Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30482867

ABSTRACT

Coenzyme Q (CoQ) deficiency has been associated with primary defects in the CoQ biosynthetic pathway or to secondary events. In some cases, the exogenous CoQ supplementation has limited efficacy. In the Coq9R239X mouse model with fatal mitochondrial encephalopathy due to CoQ deficiency, we have tested the therapeutic potential of ß-resorcylic acid (ß-RA), a structural analog of the CoQ precursor 4-hydroxybenzoic acid and the anti-inflammatory salicylic acid. ß-RA noticeably rescued the phenotypic, morphological, and histopathological signs of the encephalopathy, leading to a significant increase in the survival. Those effects were due to the decrease of the levels of demethoxyubiquinone-9 (DMQ9) and the increase of mitochondrial bioenergetics in peripheral tissues. However, neither CoQ biosynthesis nor mitochondrial function changed in the brain after the therapy, suggesting that some endocrine interactions may induce the reduction of the astrogliosis, spongiosis, and the secondary down-regulation of astrocytes-related neuroinflammatory genes. Because the therapeutic outcomes of ß-RA administration were superior to those after CoQ10 supplementation, its use in the clinic should be considered in CoQ deficiencies.


Subject(s)
Hydroxybenzoates/administration & dosage , Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Neuroprotective Agents/administration & dosage , Ubiquinone/analogs & derivatives , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Energy Metabolism , Histocytochemistry , Mice , Salicylic Acid/administration & dosage , Survival Analysis , Treatment Outcome , Ubiquinone/analysis , Ubiquinone/deficiency , Ubiquinone/genetics , Ubiquinone/metabolism
2.
Neuropharmacology ; 117: 74-84, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28161373

ABSTRACT

Mitochondrial encephalopathies are fatal, infantile neurodegenerative disorders caused by a deficit of mitochondrial functioning, for which there is urgent need to identify efficacious pharmacological treatments. Recent evidence shows that rapamycin administered both intraperitoneally or in the diet delays disease onset and enhances survival in the Ndufs4 null mouse model of mitochondrial encephalopathy. To delineate the clinical translatability of rapamycin in treatment of mitochondrial encephalopathy, we evaluated the drug's effects on disease evolution and mitochondrial parameters adopting treatment paradigms with fixed daily, oral doses starting at symptom onset in Ndufs4 knockout mice. Molecular mechanisms responsible for the pharmacodynamic effects of rapamycin were also evaluated. We found that rapamycin did not affect disease development at clinically-relevant doses (0.5 mg kg-1). Conversely, an oral dose previously adopted for intraperitoneal administration (8 mg kg-1) delayed development of neurological symptoms and increased median survival by 25%. Neurological improvement and lifespan were not further increased when the dose raised to 20 mg kg-1. Notably, rapamycin at 8 mg kg-1 did not affect the reduced expression of respiratory complex subunits, as well as mitochondrial number and mtDNA content. This treatment regimen however significantly ameliorated architecture of mitochondria cristae in motor cortex and cerebellum. However, reduction of mTOR activity by rapamycin was not consistently found within the brain of knockout mice. Overall, data show the ability of rapamycin to improve ultrastructure of dysfunctional mitochondria and corroborate its therapeutic potential in mitochondrial disorders. The non-clinical standard doses required, however, raise concerns about its rapid and safe clinical transferability.


Subject(s)
Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Sirolimus/therapeutic use , Administration, Oral , Animals , Cerebellum/metabolism , Cerebellum/pathology , DNA, Mitochondrial/metabolism , Disease Progression , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex I/genetics , Female , Male , Mice , Mice, Knockout , Mitochondria/ultrastructure , Motor Cortex/metabolism , Motor Cortex/pathology , Muscle, Skeletal/metabolism , Sirolimus/administration & dosage , Sirolimus/blood , Sirolimus/pharmacokinetics , Survival Analysis , TOR Serine-Threonine Kinases/metabolism
3.
Nature ; 524(7564): 234-8, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26176921

ABSTRACT

Mitochondria have a major role in energy production via oxidative phosphorylation, which is dependent on the expression of critical genes encoded by mitochondrial (mt)DNA. Mutations in mtDNA can cause fatal or severely debilitating disorders with limited treatment options. Clinical manifestations vary based on mutation type and heteroplasmy (that is, the relative levels of mutant and wild-type mtDNA within each cell). Here we generated genetically corrected pluripotent stem cells (PSCs) from patients with mtDNA disease. Multiple induced pluripotent stem (iPS) cell lines were derived from patients with common heteroplasmic mutations including 3243A>G, causing mitochondrial encephalomyopathy and stroke-like episodes (MELAS), and 8993T>G and 13513G>A, implicated in Leigh syndrome. Isogenic MELAS and Leigh syndrome iPS cell lines were generated containing exclusively wild-type or mutant mtDNA through spontaneous segregation of heteroplasmic mtDNA in proliferating fibroblasts. Furthermore, somatic cell nuclear transfer (SCNT) enabled replacement of mutant mtDNA from homoplasmic 8993T>G fibroblasts to generate corrected Leigh-NT1 PSCs. Although Leigh-NT1 PSCs contained donor oocyte wild-type mtDNA (human haplotype D4a) that differed from Leigh syndrome patient haplotype (F1a) at a total of 47 nucleotide sites, Leigh-NT1 cells displayed transcriptomic profiles similar to those in embryo-derived PSCs carrying wild-type mtDNA, indicative of normal nuclear-to-mitochondrial interactions. Moreover, genetically rescued patient PSCs displayed normal metabolic function compared to impaired oxygen consumption and ATP production observed in mutant cells. We conclude that both reprogramming approaches offer complementary strategies for derivation of PSCs containing exclusively wild-type mtDNA, through spontaneous segregation of heteroplasmic mtDNA in individual iPS cell lines or mitochondrial replacement by SCNT in homoplasmic mtDNA-based disease.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Haplotypes/genetics , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Mice , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mutation/genetics , Nuclear Transfer Techniques , Nucleotides/genetics , Oxygen Consumption , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, RNA , Skin/cytology
4.
Comb Chem High Throughput Screen ; 17(3): 253-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24409954

ABSTRACT

The Cell screening facility for personalized medicine (CSFPM) at Tel Aviv University in Israel is devoted to screening small molecules libraries for finding new drugs for rare diseases using human cell based models. The main strategy of the facility is based on smartly reducing the size of the compounds collection in similarity clusters and at the same time keeping high diversity of pharmacophores. This strategy allows parallel screening of several patient derived - cells in a personalized screening approach. The tested compounds are repositioned drugs derived from collections of phase III and FDA approved small molecules. In addition, the facility carries screenings using other chemical libraries and toxicological characterizations of nanomaterials.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical/methods , Rare Diseases/drug therapy , Small Molecule Libraries/pharmacology , Universities/organization & administration , Drug Discovery/organization & administration , Drug Repositioning , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/pathology , Humans , Intestinal Pseudo-Obstruction/drug therapy , Intestinal Pseudo-Obstruction/pathology , Israel , Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Muscular Dystrophy, Oculopharyngeal , Ophthalmoplegia/congenital , Precision Medicine/methods , Rare Diseases/pathology
5.
Hum Mol Genet ; 22(22): 4602-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23814040

ABSTRACT

Childhood-onset mitochondrial encephalomyopathies are severe, relentlessly progressive conditions. However, reversible infantile respiratory chain deficiency (RIRCD), due to a homoplasmic mt-tRNA(Glu) mutation, and reversible infantile hepatopathy, due to tRNA 5-methylaminomethyl-2-thiouridylate methyltransferase (TRMU) deficiency, stand out by showing spontaneous recovery, and provide the key to treatments of potential broader relevance. Modification of mt-tRNA(Glu) is a possible functional link between these two conditions, since TRMU is responsible for 2-thiouridylation of mt-tRNA(Glu), mt-tRNA(Lys) and mt-tRNA(Gln). Here we show that down-regulation of TRMU in RIRCD impairs 2-thiouridylation and exacerbates the effect of the mt-tRNA(Glu) mutation by triggering a mitochondrial translation defect in vitro. Skeletal muscle of RIRCD patients in the symptomatic phase showed significantly reduced 2-thiouridylation. Supplementation with l-cysteine, which is required for optimal TRMU function, rescued respiratory chain enzyme activities in human cell lines of patients with RIRCD as well as deficient TRMU. Our results show that l-cysteine supplementation is a potential treatment for RIRCD and for TRMU deficiency, and is likely to have broader application for the growing group of intra-mitochondrial translation disorders.


Subject(s)
Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/genetics , Protein Biosynthesis/genetics , RNA, Transfer/metabolism , tRNA Methyltransferases/genetics , Cell Line , Cysteine/metabolism , Gene Expression Regulation , Humans , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Mutation , Myoblasts/metabolism , Oxidative Phosphorylation , Protein Biosynthesis/physiology , RNA, Transfer/genetics , Thiouridine/analogs & derivatives , Thiouridine/metabolism , tRNA Methyltransferases/metabolism
6.
Am J Hum Genet ; 86(4): 639-49, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20362274

ABSTRACT

We investigated two male infant patients who were given a diagnosis of progressive mitochondrial encephalomyopathy on the basis of clinical, biochemical, and morphological features. These patients were born from monozygotic twin sisters and unrelated fathers, suggesting an X-linked trait. Fibroblasts from both showed reduction of respiratory chain (RC) cIII and cIV, but not of cI activities. We found a disease-segregating mutation in the X-linked AIFM1 gene, encoding the Apoptosis-Inducing Factor (AIF) mitochondrion-associated 1 precursor that deletes arginine 201 (R201 del). Under normal conditions, mature AIF is a FAD-dependent NADH oxidase of unknown function and is targeted to the mitochondrial intermembrane space (this form is called AIF(mit)). Upon apoptogenic stimuli, a soluble form (AIF(sol)) is released by proteolytic cleavage and migrates to the nucleus, where it induces "parthanatos," i.e., caspase-independent fragmentation of chromosomal DNA. In vitro, the AIF(R201 del) mutation decreases stability of both AIF(mit) and AIF(sol) and increases the AIF(sol) DNA binding affinity, a prerequisite for nuclear apoptosis. In AIF(R201 del) fibroblasts, staurosporine-induced parthanatos was markedly increased, whereas re-expression of AIF(wt) induced recovery of RC activities. Numerous TUNEL-positive, caspase 3-negative nuclei were visualized in patient #1's muscle, again indicating markedly increased parthanatos in the AIF(R201 del) critical tissues. We conclude that AIF(R201 del) is an unstable mutant variant associated with increased parthanatos-linked cell death. Our data suggest a role for AIF in RC integrity and mtDNA maintenance, at least in some tissues. Interestingly, riboflavin supplementation was associated with prolonged improvement of patient #1's neurological conditions, as well as correction of RC defects in mutant fibroblasts, suggesting that stabilization of the FAD binding in AIF(mit) is beneficial.


Subject(s)
Apoptosis Inducing Factor/genetics , Apoptosis , Genes, X-Linked , Mitochondrial Encephalomyopathies/genetics , Mutation/genetics , Apoptosis Inducing Factor/metabolism , Caspase 3/metabolism , Computer Simulation , DNA Primers/chemistry , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Dietary Supplements , Electron Transport/physiology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavin-Adenine Dinucleotide/metabolism , Humans , In Situ Nick-End Labeling , Infant, Newborn , Magnetic Resonance Imaging , Male , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Pedigree , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Conformation , Riboflavin/administration & dosage , Staurosporine/pharmacology , Twins, Monozygotic
7.
Neuropediatrics ; 38(4): 184-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-18058625

ABSTRACT

Folate transport to the brain depends on ATP-driven folate receptor-mediated transport across choroid plexus epithelial cells. Failure of ATP production in Kearns-Sayre syndrome syndrome provides one explanation for the finding of low spinal fluid (CSF) 5-methyltetrahydrofolate (5MTHF) levels in this condition. Therefore, we suspect the presence of reduced folate transport across the blood-spinal fluid barrier in other mitochondrial encephalopathies. In the present patient with mitochondrial complex I encephalomyopathy a low 5-methyltetrahydrofolate level was found in the CSF. Serum folate receptor autoantibodies were negative and could not explain the low spinal fluid folate levels. The epileptic seizures did not respond to primidone monotherapy, but addition of ubiquinone-10 and radical scavengers reduced seizure frequency. Add-on treatment with folinic acid led to partial clinical improvement including full control of epilepsy, followed by marked recovery from demyelination of the brainstem, thalamus, basal ganglia and white matter. Cerebral folate deficiency is not only present in Kearns-Sayre syndrome but may also be secondary to the failure of mitochondrial ATP production in other mitochondrial encephalopathies. Treatment with folinic acid in addition to supplementation with radical scavengers and cofactors of deficient respiratory enzymes can result in partial clinical improvement and reversal of abnormal myelination patterns on neuro-imaging.


Subject(s)
Mitochondrial Encephalomyopathies/cerebrospinal fluid , Tetrahydrofolates/deficiency , Child , Folic Acid/therapeutic use , Humans , Magnetic Resonance Imaging/methods , Male , Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Vitamin B Complex/therapeutic use
8.
Mov Disord ; 21(12): 2227-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17029263

ABSTRACT

A 53-year-old woman underwent several ischemic stroke-like episodes and later developed incomplete, bilateral ophthalmoplegia, left vision deterioration, and bilateral tremor. The clinical course, laboratory data, and muscle histology led to a diagnosis of mitochondrial encephalomyopathy. No other etiology could be identified in the background of her disabling bilateral postural-kinetic tremor. As this tremor did not respond to pharmacological therapy, left thalamotomy and subsequently right thalamic deep brain stimulator (DBS) implantation were performed, which resulted in an excellent clinical outcome. The Fahn-Tolosa-Marin Tremor Rating Scale improved from 110 to 11 points. This case suggests that the rare tremor caused by mitochondrial encephalopathy may be treated long-term with either thalamotomy or thalamic DBS implantation.


Subject(s)
Mitochondrial Encephalomyopathies/complications , Neurosurgical Procedures/methods , Tremor/etiology , Tremor/surgery , Electric Stimulation Therapy/methods , Female , Humans , Magnetic Resonance Imaging/methods , Middle Aged , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Encephalomyopathies/surgery , Tremor/pathology
9.
J Inherit Metab Dis ; 26(1): 55-66, 2003.
Article in English | MEDLINE | ID: mdl-12872841

ABSTRACT

The trace metal copper is an essential cofactor for a number of biological processes, including mitochondrial oxidative phosphorylation, free-radical eradication, neurotransmitter synthesis and maturation, and iron metabolism. Consequently, copper transport at the cell surface and the delivery of copper to intracellular proteins are critical events in normal cellular homeostasis. Four genes have been reported to influence the cellular uptake and the delivery of copper to specific cell compartments and proteins. These include hCTR1, which regulates cellular copper uptake; HAH1, which mediates the transfer of copper to the Menkes and Wilson disease transporters; CCS, which is related to the transfer of copper to superoxide dismutase; and hCOX17, which directs trafficking of copper to mitochondrial cytochrome-c oxidase. At present, no genetic disorders have been associated with defects in these four copper transporter genes. In this study, we test the possibility that defective copper uptake or intracellular translocation represents the basic defect in three categories of candidate phenotypes among 22 patients: ethylmalonic encephalopathy; mitochondriopathies of unknown aetiology; and neurodevelopmental abnormalities with clinical and chemical evidence of copper deficiency. Mutation analyses of the copper uptake protein, hCTR1, and the three copper chaperones were performed by direct sequencing of the whole coding regions. No causative mutations were identified for the four copper transporter genes in 22 patients. A heterozygous polymorphism (847G>A) for CCS was detected in 7 patients. For the distinct disease entity ethylmalonic encephalopathy, we additionally show normal mRNA levels for each of the four genes. The negative results notwithstanding, we encourage ongoing study of additional patients with candidate phenotypes. Further, our results are consistent with the notion that other unknown copper-related transporters could be involved in diseases.


Subject(s)
Brain Diseases, Metabolic/genetics , Cation Transport Proteins/genetics , Copper/deficiency , Copper/metabolism , Malonates/urine , Mitochondrial Encephalomyopathies/pathology , Blotting, Northern , Cells, Cultured , Child , Copper Transporter 1 , DNA Mutational Analysis , DNA, Complementary/genetics , Fatty Acids/metabolism , Female , Fibroblasts/metabolism , Humans , Intellectual Disability/genetics , Lipid Metabolism, Inborn Errors/genetics , Lymphocytes/metabolism , Male , Molecular Chaperones/genetics , Oxidation-Reduction , Phenotype
10.
Ann Clin Lab Sci ; 31(1): 25-67, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11314862

ABSTRACT

Different tissues display distinct sensitivities to defective mitochondrial oxidative phosphorylation (OXPHOS). Tissues highly dependent on oxygen such as the cardiac muscle, skeletal and smooth muscle, the central and peripheral nervous system, the kidney, and the insulin-producing pancreatic beta-cell are especially susceptible to defective OXPHOS. There is evidence that defective OXPHOS plays an important role in atherogenesis, in the pathogenesis of Alzheimer's disease, Parkinson's disease, diabetes, and aging. Defective OXPHOS may be caused by abnormal mitochondrial biosynthesis due to inherited or acquired mutations in the nuclear (n) or mitochondrial (mt) deoxyribonucleic acid (DNA). For instance, the presence of a mutation of the mtDNA in the pancreatic beta-cell impairs adenosine triphosphate (ATP) generation and insulin synthesis. The nuclear genome controls mitochondrial biosynthesis, but mtDNA has a much higher mutation rate than nDNA because it lacks histones and is exposed to the radical oxygen species (ROS) generated by the electron transport chain, and the mtDNA repair system is limited. Defective OXPHOS may be caused by insufficient fuel supply, by defective electron transport chain enzymes (Complexes I - IV), lack of the electron carrier coenzyme Q10, lack of oxygen due to ischemia or anemia, or excessive membrane leakage, resulting in insufficient mitochondrial inner membrane potential for ATP synthesis by the F0F1-ATPase. Human tissues can counteract OXPHOS defects by stimulating mitochondrial biosynthesis; however, above a certain threshold the lack of ATP causes cell death. Many agents affect OXPHOS. Several nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit or uncouple OXPHOS and induce the 'topical' phase of gastrointestinal ulcer formation. Uncoupled mitochondria reduce cell viability. The Helicobacter pylori induces uncoupling. The uncoupling that opens the membrane pores can activate apoptosis. Cholic acid in experimental atherogenic diets inhibits Complex IV, cocaine inhibits Complex I, the poliovirus inhibits Complex II, ceramide inhibits Complex III, azide, cyanide, chloroform, and methamphetamine inhibit Complex IV. Ethanol abuse and antiviral nucleoside analogue therapy inhibit mtDNA replication. By contrast, melatonin stimulates Complexes I and IV and Gingko biloba stimulates Complexes I and III. Oral Q10 supplementation is effective in treating cardiomyopathies and in restoring plasma levels reduced by the statin type of cholesterol-lowering drugs.


Subject(s)
Mitochondria/metabolism , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Myopathies/metabolism , Mitochondrial Myopathies/pathology , Oxidative Phosphorylation , Adenosine Triphosphate/metabolism , Animals , DNA, Mitochondrial/genetics , Humans , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Myopathies/genetics
SELECTION OF CITATIONS
SEARCH DETAIL