Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Brain ; 147(5): 1899-1913, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38242545

ABSTRACT

Aberrant cholesterol metabolism causes neurological disease and neurodegeneration, and mitochondria have been linked to perturbed cholesterol homeostasis via the study of pathological mutations in the ATAD3 gene cluster. However, whether the cholesterol changes were compensatory or contributory to the disorder was unclear, and the effects on cell membranes and the wider cell were also unknown. Using patient-derived cells, we show that cholesterol perturbation is a conserved feature of pathological ATAD3 variants that is accompanied by an expanded lysosome population containing membrane whorls characteristic of lysosomal storage diseases. Lysosomes are also more numerous in Drosophila neural progenitor cells expressing mutant Atad3, which exhibit abundant membrane-bound cholesterol aggregates, many of which co-localize with lysosomes. By subjecting the Drosophila Atad3 mutant to nutrient restriction and cholesterol supplementation, we show that the mutant displays heightened cholesterol dependence. Collectively, these findings suggest that elevated cholesterol enhances tolerance to pathological ATAD3 variants; however, this comes at the cost of inducing cholesterol aggregation in membranes, which lysosomal clearance only partly mitigates.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Cholesterol , Lysosomes , Membrane Proteins , Mutation , Animals , Cholesterol/metabolism , Humans , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Drosophila , Cell Membrane/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
2.
FASEB J ; 37(9): e23139, 2023 09.
Article in English | MEDLINE | ID: mdl-37584631

ABSTRACT

Mutations in POLG, the gene encoding the catalytic subunit of the mitochondrial DNA (mtDNA) polymerase gamma (Pol-γ), lead to diseases driven by defective mtDNA maintenance. Despite being the most prevalent cause of mitochondrial disease, treatments for POLG-related disorders remain elusive. In this study, we used POLG patient-induced pluripotent stem cell (iPSC)-derived neural stem cells (iNSCs), one homozygous for the POLG mutation c.2243G>C and one compound heterozygous with c.2243G>C and c.1399G>A, and treated these iNSCs with ethidium bromide (EtBr) to study the rate of depletion and repopulation of mtDNA. In addition, we investigated the effect of deoxyribonucleoside (dNs) supplementation on mtDNA maintenance during EtBr treatment and post-treatment repopulation in the same cells. EtBr-induced mtDNA depletion occurred at a similar rate in both patient and control iNSCs, however, restoration of mtDNA levels was significantly delayed in iNSCs carrying the compound heterozygous POLG mutations. In contrast, iNSC with the homozygous POLG mutation recovered their mtDNA at a rate similar to controls. When we treated cells with dNs, we found that this reduced EtBr-induced mtDNA depletion and significantly increased repopulation rates in both patient iNSCs. These observations are consistent with the hypothesis that mutations in POLG impair mtDNA repopulation also within intact neural lineage cells and suggest that those with compound heterozygous mutation have a more severe defect of mtDNA synthesis. Our findings further highlight the potential for dNs to improve mtDNA replication in the presence of POLG mutations, suggesting that this may offer a new therapeutic modality for mitochondrial diseases caused by disturbed mtDNA homeostasis.


Subject(s)
Induced Pluripotent Stem Cells , Mitochondrial Diseases , Neural Stem Cells , Humans , DNA-Directed DNA Polymerase/genetics , DNA Polymerase gamma/genetics , Ethidium/pharmacology , Mutation , DNA, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/therapy , Deoxyribonucleosides
3.
J Med Genet ; 60(1): 65-73, 2023 01.
Article in English | MEDLINE | ID: mdl-34872991

ABSTRACT

BACKGROUND: Large-scale mitochondrial DNA deletions (LMD) are a common genetic cause of mitochondrial disease and give rise to a wide range of clinical features. Lack of longitudinal data means the natural history remains unclear. This study was undertaken to describe the clinical spectrum in a large cohort of patients with paediatric disease onset. METHODS: A retrospective multicentre study was performed in patients with clinical onset <16 years of age, diagnosed and followed in seven European mitochondrial disease centres. RESULTS: A total of 80 patients were included. The average age at disease onset and at last examination was 10 and 31 years, respectively. The median time from disease onset to death was 11.5 years. Pearson syndrome was present in 21%, Kearns-Sayre syndrome spectrum disorder in 50% and progressive external ophthalmoplegia in 29% of patients. Haematological abnormalities were the hallmark of the disease in preschool children, while the most common presentations in older patients were ptosis and external ophthalmoplegia. Skeletal muscle involvement was found in 65% and exercise intolerance in 25% of the patients. Central nervous system involvement was frequent, with variable presence of ataxia (40%), cognitive involvement (36%) and stroke-like episodes (9%). Other common features were pigmentary retinopathy (46%), short stature (42%), hearing impairment (39%), cardiac disease (39%), diabetes mellitus (25%) and renal disease (19%). CONCLUSION: Our study provides new insights into the phenotypic spectrum of childhood-onset, LMD-associated syndromes. We found a wider spectrum of more prevalent multisystem involvement compared with previous studies, most likely related to a longer time of follow-up.


Subject(s)
Kearns-Sayre Syndrome , Muscular Diseases , Ophthalmoplegia, Chronic Progressive External , Child, Preschool , Humans , Child , Aged , DNA, Mitochondrial/genetics , Kearns-Sayre Syndrome/epidemiology , Kearns-Sayre Syndrome/genetics , Ophthalmoplegia, Chronic Progressive External/epidemiology , Ophthalmoplegia, Chronic Progressive External/genetics , Muscular Diseases/genetics , Disease Progression
4.
Nucleic Acids Res ; 49(9): 5230-5248, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33956154

ABSTRACT

Mutations in POLG, encoding POLγA, the catalytic subunit of the mitochondrial DNA polymerase, cause a spectrum of disorders characterized by mtDNA instability. However, the molecular pathogenesis of POLG-related diseases is poorly understood and efficient treatments are missing. Here, we generate the PolgA449T/A449T mouse model, which reproduces the A467T change, the most common human recessive mutation of POLG. We show that the mouse A449T mutation impairs DNA binding and mtDNA synthesis activities of POLγ, leading to a stalling phenotype. Most importantly, the A449T mutation also strongly impairs interactions with POLγB, the accessory subunit of the POLγ holoenzyme. This allows the free POLγA to become a substrate for LONP1 protease degradation, leading to dramatically reduced levels of POLγA in A449T mouse tissues. Therefore, in addition to its role as a processivity factor, POLγB acts to stabilize POLγA and to prevent LONP1-dependent degradation. Notably, we validated this mechanism for other disease-associated mutations affecting the interaction between the two POLγ subunits. We suggest that targeting POLγA turnover can be exploited as a target for the development of future therapies.


Subject(s)
DNA Polymerase gamma/genetics , ATP-Dependent Proteases/metabolism , Animals , Cells, Cultured , DNA Polymerase gamma/metabolism , DNA Replication , DNA, Mitochondrial/analysis , Enzyme Stability/genetics , HeLa Cells , Holoenzymes/metabolism , Humans , Mice , Mitochondrial Proteins/metabolism , Mutation
5.
Tidsskr Nor Laegeforen ; 142(10)2022 06 28.
Article in Norwegian | MEDLINE | ID: mdl-35763848

ABSTRACT

Mitochondrial disease is among the most commonly occurring metabolic disorders and is relevant for many medical specialties. This clinical review article discusses one of the most common mutations causing mitochondrial disease, namely m.3243A>G. The mutation can lead to diabetes mellitus, hearing loss, cardiac and muscle involvement, encephalopathy and epilepsy, gastric and intestinal problems and visual impairment, frequently in combination. Better knowledge of mitochondrial disease caused by the m.3243A>G mutation would improve both the diagnosis and treatment of patients who may suffer from a serious and life-threatening disease.


Subject(s)
Brain Diseases , Hearing Loss , Mitochondrial Diseases , DNA, Mitochondrial/genetics , Humans , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mitochondrial Diseases/therapy , Mutation
6.
Hum Genet ; 140(11): 1593-1609, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33835239

ABSTRACT

We investigated the clinical, genetic, and pathological characteristics of a previously unknown severe juvenile brain disorder in several litters of Parson Russel Terriers. The disease started with epileptic seizures at 6-12 weeks of age and progressed rapidly to status epilepticus and death or euthanasia. Histopathological changes at autopsy were restricted to the brain. There was severe acute neuronal degeneration and necrosis diffusely affecting the grey matter throughout the brain with extensive intraneuronal mitochondrial crowding and accumulation of amyloid-ß (Aß). Combined homozygosity mapping and genome sequencing revealed an in-frame 6-bp deletion in the nuclear-encoded pitrilysin metallopeptidase 1 (PITRM1) encoding for a mitochondrial protease involved in mitochondrial targeting sequence processing and degradation. The 6-bp deletion results in the loss of two amino acid residues in the N-terminal part of PITRM1, potentially affecting protein folding and function. Assessment of the mitochondrial function in the affected brain tissue showed a significant deficiency in respiratory chain function. The functional consequences of the mutation were modeled in yeast and showed impaired growth in permissive conditions and an impaired respiration capacity. Loss-of-function variants in human PITRM1 result in a childhood-onset progressive amyloidotic neurological syndrome characterized by spinocerebellar ataxia with behavioral, psychiatric and cognitive abnormalities. Homozygous Pitrm1-knockout mice are embryonic lethal, while heterozygotes show a progressive, neurodegenerative phenotype characterized by impairment in motor coordination and Aß deposits. Our study describes a novel early-onset PITRM1-related neurodegenerative canine brain disorder with mitochondrial dysfunction, Aß accumulation, and lethal epilepsy. The findings highlight the essential role of PITRM1 in neuronal survival and strengthen the connection between mitochondrial dysfunction and neurodegeneration.


Subject(s)
Dog Diseases/genetics , Epilepsy/veterinary , Metalloendopeptidases/genetics , Mitochondria/metabolism , Neurodegenerative Diseases/veterinary , Amyloid beta-Peptides/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Dog Diseases/pathology , Dogs , Epilepsy/genetics , Female , Male , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oxygen Consumption , Pedigree , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism
7.
J Inherit Metab Dis ; 44(2): 469-480, 2021 03.
Article in English | MEDLINE | ID: mdl-32857451

ABSTRACT

The aim of this study was to compare the value of serum biomarkers, fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15), with histological analysis of muscle in the diagnosis of mitochondrial disease. We collected 194 serum samples from patients with a suspected or known mitochondrial disease. Biomarkers were analyzed blinded using enzyme-labeled immunosorbent assay. Clinical data were collected using a structured questionnaire. Only 39% of patients with genetically verified mitochondrial disease had mitochondrial pathology in their muscle histology. In contrast, biomarkers were elevated in 62% of patients with genetically verified mitochondrial disease. Those with both biomarkers elevated had a muscle manifesting disorder and a defect affecting mitochondrial DNA expression. If at least one of the biomarkers was induced and the patient had a myopathic disease, a mitochondrial DNA expression disease was the cause with 94% probability. Among patients with biomarker analysis and muscle biopsy taken <12 months apart, a mitochondrial disorder would have been identified in 70% with analysis of FGF21 and GDF15 compared to 50% of patients whom could have been identified with muscle biopsy alone. Muscle findings were nondiagnostic in 72% (children) and 45% (adults). Induction of FGF21 and GDF15 suggest a mitochondrial etiology as an underlying cause of a muscle manifesting disease. Normal biomarker values do not, however, rule out a mitochondrial disorder, especially if the disease does not manifest in muscle. We suggest that FGF21 and GDF15 together should be first-line diagnostic investigations in mitochondrial disease complementing muscle biopsy.


Subject(s)
DNA, Mitochondrial/genetics , Fibroblast Growth Factors/genetics , Growth Differentiation Factor 15/genetics , Mitochondrial Diseases/genetics , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mitochondrial Diseases/blood , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Young Adult
8.
Cell Mol Life Sci ; 77(6): 1115-1133, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31270582

ABSTRACT

Cancers show a metabolic shift towards aerobic glycolysis. By "corrupting" their microenvironment, carcinoma cells are able to obtain energy substrates to "fuel" their mitochondrial metabolism and cell growth in an autophagy-associated, paracrine manner. However, the metabolic changes and role of normal fibroblasts in this process remain unclear. We devised a novel, indirect co-culture system to elucidate the mechanisms of metabolic coupling between stromal cells and oral squamous cell carcinoma (OSCC) cells. Here, we showed that normal oral fibroblasts (NOFs) and OSCC become metabolically coupled through several processes before acquiring an activated phenotype and without inducing senescence. We observed, for the first time, that NOFs export mitochondria towards OSCCs through both direct contact and via indirect mechanisms. NOFs are activated and are able to acquire a cancer-associated fibroblasts metabolic phenotype when co-cultivation with OSSC cells, by undergoing aerobic glycolysis, secreting more reactive oxygen species (ROS), high L-lactate and overexpressing lactate exporter MCT-4, leading to mitochondrial permeability transition pore (mPTP) opening, hypoxia, and mitophagy. On the other hand, Cav-1-low NOFs generate L-lactate to "fuel" mitochondrial metabolism and anabolic growth of OSCC. Most interestingly, the decrease in AMPK activity and PGC-1α expression might involve in regulation of ROS that functions to maintain final energy and metabolic homeostasis. This indicated, for the first time, the existence of ATP and ROS homeostasis during carcinogenesis. Our study suggests that an efficient therapeutical approach has to target the multiple mechanisms used by them to corrupt the normal surrounding stroma and metabolic homeostasis.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/metabolism , Fibroblasts/metabolism , Glycolysis , Mouth Neoplasms/metabolism , Aged , Animals , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Fibroblasts/pathology , Humans , Male , Mice, SCID , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mouth Neoplasms/pathology , Reactive Oxygen Species/metabolism
9.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070858

ABSTRACT

Variants in STUB1 cause both autosomal recessive (SCAR16) and dominant (SCA48) spinocerebellar ataxia. Reports from 18 STUB1 variants causing SCA48 show that the clinical picture includes later-onset ataxia with a cerebellar cognitive affective syndrome and varying clinical overlap with SCAR16. However, little is known about the molecular properties of dominant STUB1 variants. Here, we describe three SCA48 families with novel, dominantly inherited STUB1 variants (p.Arg51_Ile53delinsProAla, p.Lys143_Trp147del, and p.Gly249Val). All the patients developed symptoms from 30 years of age or later, all had cerebellar atrophy, and 4 had cognitive/psychiatric phenotypes. Investigation of the structural and functional consequences of the recombinant C-terminus of HSC70-interacting protein (CHIP) variants was performed in vitro using ubiquitin ligase activity assay, circular dichroism assay and native polyacrylamide gel electrophoresis. These studies revealed that dominantly and recessively inherited STUB1 variants showed similar biochemical defects, including impaired ubiquitin ligase activity and altered oligomerization properties of the CHIP. Our findings expand the molecular understanding of SCA48 but also mean that assumptions concerning unaffected carriers of recessive STUB1 variants in SCAR16 families must be re-evaluated. More investigations are needed to verify the disease status of SCAR16 heterozygotes and elucidate the molecular relationship between SCA48 and SCAR16 diseases.


Subject(s)
Frontotemporal Dementia/genetics , Genes, Dominant , Genes, Recessive , Spinocerebellar Ataxias/genetics , Ubiquitin-Protein Ligases , Adult , Age of Onset , Aged , Family , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression , Heterozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Protein Folding , Spinocerebellar Ataxias/diagnosis , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
10.
Cell Tissue Res ; 380(1): 15-30, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31925525

ABSTRACT

Mitochondria are the primary generators of ATP in eukaryotic cells through the process of oxidative phosphorylation. Mitochondria are also involved in several other important cellular functions including regulation of intracellular Ca2+, cell signaling and apoptosis. Mitochondrial dysfunction causes disease and since it is not possible to perform repeated studies in humans, models are essential to enable us to investigate the mechanisms involved. Recently, the discovery of induced pluripotent stem cells (iPSCs), made by reprogramming adult somatic cells (Takahashi and Yamanaka 2006; Yamanaka and Blau 2010), has provided a unique opportunity for studying aspects of disease mechanisms in patient-specific cells and tissues. Reprogramming cells to neuronal lineage such as neural progenitor cells (NPCs) generated from the neural induction of reprogrammed iPSCs can thus provide a useful model for investigating neurological disease mechanisms including those caused by mitochondrial dysfunction. In addition, NPCs display a huge clinical potential in drug screening and therapeutics.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Mitochondrial Diseases/genetics , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice , Stem Cells
11.
J Inherit Metab Dis ; 43(4): 726-736, 2020 07.
Article in English | MEDLINE | ID: mdl-32391929

ABSTRACT

BACKGROUND: Variants in POLG are one of the most common causes of inherited mitochondrial disease. Phenotypic classification of POLG disease has evolved haphazardly making it complicated and difficult to implement in everyday clinical practise. The aim of our study was to simplify the classification and facilitate better clinical recognition. METHODS: A multinational, retrospective study using data from 155 patients with POLG variants recruited from seven European countries. RESULTS: We describe the spectrum of clinical features associated with POLG variants in the largest known cohort of patients. While clinical features clearly form a continuum, stratifying patients simply according to age of onset-onset prior to age 12 years; onset between 12 and 40 years and onset after the age of 40 years, permitted us to identify clear phenotypic and prognostic differences. Prior to 12 years of age, liver involvement (87%), seizures (84%), and feeding difficulties (84%) were the major features. For those with onset between 12 and 40 years, ataxia (90%), peripheral neuropathy (84%), and seizures (71%) predominated, while for those with onset over 40 years, ptosis (95%), progressive external ophthalmoplegia (89%), and ataxia (58%) were the major clinical features. The earlier the onset the worse the prognosis. Patients with epilepsy and those with compound heterozygous variants carried significantly worse prognosis. CONCLUSION: Based on our data, we propose a simplified POLG disease classification, which can be used to guide diagnostic investigations and predict disease course.


Subject(s)
DNA Polymerase gamma/genetics , Genetic Predisposition to Disease/genetics , Mitochondrial Diseases/classification , Mitochondrial Diseases/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Child , Child, Preschool , Europe , Female , Humans , Infant , Male , Middle Aged , Mitochondrial Diseases/mortality , Mutation , Retrospective Studies , Survival Analysis , Young Adult
12.
J Inherit Metab Dis ; 43(4): 800-818, 2020 07.
Article in English | MEDLINE | ID: mdl-32030781

ABSTRACT

Clinical guidance is often sought when prescribing drugs for patients with primary mitochondrial disease. Theoretical considerations concerning drug safety in patients with mitochondrial disease may lead to unnecessary withholding of a drug in a situation of clinical need. The aim of this study was to develop consensus on safe medication use in patients with a primary mitochondrial disease. A panel of 16 experts in mitochondrial medicine, pharmacology, and basic science from six different countries was established. A modified Delphi technique was used to allow the panellists to consider draft recommendations anonymously in two Delphi rounds with predetermined levels of agreement. This process was supported by a review of the available literature and a consensus conference that included the panellists and representatives of patient advocacy groups. A high level of consensus was reached regarding the safety of all 46 reviewed drugs, with the knowledge that the risk of adverse events is influenced both by individual patient risk factors and choice of drug or drug class. This paper details the consensus guidelines of an expert panel and provides an important update of previously established guidelines in safe medication use in patients with primary mitochondrial disease. Specific drugs, drug groups, and clinical or genetic conditions are described separately as they require special attention. It is important to emphasise that consensus-based information is useful to provide guidance, but that decisions related to drug prescribing should always be tailored to the specific needs and risks of each individual patient. We aim to present what is current knowledge and plan to update this regularly both to include new drugs and to review those currently included.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Mitochondria/drug effects , Mitochondrial Diseases/chemically induced , Pharmaceutical Preparations , Consensus , Delphi Technique , Drug Design , Humans , Internationality , Mitochondria/metabolism , Practice Guidelines as Topic , Toxicity Tests
13.
Tidsskr Nor Laegeforen ; 140(1)2020 01 14.
Article in Norwegian | MEDLINE | ID: mdl-31948198

ABSTRACT

DNA Polymerase gamma (POLG) is an enzyme that replicates and repairs mitochondrial DNA. Mutations in the gene that codes for the catalytic subunit of the enzyme, the POLG gene, are one of the most common causes of mitochondrial disease. POLG-related disorders can have overlapping phenotypes and affect a number of organ systems, and first onset may occur at any age. The disease group can serve as a paradigm for understanding mitochondrial diseases in general.


Subject(s)
DNA Polymerase gamma , Mitochondrial Diseases , DNA Polymerase gamma/genetics , DNA, Mitochondrial/genetics , Humans , Mitochondrial Diseases/genetics , Mutation , Phenotype
14.
Genet Med ; 21(4): 1027, 2019 04.
Article in English | MEDLINE | ID: mdl-30228318

ABSTRACT

Since the online publication of the article, the authors have noted errors with Table 2; this has now been corrected in both the HTML and the PDF.

15.
J Med Genet ; 55(1): 21-27, 2018 01.
Article in English | MEDLINE | ID: mdl-29101127

ABSTRACT

BACKGROUND: Leigh syndrome is a phenotypically and genetically heterogeneous mitochondrial disorder. While some genetic defects are associated with well-described phenotypes, phenotype-genotype correlations in Leigh syndrome are not fully explored. OBJECTIVE: We aimed to identify phenotype-genotype correlations in Leigh syndrome in a large cohort of systematically evaluated patients. METHODS: We studied 96 patients with genetically confirmed Leigh syndrome diagnosed and followed in eight European centres specialising in mitochondrial diseases. RESULTS: We found that ataxia, ophthalmoplegia and cardiomyopathy were more prevalent among patients with mitochondrial DNA defects. Patients with mutations in MT-ND and NDUF genes with complex I deficiency shared common phenotypic features, such as early development of central nervous system disease, followed by high occurrence of cardiac and ocular manifestations. The cerebral cortex was affected in patients with NDUF mutations significantly more often than the rest of the cohort. Patients with the m.8993T>G mutation in MT-ATP6 gene had more severe clinical and radiological manifestations and poorer disease outcome compared with patients with the m.8993T>C mutation. CONCLUSION: Our study provides new insights into phenotype-genotype correlations in Leigh syndrome and particularly in patients with complex I deficiency and with defects in the mitochondrial ATP synthase.


Subject(s)
Genetic Association Studies , Leigh Disease/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA, Mitochondrial/genetics , Female , Follow-Up Studies , Humans , Infant , Male , Mutation/genetics , Phenotype
16.
Hum Mol Genet ; 25(5): 1031-41, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26740552

ABSTRACT

With a combined carrier frequency of 1:200, heteroplasmic mitochondrial DNA (mtDNA) mutations cause human disease in ∼1:5000 of the population. Rapid shifts in the level of heteroplasmy seen within a single generation contribute to the wide range in the severity of clinical phenotypes seen in families transmitting mtDNA disease, consistent with a genetic bottleneck during transmission. Although preliminary evidence from human pedigrees points towards a random drift process underlying the shifting heteroplasmy, some reports describe differences in segregation pattern between different mtDNA mutations. However, based on limited observations and with no direct comparisons, it is not clear whether these observations simply reflect pedigree ascertainment and publication bias. To address this issue, we studied 577 mother-child pairs transmitting the m.11778G>A, m.3460G>A, m.8344A>G, m.8993T>G/C and m.3243A>G mtDNA mutations. Our analysis controlled for inter-assay differences, inter-laboratory variation and ascertainment bias. We found no evidence of selection during transmission but show that different mtDNA mutations segregate at different rates in human pedigrees. m.8993T>G/C segregated significantly faster than m.11778G>A, m.8344A>G and m.3243A>G, consistent with a tighter mtDNA genetic bottleneck in m.8993T>G/C pedigrees. Our observations support the existence of different genetic bottlenecks primarily determined by the underlying mtDNA mutation, explaining the different inheritance patterns observed in human pedigrees transmitting pathogenic mtDNA mutations.


Subject(s)
DNA, Mitochondrial/genetics , Inheritance Patterns , Mitochondrial Diseases/genetics , Models, Genetic , Point Mutation , Bayes Theorem , Child , Female , Humans , Mitochondrial Diseases/pathology , Pedigree , Phenotype , Polymorphism, Restriction Fragment Length , Publication Bias
17.
Epilepsia ; 59(8): 1595-1602, 2018 08.
Article in English | MEDLINE | ID: mdl-29920680

ABSTRACT

OBJECTIVE: Epilepsy is common in individuals with mutations in POLG, the gene encoding the catalytic subunit of the mitochondrial DNA polymerase gamma. Early recognition and aggressive seizure management are crucial for patient survival. Disruption of the blood-brain barrier (BBB) is implicated in various neurological disorders including epilepsy. The aim of this study was to assess whether POLG-related disease is associated with BBB dysfunction and what clinical implications this has for patients. METHODS: Our retrospective study used data from 83 patients with pathogenic POLG mutations from 4 countries--Norway, Sweden, Finland, and the United Kingdom. Data were collected using a structured questionnaire. We used the presence of raised cerebrospinal fluid (CSF) protein and a raised CSF/serum ratio of albumin (Q-alb) to evaluate the integrity of the blood-CSF barrier. RESULTS: Raised CSF protein was found in 70% of patients (n = 58/83) and appeared to be associated with the most severe phenotypes. In those in whom it was measured, the Q-alb ratio was markedly elevated (n = 18). The majority of those with epilepsy (n = 50/66, 76%) had raised CSF protein, and this preceded seizure debut in 75% (n = 15/20). The median survival time from symptom onset for those with raised CSF protein was decreased (13 months) compared to those with normal CSF protein (32 months). SIGNIFICANCE: Our results indicate that there is disruption of the BBB in POLG-related disease, as evidenced by a raised CSF protein and Q-alb ratio. We also find that raised CSF protein is a common finding in patients with POLG disease. Our data suggest that the presence of BBB dysfunction predicts a poorer outcome, and elevated CSF protein may therefore be an additional biomarker both for early diagnosis and to identify those at high risk of developing epilepsy.


Subject(s)
Blood-Brain Barrier/physiopathology , Cerebrospinal Fluid Proteins/metabolism , DNA Polymerase gamma/genetics , Epilepsy , Mutation/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Epilepsy/cerebrospinal fluid , Epilepsy/diagnosis , Epilepsy/genetics , Female , Humans , Infant , Infant, Newborn , International Cooperation , Male , Middle Aged , Retrospective Studies , Young Adult
19.
Genet Med ; 19(11): 1217-1225, 2017 11.
Article in English | MEDLINE | ID: mdl-28471437

ABSTRACT

PurposeMutations in POLG, the most common single-gene cause of inherited mitochondrial disease, are diagnostically challenging owing to clinical heterogeneity and overlap between syndromes. We aimed to improve the clinical recognition of POLG-related disorders in the pediatric population.MethodsWe performed a multinational, phenotype: genotype study using patients from three centers, two Norwegian and one from the United Kingdom. Patients with age at onset <12 years and confirmed pathogenic biallelic POLG mutations were considered eligible.ResultsA total of 27 patients were identified with a median age at onset of 11 months (range 0.6-80.4). The majority presented with global developmental delay (n=24/24, 100%), hypotonia (n=22/23, 96%) and faltering growth (n=24/27, 89%). Epilepsy was common, but notably absent in patients with the myocerebrohepatopathy spectrum phenotype. We identified two novel POLG gene mutations.ConclusionOur data suggest that POLG-related disease should be suspected in any child presenting with diffuse neurological symptoms. Full POLG sequencing is recommended since targeted screening may miss mutations. Finally, we simplify the classification of POLG-related disease in children using epilepsy as the crucial defining element; we show that Alpers and myocerebrohepatopathy spectrum follow different outcomes and that they manifest different degrees of respiratory chain dysfunction.


Subject(s)
DNA Polymerase gamma/genetics , Mitochondrial Diseases/genetics , Child , Child, Preschool , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Genotype , Humans , Infant , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Muscle, Skeletal/pathology , Mutation , Phenotype , Retrospective Studies
20.
J Inherit Metab Dis ; 40(6): 861-866, 2017 11.
Article in English | MEDLINE | ID: mdl-28865037

ABSTRACT

BACKGROUND: Mitochondria play an important role in iron metabolism and haematopoietic cell homeostasis. Recent studies in mice showed that a mutation in the catalytic subunit of polymerase gamma (POLG) was associated with haematopoietic dysfunction including anaemia. The aim of this study was to analyse the frequency of anaemia in a large cohort of patients with POLG related disease. METHODS: We conducted a multi-national, retrospective study of 61 patients with confirmed, pathogenic biallelic POLG mutations from six centres, four in Norway and two in the United Kingdom. Clinical, laboratory and genetic data were collected using a structured questionnaire. Anaemia was defined as an abnormally low haemoglobin value adjusted for age and sex. Univariate survival analysis was performed using log-rank test to compare differences in survival time between categories. RESULTS: Anaemia occurred in 67% (41/61) of patients and in 23% (14/61) it was already present at clinical presentation. The frequency of anaemia in patients with early onset disease including Alpers syndrome and myocerebrohepatopathy spectrum (MCHS) was high (72%) and 35% (8/23) of these had anaemia at presentation. Survival analysis showed that the presence of anaemia was associated with a significantly worse survival (P = 0.004). CONCLUSION: Our study reveals that anaemia can be a feature of POLG-related disease. Further, we show that its presence is associated with significantly worse prognosis either because anaemia itself is impacting survival or because it reflects the presence of more serious disease. In either case, our data suggests anaemia is a marker for negative prognosis.


Subject(s)
Anemia/etiology , Anemia/genetics , DNA Polymerase gamma/genetics , Adolescent , Child , Child, Preschool , Diffuse Cerebral Sclerosis of Schilder/genetics , Female , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Pilot Projects , Retrospective Studies , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL