Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
Add more filters

Publication year range
1.
Nature ; 628(8009): 844-853, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570685

ABSTRACT

Mitochondria are critical modulators of antiviral tolerance through the release of mitochondrial RNA and DNA (mtDNA and mtRNA) fragments into the cytoplasm after infection, activating virus sensors and type-I interferon (IFN-I) response1-4. The relevance of these mechanisms for mitochondrial diseases remains understudied. Here we investigated mitochondrial recessive ataxia syndrome (MIRAS), which is caused by a common European founder mutation in DNA polymerase gamma (POLG1)5. Patients homozygous for the MIRAS variant p.W748S show exceptionally variable ages of onset and symptoms5, indicating that unknown modifying factors contribute to disease manifestation. We report that the mtDNA replicase POLG1 has a role in antiviral defence mechanisms to double-stranded DNA and positive-strand RNA virus infections (HSV-1, TBEV and SARS-CoV-2), and its p.W748S variant dampens innate immune responses. Our patient and knock-in mouse data show that p.W748S compromises mtDNA replisome stability, causing mtDNA depletion, aggravated by virus infection. Low mtDNA and mtRNA release into the cytoplasm and a slow IFN response in MIRAS offer viruses an early replicative advantage, leading to an augmented pro-inflammatory response, a subacute loss of GABAergic neurons and liver inflammation and necrosis. A population databank of around 300,000 Finnish individuals6 demonstrates enrichment of immunodeficient traits in carriers of the POLG1 p.W748S mutation. Our evidence suggests that POLG1 defects compromise antiviral tolerance, triggering epilepsy and liver disease. The finding has important implications for the mitochondrial disease spectrum, including epilepsy, ataxia and parkinsonism.


Subject(s)
Alleles , DNA Polymerase gamma , Encephalitis Viruses, Tick-Borne , Herpesvirus 1, Human , Immune Tolerance , SARS-CoV-2 , Animals , Female , Humans , Male , Mice , Age of Onset , COVID-19/immunology , COVID-19/virology , COVID-19/genetics , DNA Polymerase gamma/genetics , DNA Polymerase gamma/immunology , DNA Polymerase gamma/metabolism , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/genetics , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/virology , Founder Effect , Gene Knock-In Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Type I/immunology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/immunology , Mutation , RNA, Mitochondrial/immunology , RNA, Mitochondrial/metabolism , SARS-CoV-2/immunology
2.
J Biol Chem ; 300(5): 107269, 2024 May.
Article in English | MEDLINE | ID: mdl-38588811

ABSTRACT

Coenzyme Q10 (CoQ10) is an important cofactor and antioxidant for numerous cellular processes, and its deficiency has been linked to human disorders including mitochondrial disease, heart failure, Parkinson's disease, and hypertension. Unfortunately, treatment with exogenous CoQ10 is often ineffective, likely due to its extreme hydrophobicity and high molecular weight. Here, we show that less hydrophobic CoQ species with shorter isoprenoid tails can serve as viable substitutes for CoQ10 in human cells. We demonstrate that CoQ4 can perform multiple functions of CoQ10 in CoQ-deficient cells at markedly lower treatment concentrations, motivating further investigation of CoQ4 as a supplement for CoQ10 deficiencies. In addition, we describe the synthesis and evaluation of an initial set of compounds designed to target CoQ4 selectively to mitochondria using triphenylphosphonium. Our results indicate that select versions of these compounds can successfully be delivered to mitochondria in a cell model and be cleaved to produce CoQ4, laying the groundwork for further development.


Subject(s)
Ataxia , Mitochondria , Mitochondrial Diseases , Muscle Weakness , Ubiquinone , Humans , Mitochondria/enzymology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Muscle Weakness/enzymology , Muscle Weakness/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Hep G2 Cells
3.
Acta Pharmacol Sin ; 45(4): 777-789, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38200148

ABSTRACT

Renal fibrosis is the final pathological change in renal disease, and aging is closely related to renal fibrosis. Mitochondrial dysfunction has been reported to play an important role in aging, but the exact mechanism remains unclear. Disulfide-bond A oxidoreductase-like protein (DsbA-L) is mainly located in mitochondria and plays an important role in regulating mitochondrial function and endoplasmic reticulum (ER) stress. However, the role of DsbA-L in renal aging has not been reported. In this study, we showed a reduction in DsbA-L expression, the disruption of mitochondrial function and an increase in fibrosis in the kidneys of 12- and 24-month-old mice compared to young mice. Furthermore, the deterioration of mitochondrial dysfunction and fibrosis were observed in DsbA-L-/- mice with D-gal-induced accelerated aging. Transcriptome analysis revealed a decrease in Flt4 expression and inhibition of the PI3K-AKT signaling pathway in DsbA-L-/- mice compared to control mice. Accelerated renal aging could be alleviated by an AKT agonist (SC79) or a mitochondrial protector (MitoQ) in mice with D-gal-induced aging. In vitro, overexpression of DsbA-L in HK-2 cells restored the expression of Flt4, AKT pathway factors, SP1 and PGC-1α and alleviated mitochondrial damage and cell senescence. These beneficial effects were partially blocked by inhibiting Flt4. Finally, activating the AKT pathway or improving mitochondrial function with chemical reagents could alleviate cell senescence. Our results indicate that the DsbA-L/AKT/PGC-1α signaling pathway could be a therapeutic target for age-related renal fibrosis and is associated with mitochondrial dysfunction.


Subject(s)
Glutathione Transferase , Kidney Diseases , Kidney , Mitochondria , Animals , Mice , Aging , Fibrosis , Homeostasis , Kidney/pathology , Kidney Diseases/enzymology , Mitochondria/enzymology , Mitochondrial Diseases/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glutathione Transferase/metabolism
4.
Ann Neurol ; 91(2): 225-237, 2022 02.
Article in English | MEDLINE | ID: mdl-34954817

ABSTRACT

OBJECTIVE: ATP synthase (ATPase) is responsible for the majority of ATP production. Nevertheless, disease phenotypes associated with mutations in ATPase subunits are extremely rare. We aimed at expanding the spectrum of ATPase-related diseases. METHODS: Whole-exome sequencing in cohorts with 2,962 patients diagnosed with mitochondrial disease and/or dystonia and international collaboration were used to identify deleterious variants in ATPase-encoding genes. Findings were complemented by transcriptional and proteomic profiling of patient fibroblasts. ATPase integrity and activity were assayed using cells and tissues from 5 patients. RESULTS: We present 10 total individuals with biallelic or de novo monoallelic variants in nuclear ATPase subunit genes. Three unrelated patients showed the same homozygous missense ATP5F1E mutation (including one published case). An intronic splice-disrupting alteration in compound heterozygosity with a nonsense variant in ATP5PO was found in one patient. Three patients had de novo heterozygous missense variants in ATP5F1A, whereas another 3 were heterozygous for ATP5MC3 de novo missense changes. Bioinformatics methods and populational data supported the variants' pathogenicity. Immunohistochemistry, proteomics, and/or immunoblotting revealed significantly reduced ATPase amounts in association to ATP5F1E and ATP5PO mutations. Diminished activity and/or defective assembly of ATPase was demonstrated by enzymatic assays and/or immunoblotting in patient samples bearing ATP5F1A-p.Arg207His, ATP5MC3-p.Gly79Val, and ATP5MC3-p.Asn106Lys. The associated clinical profiles were heterogeneous, ranging from hypotonia with spontaneous resolution (1/10) to epilepsy with early death (1/10) or variable persistent abnormalities, including movement disorders, developmental delay, intellectual disability, hyperlactatemia, and other neurologic and systemic features. Although potentially reflecting an ascertainment bias, dystonia was common (7/10). INTERPRETATION: Our results establish evidence for a previously unrecognized role of ATPase nuclear-gene defects in phenotypes characterized by neurodevelopmental and neurodegenerative features. ANN NEUROL 2022;91:225-237.


Subject(s)
Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/genetics , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Neurodevelopmental Disorders/enzymology , Neurodevelopmental Disorders/genetics , Dystonia/enzymology , Dystonia/genetics , Epilepsy/genetics , Genetic Variation , Humans , Mitochondria/genetics , Mitochondrial ADP, ATP Translocases/genetics , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Models, Molecular , Mutation , Mutation, Missense , Pedigree , Phenotype , Proteomics , Exome Sequencing
5.
J Pediatr ; 228: 240-251.e2, 2021 01.
Article in English | MEDLINE | ID: mdl-32827528

ABSTRACT

OBJECTIVES: To evaluate the clinical symptoms and biochemical findings and establish the genetic etiology in a cohort of pediatric patients with combined deficiencies of the mitochondrial respiratory chain complexes. STUDY DESIGN: Clinical and biochemical data were collected from 55 children. All patients were subjected to sequence analysis of the entire mitochondrial genome, except when the causative mutations had been identified based on the clinical picture. Whole exome sequencing/whole genome sequencing (WES/WGS) was performed in 32 patients. RESULTS: Onset of disease was generally early in life (median age, 6 weeks). The most common symptoms were muscle weakness, hypotonia, and developmental delay/intellectual disability. Nonneurologic symptoms were frequent. Disease causing mutations were found in 20 different nuclear genes, and 7 patients had mutations in mitochondrial DNA. Causative variants were found in 18 of the 32 patients subjected to WES/WGS. Interestingly, many patients had low levels of coenzyme Q10 in muscle, irrespective of genetic cause. CONCLUSIONS: Children with combined enzyme defects display a diversity of clinical symptoms with varying age of presentation. We established the genetic diagnosis in 35 of the 55 patients (64%). The high diagnostic yield was achieved by the introduction of massive parallel sequencing, which also revealed novel genes and enabled elucidation of new disease mechanisms.


Subject(s)
DNA, Mitochondrial/genetics , Metabolic Diseases/genetics , Mitochondrial Diseases/genetics , Mutation , Ubiquinone/analogs & derivatives , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Humans , Infant , Infant, Newborn , Metabolic Diseases/enzymology , Mitochondrial Diseases/enzymology , Ubiquinone/blood , Exome Sequencing , Young Adult
6.
Anesth Analg ; 133(4): 924-932, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33591116

ABSTRACT

BACKGROUND: Children with mitochondrial disease undergo anesthesia for a wide array of surgical procedures. However, multiple medications used for their perioperative care can affect mitochondrial function. Defects in function of the mitochondrial electron transport chain (ETC) can lead to a profound hypersensitivity to sevoflurane in children. We studied the sensitivities to sevoflurane, during mask induction and maintenance of general anesthesia, in children presenting for muscle biopsies for diagnosis of mitochondrial disease. METHODS: In this multicenter study, 91 children, aged 6 months to 16 years, presented to the operating room for diagnostic muscle biopsy for presumptive mitochondrial disease. General anesthesia was induced by a slow increase of inhaled sevoflurane concentration. The primary end point, end-tidal (ET) sevoflurane necessary to achieve a bispectral index (BIS) of 60, was recorded. Secondary end points were maximal sevoflurane used to maintain a BIS between 40 and 60 during the case, and maximum and minimum heart rate and blood pressures. After induction, general anesthesia was maintained according to the preferences of the providers directing the cases. Primary data were analyzed comparing data from patients with complex I deficiencies to other groups using nonparametric statistics in SPSS v.27. RESULTS: The median sevoflurane concentration to reach BIS of 60 during inductions (ET sevoflurane % [BIS = 60]) was significantly lower for patients with complex I defects (0.98%; 95% confidence interval [CI], 0.5-1.4) compared to complex II (1.95%; 95% CI, 1.2-2.7; P < .001), complex III (2.0%; 95% CI, 0.7-3.5; P < .001), complex IV (2.0%; 95% CI, 1.7-3.2; P < .001), and normal groups (2.2%; 95% CI, 1.8-3.0; P < .001). The sevoflurane sensitivities of complex I patients did not reach significance when compared to patients diagnosed with mitochondrial disease but without an identifiable ETC abnormality (P = .172). Correlation of complex I activity with ET sevoflurane % (BIS = 60) gave a Spearman's coefficient of 0.505 (P < .001). The differences in sensitivities between groups were less during the maintenance of the anesthetic than during induction. CONCLUSIONS: The data indicate that patients with complex I dysfunction are hypersensitive to sevoflurane compared to normal patients. Hypersensitivity was less common in patients presenting with other mitochondrial defects or without a mitochondrial diagnosis.


Subject(s)
Anesthesia, General/adverse effects , Anesthetics, Inhalation/adverse effects , Drug Hypersensitivity/etiology , Electron Transport Complex I/deficiency , Mitochondrial Diseases/complications , Muscle, Skeletal/enzymology , Sevoflurane/adverse effects , Adolescent , Age Factors , Anesthetics, Inhalation/administration & dosage , Biopsy , Case-Control Studies , Child , Child, Preschool , Drug Hypersensitivity/diagnosis , Female , Humans , Infant , Male , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/enzymology , Muscle, Skeletal/pathology , Ohio , Risk Assessment , Risk Factors , Sevoflurane/administration & dosage , Treatment Outcome , Washington
7.
Hum Mol Genet ; 27(21): 3697-3709, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29982452

ABSTRACT

Complex I deficiency is a common cause of mitochondrial disease, resulting from mutations in genes encoding structural subunits, assembly factors or defects in mitochondrial gene expression. Advances in genetic diagnostics and sequencing have led to identification of several variants in NUBPL (nucleotide binding protein-like), encoding an assembly factor of complex I, which are potentially pathogenic. To help assign pathogenicity and learn more about the function of NUBPL, amino acid substitutions were recreated in the homologous Ind1 protein of the yeast model Yarrowia lipolytica. Leu102Pro destabilized the Ind1 protein, leading to a null-mutant phenotype. Asp103Tyr, Leu191Phe and Gly285Cys affected complex I assembly to varying degrees, whereas Gly136Asp substitution in Ind1 did not impact on complex I levels nor dNADH:ubiquinone activity. Blue-native polyacrylamide gel electrophoresis and immunolabelling of the structural subunits NUBM and NUCM revealed that all Ind1 variants accumulated a Q module intermediate of complex I. In the Ind1 Asp103Tyr variant, the matrix arm intermediate was virtually absent, indicating a dominant effect. Dysfunction of Ind1, but not absence of complex I, rendered Y. lipolytica sensitive to cold. The Ind1 Gly285Cys variant was able to support complex I assembly at 28°C, but not at 10°C. Our results indicate that Ind1 is required for progression of assembly from the Q module to the full matrix arm. Cold sensitivity could be developed as a phenotype assay to demonstrate pathogenicity of NUBPL mutations and other complex I defects.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondrial Diseases/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Yarrowia/metabolism , Amino Acid Sequence , Electron Transport Complex I/genetics , Humans , Microorganisms, Genetically-Modified , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/chemistry , Temperature , Yarrowia/genetics
8.
Hum Mol Genet ; 27(21): 3710-3719, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30085106

ABSTRACT

Mitochondrial dynamics, including mitochondrial division, fusion and transport, are integral parts of mitochondrial and cellular function. DNM1L encodes dynamin-related protein 1 (Drp1), a member of the dynamin-related protein family that is required for mitochondrial division. Several de novo mutations in DNM1L are associated with a range of disease states. Here we report the identification of five patients with pathogenic or likely pathogenic variants of DNM1L, including two novel variants. Interestingly, all of the positions identified in these Drp1 variants are fully conserved among all members of the dynamin-related protein family that are involved in membrane division and organelle division events. This work builds upon and expands the clinical spectrum associated with Drp1 variants in patients and their impact on mitochondrial division in model cells.


Subject(s)
GTP Phosphohydrolases/genetics , Microtubule-Associated Proteins/genetics , Mitochondrial Diseases/enzymology , Mitochondrial Dynamics , Mitochondrial Proteins/genetics , Mutation , Cell Line , Child , DNA Mutational Analysis , Dynamins , Female , GTP Phosphohydrolases/physiology , Humans , Infant , Male , Microtubule-Associated Proteins/physiology , Mitochondrial Diseases/physiopathology , Mitochondrial Proteins/physiology
9.
Am J Hum Genet ; 101(4): 630-637, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28965846

ABSTRACT

Hearing loss and visual impairment in childhood have mostly genetic origins, some of them being related to sensorial neuronal defects. Here, we report on eight subjects from four independent families affected by auditory neuropathy and optic atrophy. Whole-exome sequencing revealed biallelic mutations in FDXR in affected subjects of each family. FDXR encodes the mitochondrial ferredoxin reductase, the sole human ferredoxin reductase implicated in the biosynthesis of iron-sulfur clusters (ISCs) and in heme formation. ISC proteins are involved in enzymatic catalysis, gene expression, and DNA replication and repair. We observed deregulated iron homeostasis in FDXR mutant fibroblasts and indirect evidence of mitochondrial iron overload. Functional complementation in a yeast strain in which ARH1, the human FDXR ortholog, was deleted established the pathogenicity of these mutations. These data highlight the wide clinical heterogeneity of mitochondrial disorders related to ISC synthesis.


Subject(s)
Ferredoxin-NADP Reductase/genetics , Hearing Loss, Central/genetics , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Mitochondrial Diseases/genetics , Mutation , Optic Atrophy/genetics , Adolescent , Adult , Amino Acid Sequence , Child, Preschool , Female , Ferredoxin-NADP Reductase/chemistry , Ferredoxin-NADP Reductase/metabolism , Genetic Complementation Test , Hearing Loss, Central/enzymology , Hearing Loss, Central/pathology , Humans , Iron-Sulfur Proteins/genetics , Male , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Optic Atrophy/enzymology , Optic Atrophy/pathology , Pedigree , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Young Adult
10.
Nucleic Acids Res ; 46(16): 8630-8640, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30020492

ABSTRACT

Human polynucleotide phosphorylase (PNPase) is an evolutionarily conserved 3'-to-5' exoribonuclease principally located in mitochondria where it is responsible for RNA turnover and import. Mutations in PNPase impair structured RNA transport into mitochondria, resulting in mitochondrial dysfunction and disease. PNPase is a trimeric protein with a doughnut-shaped structure hosting a central channel for single-stranded RNA binding and degradation. Here, we show that the disease-linked human PNPase mutants, Q387R and E475G, form dimers, not trimers, and have significantly lower RNA binding and degradation activities compared to wild-type trimeric PNPase. Moreover, S1 domain-truncated PNPase binds single-stranded RNA but not the stem-loop signature motif of imported structured RNA, suggesting that the S1 domain is responsible for binding structured RNAs. We further determined the crystal structure of dimeric PNPase at a resolution of 2.8 Å and, combined with small-angle X-ray scattering, show that the RNA-binding K homology and S1 domains are relatively inaccessible in the dimeric assembly. Taken together, these results show that mutations at the interface of the trimeric PNPase tend to produce a dimeric protein with destructive RNA-binding surfaces, thus impairing both of its RNA import and degradation activities and leading to mitochondria disorders.


Subject(s)
Loss of Function Mutation , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mutation, Missense , Point Mutation , Polyribonucleotide Nucleotidyltransferase/chemistry , RNA Stability , RNA/metabolism , Biological Transport , Crystallography, X-Ray , Dimerization , Humans , Inverted Repeat Sequences , Mitochondrial Diseases/enzymology , Models, Molecular , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Binding , Protein Conformation , Protein Domains , RNA/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle
11.
Proc Natl Acad Sci U S A ; 114(52): E11285-E11292, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29237750

ABSTRACT

A single-nucleotide polymorphism in the human arylamine N-acetyltransferase 2 (Nat2) gene has recently been identified as associated with insulin resistance in humans. To understand the cellular and molecular mechanisms by which alterations in Nat2 activity might cause insulin resistance, we examined murine ortholog Nat1 knockout (KO) mice. Nat1 KO mice manifested whole-body insulin resistance, which could be attributed to reduced muscle, liver, and adipose tissue insulin sensitivity. Hepatic and muscle insulin resistance were associated with marked increases in both liver and muscle triglyceride (TAG) and diacylglycerol (DAG) content, which was associated with increased PKCε activation in liver and increased PKCθ activation in skeletal muscle. Nat1 KO mice also displayed reduced whole-body energy expenditure and reduced mitochondrial oxygen consumption in white adipose tissue, brown adipose tissue, and hepatocytes. Taken together, these studies demonstrate that Nat1 deletion promotes reduced mitochondrial activity and is associated with ectopic lipid-induced insulin resistance. These results provide a potential genetic link among mitochondrial dysfunction with increased ectopic lipid deposition, insulin resistance, and type 2 diabetes.


Subject(s)
Arylamine N-Acetyltransferase/deficiency , Diabetes Mellitus, Type 2 , Energy Metabolism , Insulin Resistance , Isoenzymes/deficiency , Mitochondria/enzymology , Mitochondrial Diseases , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/enzymology , Adipose Tissue, White/pathology , Animals , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diglycerides/genetics , Diglycerides/metabolism , Liver/enzymology , Liver/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Oxygen Consumption/genetics , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Triglycerides/genetics , Triglycerides/metabolism
12.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471110

ABSTRACT

The molecular pathology of diseases seen from the mitochondrial axis has become more complex with the progression of research. A variety of factors, including the failure of mitochondrial dynamics and quality control, have made it extremely difficult to narrow down drug discovery targets. We have identified MITOL (mitochondrial ubiquitin ligase: also known as MARCH5) localized on the mitochondrial outer membrane and previously reported that it is an important regulator of mitochondrial dynamics and mitochondrial quality control. In this review, we describe the pathological aspects of MITOL revealed through functional analysis and its potential as a drug discovery target.


Subject(s)
Mitochondria/metabolism , Mitochondrial Diseases/enzymology , Mitochondrial Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Endoplasmic Reticulum/metabolism , Humans , Mitochondrial Dynamics
13.
Hum Mol Genet ; 26(23): 4541-4555, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973657

ABSTRACT

Ndufc2, a subunit of the NADH: ubiquinone oxidoreductase, plays a key role in the assembly and activity of complex I within the mitochondrial OXPHOS chain. Its deficiency has been shown to be involved in diabetes, cancer and stroke. To improve our knowledge on the mechanisms underlying the increased disease risk due to Ndufc2 reduction, we performed the present in vitro study aimed at the fine characterization of the derangements in mitochondrial structure and function consequent to Ndufc2 deficiency. We found that both fibroblasts obtained from skin of heterozygous Ndufc2 knock-out rat model showed marked mitochondrial dysfunction and PBMC obtained from subjects homozygous for the TT genotype of the rs11237379/NDUFC2 variant, previously shown to associate with reduced gene expression, demonstrated increased generation of reactive oxygen species and mitochondrial damage. The latter was associated with increased oxidative stress and significant ultrastructural impairment of mitochondrial morphology with a loss of internal cristae. In both models the exposure to stress stimuli, such as high-NaCl concentration or LPS, exacerbated the mitochondrial damage and dysfunction. Resveratrol significantly counteracted the ROS generation. These findings provide additional insights on the role of an altered pattern of mitochondrial structure-function as a cause of human diseases. In particular, they contribute to underscore a potential genetic risk factor for cardiovascular diseases, including stroke.


Subject(s)
Electron Transport Complex I/deficiency , Mitochondria/enzymology , Animals , Electron Transport Complex I/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/metabolism , Metabolism, Inborn Errors/metabolism , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress/physiology , Protein Subunits , Rats , Rats, Inbred SHR , Reactive Oxygen Species/metabolism , Stroke/metabolism , Ubiquinone/metabolism
14.
Am J Hum Genet ; 99(3): 674-682, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27523597

ABSTRACT

We have used whole-exome sequencing in ten individuals from four unrelated pedigrees to identify biallelic missense mutations in the nuclear-encoded mitochondrial inorganic pyrophosphatase (PPA2) that are associated with mitochondrial disease. These individuals show a range of severity, indicating that PPA2 mutations may cause a spectrum of mitochondrial disease phenotypes. Severe symptoms include seizures, lactic acidosis, cardiac arrhythmia, and death within days of birth. In the index family, presentation was milder and manifested as cardiac fibrosis and an exquisite sensitivity to alcohol, leading to sudden arrhythmic cardiac death in the second decade of life. Comparison of normal and mutant PPA2-containing mitochondria from fibroblasts showed that the activity of inorganic pyrophosphatase was significantly reduced in affected individuals. Recombinant PPA2 enzymes modeling hypomorphic missense mutations had decreased activity that correlated with disease severity. These findings confirm the pathogenicity of PPA2 mutations and suggest that PPA2 is a cardiomyopathy-associated protein, which has a greater physiological importance in mitochondrial function than previously recognized.


Subject(s)
Death, Sudden, Cardiac/etiology , Inorganic Pyrophosphatase/deficiency , Inorganic Pyrophosphatase/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mutation, Missense/genetics , Acidosis, Lactic/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Arrhythmias, Cardiac/genetics , Cardiomyopathies/enzymology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Child , Child, Preschool , Death, Sudden, Cardiac/pathology , Ethanol/adverse effects , Exome/genetics , Female , Fibroblasts/cytology , Fibroblasts/pathology , Fibrosis/enzymology , Fibrosis/genetics , Fibrosis/pathology , Humans , Infant , Infant, Newborn , Inorganic Pyrophosphatase/chemistry , Inorganic Pyrophosphatase/metabolism , Male , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Models, Molecular , Pedigree , Phenotype , Seizures , Young Adult
15.
J Hum Genet ; 64(2): 73-85, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30401918

ABSTRACT

Mitochondrial fatty acid oxidation disorders (FAODs) are caused by defects in ß-oxidation enzymes, including very long-chain acyl-CoA dehydrogenase (VLCAD), trifunctional protein (TFP), carnitine palmitoyltransferase-2 (CPT2), carnitine-acylcarnitine translocase (CACT) and others. During prolonged fasting, infection, or exercise, patients with FAODs present with hypoglycemia, rhabdomyolysis, cardiomyopathy, liver dysfunction, and occasionally sudden death. This article describes the diagnosis, newborn screening, and treatment of long-chain FAODs with a focus on VLCAD deficiency. VLCAD deficiency is generally classified into three phenotypes based on onset time, but the classification should be comprehensively determined based on genotype, residual enzyme activity, and clinical course, due to a lack of apparent genotype-phenotype correlation. With the expansion of newborn screening for FAODs, several issues have arisen, such as missed detection, overdiagnosis (including detection of benign/asymptomatic type), and poor prognosis of the neonatal-onset form. Meanwhile, dietary management and restriction of exercise have been unnecessary for patients with the benign/asymptomatic type of VLCAD deficiency with a high fatty acid oxidation flux score. Although L-carnitine therapy for VLCAD/TFP deficiency has been controversial, supplementation with L-carnitine may be accepted for CPT2/CACT and multiple acyl-CoA dehydrogenase deficiencies. Recently, a double-blind, randomized controlled trial of triheptanoin (seven-carbon fatty acid triglyceride) versus trioctanoin (regular medium-chain triglyceride) was conducted and demonstrated improvement of cardiac functions on triheptanoin. Additionally, although the clinical efficacy of bezafibrate remains controversial, a recent open-label clinical trial showed efficacy of this drug in improving quality of life. These drugs may be promising for the treatment of FAODs, though further studies are required.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Fatty Acids/metabolism , Hypolipidemic Agents/therapeutic use , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/drug therapy , Mass Screening , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/drug therapy , Muscular Diseases/diagnosis , Muscular Diseases/drug therapy , Congenital Bone Marrow Failure Syndromes , Disease Management , Humans , Lipid Metabolism, Inborn Errors/enzymology , Mitochondrial Diseases/enzymology , Muscular Diseases/enzymology
16.
Biochim Biophys Acta Bioenerg ; 1859(8): 602-611, 2018 08.
Article in English | MEDLINE | ID: mdl-29778688

ABSTRACT

The ATP synthase which provides aerobic eukaryotes with ATP, organizes into a membrane-extrinsic catalytic domain, where ATP is generated, and a membrane-embedded FO domain that shuttles protons across the membrane. We previously identified a mutation in the mitochondrial MT-ATP6 gene (m.8969G>A) in a 14-year-old Chinese female who developed an isolated nephropathy followed by brain and muscle problems. This mutation replaces a highly conserved serine residue into asparagine at amino acid position 148 of the membrane-embedded subunit a of ATP synthase. We showed that an equivalent of this mutation in yeast (aS175N) prevents FO-mediated proton translocation. Herein we identified four first-site intragenic suppressors (aN175D, aN175K, aN175I, and aN175T), which, in light of a recently published atomic structure of yeast FO indicates that the detrimental consequences of the original mutation result from the establishment of hydrogen bonds between aN175 and a nearby glutamate residue (aE172) that was proposed to be critical for the exit of protons from the ATP synthase towards the mitochondrial matrix. Interestingly also, we found that the aS175N mutation can be suppressed by second-site suppressors (aP12S, aI171F, aI171N, aI239F, and aI200M), of which some are very distantly located (by 20-30 Å) from the original mutation. The possibility to compensate through long-range effects the aS175N mutation is an interesting observation that holds promise for the development of therapeutic molecules.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Proton-Translocating ATPases/metabolism , Mutation , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Catalytic Domain , Mitochondria/metabolism , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/genetics , Protein Conformation , Protein Subunits , Protons , Saccharomyces cerevisiae/growth & development , Sequence Homology
17.
J Cell Mol Med ; 22(3): 1601-1613, 2018 03.
Article in English | MEDLINE | ID: mdl-29265583

ABSTRACT

Methylmalonic aciduria (MMA) is a disorder of organic acid metabolism resulting from a functional defect of the mitochondrial enzyme, methylmalonyl-CoA mutase (MCM). The main treatments for MMA patients are dietary restriction of propiogenic amino acids and carnitine supplementation. Liver or combined liver/kidney transplantation has been used to treat those with the most severe clinical manifestations. Thus, therapies are necessary to help improve quality of life and prevent liver, renal and neurological complications. Previously, we successfully used the TAT-MTS-Protein approach for replacing a number of mitochondrial-mutated proteins. In this targeted system, TAT, an 11 a.a peptide, which rapidly and efficiently can cross biological membranes, is fused to a mitochondrial targeting sequence (MTS), followed by the mitochondrial mature protein which sends the protein into the mitochondria. In the mitochondria, the TAT-MTS is cleaved off and the native protein integrates into its natural complexes and is fully functional. In this study, we used heterologous MTSs of human, nuclear-encoded mitochondrial proteins, to target the human MCM protein into the mitochondria. All fusion proteins reached the mitochondria and successfully underwent processing. Treatment of MMA patient fibroblasts with these fusion proteins restored mitochondrial activity such as ATP production, mitochondrial membrane potential and oxygen consumption, indicating the importance of mitochondrial function in this disease. Treatment with the fusion proteins enhanced cell viability and most importantly reduced MMA levels. Treatment also enhanced albumin and urea secretion in a CRISPR/Cas9-engineered HepG2 MUT (-/-) liver cell line. Therefore, we suggest using this TAT-MTS-Protein approach for the treatment of MMA.


Subject(s)
Adenosine Triphosphate/biosynthesis , Fibroblasts/enzymology , Gene Products, tat/genetics , Methylmalonyl-CoA Mutase/genetics , Mitochondria/enzymology , Recombinant Fusion Proteins/genetics , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/therapy , CRISPR-Cas Systems , Escherichia coli/genetics , Escherichia coli/metabolism , Fibroblasts/pathology , Gene Expression , Gene Products, tat/metabolism , Genetic Therapy/methods , Hep G2 Cells , Humans , Liver/enzymology , Liver/pathology , Membrane Potential, Mitochondrial , Methylmalonic Acid/metabolism , Methylmalonyl-CoA Mutase/metabolism , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Diseases/therapy , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Protein Engineering/methods , Protein Sorting Signals/genetics , Recombinant Fusion Proteins/metabolism , Transfection
18.
Hum Mol Genet ; 25(4): 715-27, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26692522

ABSTRACT

Mitochondria are organelles that have their own DNA (mitochondrial DNA, mtDNA) whose maintenance is necessary for the majority of ATP production in eukaryotic cells. Defects in mtDNA maintenance or integrity are responsible for numerous diseases. The DNA polymerase γ (POLG) ensures proper mtDNA replication and repair. Mutations in POLG are a major cause of mitochondrial disorders including hepatic insufficiency, Alpers syndrome, progressive external ophthalmoplegia, sensory neuropathy and ataxia. Mutations in POLG are also associated with parkinsonism. To date, no effective therapy is available. Based on the conservation of mitochondrial function from yeast to human, we used Saccharomyces cerevisiae and Caenorhabditis elegans as first pass filters to identify a chemical that suppresses mtDNA instability in cultured fibroblasts of a POLG-deficient patient. We showed that this unsuspected compound, clofilium tosylate (CLO), belonging to a class of anti-arrhythmic agents, prevents mtDNA loss of all yeast mitochondrial polymerase mutants tested, improves behavior and mtDNA content of polg-1-deficient worms and increases mtDNA content of quiescent POLG-deficient fibroblasts. Furthermore, the mode of action of the drug seems conserved as CLO increases POLG steady-state level in yeast and human cells. Two other anti-arrhythmic agents (FDA-approved) sharing common pharmacological properties and chemical structure also show potential benefit for POLG deficiency in C. elegans. Our findings provide evidence of the first mtDNA-stabilizing compound that may be an effective pharmacological alternative for the treatment of POLG-related diseases.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Mitochondrial Diseases/drug therapy , Quaternary Ammonium Compounds/pharmacology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , DNA Polymerase I/genetics , DNA Polymerase gamma , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mutation , Phenotype , Primary Cell Culture , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
19.
Mol Genet Metab ; 125(3): 281-291, 2018 11.
Article in English | MEDLINE | ID: mdl-30177229

ABSTRACT

An increasing number of mitochondrial diseases are found to be caused by pathogenic variants in nuclear encoded mitochondrial aminoacyl-tRNA synthetases. FARS2 encodes mitochondrial phenylalanyl-tRNA synthetase (mtPheRS) which transfers phenylalanine to its cognate tRNA in mitochondria. Since the first case was reported in 2012, a total of 21 subjects with FARS2 deficiency have been reported to date with a spectrum of disease severity that falls between two phenotypes; early onset epileptic encephalopathy and a less severe phenotype characterized by spastic paraplegia. In this report, we present an additional 15 individuals from 12 families who are mostly Arabs homozygous for the pathogenic variant Y144C, which is associated with the more severe early onset phenotype. The total number of unique pathogenic FARS2 variants known to date is 21 including three different partial gene deletions reported in four individuals. Except for the large deletions, all variants but two (one in-frame deletion of one amino acid and one splice-site variant) are missense. All large deletions and the single splice-site variant are in trans with a missense variant. This suggests that complete loss of function may be incompatible with life. In this report, we also review structural, functional, and evolutionary significance of select FARS2 pathogenic variants reported here.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Phenylalanine-tRNA Ligase/genetics , Adolescent , Adult , Amino Acyl-tRNA Synthetases/deficiency , Child , Child, Preschool , Female , Gene Deletion , Humans , Male , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/deficiency , Mutation/genetics , Paraplegia/genetics , Paraplegia/pathology , Phenylalanine/genetics , Phenylalanine/metabolism , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/deficiency , Protein Isoforms/genetics , Structure-Activity Relationship , Young Adult
20.
Ann Neurol ; 81(5): 641-652, 2017 May.
Article in English | MEDLINE | ID: mdl-28318037

ABSTRACT

OBJECTIVE: Thymidine kinase 2 (TK2), a critical enzyme in the mitochondrial pyrimidine salvage pathway, is essential for mitochondrial DNA (mtDNA) maintenance. Mutations in the nuclear gene, TK2, cause TK2 deficiency, which manifests predominantly in children as myopathy with mtDNA depletion. Molecular bypass therapy with the TK2 products, deoxycytidine monophosphate (dCMP) and deoxythymidine monophosphate (dTMP), prolongs the life span of Tk2-deficient (Tk2-/- ) mice by 2- to 3-fold. Because we observed rapid catabolism of the deoxynucleoside monophosphates to deoxythymidine (dT) and deoxycytidine (dC), we hypothesized that: (1) deoxynucleosides might be the major active agents and (2) inhibition of deoxycytidine deamination might enhance dTMP+dCMP therapy. METHODS: To test these hypotheses, we assessed two therapies in Tk2-/- mice: (1) dT+dC and (2) coadministration of the deaminase inhibitor, tetrahydrouridine (THU), with dTMP+dCMP. RESULTS: We observed that dC+dT delayed disease onset, prolonged life span of Tk2-deficient mice and restored mtDNA copy number as well as respiratory chain enzyme activities and levels. In contrast, dCMP+dTMP+THU therapy decreased life span of Tk2-/- animals compared to dCMP+dTMP. INTERPRETATION: Our studies demonstrate that deoxynucleoside substrate enhancement is a novel therapy, which may ameliorate TK2 deficiency in patients. Ann Neurol 2017;81:641-652.


Subject(s)
Antimetabolites/pharmacology , Deoxycytidine Monophosphate/pharmacology , Metabolism, Inborn Errors/drug therapy , Mitochondrial Diseases/drug therapy , Tetrahydrouridine/pharmacology , Thymidine Kinase/deficiency , Thymidine/pharmacology , Animals , Antimetabolites/administration & dosage , DNA, Mitochondrial/drug effects , Deoxycytidine Monophosphate/administration & dosage , Disease Models, Animal , Drug Therapy, Combination , Metabolism, Inborn Errors/enzymology , Mice , Mice, Transgenic , Mitochondrial Diseases/enzymology , Tetrahydrouridine/administration & dosage , Thymidine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL