Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Sci Rep ; 14(1): 13655, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871974

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.


Subject(s)
Acyltransferases , Barth Syndrome , Cardiolipins , Disease Models, Animal , Mitochondria, Heart , Mitophagy , Animals , Barth Syndrome/metabolism , Barth Syndrome/genetics , Barth Syndrome/pathology , Barth Syndrome/drug therapy , Mitophagy/drug effects , Mice , Acyltransferases/metabolism , Acyltransferases/genetics , Cardiolipins/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Lysophospholipids/metabolism , Mice, Knockout , Oligopeptides
2.
Nat Metab ; 5(12): 2184-2205, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37996701

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.


Subject(s)
Barth Syndrome , Animals , Humans , Barth Syndrome/genetics , Barth Syndrome/pathology , Cytochromes c , Phospholipids , Cardiolipins , Fatty Acids, Unsaturated , Peroxidases
3.
Hum Mol Genet ; 32(24): 3353-3360, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37721533

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.


Subject(s)
Barth Syndrome , Humans , Animals , Mice , Barth Syndrome/genetics , Barth Syndrome/metabolism , Barth Syndrome/pathology , Cardiolipins/genetics , Cardiolipins/metabolism , Magnesium/metabolism , Saccharomyces cerevisiae/metabolism , Calcium/metabolism , Transcription Factors/genetics , Mitochondria/metabolism , Acyltransferases/genetics
4.
EMBO Mol Med ; 15(9): e17399, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37533404

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.


Subject(s)
Barth Syndrome , Mice , Animals , Barth Syndrome/metabolism , Barth Syndrome/pathology , Cardiolipins/metabolism , AMP-Activated Protein Kinases/metabolism , Glycolysis , Fatty Acids/metabolism , Adenosine Triphosphate
5.
Future Cardiol ; 19(4): 211-225, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37325898

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.


Subject(s)
Barth Syndrome , Heart Failure , Humans , Barth Syndrome/genetics , Barth Syndrome/pathology , Heart Failure/drug therapy , Stroke Volume , Phenotype , Cardiolipins
6.
Mol Genet Genomic Med ; 11(7): e2190, 2023 07.
Article in English | MEDLINE | ID: mdl-37186429

ABSTRACT

Barth syndrome (BTHS) is an X-linked disorder characterized by cardiomyopathy, skeletal myopathy, and 3-methylglutaconic aciduria. The causative pathogenic variants for BTHS are in TAZ, which encodes a putative acyltransferase named tafazzin and is involved in the remodeling of cardiolipin in the inner mitochondrial membranes. Pathogenic variants in TAZ result in mitochondrial structural and functional abnormalities. We report a case of infantile BTHS with severe heart failure, left ventricular noncompaction, and lactic acidosis, having a missense c.640C>T (p.His214Tyr) variant in TAZ, which is considered a pathogenic variant based on the previously reported amino acid substitution at the same site (c.641A>G, p.His214Arg). However, in this previously reported case, heart function was compensated and not entirely similar to the present case. Silico prediction analysis suggested that c.640C>T could alter the TAZ messenger RNA (mRNA) splicing process. TAZ mRNAs in isolated peripheral mononuclear cells from the patient and in vitro splicing analysis using minigenes of TAZ found an 8 bp deletion at the 3' end of exon 8, which resulted in the formation of a termination codon in the coding region of exon 9 (H214Nfs*3). These findings suggest that splicing abnormalities should always be considered in BTHS.


Subject(s)
Barth Syndrome , Cardiomyopathies , Heart Defects, Congenital , Heart Failure , Humans , Barth Syndrome/genetics , Barth Syndrome/pathology , Cardiomyopathies/genetics , Heart Defects, Congenital/genetics , Heart Failure/genetics , Transcription Factors/genetics
7.
Stem Cell Res ; 64: 102923, 2022 10.
Article in English | MEDLINE | ID: mdl-36219982

ABSTRACT

Tafazzin (TAZ), a mitochondrial transacylase located on chromosome X, is required for the production of the mitochondrial phospholipid cardiolipin. Mutations occurring in the TAZ gene will lead to Barth syndrome, an X-linked recessive disease generally presenting as cardiomyopathy affecting males. Disease modeling strategies based on pluripotent stem cells (PSCs) provide an unprecedented and powerful platform to study Barth Syndrome. However, current studies were mostly based on male PSCs, the results and conclusions of which neglected the potential distinctions existing in disease phenotypes and mechanisms between gender. In this study, based on the H9 cell line (Female), we generated a homozygous TAZ knockout (TAZ-KO) human embryonic stem cell (hESC) line by employing CRISPR/Cas9 genome editing tools. This female TAZ-KO cell line, with normal karyotype, robust pluripotency and remarkably reduced TAZ expression, would be a useful tool for further deeply studying the pathogenesis of Barth syndrome cardiomyopathy in females.


Subject(s)
Barth Syndrome , Cardiomyopathies , Human Embryonic Stem Cells , Male , Female , Humans , Barth Syndrome/genetics , Barth Syndrome/metabolism , Barth Syndrome/pathology , Cardiolipins/metabolism , Human Embryonic Stem Cells/metabolism , CRISPR-Cas Systems/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Cardiomyopathies/genetics
8.
Sci Rep ; 12(1): 9466, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35676289

ABSTRACT

Barth syndrome (BTHS) is caused by mutations in the TAZ gene encoding the cardiolipin remodeling enzyme, Tafazzin. The study objective was to quantitatively examine growth characteristics and mitochondrial morphology of transformed lymphoblast cell lines derived from five patients with BTHS relative to five healthy controls, as well as the therapeutic potential of oleoylethanolamide (OEA) and linoleoylethanolamide (LEA). These bioactive lipids both activate PPARα, which may be therapeutic. BTHS lymphoblasts grew more slowly than controls, suggesting lymphopenia merits clinical investigation. Treatment of BTHS lymphoblasts with OEA, but not LEA, significantly restored mitochondrial membrane potential, as well as colony growth in all BTHS lymphoblast lines, although a full growth rescue was not achieved. Quantification analysis of electron micrographs from three BTHS and healthy lymphoblast donors indicated similar numbers of mitochondria per cell, but lower average cristae length per mitochondrion, and higher mitochondrial density. Additionally, BTHS lymphoblasts had larger mitochondria, and a higher percentage of abnormally large mitochondria (> 1 µm2) than healthy controls. Notably, OEA treatment significantly restored mitochondrial size, without affecting density or cristae lengths. Cardiolipin total content, relative linoleic acid content and monolysocardiolipin:cardiolipin ratios were not improved by OEA, indicating that effects on growth, and mitochondrial morphology and function, occurred without resolving this deficit. However, immunoblotting showed higher levels of OPA1, a biomarker for mitochondrial fusion, in BTHS lymphoblasts, which was attenuated by OEA treatment, implicating altered mitochondrial dynamics in the pathology and treatment of BTHS.


Subject(s)
Acyltransferases/metabolism , Barth Syndrome , Cardiolipins , Lymphocytes , Acyltransferases/genetics , Barth Syndrome/genetics , Barth Syndrome/metabolism , Barth Syndrome/pathology , Cardiolipins/metabolism , Endocannabinoids , Humans , Mitochondria/metabolism , Oleic Acids , Transcription Factors/metabolism
9.
J Inherit Metab Dis ; 45(1): 7-16, 2022 01.
Article in English | MEDLINE | ID: mdl-34355402

ABSTRACT

Barth Syndrome is a rare X-linked disorder caused by pathogenic variants in the gene TAFAZZIN, which encodes for an enzyme involved in the remodeling of cardiolipin, a phospholipid primarily localized to the inner mitochondrial membrane. Barth Syndrome is characterized by cardiomyopathy, skeletal myopathy, neutropenia, and growth abnormalities, among other features. In this review, we will discuss the clinical presentation and natural history of Barth Syndrome, review key features of this disease, and introduce less common clinical associations. Recognition and understanding of the natural history of Barth Syndrome are important for ongoing patient management and developing endpoints for the demonstration of efficacy of new and emerging therapies.


Subject(s)
Barth Syndrome/metabolism , Barth Syndrome/pathology , Acyltransferases/genetics , Barth Syndrome/genetics , Barth Syndrome/therapy , Cardiolipins/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Humans , Mitochondrial Membranes/metabolism , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation , Neutropenia/metabolism , Neutropenia/pathology
10.
J Inherit Metab Dis ; 45(1): 38-50, 2022 01.
Article in English | MEDLINE | ID: mdl-34494285

ABSTRACT

Deficiency of the transacylase tafazzin due to loss of function variants in the X-chromosomal TAFAZZIN gene causes Barth syndrome (BTHS) with severe neonatal or infantile cardiomyopathy, neutropenia, myopathy, and short stature. The condition is characterized by drastic changes in the composition of cardiolipins, a mitochondria-specific class of phospholipids. Studies examining the impact of tafazzin deficiency on the metabolism of other phospholipids have so far generated inhomogeneous and partly conflicting results. Recent studies showed that the cardiolipin composition in cells and different murine tissues is highly dependent on the surrounding lipid environment. In order to study the relevance of different lipid states and tafazzin function for cardiolipin and phospholipid homeostasis we conducted systematic modulation experiments in a CRISPR/Cas9 knock-out model for BTHS. We found that-irrespective of tafazzin function-the composition of cardiolipins strongly depends on the nutritionally available lipid pool. Tafazzin deficiency causes a consistent shift towards cardiolipin species with more saturated and shorter acyl chains. Interestingly, the typical biochemical BTHS phenotype in phospholipid profiles of HEK 293T TAZ knock-out cells strongly depends on the cellular lipid context. In response to altered nutritional lipid compositions, we measured more pronounced changes on phospholipids that were largely masked under standard cell culturing conditions, therewith giving a possible explanation for the conflicting results reported so far on BTHS lipid phenotypes.


Subject(s)
Acyltransferases/genetics , Barth Syndrome/metabolism , Cardiolipins/metabolism , Phospholipids/metabolism , Barth Syndrome/genetics , Barth Syndrome/pathology , CRISPR-Cas Systems , Gene Knockout Techniques , HEK293 Cells , Humans , Mitochondria/metabolism , Mutation
11.
Mitochondrion ; 61: 188-195, 2021 11.
Article in English | MEDLINE | ID: mdl-34197965

ABSTRACT

Barth syndrome is a rare X-linked multisystem mitochondrial disease that is caused by variants in the tafazzin gene leading to deficient and abnormal cardiolipin. Previous research has focused on the cardiomyopathy and neutropenia in individuals with Barth syndrome, yet just as common are the least explored neurological aspects of Barth syndrome. This review focuses on the major neuropsychological and neurophysiological phenotypes that affect the quality of life of individuals with Barth syndrome, including difficulties in sensory perception and feeding, fatigue, and cognitive and psychological challenges. We propose selected pathogenetic mechanisms underlying these phenotypes and draw parallels to other relevant disorders. Finally, avenues for future research are also suggested.


Subject(s)
Acyltransferases/genetics , Barth Syndrome/pathology , Barth Syndrome/genetics , Humans
12.
Mol Cell Biochem ; 476(3): 1605-1629, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33415565

ABSTRACT

Barth syndrome is a rare X-linked genetic disease classically characterized by cardiomyopathy, skeletal myopathy, growth retardation, neutropenia, and 3-methylglutaconic aciduria. It is caused by mutations in the tafazzin gene localized to chromosome Xq28.12. Mutations in tafazzin may result in alterations in the level and molecular composition of the mitochondrial phospholipid cardiolipin and result in large elevations in the lysophospholipid monolysocardiolipin. The increased monolysocardiolipin:cardiolipin ratio in blood is diagnostic for the disease, and it leads to disruption in mitochondrial bioenergetics. In this review, we discuss cardiolipin structure, synthesis, and function and provide an overview of the clinical and cellular pathophysiology of Barth Syndrome. We highlight known pharmacological management for treatment of the major pathological features associated with the disease. In addition, we discuss non-pharmacological management. Finally, we highlight the most recent promising therapeutic options for this rare mitochondrial disease including lipid replacement therapy, peroxisome proliferator-activated receptor agonists, tafazzin gene replacement therapy, induced pluripotent stem cells, mitochondria-targeted antioxidants and peptides, and the polyphenolic compound resveratrol.


Subject(s)
Acyltransferases/genetics , Barth Syndrome/pathology , Barth Syndrome/therapy , Mutation , Animals , Antioxidants/metabolism , Barth Syndrome/genetics , Cardiolipins/metabolism , Cardiomyopathies/metabolism , Cholesterol/blood , Cognition , HEK293 Cells , Humans , Lysophospholipids/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Myocardium/metabolism , Neutropenia/metabolism , Oxidative Phosphorylation , Phenotype , Transcription Factors/genetics
13.
FEBS Lett ; 595(3): 415-432, 2021 02.
Article in English | MEDLINE | ID: mdl-33112430

ABSTRACT

Barth syndrome (BTHS) is a rare X-linked genetic disorder caused by mutations in the gene encoding the transacylase tafazzin and characterized by loss of cardiolipin and severe cardiomyopathy. Mitochondrial oxidants have been implicated in the cardiomyopathy in BTHS. Eleven mitochondrial sites produce superoxide/hydrogen peroxide (H2 O2 ) at significant rates. Which of these sites generate oxidants at excessive rates in BTHS is unknown. Here, we measured the maximum capacity of superoxide/H2 O2 production from each site and the ex vivo rate of superoxide/H2 O2 production in the heart and skeletal muscle mitochondria of the tafazzin knockdown mice (tazkd) from 3 to 12 months of age. Despite reduced oxidative capacity, superoxide/H2 O2 production was indistinguishable between tazkd mice and wild-type littermates. These observations raise questions about the involvement of mitochondrial oxidants in BTHS pathology.


Subject(s)
Acyltransferases/genetics , Barth Syndrome/genetics , Mitochondria, Heart/enzymology , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Myocardium/enzymology , Acyltransferases/deficiency , Animals , Barth Syndrome/enzymology , Barth Syndrome/pathology , Cardiolipins/metabolism , Disease Models, Animal , Electron Transport Chain Complex Proteins , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Knockout , Mitochondria, Heart/pathology , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Myocardium/pathology , NAD/metabolism , Oxygen Consumption/genetics , Superoxides/metabolism
14.
Cell Metab ; 32(6): 981-995.e7, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33264603

ABSTRACT

Mitochondria constantly adapt to the metabolic needs of a cell. This mitochondrial plasticity is critical to T cells, which modulate metabolism depending on antigen-driven signals and environment. We show here that de novo synthesis of the mitochondrial membrane-specific lipid cardiolipin maintains CD8+ T cell function. T cells deficient for the cardiolipin-synthesizing enzyme PTPMT1 had reduced cardiolipin and responded poorly to antigen because basal cardiolipin levels were required for activation. However, neither de novo cardiolipin synthesis, nor its Tafazzin-dependent remodeling, was needed for T cell activation. In contrast, PTPMT1-dependent cardiolipin synthesis was vital when mitochondrial fitness was required, most notably during memory T cell differentiation or nutrient stress. We also found CD8+ T cell defects in a small cohort of patients with Barth syndrome, where TAFAZZIN is mutated, and in a Tafazzin-deficient mouse model. Thus, the dynamic regulation of a single mitochondrial lipid is crucial for CD8+ T cell immunity.


Subject(s)
Acyltransferases/immunology , Barth Syndrome/immunology , CD8-Positive T-Lymphocytes/immunology , Cardiolipins/immunology , Mitochondria/immunology , PTEN Phosphohydrolase/immunology , Animals , Barth Syndrome/pathology , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Cells ; 9(10)2020 10 21.
Article in English | MEDLINE | ID: mdl-33096711

ABSTRACT

Tafazzin is a phospholipid transacylase that catalyzes the remodeling of cardiolipin, a mitochondrial phospholipid required for oxidative phosphorylation. Mutations of the tafazzin gene cause Barth syndrome, which is characterized by mitochondrial dysfunction and dilated cardiomyopathy, leading to premature death. However, the molecular mechanisms underlying the cause of mitochondrial dysfunction in Barth syndrome remain poorly understood. We again highlight the fact that the tafazzin deficiency is also linked to defective oxidative phosphorylation associated with oxidative stress. All the mitochondrial events are positioned in a context where mitophagy is a key element in mitochondrial quality control. Here, we investigated the role of tafazzin in mitochondrial homeostasis dysregulation and mitophagy alteration. Using a HeLa cell model of tafazzin deficiency, we show that dysregulation of tafazzin in HeLa cells induces alteration of mitophagy. Our findings provide some additional insights into mitochondrial dysfunction associated with Barth syndrome, but also show that mitophagy inhibition is concomitant with apoptosis dysfunction through the inability of abnormal mitochondrial cardiolipin to assume its role in cytoplasmic signal transduction. Our work raises hope that pharmacological manipulation of the mitophagic pathway together with mitochondrially targeted antioxidants may provide new insights leading to promising treatment for these highly lethal conditions.


Subject(s)
Barth Syndrome/genetics , Cardiolipins/metabolism , Mitochondria/metabolism , Mitophagy/genetics , Mutation/genetics , Superoxides/metabolism , Transcription Factors/genetics , Acyltransferases , Adenylate Kinase/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Barth Syndrome/pathology , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Energy Metabolism/drug effects , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/ultrastructure , Mitophagy/drug effects , Organelle Biogenesis , Sirolimus/pharmacology
16.
J Biol Chem ; 295(35): 12485-12497, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32665401

ABSTRACT

Barth syndrome is a mitochondrial myopathy resulting from mutations in the tafazzin (TAZ) gene encoding a phospholipid transacylase required for cardiolipin remodeling. Cardiolipin is a phospholipid of the inner mitochondrial membrane essential for the function of numerous mitochondrial proteins and processes. However, it is unclear how tafazzin deficiency impacts cardiac mitochondrial metabolism. To address this question while avoiding confounding effects of cardiomyopathy on mitochondrial phenotype, we utilized Taz-shRNA knockdown (TazKD ) mice, which exhibit defective cardiolipin remodeling and respiratory supercomplex instability characteristic of human Barth syndrome but normal cardiac function into adulthood. Consistent with previous reports from other models, mitochondrial H2O2 emission and oxidative damage were greater in TazKD than in wild-type (WT) hearts, but there were no differences in oxidative phosphorylation coupling efficiency or membrane potential. Fatty acid and pyruvate oxidation capacities were 40-60% lower in TazKD mitochondria, but an up-regulation of glutamate oxidation supported respiration rates approximating those with pyruvate and palmitoylcarnitine in WT. Deficiencies in mitochondrial CoA and shifts in the cardiac acyl-CoA profile paralleled changes in fatty acid oxidation enzymes and acyl-CoA thioesterases, suggesting limitations of CoA availability or "trapping" in TazKD mitochondrial metabolism. Incubation of TazKD mitochondria with exogenous CoA partially rescued pyruvate and palmitoylcarnitine oxidation capacities, implicating dysregulation of CoA-dependent intermediary metabolism rather than respiratory chain defects in the bioenergetic impacts of tafazzin deficiency. These findings support links among cardiolipin abnormalities, respiratory supercomplex instability, and mitochondrial oxidant production and shed new light on the distinct metabolic consequences of tafazzin deficiency in the mammalian heart.


Subject(s)
Barth Syndrome/metabolism , Coenzyme A/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Transcription Factors/deficiency , Acyltransferases , Animals , Barth Syndrome/genetics , Barth Syndrome/pathology , Coenzyme A/genetics , Electron Transport , Female , Humans , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Myocardium/pathology , Oxidation-Reduction , Transcription Factors/metabolism
17.
J Biol Chem ; 295(33): 11928-11937, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32636300

ABSTRACT

Cardiolipin (CL) is the signature phospholipid of mitochondrial membranes, where it is synthesized locally and plays an important role in mitochondrial bioenergetics. Previous studies in the yeast model have indicated that CL is required for optimal iron homeostasis, which is disrupted by a mechanism not yet determined in the yeast CL mutant, crd1Δ. This finding has implications for the severe genetic disorder, Barth syndrome (BTHS), in which CL metabolism is perturbed because of mutations in the CL-remodeling enzyme, tafazzin. Here, we investigate the effects of tafazzin deficiency on iron homeostasis in the mouse myoblast model of BTHS tafazzin knockout (TAZ-KO) cells. Similarly to CL-deficient yeast cells, TAZ-KO cells exhibited elevated sensitivity to iron, as well as to H2O2, which was alleviated by the iron chelator deferoxamine. TAZ-KO cells exhibited increased expression of the iron exporter ferroportin and decreased expression of the iron importer transferrin receptor, likely reflecting a regulatory response to elevated mitochondrial iron. Reduced activities of mitochondrial iron-sulfur cluster enzymes suggested that the mechanism underlying perturbation of iron homeostasis was defective iron-sulfur biogenesis. We observed decreased levels of Yfh1/frataxin, an essential component of the iron-sulfur biogenesis machinery, in mitochondria from TAZ-KO mouse cells and in CL-deleted yeast crd1Δ cells, indicating that the role of CL in iron-sulfur biogenesis is highly conserved. Yeast crd1Δ cells exhibited decreased processing of the Yfh1 precursor upon import, which likely contributes to the iron homeostasis defects. Implications for understanding the pathogenesis of BTHS are discussed.


Subject(s)
Barth Syndrome/metabolism , Cardiolipins/metabolism , Iron-Binding Proteins/metabolism , Iron/metabolism , Myoblasts/metabolism , Acyltransferases , Animals , Barth Syndrome/genetics , Barth Syndrome/pathology , Cardiolipins/genetics , Cell Line , Gene Deletion , Gene Knockout Techniques , Iron-Binding Proteins/genetics , Mice , Myoblasts/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Frataxin
18.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165803, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32348916

ABSTRACT

Mitochondria play a prominent role in cardiac energy metabolism, and their function is critically dependent on the integrity of mitochondrial membranes. Disorders characterized by mitochondrial dysfunction are commonly associated with cardiac disease. The mitochondrial phospholipid cardiolipin directly interacts with a number of essential protein complexes in the mitochondrial membranes including the respiratory chain, mitochondrial metabolite carriers, and proteins critical for mitochondrial morphology. Barth syndrome is an X-linked disorder caused by an inherited defect in the biogenesis of the mitochondrial phospholipid cardiolipin. How cardiolipin deficiency impacts on mitochondrial function and how mitochondrial dysfunction causes cardiomyopathy has been intensively studied in cellular and animal models of Barth syndrome. These findings may also have implications for the molecular mechanisms underlying other inherited disorders associated with defects in cardiolipin, such as Sengers syndrome and dilated cardiomyopathy with ataxia (DCMA).


Subject(s)
Barth Syndrome/metabolism , Cardiolipins/metabolism , Cardiomyopathies/metabolism , Cardiomyopathy, Dilated/metabolism , Cataract/metabolism , Cerebellar Ataxia/metabolism , Metabolism, Inborn Errors/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Barth Syndrome/genetics , Barth Syndrome/pathology , Biological Transport , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cataract/genetics , Cataract/pathology , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Electron Transport/genetics , Humans , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/pathology , Mitochondria/pathology , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitophagy/genetics , Myocardium/metabolism , Myocardium/pathology , Phosphatidic Acids/metabolism
19.
Circ Res ; 126(8): 1024-1039, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32146862

ABSTRACT

RATIONALE: Barth syndrome is an X-linked cardiac and skeletal myopathy caused by mutation of the gene Tafazzin (TAZ). Currently, there is no targeted treatment for Barth syndrome. Lack of a proper genetic animal model that recapitulates the features of Barth syndrome has hindered understanding of disease pathogenesis and therapeutic development. OBJECTIVE: We characterized murine germline TAZ knockout mice (TAZ-KO) and cardiomyocyte-specific TAZ knockout mice models and tested the efficacy of adeno-associated virus (AAV)-mediated gene replacement therapy with human TAZ (hTAZ). METHODS AND RESULTS: TAZ-KO caused embryonic and neonatal lethality, impaired growth, dilated cardiomyopathy, and skeletal myopathy. TAZ-KO mice that survived the neonatal period developed progressive, severe cardiac dysfunction, and fibrosis. Cardiomyocyte-specific inactivation of floxed Taz in cardiomyocytes using Myh6-Cre caused progressive dilated cardiomyopathy without fetal or perinatal loss. Using both constitutive and conditional knockout models, we tested the efficacy and durability of Taz replacement by AAV gene therapy. Neonatal AAV-hTAZ rescued neonatal death, cardiac dysfunction, and fibrosis in TAZ-KO mice, and both prevented and reversed established cardiac dysfunction in TAZ-KO and cardiomyocyte-specific TAZ knockout mice models. However, both neonatal and adult therapies required high cardiomyocyte transduction (≈70%) for durable efficacy. CONCLUSIONS: TAZ-KO and cardiomyocyte-specific TAZ knockout mice recapitulate many of the key clinical features of Barth syndrome. AAV-mediated gene replacement is efficacious when a sufficient fraction of cardiomyocytes are transduced.


Subject(s)
Barth Syndrome/genetics , Barth Syndrome/therapy , Dependovirus/genetics , Genetic Therapy/methods , Heart Failure/genetics , Heart Failure/therapy , Animals , Barth Syndrome/pathology , Disease Models, Animal , Heart Failure/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology
20.
Am J Med Genet A ; 182(1): 64-70, 2020 01.
Article in English | MEDLINE | ID: mdl-31729175

ABSTRACT

Infantile onset cardiomyopathies are highly heterogeneous with several phenocopies compared with adult cardiomyopathies. Multidisciplinary management is essential in determining the underlying etiology in children's cardiomyopathy. Elevated urinary excretion of 3-methylglutaconic acid (3-MGA) is a useful tool in identifying the etiology in some metabolic cardiomyopathy. Here, we report the delayed appearance of 3-MGA-uria, between 6 and 18 months in three patients (out of 100 childhood onset cardiomyopathy) with neonatal onset cardiomyopathy, secondary to TMEM70 mutations and TAZ mutations (Barth syndrome), in whom extensive metabolic investigations, performed in the first weeks of life, did not display 3-MGA-uria. Serial retrospective evaluations showed full characteristic features of TMEM70 and TAZ mutations (Barth syndrome) in these three patients, including a clearly abnormal monolysocardiolipin/cardiolipin ratio in the two Barth syndrome patients. Serially repeated metabolic investigations finally discovered the 3-MGA-uria biomarker in all three patients between the age of 6 and 18 months. Our observation provides novel insights into the temporal appearance of 3-MGA-uria in TMEM70 and TAZ mutations (Barth syndrome) and focus the importance of multidisciplinary management and careful evaluation of family history and red flag signs for phenocopies in infantile onset cardiomyopathies.


Subject(s)
Barth Syndrome/genetics , Membrane Proteins/genetics , Metabolic Syndrome/genetics , Metabolism, Inborn Errors/genetics , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Acyltransferases , Adult , Age of Onset , Barth Syndrome/pathology , Barth Syndrome/urine , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Child , Female , Glutarates/metabolism , Glutarates/urine , Humans , Infant , Infant, Newborn , Male , Metabolic Syndrome/pathology , Metabolic Syndrome/urine , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/urine , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...