Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Circulation ; 144(21): 1694-1713, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34648376

ABSTRACT

BACKGROUND: Barth syndrome (BTHS) is caused by mutations of the gene encoding tafazzin, which catalyzes maturation of mitochondrial cardiolipin and often manifests with systolic dysfunction during early infancy. Beyond the first months of life, BTHS cardiomyopathy typically transitions to a phenotype of diastolic dysfunction with preserved ejection fraction, blunted contractile reserve during exercise, and arrhythmic vulnerability. Previous studies traced BTHS cardiomyopathy to mitochondrial formation of reactive oxygen species (ROS). Because mitochondrial function and ROS formation are regulated by excitation-contraction coupling, integrated analysis of mechano-energetic coupling is required to delineate the pathomechanisms of BTHS cardiomyopathy. METHODS: We analyzed cardiac function and structure in a mouse model with global knockdown of tafazzin (Taz-KD) compared with wild-type littermates. Respiratory chain assembly and function, ROS emission, and Ca2+ uptake were determined in isolated mitochondria. Excitation-contraction coupling was integrated with mitochondrial redox state, ROS, and Ca2+ uptake in isolated, unloaded or preloaded cardiac myocytes, and cardiac hemodynamics analyzed in vivo. RESULTS: Taz-KD mice develop heart failure with preserved ejection fraction (>50%) and age-dependent progression of diastolic dysfunction in the absence of fibrosis. Increased myofilament Ca2+ affinity and slowed cross-bridge cycling caused diastolic dysfunction, in part, compensated by accelerated diastolic Ca2+ decay through preactivated sarcoplasmic reticulum Ca2+-ATPase. Taz deficiency provoked heart-specific loss of mitochondrial Ca2+ uniporter protein that prevented Ca2+-induced activation of the Krebs cycle during ß-adrenergic stimulation, oxidizing pyridine nucleotides and triggering arrhythmias in cardiac myocytes. In vivo, Taz-KD mice displayed prolonged QRS duration as a substrate for arrhythmias, and a lack of inotropic response to ß-adrenergic stimulation. Cellular arrhythmias and QRS prolongation, but not the defective inotropic reserve, were restored by inhibiting Ca2+ export through the mitochondrial Na+/Ca2+ exchanger. All alterations occurred in the absence of excess mitochondrial ROS in vitro or in vivo. CONCLUSIONS: Downregulation of mitochondrial Ca2+ uniporter, increased myofilament Ca2+ affinity, and preactivated sarcoplasmic reticulum Ca2+-ATPase provoke mechano-energetic uncoupling that explains diastolic dysfunction and the lack of inotropic reserve in BTHS cardiomyopathy. Furthermore, defective mitochondrial Ca2+ uptake provides a trigger and a substrate for ventricular arrhythmias. These insights can guide the ongoing search for a cure of this orphaned disease.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/etiology , Barth Syndrome/complications , Barth Syndrome/genetics , Calcium Channels/deficiency , Myocardial Contraction/genetics , Adenosine Triphosphate/biosynthesis , Animals , Barth Syndrome/metabolism , Biomarkers , Brain/metabolism , Calcium/metabolism , Diastole , Disease Models, Animal , Disease Susceptibility , Excitation Contraction Coupling/genetics , Heart Function Tests , Humans , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , NADP/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Stroke Volume , Systole
3.
J Pediatr Endocrinol Metab ; 34(7): 951-955, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-33851526

ABSTRACT

OBJECTIVES: To describe the metabolic and endocrine features of a patient with Barth syndrome who showed evidence of growth hormone resistance. CASE PRESENTATION: A male proband deteriorated rapidly with lactic acidosis after a circumcision at age three weeks and was found to have severe dilated cardiomyopathy. A cardiomyopathy gene panel led to the diagnosis of TAZ-deficiency Barth syndrome. He subsequently experienced hypotonia and gross motor delay, feeding difficulties for the first four years, constitutional growth delay and one episode of ketotic hypoglycaemia. Cardiomyopathy resolved on oral anti-failure therapy by age three years. He had a hormonal pattern of growth hormone resistance, and growth hormone treatment was considered, however height velocity improved spontaneously after age 3½ years. He also had biochemical primary hypothyroidism. CONCLUSIONS: With careful metabolic management with l-arginine supplementation, overnight corn starch, and a prescribed exercise program, our patient's strength, endurance, level of physical activity and body composition improved significantly by age six years.


Subject(s)
Barth Syndrome/complications , Cardiomyopathy, Dilated/etiology , Growth Hormone/pharmacology , Arginine/administration & dosage , Body Height , Child , Humans , Male
6.
Orphanet J Rare Dis ; 8: 70, 2013 May 08.
Article in English | MEDLINE | ID: mdl-23656970

ABSTRACT

BACKGROUND: This study describes the natural history of Barth syndrome (BTHS). METHODS: The medical records of all patients with BTHS living in France were identified in multiple sources and reviewed. RESULTS: We identified 16 BTHS pedigrees that included 22 patients. TAZ mutations were observed in 15 pedigrees. The estimated incidence of BTHS was 1.5 cases per million births (95%CI: 0.2-2.3). The median age at presentation was 3.1 weeks (range, 0-1.4 years), and the median age at last follow-up was 4.75 years (range, 3-15 years). Eleven patients died at a median age of 5.1 months; 9 deaths were related to cardiomyopathy and 2 to sepsis. The 5-year survival rate was 51%, and no deaths were observed in patients ≥3 years. Fourteen patients presented with cardiomyopathy, and cardiomyopathy was documented in 20 during follow-up. Left ventricular systolic function was very poor during the first year of life and tended to normalize over time. Nineteen patients had neutropenia. Metabolic investigations revealed inconstant moderate 3-methylglutaconic aciduria and plasma arginine levels that were reduced or in the low-normal range. Survival correlated with two prognostic factors: severe neutropenia at diagnosis (<0.5 × 109/L) and birth year. Specifically, the survival rate was 70% for patients born after 2000 and 20% for those born before 2000. CONCLUSIONS: This survey found that BTHS outcome was affected by cardiac events and by a risk of infection that was related to neutropenia. Modern management of heart failure and prevention of infection in infancy may improve the survival of patients with BTHS without the need for heart transplantation.


Subject(s)
Barth Syndrome/mortality , Barth Syndrome/physiopathology , Acyltransferases , Adolescent , Barth Syndrome/complications , Barth Syndrome/genetics , Cardiomyopathies/complications , Child , Child, Preschool , Cohort Studies , Female , France , Humans , Male , Mutation , Neutropenia/complications , Pedigree , Survival Rate , Transcription Factors/genetics
7.
Orphanet J Rare Dis ; 8: 23, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23398819

ABSTRACT

First described in 1983, Barth syndrome (BTHS) is widely regarded as a rare X-linked genetic disease characterised by cardiomyopathy (CM), skeletal myopathy, growth delay, neutropenia and increased urinary excretion of 3-methylglutaconic acid (3-MGCA). Fewer than 200 living males are known worldwide, but evidence is accumulating that the disorder is substantially under-diagnosed. Clinical features include variable combinations of the following wide spectrum: dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), endocardial fibroelastosis (EFE), left ventricular non-compaction (LVNC), ventricular arrhythmia, sudden cardiac death, prolonged QTc interval, delayed motor milestones, proximal myopathy, lethargy and fatigue, neutropenia (absent to severe; persistent, intermittent or perfectly cyclical), compensatory monocytosis, recurrent bacterial infection, hypoglycaemia, lactic acidosis, growth and pubertal delay, feeding problems, failure to thrive, episodic diarrhoea, characteristic facies, and X-linked family history. Historically regarded as a cardiac disease, BTHS is now considered a multi-system disorder which may be first seen by many different specialists or generalists. Phenotypic breadth and variability present a major challenge to the diagnostician: some children with BTHS have never been neutropenic, whereas others lack increased 3-MGCA and a minority has occult or absent CM. Furthermore, BTHS was first described in 2010 as an unrecognised cause of fetal death. Disabling mutations or deletions of the tafazzin (TAZ) gene, located at Xq28, cause the disorder by reducing remodeling of cardiolipin, a principal phospholipid of the inner mitochondrial membrane. A definitive biochemical test, based on detecting abnormal ratios of different cardiolipin species, was first described in 2008. Key areas of differential diagnosis include metabolic and viral cardiomyopathies, mitochondrial diseases, and many causes of neutropenia and recurrent male miscarriage and stillbirth. Cardiolipin testing and TAZ sequencing now provide relatively rapid diagnostic testing, both prospectively and retrospectively, from a range of fresh or stored tissues, blood or neonatal bloodspots. TAZ sequencing also allows female carrier detection and antenatal screening. Management of BTHS includes medical therapy of CM, cardiac transplantation (in 14% of patients), antibiotic prophylaxis and granulocyte colony-stimulating factor (G-CSF) therapy. Multidisciplinary teams/clinics are essential for minimising hospital attendances and allowing many more individuals with BTHS to live into adulthood.


Subject(s)
Barth Syndrome/genetics , Barth Syndrome/complications , Barth Syndrome/diagnosis , Barth Syndrome/physiopathology , Heart Diseases/complications , Humans , Male
8.
J Inherit Metab Dis ; 36(6): 929-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23361305

ABSTRACT

Barth syndrome (BTHS) is an X-linked mitochondrial defect characterised by dilated cardiomyopathy, neutropaenia and 3-methylglutaconic aciduria (3-MGCA). We report on two affected brothers with c.646G > A (p.G216R) TAZ gene mutations. The pathogenicity of the mutation, as indicated by the structure-based functional analyses, was further confirmed by abnormal monolysocardiolipin/cardiolipin ratio in dry blood spots of the patients as well as the occurrence of this mutation in another reported BTHS proband. In both brothers, 2D-echocardiography revealed some features of left ventricular noncompaction (LVNC) despite marked differences in the course of the disease; the eldest child presented with isolated cardiomyopathy from late infancy, whereas the youngest showed severe lactic acidosis without 3-MGCA during the neonatal period. An examination of the patients' fibroblast cultures revealed that extremely low mitochondrial membrane potentials (mtΔΨ about 50 % of the control value) dominated other unspecific mitochondrial changes detected (respiratory chain dysfunction, abnormal ROS production and depressed antioxidant defense). 1) Our studies confirm generalised mitochondrial dysfunction in the skeletal muscle and the fibroblasts of BTHS patients, especially a severe impairment in the mtΔΨ and the inhibition of complex V activity. It can be hypothesised that impaired mtΔΨ and mitochondrial ATP synthase activity may contribute to episodes of cardiac arrhythmia that occurred unexpectedly in BTHS patients. 2) Severe lactic acidosis without 3-methylglutaconic aciduria in male neonates as well as an asymptomatic mild left ventricular noncompaction may characterise the ranges of natural history of Barth syndrome.


Subject(s)
Barth Syndrome/complications , Barth Syndrome/physiopathology , Membrane Potential, Mitochondrial , Barth Syndrome/diagnosis , Barth Syndrome/etiology , Cells, Cultured , Child , Child, Preschool , Humans , Male , Muscle, Skeletal/pathology , Siblings
11.
Pediatr Transplant ; 17(2): E46-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23190323

ABSTRACT

Barth syndrome is an X-linked recessive disorder that is characterized by cardiomyopathy, variable neutropenia, skeletal myopathy, growth delay, and organic aciduria. The cardiac involvement typically results in a high risk of severe heart failure in infancy or early childhood. While Berlin Heart EXCOR is widely accepted as ventricular assistance in pediatric patients with end-stage cardiac failure, infections remain a frequent and potentially severe complication. Therefore, the extended use of the device in the setting of intermittent or severe neutropenia is challenging. We present the case of a three-yr child with Barth syndrome who was bridged successfully to transplant with a Berlin Heart EXCOR assist device for eight months (251 days) without major infectious complication, despite several episodes of severe neutropenia. This case demonstrates that prolonged mechanical circulatory support for a patient with neutropenia is feasible without important morbidity, with careful monitoring and a multidisciplinary approach. G-CSF provides an excellent support in managing neutropenia.


Subject(s)
Barth Syndrome/surgery , Heart-Assist Devices , Neutropenia/etiology , Barth Syndrome/complications , Child, Preschool , Humans , Male
12.
Eur J Haematol ; 88(3): 195-209, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22023389

ABSTRACT

Barth syndrome (BTHS), a rare, X-linked, recessive disease, is characterized by neutropenia and cardiomyopathy. BTHS is caused by loss-of-function mutations of the tafazzin (TAZ) gene. We developed a model of BTHS by transfecting human HL60 myeloid progenitor cells with TAZ-specific shRNAs. Results demonstrate a significant downregulation in TAZ expression, mimicking the effects of naturally occurring truncation mutations in TAZ. Flow cytometry analyses of cells with TAZ-specific, but not scrambled, shRNAs demonstrate nearly twofold increase in the proportion of annexin V-positive cells and significantly increased dissipation of mitochondrial membrane potential as determined by DIOC6 staining. Transfection of TAZ-specific shRNA had similar effects in U937 myeloid cells but not in lymphoid cell lines. Further studies in HL60 myeloid progenitor cells revealed aberrant release of cytochrome c from mitochondria and significantly elevated levels of activated caspase-3 in response to TAZ knockdown. Treatment with caspase-specific inhibitor zVAD-fmk resulted in substantially reduced apoptosis to near-normal levels. These data suggest that neutropenia in BTHS is attributable to increased dissipation of mitochondrial membrane potential, aberrant release of cytochrome c, activation of caspase-3, and accelerated apoptosis of myeloid progenitor cells, and that this defect can be partially restored in vitro by treatment with caspase-specific inhibitors.


Subject(s)
Barth Syndrome/complications , Neutropenia/etiology , Neutropenia/metabolism , Acyltransferases , Apoptosis/genetics , Cardiolipins/metabolism , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation , Gene Silencing , HL-60 Cells , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/genetics , Myeloid Progenitor Cells/metabolism , Precursor Cells, T-Lymphoid/metabolism , RNA, Small Interfering , Transcription Factors/genetics , U937 Cells
13.
Am J Physiol Heart Circ Physiol ; 301(5): H2122-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873497

ABSTRACT

Barth syndrome (BTHS) is a mitochondrial myopathy characterized by reports of exercise intolerance. We sought to determine if 1) BTHS leads to abnormalities of skeletal muscle O(2) extraction/utilization and 2) exercise intolerance in BTHS is related to impaired O(2) extraction/utilization, impaired cardiac function, or both. Participants with BTHS (age: 17 ± 5 yr, n = 15) and control participants (age: 13 ± 4 yr, n = 9) underwent graded exercise testing on a cycle ergometer with continuous ECG and metabolic measurements. Echocardiography was performed at rest and at peak exercise. Near-infrared spectroscopy of the vastus lateralis muscle was continuously recorded for measurements of skeletal muscle O(2) extraction. Adjusting for age, peak O(2) consumption (16.5 ± 4.0 vs. 39.5 ± 12.3 ml·kg(-1)·min(-1), P < 0.001) and peak work rate (58 ± 19 vs. 166 ± 60 W, P < 0.001) were significantly lower in BTHS than control participants. The percent increase from rest to peak exercise in ejection fraction (BTHS: 3 ± 10 vs. control: 19 ± 4%, P < 0.01) was blunted in BTHS compared with control participants. The muscle tissue O(2) saturation change from rest to peak exercise was paradoxically opposite (BTHS: 8 ± 16 vs. control: -5 ± 9, P < 0.01), and the deoxyhemoglobin change was blunted (BTHS: 0 ± 12 vs. control: 10 ± 8, P < 0.09) in BTHS compared with control participants, indicating impaired skeletal muscle extraction in BTHS. In conclusion, severe exercise intolerance in BTHS is due to both cardiac and skeletal muscle impairments that are consistent with cardiac and skeletal mitochondrial myopathy. These findings provide further insight to the pathophysiology of BTHS.


Subject(s)
Barth Syndrome/complications , Cardiomyopathy, Dilated/etiology , Exercise Tolerance , Muscle Contraction , Oxygen Consumption , Oxygen/metabolism , Quadriceps Muscle/metabolism , Adolescent , Adrenergic beta-Antagonists/therapeutic use , Analysis of Variance , Barth Syndrome/diagnosis , Barth Syndrome/metabolism , Barth Syndrome/physiopathology , Biomarkers/blood , Blood Pressure , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Case-Control Studies , Child , Cross-Sectional Studies , Echocardiography , Electrocardiography , Exercise Test , Heart Rate , Hemoglobins/metabolism , Humans , Male , Oxygen/blood , Quadriceps Muscle/physiopathology , Respiratory Mechanics , Spectroscopy, Near-Infrared , Ventricular Function, Left , Young Adult
14.
Heart Fail Clin ; 6(4): 453-69, viii, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20869646

ABSTRACT

In this article the newly classified cardiomyopathy known as left ventricular noncompaction is discussed. This genetic inherited form of heart disease has substantial risk of heart failure, stroke, metabolic derangement, arrhythmias, and sudden cardiac death. The disorder seems to occur because of an arrest of the normal process of development, and the genes identified to date seem to encode for cytoskeletal or sarcomeric proteins. These features are outlined.


Subject(s)
Cardiomyopathies/diagnosis , Cardiomyopathies/therapy , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/therapy , Adaptor Proteins, Signal Transducing/genetics , Animals , Arrhythmias, Cardiac/complications , Barth Syndrome/complications , Cardiomyopathies/complications , Carrier Proteins/genetics , DNA, Mitochondrial/genetics , Disease Models, Animal , Echocardiography , Electrocardiography , Heart Defects, Congenital/complications , Heart Ventricles/pathology , Homeodomain Proteins/genetics , Humans , Incidence , Kearns-Sayre Syndrome/complications , LIM Domain Proteins , MERRF Syndrome/complications , Magnetic Resonance Imaging, Cine , Mitochondrial Myopathies/complications , Muscular Diseases/genetics , Mutation , Myocardium/cytology , NFATC Transcription Factors/genetics , TOR Serine-Threonine Kinases/genetics , Ventricular Dysfunction, Left/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...