARY: This study investigates the dose-dependent effect of valproate, a mood-stabilizing drug, on mitochondrial function. The therapeutic concentration reduced overall cellular metabolic activity, while a subtherapeutic concentration notably improved the function of cardiolipin-dependent proteins within mitochondria. These findings shed light on novel aspects of valproate's effect and suggest potential practical applications for its use. By elucidating the differential effects of valproate doses on mitochondrial activity, this research underscores the drug's multifaceted role in cellular metabolism and highlights avenues for further exploration in therapeutic interventions.
Collapse
Affiliation(s)
- Paulina Horonyova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ivana Durisova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Petra Cermakova
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Lenka Babelova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbora Buckova
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Lucia Sofrankova
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Slovakia
| | - Martin Valachovic
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Maria Balazova
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
3
|
Che M, Li F, Jia Y, Liu Q, Hu J, Zhang J, Liu S. Case Report: A Chinese child with Barth syndrome caused by a novel TAFAZZIN mutation. Front Cardiovasc Med 2024; 11:1465912. [PMID: 39309604 PMCID: PMC11412893 DOI: 10.3389/fcvm.2024.1465912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Barth syndrome (BTHS) is a rare X-linked recessive genetic disorder characterized by a broad spectrum of clinical features including cardiomyopathy, skeletal myopathy, neutropenia, growth delay, and 3-methylglutaconic aciduria. This disease is caused by loss-of-function mutations in the TAFAZZIN gene located on chromosome Xq28, resulting in cardiolipin deficiency. Most patients are diagnosed in childhood, and the mortality rate is highest in the early years. We report a case of acute, life-threatening metabolic decompensation occurring one day after birth. A novel TAFAZZIN splice site mutation was identified in the patient, marking the first reported case of such a mutation in BTHS identified in China. The report aims to expand our understanding of the spectrum of TAFAZZIN mutations in BTHS.
Collapse
Affiliation(s)
- Mingxuan Che
- Cardiovascular Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fuhai Li
- Cardiovascular Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaning Jia
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qingzheng Liu
- Cardiovascular Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jian Hu
- Cardiovascular Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jidong Zhang
- Cardiovascular Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Decker ST, Funai K. Mitochondrial membrane lipids in the regulation of bioenergetic flux. Cell Metab 2024; 36:1963-1978. [PMID: 39178855 PMCID: PMC11374467 DOI: 10.1016/j.cmet.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Oxidative phosphorylation (OXPHOS) occurs through and across the inner mitochondrial membrane (IMM). Mitochondrial membranes contain a distinct lipid composition, aided by lipid biosynthetic machinery localized in the IMM and class-specific lipid transporters that limit lipid traffic in and out of mitochondria. This unique lipid composition appears to be essential for functions of mitochondria, particularly OXPHOS, by its effects on direct lipid-to-protein interactions, membrane properties, and cristae ultrastructure. This review highlights the biological significance of mitochondrial lipids, with a particular spotlight on the role of lipids in mitochondrial bioenergetics. We describe pathways for the biosynthesis of mitochondrial lipids and provide evidence for their roles in physiology, their implications in human disease, and the mechanisms by which they regulate mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Stephen Thomas Decker
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
5
|
Balderas E, Lee SHJ, Rai NK, Mollinedo DM, Duron HE, Chaudhuri D. Mitochondrial Calcium Regulation of Cardiac Metabolism in Health and Disease. Physiology (Bethesda) 2024; 39:0. [PMID: 38713090 PMCID: PMC11460536 DOI: 10.1152/physiol.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
Oxidative phosphorylation is regulated by mitochondrial calcium (Ca2+) in health and disease. In physiological states, Ca2+ enters via the mitochondrial Ca2+ uniporter and rapidly enhances NADH and ATP production. However, maintaining Ca2+ homeostasis is critical: insufficient Ca2+ impairs stress adaptation, and Ca2+ overload can trigger cell death. In this review, we delve into recent insights further defining the relationship between mitochondrial Ca2+ dynamics and oxidative phosphorylation. Our focus is on how such regulation affects cardiac function in health and disease, including heart failure, ischemia-reperfusion, arrhythmias, catecholaminergic polymorphic ventricular tachycardia, mitochondrial cardiomyopathies, Barth syndrome, and Friedreich's ataxia. Several themes emerge from recent data. First, mitochondrial Ca2+ regulation is critical for fuel substrate selection, metabolite import, and matching of ATP supply to demand. Second, mitochondrial Ca2+ regulates both the production and response to reactive oxygen species (ROS), and the balance between its pro- and antioxidant effects is key to how it contributes to physiological and pathological states. Third, Ca2+ exerts localized effects on the electron transport chain (ETC), not through traditional allosteric mechanisms but rather indirectly. These effects hinge on specific transporters, such as the uniporter or the Na+/Ca2+ exchanger, and may not be noticeable acutely, contributing differently to phenotypes depending on whether Ca2+ transporters are acutely or chronically modified. Perturbations in these novel relationships during disease states may either serve as compensatory mechanisms or exacerbate impairments in oxidative phosphorylation. Consequently, targeting mitochondrial Ca2+ holds promise as a therapeutic strategy for a variety of cardiac diseases characterized by contractile failure or arrhythmias.
Collapse
Affiliation(s)
- Enrique Balderas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Sandra H J Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Neeraj K Rai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - David M Mollinedo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Hannah E Duron
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, Biochemistry, Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
6
|
Stańczyk M, Szubart N, Maslanka R, Zadrag-Tecza R. Mitochondrial Dysfunctions: Genetic and Cellular Implications Revealed by Various Model Organisms. Genes (Basel) 2024; 15:1153. [PMID: 39336744 PMCID: PMC11431519 DOI: 10.3390/genes15091153] [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: 07/30/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondria play a crucial role in maintaining the energy status and redox homeostasis of eukaryotic cells. They are responsible for the metabolic efficiency of cells, providing both ATP and intermediate metabolic products. They also regulate cell survival and death under stress conditions by controlling the cell response or activating the apoptosis process. This functional diversity of mitochondria indicates their great importance for cellular metabolism. Hence, dysfunctions of these structures are increasingly recognized as an element of the etiology of many human diseases and, therefore, an extremely promising therapeutic target. Mitochondrial dysfunctions can be caused by mutations in both nuclear and mitochondrial DNA, as well as by stress factors or replication errors. Progress in knowledge about the biology of mitochondria, as well as the consequences for the efficiency of the entire organism resulting from the dysfunction of these structures, is achieved through the use of model organisms. They are an invaluable tool for analyzing complex cellular processes, leading to a better understanding of diseases caused by mitochondrial dysfunction. In this work, we review the most commonly used model organisms, discussing both their advantages and limitations in modeling fundamental mitochondrial processes or mitochondrial diseases.
Collapse
Affiliation(s)
| | | | | | - Renata Zadrag-Tecza
- Institute of Biology, College of Natural Sciences, University of Rzeszow, 35-959 Rzeszow, Poland; (M.S.); (N.S.); (R.M.)
| |
Collapse
|
7
|
Venkatraman K, Budin I. Cardiolipin remodeling maintains the inner mitochondrial membrane in cells with saturated lipidomes. J Lipid Res 2024; 65:100601. [PMID: 39038656 PMCID: PMC11381790 DOI: 10.1016/j.jlr.2024.100601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Cardiolipin (CL) is a unique, four-chain phospholipid synthesized in the inner mitochondrial membrane (IMM). The acyl chain composition of CL is regulated through a remodeling pathway, whose loss causes mitochondrial dysfunction in Barth syndrome (BTHS). Yeast has been used extensively as a model system to characterize CL metabolism, but mutants lacking its two remodeling enzymes, Cld1p and Taz1p, exhibit mild structural and respiratory phenotypes compared to mammalian cells. Here, we show an essential role for CL remodeling in the structure and function of the IMM in yeast grown under reduced oxygenation. Microaerobic fermentation, which mimics natural yeast environments, caused the accumulation of saturated fatty acids and, under these conditions, remodeling mutants showed a loss of IMM ultrastructure. We extended this observation to HEK293 cells, where phospholipase A2 inhibition by Bromoenol lactone resulted in respiratory dysfunction and cristae loss upon mild treatment with exogenous saturated fatty acids. In microaerobic yeast, remodeling mutants accumulated unremodeled, saturated CL, but also displayed reduced total CL levels, highlighting the interplay between saturation and CL biosynthesis and/or breakdown. We identified the mitochondrial phospholipase A1 Ddl1p as a regulator of CL levels, and those of its precursors phosphatidylglycerol and phosphatidic acid, under these conditions. Loss of Ddl1p partially rescued IMM structure in cells unable to initiate CL remodeling and had differing lipidomic effects depending on oxygenation. These results introduce a revised yeast model for investigating CL remodeling and suggest that its structural functions are dependent on the overall lipid environment in the mitochondrion.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Snider PL, Sierra Potchanant EA, Sun Z, Edwards DM, Chan KK, Matias C, Awata J, Sheth A, Pride PM, Payne RM, Rubart M, Brault JJ, Chin MT, Nalepa G, Conway SJ. A Barth Syndrome Patient-Derived D75H Point Mutation in TAFAZZIN Drives Progressive Cardiomyopathy in Mice. Int J Mol Sci 2024; 25:8201. [PMID: 39125771 PMCID: PMC11311365 DOI: 10.3390/ijms25158201] [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: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiomyopathy is the predominant defect in Barth syndrome (BTHS) and is caused by a mutation of the X-linked Tafazzin (TAZ) gene, which encodes an enzyme responsible for remodeling mitochondrial cardiolipin. Despite the known importance of mitochondrial dysfunction in BTHS, how specific TAZ mutations cause diverse BTHS heart phenotypes remains poorly understood. We generated a patient-tailored CRISPR/Cas9 knock-in mouse allele (TazPM) that phenocopies BTHS clinical traits. As TazPM males express a stable mutant protein, we assessed cardiac metabolic dysfunction and mitochondrial changes and identified temporally altered cardioprotective signaling effectors. Specifically, juvenile TazPM males exhibit mild left ventricular dilation in systole but have unaltered fatty acid/amino acid metabolism and normal adenosine triphosphate (ATP). This occurs in concert with a hyperactive p53 pathway, elevation of cardioprotective antioxidant pathways, and induced autophagy-mediated early senescence in juvenile TazPM hearts. However, adult TazPM males exhibit chronic heart failure with reduced growth and ejection fraction, cardiac fibrosis, reduced ATP, and suppressed fatty acid/amino acid metabolism. This biphasic changeover from a mild-to-severe heart phenotype coincides with p53 suppression, downregulation of cardioprotective antioxidant pathways, and the onset of terminal senescence in adult TazPM hearts. Herein, we report a BTHS genotype/phenotype correlation and reveal that absent Taz acyltransferase function is sufficient to drive progressive cardiomyopathy.
Collapse
Affiliation(s)
- Paige L. Snider
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Elizabeth A. Sierra Potchanant
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Zejin Sun
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Donna M. Edwards
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Ka-Kui Chan
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Catalina Matias
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (C.M.); (J.J.B.)
| | - Junya Awata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (J.A.); (M.T.C.)
| | - Aditya Sheth
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - P. Melanie Pride
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - R. Mark Payne
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Michael Rubart
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Jeffrey J. Brault
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (C.M.); (J.J.B.)
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (J.A.); (M.T.C.)
| | - Grzegorz Nalepa
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46033, USA; (P.L.S.); (E.A.S.P.); (Z.S.); (D.M.E.); (K.-K.C.); (A.S.); (P.M.P.); (R.M.P.); (M.R.); (G.N.)
| |
Collapse
|
9
|
Thompson WR, Manuel R, Abbruscato A, Carr J, Campbell J, Hornby B, Vaz FM, Vernon HJ. Long-term efficacy and safety of elamipretide in patients with Barth syndrome: 168-week open-label extension results of TAZPOWER. Genet Med 2024; 26:101138. [PMID: 38602181 DOI: 10.1016/j.gim.2024.101138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
PURPOSE Evaluate long-term efficacy and safety of elamipretide during the open-label extension (OLE) of the TAZPOWER trial in individuals with Barth syndrome (BTHS). METHODS TAZPOWER was a 28-week randomized, double-blind, and placebo-controlled trial followed by a 168-week OLE. Patients entering the OLE continued elamipretide 40 mg subcutaneous daily. OLE primary endpoints were safety and tolerability; secondary endpoints included change from baseline in the 6-minute walk test (6MWT) and BarTH Syndrome Symptom Assessment (BTHS-SA) Total Fatigue score. Muscle strength, physician- and patient-assessed outcomes, echocardiographic parameters, and biomarkers, including cardiolipin (CL) and monolysocardiolipin (MLCL), were assessed. RESULTS Ten patients entered the OLE; 8 reached the week 168 visit. Elamipretide was well tolerated, with injection-site reactions being the most common adverse events. Significant improvements from OLE baseline on 6MWT occurred at all OLE time points (cumulative 96.1 m of improvement [week 168, P = .003]). Mean BTHS-SA Total Fatigue scores were below baseline (improved) at all OLE time points. Three-dimensional (3D) left ventricular stroke, end-diastolic, and end-systolic volumes improved, showing significant trends for improvement from baseline to week 168. MLCL/CL values showed improvement, correlating to important clinical outcomes. CONCLUSION Elamipretide was associated with sustained long-term tolerability and efficacy, with improvements in functional assessments and cardiac function in BTHS.
Collapse
Affiliation(s)
- William R Thompson
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ryan Manuel
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Jim Carr
- Stealth BioTherapeutics, Inc., Newton, MA
| | | | - Brittany Hornby
- Department of Physical Therapy, Kennedy Krieger, Baltimore, MD
| | - Frédéric M Vaz
- Amsterdam UMC Location University of Amsterdam, Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
10
|
Senoo N, Chinthapalli DK, Baile MG, Golla VK, Saha B, Oluwole AO, Ogunbona OB, Saba JA, Munteanu T, Valdez Y, Whited K, Sheridan MS, Chorev D, Alder NN, May ER, Robinson CV, Claypool SM. Functional diversity among cardiolipin binding sites on the mitochondrial ADP/ATP carrier. EMBO J 2024; 43:2979-3008. [PMID: 38839991 PMCID: PMC11251061 DOI: 10.1038/s44318-024-00132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Lipid-protein interactions play a multitude of essential roles in membrane homeostasis. Mitochondrial membranes have a unique lipid-protein environment that ensures bioenergetic efficiency. Cardiolipin (CL), the signature mitochondrial lipid, plays multiple roles in promoting oxidative phosphorylation (OXPHOS). In the inner mitochondrial membrane, the ADP/ATP carrier (AAC in yeast; adenine nucleotide translocator, ANT in mammals) exchanges ADP and ATP, enabling OXPHOS. AAC/ANT contains three tightly bound CLs, and these interactions are evolutionarily conserved. Here, we investigated the role of these buried CLs in AAC/ANT using a combination of biochemical approaches, native mass spectrometry, and molecular dynamics simulations. We introduced negatively charged mutations into each CL-binding site of yeast Aac2 and established experimentally that the mutations disrupted the CL interactions. While all mutations destabilized Aac2 tertiary structure, transport activity was impaired in a binding site-specific manner. Additionally, we determined that a disease-associated missense mutation in one CL-binding site in human ANT1 compromised its structure and transport activity, resulting in OXPHOS defects. Our findings highlight the conserved significance of CL in AAC/ANT structure and function, directly tied to specific lipid-protein interactions.
Collapse
Affiliation(s)
- Nanami Senoo
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Dinesh K Chinthapalli
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Oxford, OX1 3QU, UK
| | - Matthew G Baile
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vinaya K Golla
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Bodhisattwa Saha
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, OX1 3QU, UK
| | - Abraham O Oluwole
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Oxford, OX1 3QU, UK
| | - Oluwaseun B Ogunbona
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - James A Saba
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Teona Munteanu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yllka Valdez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kevin Whited
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Macie S Sheridan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Dror Chorev
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, OX1 3QU, UK
| | - Nathan N Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - Eric R May
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - Carol V Robinson
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Oxford, OX1 3QU, UK
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
11
|
Russo S, De Rasmo D, Rossi R, Signorile A, Lobasso S. SS-31 treatment ameliorates cardiac mitochondrial morphology and defective mitophagy in a murine model of Barth syndrome. Sci Rep 2024; 14:13655. [PMID: 38871974 DOI: 10.1038/s41598-024-64368-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Barth syndrome (BTHS) is a lethal rare genetic disorder, which results in cardiac dysfunction, severe skeletal muscle weakness, immune issues and growth delay. Mutations in the TAFAZZIN gene, which is responsible for the remodeling of the phospholipid cardiolipin (CL), lead to abnormalities in mitochondrial membrane, including alteration of mature CL acyl composition and the presence of monolysocardiolipin (MLCL). The dramatic increase in the MLCL/CL ratio is the hallmark of patients with BTHS, which is associated with mitochondrial bioenergetics dysfunction and altered membrane ultrastructure. There are currently no specific therapies for BTHS. Here, we showed that cardiac mitochondria isolated from TAFAZZIN knockdown (TazKD) mice presented abnormal ultrastructural membrane morphology, accumulation of vacuoles, pro-fission conditions and defective mitophagy. Interestingly, we found that in vivo treatment of TazKD mice with a CL-targeted small peptide (named SS-31) was able to restore mitochondrial morphology in tafazzin-deficient heart by affecting specific proteins involved in dynamic process and mitophagy. This agrees with our previous data showing an improvement in mitochondrial respiratory efficiency associated with increased supercomplex organization in TazKD mice under the same pharmacological treatment. Taken together our findings confirm the beneficial effect of SS-31 in the amelioration of tafazzin-deficient dysfunctional mitochondria in a BTHS animal model.
Collapse
Affiliation(s)
- Silvia Russo
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Pl. G. Cesare 11, 70124, Bari, Italy
| | - Domenico De Rasmo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) , National Research Council (CNR), Bari, Italy
| | - Roberta Rossi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Anna Signorile
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Pl. G. Cesare 11, 70124, Bari, Italy.
| | - Simona Lobasso
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Pl. G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
12
|
Zhu S, Pang J, Nguyen A, Huynh H, Lee S, Gu Y, Vaz FM, Fang X. Dietary linoleic acid supplementation fails to rescue established cardiomyopathy in Barth syndrome. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100076. [PMID: 38974772 PMCID: PMC11225933 DOI: 10.1016/j.jmccpl.2024.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Barth syndrome (BTHS) is a mitochondrial lipid disorder caused by mutations in TAFAZZIN (TAZ), required for cardiolipin (CL) remodeling. Cardiomyopathy is a major clinical feature, with no curative therapy. Linoleic acid (LA) supplementation is proposed to ameliorate BTHS cardiomyopathy by enhancing linoleoyl group incorporation into CL. While the beneficial effect of dietary LA supplementation in delaying the development of BTHS cardiomyopathy has been recently tested, its potential to reverse established BTHS cardiomyopathy remains unclear. Our study revealed that LA supplementation cannot rescue established BTHS cardiomyopathy in mice, highlighting the importance of early initiation of LA supplementation for maximum benefits.
Collapse
Affiliation(s)
- Siting Zhu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Pang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anh Nguyen
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Helen Huynh
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sharon Lee
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Frederic 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, the Netherlands
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
Tong Q, Miao Y, Yin H. Echocardiographic manifestations of mitochondrial disease with GTPBP3 gene mutations: A case report. Medicine (Baltimore) 2024; 103:e37847. [PMID: 38701254 PMCID: PMC11062751 DOI: 10.1097/md.0000000000037847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
RATIONALE Mitochondrial diseases are a group of disorders in which mutations in mitochondrial DNA or nuclear DNA lead to dysfunctional oxidative phosphorylation of cells, with mutations in mitochondrial DNA being the most common cause of mitochondrial disease, and mutations in nuclear genes being rarely reported. The echocardiographic findings of mitochondrial diseases with nuclear gene mutations in children's hearts are even rarer. Even more valuable is that we followed up the patient for 4 years and dynamically observed the cardiac echocardiographic manifestations of mitochondrial disease. Provide ideas for the clinical diagnosis and prognosis of mitochondrial diseases. PATIENT CONCERNS The patient was seen in the pediatric outpatient clinic for poor strength and mental retardation. echocardiography: mild left ventricular (LV) enlargement and LV wall thickening. Nuclear genetic testing: uanosine triphosphate binding protein 3 (GTPBP3) gene mutation. Diagnosis of mitochondrial disease. DIAGNOSES Mitochondrial disease with GTPBP3 gene mutations. OUTCOMES After receiving drug treatment, the patient exhibited a reduction in lactate levels, an enhanced physical condition compared to prior assessments, and demonstrated average intellectual development. LESSONS SUBSECTIONS For echocardiographic indications of LV wall thickening and LV enlargement, one needs to be alert to the possibility of hereditary cardiomyopathy, especially in children.
Collapse
Affiliation(s)
- Qiaoli Tong
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yajing Miao
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongning Yin
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Sniezek Carney O, Harris KW, Wohlfarter Y, Lee K, Butschek G, Anzmann A, Claypool SM, Hamacher-Brady A, Keller M, Vernon HJ. Stem cell models of TAFAZZIN deficiency reveal novel tissue-specific pathologies in Barth Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591534. [PMID: 38746168 PMCID: PMC11092433 DOI: 10.1101/2024.04.28.591534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Barth syndrome (BTHS) is a rare mitochondrial disease caused by pathogenic variants in the gene TAFAZZIN, which leads to abnormal cardiolipin (CL) metabolism on the inner mitochondrial membrane. Although TAFAZZIN is ubiquitously expressed, BTHS involves a complex combination of tissue specific phenotypes including cardiomyopathy, neutropenia, skeletal myopathy, and growth delays, with a relatively minimal neurological burden. To understand both the developmental and functional effects of TAZ-deficiency in different tissues, we generated isogenic TAZ knockout (TAZ- KO) and WT cardiomyocytes (CMs) and neural progenitor cells (NPCs) from CRISPR-edited induced pluripotent stem cells (iPSCs). In TAZ-KO CMs we discovered evidence of dysregulated mitophagy including dysmorphic mitochondria and mitochondrial cristae, differential expression of key autophagy-associated genes, and an inability of TAZ-deficient CMs to properly initiate stress-induced mitophagy. In TAZ-deficient NPCs we identified novel phenotypes including a reduction in CIV abundance and CIV activity in the CIII2&CIV2 intermediate complex. Interestingly, while CL acyl chain manipulation was unable to alter mitophagy defects in TAZ-KO CMs, we found that linoleic acid or oleic acid supplementation was able to partially restore CIV abundance in TAZ-deficient NPCs. Taken together, our results have implications for understanding the tissue-specific pathology of BTHS and potential for tissue-specific therapeutic targeting. Moreover, our results highlight an emerging role for mitophagy in the cardiac pathophysiology of BTHS and reveal a potential neuron-specific bioenergetic phenotype.
Collapse
|
15
|
Hachmann M, Gülcan G, Rajendran R, Höring M, Liebisch G, Bachhuka A, Kohlhaas M, Maack C, Ergün S, Dudek J, Karnati S. Tafazzin deficiency causes substantial remodeling in the lipidome of a mouse model of Barth Syndrome cardiomyopathy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1389456. [PMID: 39086433 PMCID: PMC11285559 DOI: 10.3389/fmmed.2024.1389456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/09/2024] [Indexed: 08/02/2024]
Abstract
Barth Syndrome (BTHS) is a rare X-linked disease, characterized clinically by cardiomyopathy, skeletal myopathy, neutropenia, and growth retardation. BTHS is caused by mutations in the phospholipid acyltransferase tafazzin (Gene: TAFAZZIN, TAZ). Tafazzin catalyzes the final step in the remodeling of cardiolipin (CL), a glycerophospholipid located in the inner mitochondrial membrane. As the phospholipid composition strongly determines membrane properties, correct biosynthesis of CL and other membrane lipids is essential for mitochondrial function. Mitochondria provide 95% of the energy demand in the heart, particularly due to their role in fatty acid oxidation. Alterations in lipid homeostasis in BTHS have an impact on mitochondrial membrane proteins and thereby contribute to cardiomyopathy. We analyzed a transgenic TAFAZZIN-knockdown (TAZ-KD) BTHS mouse model and determined the distribution of 193 individual lipid species in TAZ-KD and WT hearts at 10 and 50 weeks of age, using electrospray ionization tandem mass spectrometry (ESI-MS/MS). Our results revealed significant lipid composition differences between the TAZ-KD and WT groups, indicating genotype-dependent alterations in most analyzed lipid species. Significant changes in the myocardial lipidome were identified in both young animals without cardiomyopathy and older animals with heart failure. Notable alterations were found in phosphatidylcholine (PC), phosphatidylethanolamine (PE), lysophosphatidylethanolamine (LPE), lysophosphatidylcholine (LPC) and plasmalogen species. PC species with 2-4 double bonds were significantly increased, while polyunsaturated PC species showed a significant decrease in TAZ-KD mice. Furthermore, Linoleic acid (LA, 18:2) containing PC and PE species, as well as arachidonic acid (AA, 20:4) containing PE 38:4 species are increased in TAZ-KD. We found higher levels of AA containing LPE and PE-based plasmalogens (PE P-). Furthermore, we are the first to show significant changes in sphingomyelin (SM) and ceramide (Cer) lipid species Very long-chained SM species are accumulating in TAZ-KD hearts, whereas long-chained Cer and several hexosyl ceramides (HexCer) species accumulate only in 50-week-old TAZ-KD hearts These findings offer potential avenues for the diagnosis and treatment of BTHS, presenting new possibilities for therapeutic approaches.
Collapse
Affiliation(s)
- Malte Hachmann
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Güntas Gülcan
- Department of Medical Biochemistry, Faculty of Medicine, Atlas University, Istanbul, Turkey
| | - Ranjithkumar Rajendran
- Experimental Neurology, Department of Neurology, Justus Liebig University, Giessen, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, Regensburg, Germany
| | - Akash Bachhuka
- Department of Electronics, Electric, and Automatic Engineering, Rovira I Virgili University, Tarragona, Spain
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Medical Clinic 1, University Hospital Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Srikanth Karnati
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Choi Y. Association of neutrophil defects with oral ulcers but undetermined role of neutrophils in recurrent aphthous stomatitis. Heliyon 2024; 10:e26740. [PMID: 38439826 PMCID: PMC10911260 DOI: 10.1016/j.heliyon.2024.e26740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Objective Recurrent oral ulcers and severe periodontal diseases in patients with quantitative or qualitative neutrophil defects highlight the important role of neutrophils in maintaining oral mucosal barrier homeostasis. Recurrent aphthous stomatitis (RAS) is a common oral mucosal disease affecting up to 25% of the population, yet its etiopathogenesis remains unclear, and management is unsatisfactory. This review aims to gain insight into the pathogenesis of RAS. Design This narrative review examines the characteristics of oral and blood neutrophils, the associations between neutrophil defects and the occurrence of oral ulcers, and the evidence for the involvement of neutrophils in RAS. To conduct the review, relevant literature was searched in PubMed and Google Scholar, which was then thoroughly reviewed and critically appraised. Results Neutropenia, specifically a decrease in the number of oral neutrophils, impaired extravasation, and defective ROS production appear to be associated with oral ulcers, while defects in granule enzymes or NETosis are unlikely to have a link to oral ulcers. The review of the histopathology of RAS shows that neutrophils are concentrated in the denuded area but are latecomers to the scene and early leavers. However, the evidence for the involvement of neutrophils in the pathogenesis of RAS is inconsistent, leading to the proposal of two different scenarios involving either impaired or hyperactive neutrophils in the pathogenesis of RAS.
Collapse
Affiliation(s)
- Youngnim Choi
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Kaura V, Hopkins P. Recent advances in skeletal muscle physiology. BJA Educ 2024; 24:84-90. [PMID: 38375493 PMCID: PMC10874741 DOI: 10.1016/j.bjae.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 02/21/2024] Open
Affiliation(s)
- V. Kaura
- Leeds Institute of Medical Research at St James's, University of Leeds, UK
| | - P.M. Hopkins
- Leeds Institute of Medical Research at St James's, University of Leeds, UK
| |
Collapse
|
18
|
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. LAB ON A CHIP 2024; 24:1494-1528. [PMID: 38318723 DOI: 10.1039/d3lc00829k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, casting a substantial economic footprint and burdening the global healthcare system. Historically, pre-clinical CVD modeling and therapeutic screening have been performed using animal models. Unfortunately, animal models oftentimes fail to adequately mimic human physiology, leading to a poor translation of therapeutics from pre-clinical trials to consumers. Even those that make it to market can be removed due to unforeseen side effects. As such, there exists a clinical, technological, and economical need for systems that faithfully capture human (patho)physiology for modeling CVD, assessing cardiotoxicity, and evaluating drug efficacy. Heart-on-a-chip (HoC) systems are a part of the broader organ-on-a-chip paradigm that leverages microfluidics, tissue engineering, microfabrication, electronics, and gene editing to create human-relevant models for studying disease, drug-induced side effects, and therapeutic efficacy. These compact systems can be capable of real-time measurements and on-demand characterization of tissue behavior and could revolutionize the drug development process. In this review, we highlight the key components that comprise a HoC system followed by a review of contemporary reports of their use in disease modeling, drug toxicity and efficacy assessment, and as part of multi-organ-on-a-chip platforms. We also discuss future perspectives and challenges facing the field, including a discussion on the role that standardization is expected to play in accelerating the widespread adoption of these platforms.
Collapse
Affiliation(s)
- Derrick Butler
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | - Darwin R Reyes
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
19
|
Oyarbide U, Crane GM, Corey SJ. The metabolic basis of inherited neutropenias. Br J Haematol 2024; 204:45-55. [PMID: 38049194 DOI: 10.1111/bjh.19192] [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: 08/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Neutrophils are the shortest-lived blood cells, which requires a prodigious degree of proliferation and differentiation to sustain physiologically sufficient numbers and be poised to respond quickly to infectious emergencies. More than 107 neutrophils are produced every minute in an adult bone marrow-a process that is tightly regulated by a small group of cytokines and chemical mediators and dependent on nutrients and energy. Like granulocyte colony-stimulating factor, the primary growth factor for granulopoiesis, they stimulate signalling pathways, some affecting metabolism. Nutrient or energy deficiency stresses the survival, proliferation, and differentiation of neutrophils and their precursors. Thus, it is not surprising that monogenic disorders related to metabolism exist that result in neutropenia. Among these are pathogenic mutations in HAX1, G6PC3, SLC37A4, TAFAZZIN, SBDS, EFL1 and the mitochondrial disorders. These mutations perturb carbohydrate, lipid and/or protein metabolism. We hypothesize that metabolic disturbances may drive the pathogenesis of a subset of inherited neutropenias just as defects in DNA damage response do in Fanconi anaemia, telomere maintenance in dyskeratosis congenita and ribosome formation in Diamond-Blackfan anaemia. Greater understanding of metabolic pathways in granulopoiesis will identify points of vulnerability in production and may point to new strategies for the treatment of neutropenias.
Collapse
Affiliation(s)
- Usua Oyarbide
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| | - Genevieve M Crane
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Seth J Corey
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Makio T, Simmen T. Not So Rare: Diseases Based on Mutant Proteins Controlling Endoplasmic Reticulum-Mitochondria Contact (MERC) Tethering. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241261228. [PMID: 39070058 PMCID: PMC11273598 DOI: 10.1177/25152564241261228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024]
Abstract
Mitochondria-endoplasmic reticulum contacts (MERCs), also called endoplasmic reticulum (ER)-mitochondria contact sites (ERMCS), are the membrane domains, where these two organelles exchange lipids, Ca2+ ions, and reactive oxygen species. This crosstalk is a major determinant of cell metabolism, since it allows the ER to control mitochondrial oxidative phosphorylation and the Krebs cycle, while conversely, it allows the mitochondria to provide sufficient ATP to control ER proteostasis. MERC metabolic signaling is under the control of tethers and a multitude of regulatory proteins. Many of these proteins have recently been discovered to give rise to rare diseases if their genes are mutated. Surprisingly, these diseases share important hallmarks and cause neurological defects, sometimes paired with, or replaced by skeletal muscle deficiency. Typical symptoms include developmental delay, intellectual disability, facial dysmorphism and ophthalmologic defects. Seizures, epilepsy, deafness, ataxia, or peripheral neuropathy can also occur upon mutation of a MERC protein. Given that most MERC tethers and regulatory proteins have secondary functions, some MERC protein-based diseases do not fit into this categorization. Typically, however, the proteins affected in those diseases have dominant functions unrelated to their roles in MERCs tethering or their regulation. We are discussing avenues to pharmacologically target genetic diseases leading to MERC defects, based on our novel insight that MERC defects lead to common characteristics in rare diseases. These shared characteristics of MERCs disorders raise the hope that they may allow for similar treatment options.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Lim Y, Hong I, Han A. The Impact of Raising Children with Barth Syndrome on Parental Health-Related Quality of Life and Family Functioning: Preliminary Reliability and Validity of the PedsQL™ Family Impact Module. Occup Ther Int 2023; 2023:5588935. [PMID: 38187035 PMCID: PMC10771332 DOI: 10.1155/2023/5588935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/02/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024] Open
Abstract
Objective This study examined the preliminary reliability and validity of the PedsQL™ Family Impact Module (PedsQL FIM) in families of children with Barth syndrome (BTHS). Method A total of 72 parents with children or youth between the ages of 5 and 19 participated in this study. Thirty-three parents of children with BTHS and 39 parents of unaffected children completed the PedsQL FIM and a demographic information form. Internal consistency reliability and item-total correlations were calculated to test the reliability of the PedsQL FIM. Construct validity was examined using the known-groups method. We estimated the mean score differences of the PedsQL FIM between the two groups using three different models, including unadjusted, multivariate regression, and propensity score matching with inverse probability of treatment weighting (PS-IPTW) models. Results The Cronbach's alpha coefficients were greater than 0.70 for all scales of the PedsQL FIM, except for the communication scale. The item-total correlations were significant for all scales with moderate to high correlations (p < .05). In construct validity, the mean scores of the PedsQL FIM between the two groups were significantly different (p < .05) for all scales and total score in the unadjusted and PS-IPTW models. However, in the multivariate regression model, the family relationships scale was not significant between the two groups. Conclusion The PedsQL FIM demonstrated adequate measurement properties of preliminary reliability and validity in assessing the impact of children with BTHS on parental health-related quality of life (HRQoL) and family functioning. Further research needs to be conducted to examine the psychometric properties of the PedsQL FIM with a large sample of BTHS and with other pediatric rare diseases.
Collapse
Affiliation(s)
- Yoonjeong Lim
- Division of Occupational Therapy, Binghamton University, Johnson City, NY 13790, USA
| | - Ickpyo Hong
- Department of Occupational Therapy, Yonsei University, Wonju 26493, Republic of Korea
| | - Areum Han
- Department of Occupational Therapy, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
22
|
Kagan VE, Tyurina YY, Mikulska-Ruminska K, Damschroder D, Vieira Neto E, Lasorsa A, Kapralov AA, Tyurin VA, Amoscato AA, Samovich SN, Souryavong AB, Dar HH, Ramim A, Liang Z, Lazcano P, Ji J, Schmidtke MW, Kiselyov K, Korkmaz A, Vladimirov GK, Artyukhova MA, Rampratap P, Cole LK, Niyatie A, Baker EK, Peterson J, Hatch GM, Atkinson J, Vockley J, Kühn B, Wessells R, van der Wel PCA, Bahar I, Bayir H, Greenberg ML. Anomalous peroxidase activity of cytochrome c is the primary pathogenic target in Barth syndrome. Nat Metab 2023; 5:2184-2205. [PMID: 37996701 PMCID: PMC11213643 DOI: 10.1038/s42255-023-00926-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Barth syndrome (BTHS) is a life-threatening genetic disorder with unknown pathogenicity caused by mutations in TAFAZZIN (TAZ) that affect remodeling of mitochondrial cardiolipin (CL). TAZ deficiency leads to accumulation of mono-lyso-CL (MLCL), which forms a peroxidase complex with cytochrome c (cyt c) capable of oxidizing polyunsaturated fatty acid-containing lipids. We hypothesized that accumulation of MLCL facilitates formation of anomalous MLCL-cyt c peroxidase complexes and peroxidation of polyunsaturated fatty acid phospholipids as the primary BTHS pathogenic mechanism. Using genetic, biochemical/biophysical, redox lipidomic and computational approaches, we reveal mechanisms of peroxidase-competent MLCL-cyt c complexation and increased phospholipid peroxidation in different TAZ-deficient cells and animal models and in pre-transplant biopsies from hearts of patients with BTHS. A specific mitochondria-targeted anti-peroxidase agent inhibited MLCL-cyt c peroxidase activity, prevented phospholipid peroxidation, improved mitochondrial respiration of TAZ-deficient C2C12 myoblasts and restored exercise endurance in a BTHS Drosophila model. Targeting MLCL-cyt c peroxidase offers therapeutic approaches to BTHS treatment.
Collapse
Affiliation(s)
- Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Karolina Mikulska-Ruminska
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Deena Damschroder
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eduardo Vieira Neto
- Department of Pediatrics, Genetic and Genomic Medicine Division, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alessia Lasorsa
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Alexander A Kapralov
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A Amoscato
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Svetlana N Samovich
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Austin B Souryavong
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haider H Dar
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Abu Ramim
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Zhuqing Liang
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Pablo Lazcano
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Jiajia Ji
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | | | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aybike Korkmaz
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Georgy K Vladimirov
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margarita A Artyukhova
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pushpa Rampratap
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Laura K Cole
- Department of Pharmacology and Therapeutics, University of Manitoba, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Ammanamanchi Niyatie
- Department of Pediatrics, Pediatric Institute for Heart Regeneration and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emma-Kate Baker
- Department of Chemistry & Centre for Biotechnology, Brock University, St Catharines, Ontario, Canada
| | - Jim Peterson
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, School of Public Health, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Grant M Hatch
- Department of Pharmacology and Therapeutics, University of Manitoba, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jeffrey Atkinson
- Department of Chemistry & Centre for Biotechnology, Brock University, St Catharines, Ontario, Canada
| | - Jerry Vockley
- Department of Pediatrics, Genetic and Genomic Medicine Division, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bernhard Kühn
- Department of Pediatrics, Pediatric Institute for Heart Regeneration and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert Wessells
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Patrick C A van der Wel
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Ivet Bahar
- Laufer Center for Physical Quantitative Biology and Department of Biochemistry and Cell Biology, School of Medicine, Stony Brook University, New York, NY, USA
| | - Hülya Bayir
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
23
|
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
|
24
|
Conti F, Di Martino S, Drago F, Bucolo C, Micale V, Montano V, Siciliano G, Mancuso M, Lopriore P. Red Flags in Primary Mitochondrial Diseases: What Should We Recognize? Int J Mol Sci 2023; 24:16746. [PMID: 38069070 PMCID: PMC10706469 DOI: 10.3390/ijms242316746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Primary mitochondrial diseases (PMDs) are complex group of metabolic disorders caused by genetically determined impairment of the mitochondrial oxidative phosphorylation (OXPHOS). The unique features of mitochondrial genetics and the pivotal role of mitochondria in cell biology explain the phenotypical heterogeneity of primary mitochondrial diseases and the resulting diagnostic challenges that follow. Some peculiar features ("red flags") may indicate a primary mitochondrial disease, helping the physician to orient in this diagnostic maze. In this narrative review, we aimed to outline the features of the most common mitochondrial red flags offering a general overview on the topic that could help physicians to untangle mitochondrial medicine complexity.
Collapse
Affiliation(s)
- Federica Conti
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Filippo Drago
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95213 Catania, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Vincenzo Montano
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Gabriele Siciliano
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| |
Collapse
|
25
|
Liu O, Chinni BK, Manlhiot C, Vernon HJ. FGF21 and GDF15 are elevated in Barth Syndrome and are correlated to important clinical measures. Mol Genet Metab 2023; 140:107676. [PMID: 37549445 DOI: 10.1016/j.ymgme.2023.107676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Barth Syndrome (BTHS) is a rare X-linked disorder that is caused by defects TAFAZZIN, which leads to an abnormal cardiolipin (CL) profile of the inner mitochondrial membrane and clinical features including cardiomyopathy, neutropenia and skeletal myopathy. The ratio of monolysocardiolipin (MLCL, the remodeling intermediate of cardiolipin) to remodeled CL is always abnormal in Barth Syndrome and 3-methylglutaconic acid is often elevated affected patients, however neither of these biomarkers has been shown to temporally correlate to clinical status. In this study, we measured plasma FGF21 and GDF15 levels in 16 individuals with Barth Syndrome and evaluated whether these biomarkers were correlated to the MLCL/CL ratio in patient bloodspots and clinical laboratory parameters indicative of organ involvement in Barth Syndrome including: neutrophil and monocyte counts, liver function, and cardiac function (NT-proBNP). We found that FGF21 and GDF15 were elevated in all 16 patients and that FGF21 was significantly correlated to AST, ALT GGT, percentage of neutrophils comprising total white blood cells, percent monocytes comprising total white blood cells, and NT-proBNP levels. GDF-15 was significantly positively associated with NT-proBNP. We conclude that clinical measurements of FGF21 and GDF-15 may be relevant in the monitoring multi-organ system involvement in Barth Syndrome.
Collapse
Affiliation(s)
- Olivia Liu
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Bhargava Kumar Chinni
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Cedric Manlhiot
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA..
| |
Collapse
|
26
|
Tomczewski MV, Chan JZ, Al-Majmaie DM, Liu MR, Cocco AD, Stark KD, Strathdee D, Duncan RE. Phenotypic Characterization of Female Carrier Mice Heterozygous for Tafazzin Deletion. BIOLOGY 2023; 12:1238. [PMID: 37759637 PMCID: PMC10525480 DOI: 10.3390/biology12091238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Barth syndrome (BTHS) is caused by mutations in tafazzin resulting in deficits in cardiolipin remodeling that alter major metabolic processes. The tafazzin gene is encoded on the X chromosome, and therefore BTHS primarily affects males. Female carriers are typically considered asymptomatic, but age-related changes have been reported in female carriers of other X-linked disorders. Therefore, we examined the phenotype of female mice heterozygous for deletion of the tafazzin gene (Taz-HET) at 3 and 12 months of age. Food intakes, body masses, lean tissue and adipose depot weights, daily activity levels, metabolic measures, and exercise capacity were assessed. Age-related changes in mice resulted in small but significant genotype-specific differences in Taz-HET mice compared with their female Wt littermates. By 12 months, Taz-HET mice weighed less than Wt controls and had smaller gonadal, retroperitoneal, and brown adipose depots and liver and brain masses, despite similar food consumption. Daily movement, respiratory exchange ratio, and total energy expenditure did not vary significantly between the age-matched genotypes. Taz-HET mice displayed improved glucose tolerance and insulin sensitivity at 12 months compared with their Wt littermates but had evidence of slightly reduced exercise capacity. Tafazzin mRNA levels were significantly reduced in the cardiac muscle of 12-month-old Taz-HET mice, which was associated with minor but significant alterations in the heart cardiolipin profile. This work is the first to report the characterization of a model of female carriers of heterozygous tafazzin deficiency and suggests that additional study, particularly with advancing age, is warranted.
Collapse
Affiliation(s)
- Michelle V. Tomczewski
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - John Z. Chan
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - Duaa M. Al-Majmaie
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - Ming Rong Liu
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - Alex D. Cocco
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - Ken D. Stark
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| | - Douglas Strathdee
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, Scotland, UK;
| | - Robin E. Duncan
- Department of Kinesiology and Health Sciences, Faculty of Health, University of Waterloo, 200 University Ave W., BMH1044, Waterloo, ON N2L 3G1, Canada; (M.V.T.); (J.Z.C.); (D.M.A.-M.); (M.R.L.); (K.D.S.)
| |
Collapse
|
27
|
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
|
28
|
Tovaglieri N, Russo S, Micaglio E, Corcelli A, Lobasso S. Case report: Variability in clinical features as a potential pitfall for the diagnosis of Barth syndrome. Front Pediatr 2023; 11:1250772. [PMID: 37654687 PMCID: PMC10467424 DOI: 10.3389/fped.2023.1250772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
Background Barth syndrome is a rare genetic disease characterized by cardiomyopathy, skeletal muscle weakness, neutropenia, growth retardation and organic aciduria. This variable phenotype is caused by pathogenic hemizygous variants of the TAFAZZIN gene on the X chromosome, which impair metabolism of the mitochondrial phospholipid cardiolipin. Although most patients are usually diagnosed in the first years of life, the extremely variable clinical picture and the wide range of clinical presentations may both delay diagnosis. This is the case reported here of a man affected with severe neutropenia, who was not diagnosed with Barth syndrome until adulthood. Case presentation We describe herein a family case, specifically two Caucasian male cousins sharing the same mutation in the TAFAZZIN gene with a wide phenotypic variability: an infant who was early diagnosed with Barth syndrome due to heart failure, and his maternal cousin with milder and extremely different clinical features who has received the same diagnosis only at 33 years of age. Conclusions Our report supports the underestimation of the prevalence of Barth syndrome, which should be always considered in the differential diagnosis of male patients with recurrent neutropenia with or without signs and symptoms of cardiomyopathy.
Collapse
Affiliation(s)
| | - Silvia Russo
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Emanuele Micaglio
- Department of Arrhythmology and Clinical Electrophysiology, Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinic San Donato, Milan, Italy
| | - Angela Corcelli
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Simona Lobasso
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
29
|
Muñoz JP, Basei FL, Rojas ML, Galvis D, Zorzano A. Mechanisms of Modulation of Mitochondrial Architecture. Biomolecules 2023; 13:1225. [PMID: 37627290 PMCID: PMC10452872 DOI: 10.3390/biom13081225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial network architecture plays a critical role in cellular physiology. Indeed, alterations in the shape of mitochondria upon exposure to cellular stress can cause the dysfunction of these organelles. In this scenario, mitochondrial dynamics proteins and the phospholipid composition of the mitochondrial membrane are key for fine-tuning the modulation of mitochondrial architecture. In addition, several factors including post-translational modifications such as the phosphorylation, acetylation, SUMOylation, and o-GlcNAcylation of mitochondrial dynamics proteins contribute to shaping the plasticity of this architecture. In this regard, several studies have evidenced that, upon metabolic stress, mitochondrial dynamics proteins are post-translationally modified, leading to the alteration of mitochondrial architecture. Interestingly, several proteins that sustain the mitochondrial lipid composition also modulate mitochondrial morphology and organelle communication. In this context, pharmacological studies have revealed that the modulation of mitochondrial shape and function emerges as a potential therapeutic strategy for metabolic diseases. Here, we review the factors that modulate mitochondrial architecture.
Collapse
Affiliation(s)
- Juan Pablo Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Fernanda Luisa Basei
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, 13083-871 Campinas, SP, Brazil
| | - María Laura Rojas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - David Galvis
- Programa de Química Farmacéutica, Universidad CES, Medellín 050031, Colombia
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
30
|
Perera G, Power L, Larson A, Codden CJ, Awata J, Batorsky R, Strathdee D, Chin MT. Single Cell Transcriptomic Analysis in a Mouse Model of Barth Syndrome Reveals Cell-Specific Alterations in Gene Expression and Intercellular Communication. Int J Mol Sci 2023; 24:11594. [PMID: 37511352 PMCID: PMC10380964 DOI: 10.3390/ijms241411594] [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: 06/21/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Barth Syndrome, a rare X-linked disorder affecting 1:300,000 live births, results from defects in Tafazzin, an acyltransferase that remodels cardiolipin and is essential for mitochondrial respiration. Barth Syndrome patients develop cardiomyopathy, muscular hypotonia and cyclic neutropenia during childhood, rarely surviving to middle age. At present, no effective therapy exists, and downstream transcriptional effects of Tafazzin dysfunction are incompletely understood. To identify novel, cell-specific, pathological pathways that mediate heart dysfunction, we performed single-nucleus RNA-sequencing (snRNA-seq) on wild-type (WT) and Tafazzin-knockout (Taz-KO) mouse hearts. We determined differentially expressed genes (DEGs) and inferred predicted cell-cell communication networks from these data. Surprisingly, DEGs were distributed heterogeneously across the cell types, with fibroblasts, cardiomyocytes, endothelial cells, macrophages, adipocytes and pericytes exhibiting the greatest number of DEGs between genotypes. One differentially expressed gene was detected for the lymphatic endothelial and mesothelial cell types, while no significant DEGs were found in the lymphocytes. A Gene Ontology (GO) analysis of these DEGs showed cell-specific effects on biological processes such as fatty acid metabolism in adipocytes and cardiomyocytes, increased translation in cardiomyocytes, endothelial cells and fibroblasts, in addition to other cell-specific processes. Analysis of ligand-receptor pair expression, to infer intercellular communication patterns, revealed the strongest dysregulated communication involved adipocytes and cardiomyocytes. For the knockout hearts, there was a strong loss of ligand-receptor pair expression involving adipocytes, and cardiomyocyte expression of ligand-receptor pairs underwent reorganization. These findings suggest that adipocyte and cardiomyocyte mitochondria may be most sensitive to mitochondrial Tafazzin deficiency and that rescuing adipocyte mitochondrial dysfunction, in addition to cardiomyocyte mitochondrial dysfunction, may provide therapeutic benefit in Barth Syndrome patients.
Collapse
Affiliation(s)
- Gayani Perera
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (G.P.); (A.L.); (C.J.C.); (J.A.)
| | - Liam Power
- Medical Scientist Training Program, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Amy Larson
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (G.P.); (A.L.); (C.J.C.); (J.A.)
| | - Christina J. Codden
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (G.P.); (A.L.); (C.J.C.); (J.A.)
| | - Junya Awata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (G.P.); (A.L.); (C.J.C.); (J.A.)
| | - Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA;
| | | | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (G.P.); (A.L.); (C.J.C.); (J.A.)
| |
Collapse
|
31
|
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
|
32
|
Zegallai HM, Duan K, Hatch GM. Reduction in mRNA Expression of the Neutrophil Chemoattract Factor CXCL1 in Pseudomonas aeruginosa Treated Barth Syndrome B Lymphoblasts. BIOLOGY 2023; 12:biology12050730. [PMID: 37237543 DOI: 10.3390/biology12050730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Barth Syndrome (BTHS) is a rare X-linked genetic disease caused by a mutation in the TAFAZZIN gene, which codes for the protein tafazzin involved in cardiolipin remodeling. Approximately 70% of patients with BTHS exhibit severe infections due to neutropenia. However, neutrophils from BTHS patients have been shown to exhibit normal phagocytosis and killing activity. B lymphocytes play a crucial role in the regulation of the immune system and, when activated, secrete cytokines known to attract neutrophils to sites of infection. We examined the expression of chemokine (C-X-C motif) ligand 1 (CXCL1), a known chemotactic for neutrophils, in Epstein-Barr virus transformed control and BTHS B lymphoblasts. Age-matched control and BTHS B lymphoblasts were incubated with Pseudomonas aeruginosa for 24 h and then cell viability, CD27+, CD24+, CD38+, CD138+ and PD1+ surface marker expression and CXCL1 mRNA expression determined. Cell viability was maintained in lymphoblasts incubated in a ratio of 50:1 bacteria:B cells. Surface marker expression was unaltered between control and BTHS B lymphoblasts. In contrast, CXCL1 mRNA expression was reduced approximately 70% (p < 0.05) in untreated BTHS B lymphoblasts compared to control and approximately 90% (p < 0.05) in bacterial treated BTHS B lymphoblasts compared to the control. Thus, naïve and bacterial-activated BTHS B lymphoblasts exhibit reduced mRNA expression of the neutrophil chemoattractant factor CXCL1. We suggest that impaired bacterial activation of B cells in some BTHS patients could influence neutrophil function via impairing neutrophil recruitment to sites of infection and this could potentially contribute to these infections.
Collapse
Affiliation(s)
- Hana M Zegallai
- Department of Pharmacology & Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba 753 McDermot Avenue, Winnipeg, MB R3E0T6, Canada
| | - Kangmin Duan
- Department of Oral Biology, University of Manitoba, Winnipeg, MB R3E0T6, Canada
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba 753 McDermot Avenue, Winnipeg, MB R3E0T6, Canada
| |
Collapse
|
33
|
Van den Eynde J, Chinni B, Vernon H, Thompson WR, Hornby B, Kutty S, Manlhiot C. Identifying responders to elamipretide in Barth syndrome: Hierarchical clustering for time series data. Orphanet J Rare Dis 2023; 18:76. [PMID: 37041653 PMCID: PMC10088720 DOI: 10.1186/s13023-023-02676-8] [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: 01/06/2023] [Accepted: 03/11/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Barth syndrome (BTHS) is a rare genetic disease that is characterized by cardiomyopathy, skeletal myopathy, neutropenia, and growth abnormalities and often leads to death in childhood. Recently, elamipretide has been tested as a potential first disease-modifying drug. This study aimed to identify patients with BTHS who may respond to elamipretide, based on continuous physiological measurements acquired through wearable devices. RESULTS Data from a randomized, double-blind, placebo-controlled crossover trial of 12 patients with BTHS were used, including physiological time series data measured using a wearable device (heart rate, respiratory rate, activity, and posture) and functional scores. The latter included the 6-minute walk test (6MWT), Patient-Reported Outcomes Measurement Information System (PROMIS) fatigue score, SWAY Balance Mobile Application score (SWAY balance score), BTHS Symptom Assessment (BTHS-SA) Total Fatigue score, muscle strength by handheld dynamometry, 5 times sit-and-stand test (5XSST), and monolysocardiolipin to cardiolipin ratio (MLCL:CL). Groups were created through median split of the functional scores into "highest score" and "lowest score", and "best response to elamipretide" and "worst response to elamipretide". Agglomerative hierarchical clustering (AHC) models were implemented to assess whether physiological data could classify patients according to functional status and distinguish non-responders from responders to elamipretide. AHC models clustered patients according to their functional status with accuracies of 60-93%, with the greatest accuracies for 6MWT (93%), PROMIS (87%), and SWAY balance score (80%). Another set of AHC models clustered patients with respect to their response to treatment with elamipretide with perfect accuracy (all 100%). CONCLUSIONS In this proof-of-concept study, we demonstrated that continuously acquired physiological measurements from wearable devices can be used to predict functional status and response to treatment among patients with BTHS.
Collapse
Affiliation(s)
- Jef Van den Eynde
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Bhargava Chinni
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hilary Vernon
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - W Reid Thompson
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Brittany Hornby
- Department of Physical Therapy, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Shelby Kutty
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Cedric Manlhiot
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
34
|
San-Millán I. The Key Role of Mitochondrial Function in Health and Disease. Antioxidants (Basel) 2023; 12:antiox12040782. [PMID: 37107158 PMCID: PMC10135185 DOI: 10.3390/antiox12040782] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
The role of mitochondrial function in health and disease has become increasingly recognized, particularly in the last two decades. Mitochondrial dysfunction as well as disruptions of cellular bioenergetics have been shown to be ubiquitous in some of the most prevalent diseases in our society, such as type 2 diabetes, cardiovascular disease, metabolic syndrome, cancer, and Alzheimer's disease. However, the etiology and pathogenesis of mitochondrial dysfunction in multiple diseases have yet to be elucidated, making it one of the most significant medical challenges in our history. However, the rapid advances in our knowledge of cellular metabolism coupled with the novel understanding at the molecular and genetic levels show tremendous promise to one day elucidate the mysteries of this ancient organelle in order to treat it therapeutically when needed. Mitochondrial DNA mutations, infections, aging, and a lack of physical activity have been identified to be major players in mitochondrial dysfunction in multiple diseases. This review examines the complexities of mitochondrial function, whose ancient incorporation into eukaryotic cells for energy purposes was key for the survival and creation of new species. Among these complexities, the tightly intertwined bioenergetics derived from the combustion of alimentary substrates and oxygen are necessary for cellular homeostasis, including the production of reactive oxygen species. This review discusses different etiological mechanisms by which mitochondria could become dysregulated, determining the fate of multiple tissues and organs and being a protagonist in the pathogenesis of many non-communicable diseases. Finally, physical activity is a canonical evolutionary characteristic of humans that remains embedded in our genes. The normalization of a lack of physical activity in our modern society has led to the perception that exercise is an "intervention". However, physical activity remains the modus vivendi engrained in our genes and being sedentary has been the real intervention and collateral effect of modern societies. It is well known that a lack of physical activity leads to mitochondrial dysfunction and, hence, it probably becomes a major etiological factor of many non-communicable diseases affecting modern societies. Since physical activity remains the only stimulus we know that can improve and maintain mitochondrial function, a significant emphasis on exercise promotion should be imperative in order to prevent multiple diseases. Finally, in populations with chronic diseases where mitochondrial dysfunction is involved, an individualized exercise prescription should be crucial for the "metabolic rehabilitation" of many patients. From lessons learned from elite athletes (the perfect human machines), it is possible to translate and apply multiple concepts to the betterment of populations with chronic diseases.
Collapse
Affiliation(s)
- Iñigo San-Millán
- Department of Human Physiology and Nutrition, University of Colorado, Colorado Springs, CO 80198, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
35
|
Sabbah HN, Taylor C, Vernon HJ. Temporal evolution of the heart failure phenotype in Barth syndrome and treatment with elamipretide. Future Cardiol 2023; 19:211-225. [PMID: 37325898 DOI: 10.2217/fca-2023-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/19/2023] [Indexed: 06/17/2023] Open
Abstract
Barth syndrome (BTHS) is a rare genetic disorder caused by pathogenic variants in TAFAZZIN leading to reduced remodeled cardiolipin (CL), a phospholipid essential to mitochondrial function and structure. Cardiomyopathy presents in most patients with BTHS, typically appearing as dilated cardiomyopathy (DCM) in infancy and evolving to hypertrophic cardiomyopathy (HCM) resembling heart failure (HF) with preserved ejection fraction (HFpEF) in some patients ≥12 years. Elamipretide localizes to the inner mitochondrial membrane where it associates with CL, improving mitochondrial function, structure and bioenergetics, including ATP synthesis. Numerous preclinical and clinical studies in BTHS and other forms of HF have demonstrated that elamipretide improves left ventricular relaxation by ameliorating mitochondrial dysfunction, making it well suited for therapeutic use in adolescent and adult patients with BTHS.
Collapse
Affiliation(s)
- Hani N Sabbah
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Henry Ford Health, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Carolyn Taylor
- Department of Pediatrics, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
36
|
Tsujimoto SI, Sakamoto K, Nakano Y, Mizuno T, Shindo T, Watanabe J, Sato-Otsubo A, Osumi T, Matsumoto K, Tomizawa D, Kato M. Myelodysplastic syndrome in a patient with Barth syndrome (3-methylglutaconic aciduria type II). Pediatr Blood Cancer 2023; 70:e30033. [PMID: 36184828 DOI: 10.1002/pbc.30033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 01/25/2023]
Affiliation(s)
- Shin-Ichi Tsujimoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kenichi Sakamoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Yoshiko Nakano
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
| | - Takanori Mizuno
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Shindo
- Division of Cardiology, National Center for Child Health and Development, Tokyo, Japan
| | - Junichi Watanabe
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Aiko Sato-Otsubo
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoo Osumi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.,Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
37
|
Deng J, Jiang Y, Chen ZB, Rhee JW, Deng Y, Wang ZV. Mitochondrial Dysfunction in Cardiac Arrhythmias. Cells 2023; 12:679. [PMID: 36899814 PMCID: PMC10001005 DOI: 10.3390/cells12050679] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Electrophysiological and structural disruptions in cardiac arrhythmias are closely related to mitochondrial dysfunction. Mitochondria are an organelle generating ATP, thereby satisfying the energy demand of the incessant electrical activity in the heart. In arrhythmias, the homeostatic supply-demand relationship is impaired, which is often accompanied by progressive mitochondrial dysfunction leading to reduced ATP production and elevated reactive oxidative species generation. Furthermore, ion homeostasis, membrane excitability, and cardiac structure can be disrupted through pathological changes in gap junctions and inflammatory signaling, which results in impaired cardiac electrical homeostasis. Herein, we review the electrical and molecular mechanisms of cardiac arrhythmias, with a particular focus on mitochondrial dysfunction in ionic regulation and gap junction action. We provide an update on inherited and acquired mitochondrial dysfunction to explore the pathophysiology of different types of arrhythmias. In addition, we highlight the role of mitochondria in bradyarrhythmia, including sinus node dysfunction and atrioventricular node dysfunction. Finally, we discuss how confounding factors, such as aging, gut microbiome, cardiac reperfusion injury, and electrical stimulation, modulate mitochondrial function and cause tachyarrhythmia.
Collapse
Affiliation(s)
- Jielin Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yunqiu Jiang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Zhen Bouman Chen
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - June-Wha Rhee
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Department of Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Zhao V. Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
38
|
Phenotypic Characterization of Male Tafazzin-Knockout Mice at 3, 6, and 12 Months of Age. Biomedicines 2023; 11:biomedicines11020638. [PMID: 36831174 PMCID: PMC9953241 DOI: 10.3390/biomedicines11020638] [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: 01/28/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Barth syndrome (BTHS) is an X-linked mitochondrial disease caused by mutations in the gene encoding for tafazzin (TAZ), a key enzyme in the remodeling of cardiolipin. Mice with a germline deficiency in Taz have been generated (Taz-KO) but not yet fully characterized. We performed physiological assessments of 3-, 6-, and 12-month-old male Taz-KO mice, including measures of perinatal survival, growth, lifespan, gross anatomy, whole-body energy and substrate metabolism, glucose homeostasis, and exercise capacity. Taz-KO mice displayed reduced viability, with lower-than-expected numbers of mice recorded at 4 weeks of age, and a shortened lifespan due to disease progression. At all ages, Taz-KO mice had lower body weights compared with wild-type (Wt) littermates despite similar absolute food intakes. This finding was attributed to reduced adiposity and diminutive organs and tissues, including heart and skeletal muscles. Although there were no differences in basal levels of locomotion between age-matched genotypes, indirect calorimetry studies showed higher energy expenditure measures and respiratory exchange ratios in Taz-KO mice. At the youngest age, Taz-KO mice had comparable glucose tolerance and insulin action to Wt mice, but while these measures indicated metabolic impairments in Wt mice with advancing age that were likely associated with increasing adiposity, Taz-KO mice were protected. Comparisons across the three age-cohorts revealed a significant and more severe deterioration of exercise capacity in Taz-KO mice than in their Wt littermate controls. The Taz-KO mouse model faithfully recapitulates important aspects of BTHS, and thus provides an important new tool to investigate pathophysiological mechanisms and potential therapies.
Collapse
|
39
|
Towheed A, Goldstein AC. Genetics of Mitochondrial Cardiomyopathy. CURRENT CARDIOVASCULAR RISK REPORTS 2023. [DOI: 10.1007/s12170-023-00715-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
40
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea,Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea,Corresponding author. Tel: +82-2-2123-5662; Fax: +82-2-362-7265; E-mail:
| |
Collapse
|
41
|
Jett KA, Baker ZN, Hossain A, Boulet A, Cobine PA, Ghosh S, Ng P, Yilmaz O, Barreto K, DeCoteau J, Mochoruk K, Ioannou GN, Savard C, Yuan S, Abdalla OH, Lowden C, Kim BE, Cheng HYM, Battersby BJ, Gohil VM, Leary SC. Mitochondrial dysfunction reactivates α-fetoprotein expression that drives copper-dependent immunosuppression in mitochondrial disease models. J Clin Invest 2023; 133:e154684. [PMID: 36301669 PMCID: PMC9797342 DOI: 10.1172/jci154684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/26/2022] [Indexed: 02/04/2023] Open
Abstract
Signaling circuits crucial to systemic physiology are widespread, yet uncovering their molecular underpinnings remains a barrier to understanding the etiology of many metabolic disorders. Here, we identified a copper-linked signaling circuit activated by disruption of mitochondrial function in the murine liver or heart that resulted in atrophy of the spleen and thymus and caused a peripheral white blood cell deficiency. We demonstrated that the leukopenia was caused by α-fetoprotein, which required copper and the cell surface receptor CCR5 to promote white blood cell death. We further showed that α-fetoprotein expression was upregulated in several cell types upon inhibition of oxidative phosphorylation. Collectively, our data argue that α-fetoprotein may be secreted by bioenergetically stressed tissue to suppress the immune system, an effect that may explain the recurrent or chronic infections that are observed in a subset of mitochondrial diseases or in other disorders with secondary mitochondrial dysfunction.
Collapse
Affiliation(s)
- Kimberly A. Jett
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Zakery N. Baker
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Amzad Hossain
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Aren Boulet
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Paul A. Cobine
- Department of Biological Sciences, Auburn University, Auburn, Alabama, USA
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Philip Ng
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Orhan Yilmaz
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Kris Barreto
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - John DeCoteau
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Karen Mochoruk
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - George N. Ioannou
- Division of Gastroenterology
- Research and Development, Veterans Affairs Puget Sound Health Care System and the
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Christopher Savard
- Division of Gastroenterology
- Research and Development, Veterans Affairs Puget Sound Health Care System and the
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Sai Yuan
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - Osama H.M.H. Abdalla
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Christopher Lowden
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | | | - Vishal M. Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Scot C. Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
42
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
43
|
Koenig MK, Russo SN, McBride KL, Bjornsson HT, Gunnarsdottir BB, Goldstein A, Falk SA. Use of Elamipretide in patients assigned treatment in the compassionate use program: Case series in pediatric patients with rare orphan diseases. JIMD Rep 2023; 64:65-70. [PMID: 36636586 PMCID: PMC9830009 DOI: 10.1002/jmd2.12335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 01/16/2023] Open
Abstract
Several mitochondrial diseases are caused by pathogenic variants that impair membrane phospholipid remodeling, with no FDA-approved therapies. Elamipretide targets the inner mitochondrial membrane where it binds to cardiolipin, resulting in improved membrane stability, cellular respiration, and ATP production. In clinical trials, elamipretide produced clinical and functional improvements in adults and adolescents with mitochondrial disorders, such as primary mitochondrial myopathy and Barth syndrome; however, experience in younger patients is limited and to our knowledge, these are the first case reports on the safety and efficacy of elamipretide treatment in children under 12 years of age. We describe the use of elamipretide in patients with mitochondrial disorders to provide dosing parameters in patients aged <12 years.
Collapse
Affiliation(s)
- Mary Kay Koenig
- The University of Texas McGovern Medical School, Center for the Treatment of Pediatric Neurodegenerative DiseaseHoustonTexasUSA
| | - Sam Nick Russo
- The University of Texas McGovern Medical School, Center for the Treatment of Pediatric Neurodegenerative DiseaseHoustonTexasUSA
| | - Kim L. McBride
- Division of Genetic and Genomic Medicine and the Heart Center Nationwide Children's Hospital, Department of Pediatrics College of MedicineOhio State UniversityColumbusOhioUSA
| | - Hans Tomas Bjornsson
- Landspitali University HospitalReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
- Mckusick‐Nathans Department of Genetic MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Amy Goldstein
- Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Scott A. Falk
- Perelman School of Medicine of the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
44
|
Abstract
Mitochondrial dysfunction, especially perturbation of oxidative phosphorylation and adenosine triphosphate (ATP) generation, disrupts cellular homeostasis and is a surprisingly frequent cause of central and peripheral nervous system pathology. Mitochondrial disease is an umbrella term that encompasses a host of clinical syndromes and features caused by in excess of 300 different genetic defects affecting the mitochondrial and nuclear genomes. Patients with mitochondrial disease can present at any age, ranging from neonatal onset to late adult life, with variable organ involvement and neurological manifestations including neurodevelopmental delay, seizures, stroke-like episodes, movement disorders, optic neuropathy, myopathy, and neuropathy. Until relatively recently, analysis of skeletal muscle biopsy was the focus of diagnostic algorithms, but step-changes in the scope and availability of next-generation sequencing technology and multiomics analysis have revolutionized mitochondrial disease diagnosis. Currently, there is no specific therapy for most types of mitochondrial disease, although clinical trials research in the field is gathering momentum. In that context, active management of epilepsy, stroke-like episodes, dystonia, brainstem dysfunction, and Parkinsonism are all the more important in improving patient quality of life and reducing mortality.
Collapse
Affiliation(s)
- Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Robert McFarland
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
45
|
Liu L, Zhou K, Liu X, Hua Y, Wang H, Li Y. The interplay between cardiac dyads and mitochondria regulated the calcium handling in cardiomyocytes. Front Physiol 2022; 13:1013817. [PMID: 36531185 PMCID: PMC9755166 DOI: 10.3389/fphys.2022.1013817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/24/2022] [Indexed: 11/15/2023] Open
Abstract
Calcium mishandling and mitochondrial dysfunction have been increasingly recognized as significant factors involved in the progression procedure of cardiomyopathy. Ca2+ mishandling could cause calcium-triggered arrhythmias, which could enhance force development and ATP consumption. Mitochondrial disorganization and dysfunction in cardiomyopathy could disturb the balance of energy catabolic and anabolic procedure. Close spatial localization and arrangement of structural among T-tubule, sarcoplasmic reticulum, mitochondria are important for Ca2+ handling. So that, we illustrate the regulating network between calcium handling and mitochondrial homeostasis, as well as its intracellular mechanisms in this review, which would be worthy to develop novel therapeutic strategy and restore the function of injured cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | | | - Hua Wang
- 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, 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, China
| |
Collapse
|
46
|
Zegallai HM, Abu-El-Rub E, Mejia EM, Sparagna GC, Cole LK, Marshall AJ, Hatch GM. Tafazzin deficiency attenuates anti-cluster of differentiation 40 and interleukin-4 activation of mouse B lymphocytes. Cell Tissue Res 2022; 390:429-439. [PMID: 36129532 DOI: 10.1007/s00441-022-03692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
Barth syndrome (BTHS) is a rare X-linked genetic disease caused by mutations in TAFAZZIN. The tafazzin (Taz) protein is a cardiolipin remodeling enzyme required for maintaining mitochondrial function. Patients with BTHS exhibit impaired mitochondrial respiratory chain and metabolic function and are susceptible to serious infections. B lymphocytes (B cells) play a vital role in humoral immunity required to eradicate circulating antigens from pathogens. Intact mitochondrial respiration is required for proper B-cell function. We investigated whether Taz deficiency in mouse B cells altered their response to activation by anti-cluster of differentiation 40 (anti-CD40) + interleukin-4 (IL-4). B cells were isolated from 3-4-month-old wild type (WT) or tafazzin knockdown (TazKD) mice and were stimulated with anti-CD40 + IL-4 for 24 h and cellular bioenergetics, surface marker expression, proliferation, antibody production, and proteasome and immunoproteasome activities determined. TazKD B cells exhibited reduced mRNA expression of Taz, lowered levels of cardiolipin, and impairment in both oxidative phosphorylation and glycolysis compared to WT B cells. In addition, anti-CD40 + IL-4 stimulated TazKD B cells expressed lower levels of the immunogenic surface markers, cluster of differentiation 86 (CD86) and cluster of differentiation 69 (CD69), exhibited a lower proliferation rate, reduced production of immunoglobulin M and immunoglobulin G, and reduced proteasome and immunoproteasome proteolytic activities compared to WT B cells stimulated with anti-CD40 + IL-4. The results indicate that Taz is required to support T-cell-dependent signaling activation of mouse B cells.
Collapse
Affiliation(s)
- Hana M Zegallai
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Ejlal Abu-El-Rub
- Physiology and Pathophysiology, Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
- Physiology and Pathophysiology, Rady Faculty of Health Sciences, Regenerative Medicine, University of Manitoba, Winnipeg, Canada
| | - Edgard M Mejia
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Center, Aurora, Denver CO, USA
| | - Laura K Cole
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Aaron J Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Grant M Hatch
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
47
|
Starosta RT, Shinawi M. Primary Mitochondrial Disorders in the Neonate. Neoreviews 2022; 23:e796-e812. [PMID: 36450643 DOI: 10.1542/neo.23-12-e796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Primary mitochondrial disorders (PMDs) are a heterogeneous group of disorders characterized by functional or structural abnormalities in the mitochondria that lead to a disturbance of cellular energy, reactive oxygen species, and free radical production, as well as impairment of other intracellular metabolic functions, causing single- or multiorgan dysfunction. PMDs are caused by pathogenic variants in nuclear and mitochondrial genes, resulting in distinct modes of inheritance. Onset of disease is variable and can occur in the neonatal period, with a high morbidity and mortality. In this article, we review the most common methods used for the diagnosis of PMDs, as well as their prenatal and neonatal presentations. We highlight the shift in the diagnostic approach for PMDs since the introduction of nontargeted molecular tests into clinical practice, which has significantly reduced the use of invasive studies. We discuss common PMDs that can present in the neonate, including general, nonsyndromic presentations as well as specific syndromic disorders. We also review current treatment advances, including the use of mitochondrial "cocktails" based on limited scientific evidence and theoretical reasoning, as well as the impending arrival of personalized mitochondrial-specific treatments.
Collapse
Affiliation(s)
| | - Marwan Shinawi
- Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
48
|
Russo S, De Rasmo D, Signorile A, Corcelli A, Lobasso S. Beneficial effects of SS-31 peptide on cardiac mitochondrial dysfunction in tafazzin knockdown mice. Sci Rep 2022; 12:19847. [PMID: 36400945 PMCID: PMC9674582 DOI: 10.1038/s41598-022-24231-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Barth Syndrome (BTHS), a genetic disease associated with early-onset cardioskeletal myopathy, is caused by loss-of-function mutations of the TAFAZZIN gene, which is responsible for remodeling the mitochondrial phospholipid cardiolipin (CL). Deregulation of CL biosynthesis and maturation in BTHS mitochondria result in a dramatically increased monolysocardiolipin (MLCL)/CL ratio associated with bioenergetic dysfunction. One of the most promising therapeutic approaches for BTHS includes the mitochondria-targeted tetrapeptide SS-31, which interacts with CL. Here, we used TAFAZZIN knockdown (TazKD) mice to investigate for the first time whether in vivo administration of SS-31 could affect phospholipid profiles and mitochondrial dysfunction. The CL fingerprinting of TazKD cardiac mitochondria obtained by MALDI-TOF/MS revealed the typical lipid changes associated with BTHS. TazKD mitochondria showed lower respiratory rates in state 3 and 4 together with a decreased in maximal respiratory rates. Treatment of TazKD mice with SS-31 improved mitochondrial respiratory capacity and promoted supercomplex organization, without affecting the MLCL/CL ratio. We hypothesize that SS-31 exerts its effect by influencing the function of the respiratory chain rather than affecting CL directly. In conclusion, our results indicate that SS-31 have beneficial effects on improving cardiac mitochondrial dysfunction in a BTHS animal model, suggesting the peptide as future pharmacologic agent for therapy.
Collapse
Affiliation(s)
- Silvia Russo
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Domenico De Rasmo
- grid.503043.1CNR-Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| | - Anna Signorile
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Angela Corcelli
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Simona Lobasso
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
49
|
Nasr W, Filippi MD. Acquired and hereditary bone marrow failure: A mitochondrial perspective. Front Oncol 2022; 12:1048746. [PMID: 36408191 PMCID: PMC9666693 DOI: 10.3389/fonc.2022.1048746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
The disorders known as bone marrow failure syndromes (BMFS) are life-threatening disorders characterized by absence of one or more hematopoietic lineages in the peripheral blood. Myelodysplastic syndromes (MDS) are now considered BMF disorders with associated cellular dysplasia. BMFs and MDS are caused by decreased fitness of hematopoietic stem cells (HSC) and poor hematopoiesis. BMF and MDS can occur de novo or secondary to hematopoietic stress, including following bone marrow transplantation or myeloablative therapy. De novo BMF and MDS are usually associated with specific genetic mutations. Genes that are commonly mutated in BMF/MDS are in DNA repair pathways, epigenetic regulators, heme synthesis. Despite known and common gene mutations, BMF and MDS are very heterogenous in nature and non-genetic factors contribute to disease phenotype. Inflammation is commonly found in BMF and MDS, and contribute to ineffective hematopoiesis. Another common feature of BMF and MDS, albeit less known, is abnormal mitochondrial functions. Mitochondria are the power house of the cells. Beyond energy producing machinery, mitochondrial communicate with the rest of the cells via triggering stress signaling pathways and by releasing numerous metabolite intermediates. As a result, mitochondria play significant roles in chromatin regulation and innate immune signaling pathways. The main goal of this review is to investigate BMF processes, with a focus mitochondria-mediated signaling in acquired and inherited BMF.
Collapse
Affiliation(s)
- Waseem Nasr
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, OH, United States,University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, OH, United States,University of Cincinnati College of Medicine, Cincinnati, OH, United States,*Correspondence: Marie-Dominique Filippi,
| |
Collapse
|
50
|
Fox CA, Ryan RO. Studies of the cardiolipin interactome. Prog Lipid Res 2022; 88:101195. [DOI: 10.1016/j.plipres.2022.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
|