Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Cell ; 74(5): 877-890.e6, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31023583

ABSTRACT

Endoplasmic reticulum (ER) stress and unfolded protein response are energetically challenging under nutrient stress conditions. However, the regulatory mechanisms that control the energetic demand under nutrient and ER stress are largely unknown. Here we show that ER stress and glucose deprivation stimulate mitochondrial bioenergetics and formation of respiratory supercomplexes (SCs) through protein kinase R-like ER kinase (PERK). Genetic ablation or pharmacological inhibition of PERK suppresses nutrient and ER stress-mediated increases in SC levels and reduces oxidative phosphorylation-dependent ATP production. Conversely, PERK activation augments respiratory SCs. The PERK-eIF2α-ATF4 axis increases supercomplex assembly factor 1 (SCAF1 or COX7A2L), promoting SCs and enhanced mitochondrial respiration. PERK activation is sufficient to rescue bioenergetic defects caused by complex I missense mutations derived from mitochondrial disease patients. These studies have identified an energetic communication between ER and mitochondria, with implications in cell survival and diseases associated with mitochondrial failures.


Subject(s)
Activating Transcription Factor 4/genetics , Energy Metabolism/genetics , Eukaryotic Initiation Factor-2/genetics , Mitochondria/genetics , eIF-2 Kinase/genetics , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Line , Cell Survival/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Glucose/metabolism , Humans , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation, Missense/genetics , Nutrients/metabolism , Phosphorylation , Serine-Arginine Splicing Factors/genetics , Signal Transduction
2.
Mol Ther Methods Clin Dev ; 13: 167-179, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-30788385

ABSTRACT

Barth syndrome (BTHS) is a rare mitochondrial disease that causes severe cardiomyopathy and has no disease-modifying therapy. It is caused by recessive mutations in the gene tafazzin (TAZ), which encodes tafazzin-an acyltransferase that remodels the inner mitochondrial membrane lipid cardiolipin. To identify novel mechanistic pathways involved in BTHS and evaluate the effects of gene therapy on proteomic profiles, we performed a multiplex tandem mass tagging (TMT) quantitative proteomics analysis to compare protein expression profiles from heart lysates isolated from BTHS, healthy wild-type (WT), and BTHS treated with adeno-associated virus serotype 9 (AAV9)-TAZ gene replacement as neonates or adults. 197 proteins with ≥2 unique peptides were identified. Of these, 91 proteins were significantly differentially expressed in BTHS compared to WT controls. Cause-effect relationships between tafazzin deficiency and altered protein profiles were confirmed through demonstrated significant improvements in expression levels following administration of AAV9-TAZ. The importance of TMEM65 in Cx43 localization to cardiac intercalated discs was revealed as a novel consequence of tafazzin deficiency that was improved following gene therapy. This study identifies novel mechanistic pathways involved in the pathophysiology of BTHS, demonstrates the ability of gene delivery to improve protein expression profiles, and provides support for clinical translation of AAV9-TAZ gene therapy.

3.
Hum Gene Ther ; 30(2): 139-154, 2019 02.
Article in English | MEDLINE | ID: mdl-30070157

ABSTRACT

Barth syndrome (BTHS) is a rare mitochondrial disease that affects heart and skeletal muscle and has no curative treatment. It is caused by recessive mutations in the X-linked gene TAZ, which encodes tafazzin. To develop a clinically relevant gene therapy to restore tafazzin function and treat BTHS, three different adeno-associated virus serotype 9 vectors were tested and compared to identify the optimal promoter-cytomegalovirus (CMV), desmin (Des), or a native tafazzin promoter (Taz)-for TAZ expression following intravenous administration of 1 × 1013 vector genomes/kilogram to a mouse model of BTHS as either neonates (1-2 days of age) or adults (3 months of age). At 5 months of age, evaluations of biodistribution and TAZ expression levels, mouse activity assessments, fatigue in response to exercise, muscle strength, cardiac function, mitochondrial structure, oxygen consumption, and electron transport chain complex activity assays were performed to measure the extent of improvement in treated mice. Each promoter was scored for significant improvement over untreated control mice and significant improvement compared with the other two promoters for every measurement and within each age of administration. All three of the promoters resulted in significant improvements in a majority of the assessments compared with untreated BTHS controls. When scored for overall effectiveness as a gene therapy, the Des promoter was found to provide improvement in the most assessments, followed by the CMV promoter, and finally Taz regardless of injection age. This study provides substantial support for translation of an adeno-associated virus serotype 9-mediated TAZ gene replacement strategy using a Des promoter for human BTHS patients in the clinic.


Subject(s)
Barth Syndrome , Dependovirus , Genetic Therapy , Genetic Vectors , Transcription Factors , Transduction, Genetic , Acyltransferases , Animals , Barth Syndrome/genetics , Barth Syndrome/metabolism , Barth Syndrome/physiopathology , Barth Syndrome/therapy , Female , Humans , Male , Mice , Mice, Transgenic , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Recovery of Function/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
4.
Cell Death Dis ; 9(6): 658, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855477

ABSTRACT

Mitochondrial mutations cause bioenergetic defects associated with failures to use the electron transfer chain and oxidize substrates. These defects are exacerbated under energetic stress conditions and ultimately cause cell deterioration and death. However, little is known about cellular strategies that rescue mitochondrial stress failures and maintain cell survival under these conditions. Here, we have designed and performed a high-throughput chemical screen to identify small molecules that rescue human mitochondrial complex I mutations from energetic stress-induced cell death. The top positive hits were a series of sulfonylureas that efficiently maintain prolonged cell survival and growth under energetic stress conditions. The addition of galactose instead of glucose, to experimentally force mitochondrial respiration, triggered an initial ER stress response that was associated with IRE1α-dependent inflammatory signals including JNK and p38 MAP kinases in mutant cells. Sulfonylureas, similar to inhibition of IRE1α and p38 MAP kinase, potently blocked this ER stress inflammatory and cell death pathway and maintained viability and cell growth under severe energetic stress conditions. These studies reveal that sulfonylureas and specific inhibition of the IRE1α inflammatory pathway protect against cell death and can be used to rescue bioenergetic failures in mitochondrial complex I-mutated cells under stress conditions.


Subject(s)
Apoptosis , Cytoprotection , Electron Transport Complex I/genetics , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Inflammation/pathology , Mitochondria/metabolism , Mutation/genetics , Protein Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Cytoprotection/drug effects , Electron Transport Complex I/metabolism , Endoplasmic Reticulum Stress/drug effects , Galactose , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Sulfonylurea Compounds/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Mol Cell ; 64(1): 163-175, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27666594

ABSTRACT

Mitochondrial diseases comprise a heterogeneous group of genetically inherited disorders that cause failures in energetic and metabolic function. Boosting residual oxidative phosphorylation (OXPHOS) activity can partially correct these failures. Herein, using a high-throughput chemical screen, we identified the bromodomain inhibitor I-BET 525762A as one of the top hits that increases COX5a protein levels in complex I (CI) mutant cybrid cells. In parallel, bromodomain-containing protein 4 (BRD4), a target of I-BET 525762A, was identified using a genome-wide CRISPR screen to search for genes whose loss of function rescues death of CI-impaired cybrids grown under conditions requiring OXPHOS activity for survival. We show that I-BET525762A or loss of BRD4 remodeled the mitochondrial proteome to increase the levels and activity of OXPHOS protein complexes, leading to rescue of the bioenergetic defects and cell death caused by mutations or chemical inhibition of CI. These studies show that BRD4 inhibition may have therapeutic implications for the treatment of mitochondrial diseases.


Subject(s)
Benzodiazepines/pharmacology , Cytochrome c Group/genetics , Electron Transport Complex I/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Cell Cycle Proteins , Cell Fusion , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Cytochrome c Group/metabolism , Electron Transport Complex I/deficiency , Electron Transport Complex IV , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Screening Assays , Humans , Metabolome , Metabolomics , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
6.
Mol Cell Biol ; 36(1): 184-96, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26503783

ABSTRACT

Mitochondrial oxidative and thermogenic functions in brown and beige adipose tissues modulate rates of energy expenditure. It is unclear, however, how beige or white adipose tissue contributes to brown fat thermogenic function or compensates for partial deficiencies in this tissue and protects against obesity. Here, we show that the transcription factor Yin Yang 1 (YY1) in brown adipose tissue activates the canonical thermogenic and uncoupling gene expression program. In contrast, YY1 represses a series of secreted proteins, including fibroblast growth factor 21 (FGF21), bone morphogenetic protein 8b (BMP8b), growth differentiation factor 15 (GDF15), angiopoietin-like 6 (Angptl6), neuromedin B, and nesfatin, linked to energy expenditure. Despite substantial decreases in mitochondrial thermogenic proteins in brown fat, mice lacking YY1 in this tissue are strongly protected against diet-induced obesity and exhibit increased energy expenditure and oxygen consumption in beige and white fat depots. The increased expression of secreted proteins correlates with elevation of energy expenditure and promotion of beige and white fat activation. These results indicate that YY1 in brown adipose tissue controls antagonistic gene expression programs associated with energy balance and maintenance of body weight.


Subject(s)
Adipose Tissue, Brown/metabolism , Diet , Energy Metabolism/physiology , Obesity/metabolism , Obesity/prevention & control , YY1 Transcription Factor/metabolism , Adipose Tissue, White/metabolism , Adiposity/genetics , Adiposity/physiology , Animals , Body Weight/physiology , Energy Metabolism/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Thermogenesis/genetics , YY1 Transcription Factor/deficiency
7.
Mol Ther Methods Clin Dev ; 2: 15007, 2015.
Article in English | MEDLINE | ID: mdl-26029718

ABSTRACT

Pompe disease is an autosomal recessive genetic disorder characterized by a deficiency of the enzyme responsible for degradation of lysosomal glycogen (acid α-glucosidase (GAA)). Cardiac dysfunction and respiratory muscle weakness are primary features of this disorder. To attenuate the progressive and rapid accumulation of glycogen resulting in cardiorespiratory dysfunction, adult Gaa (-/-) mice were administered a single systemic injection of rAAV2/9-DES-hGAA (AAV9-DES) or bimonthly injections of recombinant human GAA (enzyme replacement therapy (ERT)). Assessment of cardiac function and morphology was measured 1 and 3 months after initiation of treatment while whole-body plethysmography and diaphragmatic contractile function was evaluated at 3 months post-treatment in all groups. Gaa (-/-) animals receiving either AAV9-DES or ERT demonstrated a significant improvement in cardiac function and diaphragmatic contractile function as compared to control animals. AAV9-DES treatment resulted in a significant reduction in cardiac dimension (end diastolic left ventricular mass/gram wet weight; EDMc) at 3 months postinjection. Neither AAV nor ERT therapy altered minute ventilation during quiet breathing (eupnea). However, breathing frequency and expiratory time were significantly improved in AAV9-DES animals. These results indicate systemic delivery of either strategy improves cardiac function but AAV9-DES alone improves respiratory parameters at 3 months post-treatment in a murine model of Pompe disease.

8.
J Inherit Metab Dis ; 38(5): 915-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25860817

ABSTRACT

Barth syndrome (BTHS) is an X-linked metabolic disorder that causes cardiomyopathy in infancy and is linked to mutations within the Tafazzin (TAZ) gene. The first mouse model, a TAZ knockdown model (TAZKD), has been generated to further understand the bioenergetics leading to cardiomyopathy. However, the TAZKD model does not show early signs of cardiomyopathy, and cardiac pathophysiology has not been documented until 7-8 months of age. Here we sought to determine the impact of endurance training on the cardiac and skeletal muscle phenotype in young TAZKD mice. TAZKD exercise trained (TAZKD-ET) and control exercise trained (CON-ET) mice underwent a 35-day swimming protocol. Non-trained aged matched TAZKD and CON mice were used as controls. At the end of the protocol, cardiac MRI was used to assess cardiac parameters. Cardiac MRI showed that training resulted in cardiac hypertrophy within both groups and did not result in a decline of ejection fraction. TAZKD mice exhibited a decrease in respiratory complex I, III, and IV enzymatic activity in cardiac tissue compared to control mice; however, training led to an increase in complex III activity in TAZKD-ET mice resulting in similar levels to those of CON-ET mice. (31)P magnetic resonance spectroscopy of the gastrocnemius showed a significantly lowered pH in TAZKD-ET mice post electrical-stimulation compared to CON-ET mice. Endurance training does not accelerate cardiac dysfunction in young TAZKD mice, but results in beneficial physiological effects. Furthermore, our results suggest that a significant drop in intracellular pH levels may contribute to oxidative phosphorylation defects during exercise.


Subject(s)
Barth Syndrome/pathology , Barth Syndrome/therapy , Disease Models, Animal , Electron Transport Complex III/deficiency , Physical Conditioning, Animal/physiology , Physical Endurance , Transcription Factors/genetics , Acyltransferases , Animals , Barth Syndrome/genetics , Electron Transport Complex III/genetics , Exercise/physiology , Exercise Tolerance/genetics , Humans , Mice , Mice, Knockout , Physical Endurance/genetics , Physical Endurance/physiology , Reactive Oxygen Species/metabolism
9.
EMBO J ; 34(8): 981-3, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25736377

ABSTRACT

Obesity is a global concern, which has been linked to increased risk for cardiovascular disease, type 2 diabetes, atherosclerosis,non-alcoholic fatty liver, and cancer. In this issue of The EMBO Journal,Fujita et al (2015) describe the role of an endoplasmic reticulum (ER)-resident E3ubiquitin ligase, synoviolin, and its ability to control body weight and energy expenditure by targeting PGC-1b, a transcriptional modulator of mitochondrial oxidative metabolism.


Subject(s)
Body Weight/genetics , Mitochondria/physiology , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Humans
10.
Hum Mol Genet ; 24(3): 625-36, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25217571

ABSTRACT

Pompe disease is a systemic metabolic disorder characterized by lack of acid-alpha glucosidase (GAA) resulting in ubiquitous lysosomal glycogen accumulation. Respiratory and ambulatory dysfunction are prominent features in patients with Pompe yet the mechanism defining the development of muscle weakness is currently unclear. Transgenic animal models of Pompe disease mirroring the patient phenotype have been invaluable in mechanistic and therapeutic study. Here, we demonstrate significant pathological alterations at neuromuscular junctions (NMJs) of the diaphragm and tibialis anterior muscle as prominent features of disease pathology in Gaa knockout mice. Postsynaptic defects including increased motor endplate area and fragmentation were readily observed in Gaa(-/-) but not wild-type mice. Presynaptic neuropathic changes were also evident, as demonstrated by significant reduction in the levels of neurofilament proteins, and alterations in axonal fiber diameter and myelin thickness within the sciatic and phrenic nerves. Our data suggest the loss of NMJ integrity is a primary contributor to the decline in respiratory and ambulatory function in Pompe and arises from both pre- and postsynaptic pathology. These observations highlight the importance of systemic phenotype correction, specifically restoration of GAA to skeletal muscle and the nervous system for treatment of Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/pathology , Membrane Glycoproteins/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Phrenic Nerve/pathology , Animals , Diaphragm/metabolism , Diaphragm/pathology , Disease Models, Animal , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Phrenic Nerve/metabolism , Tibia/metabolism , Tibia/pathology
11.
Hum Gene Ther ; 24(11): 928-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24164240

ABSTRACT

Metabolic myopathies are a diverse group of rare diseases in which impaired breakdown of stored energy leads to profound muscle dysfunction ranging from exercise intolerance to severe muscle wasting. Metabolic myopathies are largely caused by functional deficiency of a single gene and are generally subcategorized into three major types of metabolic disease: mitochondrial, lipid, or glycogen. Treatment varies greatly depending on the biochemical nature of the disease, and unfortunately no definitive treatments exist for metabolic myopathy. Since this group of diseases is inherited, gene therapy is being explored as an approach to personalized medical treatment. Adeno-associated virus-based vectors in particular have shown to be promising in the treatment of several forms of metabolic myopathy. This review will discuss the most recent advances in gene therapy efforts for the treatment of metabolic myopathies.


Subject(s)
Dependovirus/genetics , Glycogen Storage Disease/therapy , Lipid Metabolism Disorders/therapy , Mitochondrial Myopathies/therapy , Animals , Genetic Therapy , Genetic Vectors , Humans
12.
Mol Ther ; 21(9): 1661-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23732990

ABSTRACT

Pompe disease is a neuromuscular disease resulting from deficiency in acid α-glucosidase (GAA), results in cardiac, skeletal muscle, and central nervous system (CNS) pathology. Enzyme replacement therapy (ERT) has been shown to partially correct cardiac and skeletal muscle dysfunction. However, ERT does not cross the blood-brain barrier and progressive CNS pathology ensues. We tested the hypothesis that intrapleural administration of recombinant adeno-associated virus (rAAV9)-GAA driven by a cytomegalovirus (CMV) or desmin (DES) promoter would improve cardiac and respiratory function in Gaa(-/-) mice through a direct effect and retrograde transport to motoneurons. Cardiac magnetic resonance imaging revealed significant improvement in ejection fraction in rAAV9-GAA-treated animals. Inspiratory phrenic and diaphragm activity was examined at baseline and during hypercapnic respiratory challenge. Mice treated with AAV9 had greater relative inspiratory burst amplitude during baseline conditions when compared with Gaa(-/-). In addition, efferent phrenic burst amplitude was significantly correlated with diaphragm activity in both AAV9-DES and AAV9-CMV groups but not in Gaa(-/-). This is the first study to indicate improvements in cardiac, skeletal muscle, and respiratory neural output following rAAV administration in Pompe disease. These results further implicate a role for the CNS in Pompe disease pathology and the critical need to target the neurologic aspects in developing therapeutic strategies.


Subject(s)
Dependovirus/genetics , Glycogen Storage Disease Type II/physiopathology , Glycogen Storage Disease Type II/therapy , Heart/physiology , Phrenic Nerve/physiology , Respiratory Muscles/physiology , alpha-Glucosidases/genetics , Animals , Dependovirus/metabolism , Diaphragm/physiology , Disease Models, Animal , Genetic Vectors , Glycogen Storage Disease Type II/genetics , Humans , Mice , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Myocardium/metabolism , Myocardium/pathology , Pleura , Random Allocation , Spinal Cord/metabolism , Transduction, Genetic , alpha-Glucosidases/metabolism
13.
Hum Gene Ther ; 22(7): 865-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21091282

ABSTRACT

Barth's syndrome (BTHS) is an X-linked mitochondrial disease that is due to a mutation in the Tafazzin (TAZ) gene. Based on sequence homology, TAZ has been characterized as an acyltransferase involved in the metabolism of cardiolipin (CL), a unique phospholipid almost exclusively located in the mitochondrial inner membrane. Yeast, Drosophila, and zebrafish models have been invaluable in elucidating the role of TAZ in BTHS, but until recently a mammalian model to study the disease has been lacking. Based on in vitro evidence of RNA-mediated TAZ depletion, an inducible short hairpin RNA (shRNA)-mediated TAZ knockdown (TAZKD) mouse model has been developed (TaconicArtemis GmbH, Cologne, Germany), and herein we describe the assessment of this mouse line as a model of BTHS. Upon induction of the TAZ-specific shRNA in vivo, transgenic mouse TAZ mRNA levels were reduced by >89% in cardiac and skeletal muscle. TAZ deficiency led to the absence of tetralineoyl-CL and accumulation of monolyso-CL in cardiac muscle. Furthermore, mitochondrial morphology from cardiac and skeletal muscle was altered. Skeletal muscle mitochondria demonstrated disrupted cristae, and cardiac mitochondria were significantly enlarged and displace neighboring myofibrils. Physiological measurements demonstrated a reduction in isometric contractile strength of the soleus and a reduction in cardiac left ventricular ejection fraction of TAZKD mice compared with control animals. Therefore, the inducible TAZ-deficient model exhibits some of the molecular and clinical characteristics of BTHS patients and may ultimately help to improve our understanding of BTHS-related cardioskeletal myopathy as well as serve as an important tool in developing therapeutic strategies for BTHS.


Subject(s)
Barth Syndrome/genetics , Disease Models, Animal , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/deficiency , Acyltransferases , Animals , Animals, Genetically Modified , Barth Syndrome/metabolism , Electrocardiography , Female , Gene Knockdown Techniques/methods , Genetic Loci , Genotype , Magnetic Resonance Imaging , Mice , Microscopy, Electron , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Mutation , Myocardium/metabolism , Myocardium/pathology , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...