Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Mod Pathol ; 35(2): 177-185, 2022 02.
Article in English | MEDLINE | ID: mdl-34404905

ABSTRACT

Next-generation sequencing of oral squamous cell carcinoma (OSCC) has revealed TP53 as the most frequently mutated gene in OSCC mutually exclusive with human papillomavirus infection. Oral epithelial dysplasia (OED) is defined as a precancerous lesion of OSCC by the current World Health Organization (WHO) classification; therefore, it is assumed that TP53 mutations occur in early precancerous conditions such as OED. Here, we conducted an integrated analysis of TP53, including whole coding sequencing of TP53, FISH analysis of the 17p13.1 locus, and immunohistochemical analysis for p53 (p53-IHC), in 40 OED cases. We detected 20 mutations in 16 (40%) OED cases, and four cases, each harbored two mutations. FISH analysis revealed six of 24 cases (25%) had a deletion on 17p13.1, and four cases had concurrent TP53 mutations and 17p13.1 deletion (2-hit). Also, the increased frequency of TP53 mutations in higher degrees of OED implies acquisition of the mutation is a major event toward OSCC. p53-IHC revealed that overall cases could be categorized into four patterns that correlate well with the mutational status of TP53. Especially, two patterns, broad p53 expression type (pattern HI) and p53 null type (pattern LS), strongly correlated with a missense mutation and nonsense mutation, respectively. Furthermore, seven of the 40 cases progressed to SCC, and six of these seven cases presented pattern HI or LS. Therefore, patterns HI and LS have a high risk for malignant transformation if excisional treatment is not performed irrespective of the dysplasia grade. Although the current WHO classification mainly focuses on morphological criteria for the diagnosis of OED, interobserver discrepancy appears in some instances of the OED diagnosis. Our immunohistochemical analysis supports a more accurate pathological diagnosis for OED in cases of low dysplastic changes or of differential diagnosis with non-dysplastic lesions.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/pathology , Humans , Immunohistochemistry , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mutation , Staining and Labeling , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Hum Mutat ; 42(11): 1422-1428, 2021 11.
Article in English | MEDLINE | ID: mdl-34405929

ABSTRACT

Isolated complex I deficiency is the most common cause of pediatric mitochondrial disease. Exome sequencing (ES) has revealed many complex I causative genes. However, there are limitations associated with identifying causative genes by ES analysis. In this study, we performed multiomics analysis to reveal the causal variants. We here report two cases with mitochondrial complex I deficiency. In both cases, ES identified a novel c.580G>A (p.Glu194Lys) variant in NDUFV2. One case additionally harbored c.427C>T (p.Arg143*), but no other variants were observed in the other case. RNA sequencing showed aberrant exon splicing of NDUFV2 in the unsolved case. Genome sequencing revealed a novel heterozygous deletion in NDUFV2, which included one exon and resulted in exon skipping. Detailed examination of the breakpoint revealed that an Alu insertion-mediated rearrangement caused the deletion. Our report reveals that combined use of transcriptome sequencing and GS was effective for diagnosing cases that were unresolved by ES.


Subject(s)
Alu Elements , Electron Transport Complex I/deficiency , Gene Deletion , Genome, Human , INDEL Mutation , Mitochondrial Diseases/genetics , NADH Dehydrogenase/genetics , Sequence Analysis, RNA/methods , Electron Transport Complex I/genetics , Female , Humans , Infant , Male , Mitochondrial Diseases/diagnosis , Pedigree
4.
Am J Hum Genet ; 103(2): 221-231, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30057030

ABSTRACT

Bloom syndrome, caused by biallelic mutations in BLM, is characterized by prenatal-onset growth deficiency, short stature, an erythematous photosensitive malar rash, and increased cancer predisposition. Diagnostically, a hallmark feature is the presence of increased sister chromatid exchanges (SCEs) on cytogenetic testing. Here, we describe biallelic mutations in TOP3A in ten individuals with prenatal-onset growth restriction and microcephaly. TOP3A encodes topoisomerase III alpha (TopIIIα), which binds to BLM as part of the BTRR complex, and promotes dissolution of double Holliday junctions arising during homologous recombination. We also identify a homozygous truncating variant in RMI1, which encodes another component of the BTRR complex, in two individuals with microcephalic dwarfism. The TOP3A mutations substantially reduce cellular levels of TopIIIα, and consequently subjects' cells demonstrate elevated rates of SCE. Unresolved DNA recombination and/or replication intermediates persist into mitosis, leading to chromosome segregation defects and genome instability that most likely explain the growth restriction seen in these subjects and in Bloom syndrome. Clinical features of mitochondrial dysfunction are evident in several individuals with biallelic TOP3A mutations, consistent with the recently reported additional function of TopIIIα in mitochondrial DNA decatenation. In summary, our findings establish TOP3A mutations as an additional cause of prenatal-onset short stature with increased cytogenetic SCEs and implicate the decatenation activity of the BTRR complex in their pathogenesis.

5.
Am J Hum Genet ; 101(4): 525-538, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28942965

ABSTRACT

Complement component 1 Q subcomponent-binding protein (C1QBP; also known as p32) is a multi-compartmental protein whose precise function remains unknown. It is an evolutionary conserved multifunctional protein localized primarily in the mitochondrial matrix and has roles in inflammation and infection processes, mitochondrial ribosome biogenesis, and regulation of apoptosis and nuclear transcription. It has an N-terminal mitochondrial targeting peptide that is proteolytically processed after import into the mitochondrial matrix, where it forms a homotrimeric complex organized in a doughnut-shaped structure. Although C1QBP has been reported to exert pleiotropic effects on many cellular processes, we report here four individuals from unrelated families where biallelic mutations in C1QBP cause a defect in mitochondrial energy metabolism. Infants presented with cardiomyopathy accompanied by multisystemic involvement (liver, kidney, and brain), and children and adults presented with myopathy and progressive external ophthalmoplegia. Multiple mitochondrial respiratory-chain defects, associated with the accumulation of multiple deletions of mitochondrial DNA in the later-onset myopathic cases, were identified in all affected individuals. Steady-state C1QBP levels were decreased in all individuals' samples, leading to combined respiratory-chain enzyme deficiency of complexes I, III, and IV. C1qbp-/- mouse embryonic fibroblasts (MEFs) resembled the human disease phenotype by showing multiple defects in oxidative phosphorylation (OXPHOS). Complementation with wild-type, but not mutagenized, C1qbp restored OXPHOS protein levels and mitochondrial enzyme activities in C1qbp-/- MEFs. C1QBP deficiency represents an important mitochondrial disorder associated with a clinical spectrum ranging from infantile lactic acidosis to childhood (cardio)myopathy and late-onset progressive external ophthalmoplegia.


Subject(s)
Cardiomyopathies/genetics , Carrier Proteins/genetics , Electron Transport/physiology , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mutation , Adult , Age of Onset , Aged , Alleles , Amino Acid Sequence , Animals , Cardiomyopathies/complications , Cardiomyopathies/pathology , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cells, Cultured , Child, Preschool , Cohort Studies , DNA, Mitochondrial , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant, Newborn , Male , Mice , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Pedigree , Protein Conformation , Sequence Homology , Severity of Illness Index , Young Adult
6.
J Inherit Metab Dis ; 43(4): 819-826, 2020 07.
Article in English | MEDLINE | ID: mdl-31967322

ABSTRACT

Leigh syndrome is a major phenotype of mitochondrial diseases in children. With new therapeutic options being proposed, assessing the mortality and clinical condition of Leigh syndrome patients is crucial for evaluating therapeutics. As data are scarce in Japan, we analysed the mortality rate and clinical condition of Japanese Leigh syndrome patients that we diagnosed since 2007. Data from 166 Japanese patients diagnosed with Leigh syndrome from 2007 to 2017 were reviewed. Patients' present status, method of ventilation and feeding, and degree of disability as of April 2018 was analysed. Overall, 124 (74.7%) were living, 40 (24.1%) were deceased, and 2 (1.2%) were lost to follow-up. Median age of living patients was 8 years (1-39 years). Median length of disease course was 91 months for living patients and 23.5 months for deceased patients. Nearly 90% of deaths occurred by age 6. Mortality rate of patients with onset before 6 months of age was significantly higher than that of onset after 6 months. All patients with neonatal onset were either deceased or bedridden. MT-ATP6 deficiency caused by m.8993T>G mutation and MT-ND5 deficiency induced a severe form of Leigh syndrome. Patients with NDUFAF6, ECHS1, and SURF1 deficiency had relatively mild symptoms and better survival. The impact of onset age on prognosis varied across the genetic diagnoses. The clinical condition of many patients was poor; however, few did not require mechanical ventilation or tube-feeding and were not physically dependent. Early disease onset and genetic diagnosis may have prognostic value.


Subject(s)
Leigh Disease/genetics , Leigh Disease/mortality , Adolescent , Adult , Age of Onset , Child , Child, Preschool , DNA/genetics , DNA, Mitochondrial/genetics , Female , Humans , Infant , Japan/epidemiology , Kaplan-Meier Estimate , Leigh Disease/diagnosis , Magnetic Resonance Imaging , Male , Mutation/genetics , Phenotype , Survival Rate , Young Adult
7.
Neurogenetics ; 20(1): 9-25, 2019 03.
Article in English | MEDLINE | ID: mdl-30607703

ABSTRACT

Pentatricopeptide repeat domain proteins are a large family of RNA-binding proteins involved in mitochondrial RNA editing, stability, and translation. Mitochondrial translation machinery defects are an expanding group of genetic diseases in humans. We describe a patient who presented with low birth weight, mental retardation, and optic atrophy. Brain MRI showed abnormal bilateral signals at the basal ganglia and brainstem, and the patient was diagnosed as Leigh syndrome. Exome sequencing revealed two potentially loss-of-function variants [c.415-2A>G, and c.1747_1748insCT (p.Phe583Serfs*3)] in PTCD3 (also known as MRPS39). PTCD3, a member of the pentatricopeptide repeat domain protein family, is a component of the small mitoribosomal subunit. The patient had marked decreases in mitochondrial complex I and IV levels and activities, oxygen consumption and ATP biosynthesis, and generalized mitochondrial translation defects in fibroblasts. Quantitative proteomic analysis revealed decreased levels of the small mitoribosomal subunits. Complementation experiments rescued oxidative phosphorylation complex I and IV levels and activities, ATP biosynthesis, and MT-RNR1 rRNA transcript level, providing functional validation of the pathogenicity of identified variants. This is the first report of an association of PTCD3 mutations with Leigh syndrome along with combined oxidative phosphorylation deficiencies caused by defects in the mitochondrial translation machinery.


Subject(s)
Arabidopsis Proteins/genetics , Leigh Disease/genetics , Mutation/genetics , Oxidative Phosphorylation , RNA-Binding Proteins/genetics , Female , Humans , Mitochondria/genetics , Pedigree
8.
Stem Cells ; 36(9): 1355-1367, 2018 09.
Article in English | MEDLINE | ID: mdl-29761578

ABSTRACT

Embryonic stem cells (ESCs) exhibit two salient features beneficial for regenerative medicine: unlimited self-renewal and pluripotency. Methyl-CpG-binding domain protein 3 (Mbd3), a scaffolding component of the nucleosome remodeling deacetylase complex, is a specific regulator of pluripotency, as ESCs lacking Mbd3 are defective for lineage commitment potential but retain normal self-renewal properties. However, functional similarities and dissimilarities among the three Mbd3 isoforms (a, b, and c) have not been intensively explored. Herein, we demonstrated that Mbd3c, which lacks an entire portion of the MBD domain, exerted equivalent activity for counteracting the defective lineage commitment potential of Mbd3-knockout ESCs. Our analyses also revealed that the coiled-coil domain common to all three MBD3 isoforms, but not the MBD domain, plays a crucial role in this activity. Mechanistically, our data demonstrate that the activity of the coiled-coil domain is exerted, at least in part, through recruitment of polycomb repressive complex 2 to a subset of genes linked to development and organogenesis, thus establishing stable transcriptional repression. Stem Cells 2018;36:1355-1367.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Gene Expression Profiling , Gene Knockout Techniques , Mice , Protein Domains , Protein Isoforms , Transcription Factors/chemistry , Transcription Factors/genetics
9.
PLoS Genet ; 12(1): e1005679, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26741492

ABSTRACT

Mitochondrial disorders have the highest incidence among congenital metabolic disorders characterized by biochemical respiratory chain complex deficiencies. It occurs at a rate of 1 in 5,000 births, and has phenotypic and genetic heterogeneity. Mutations in about 1,500 nuclear encoded mitochondrial proteins may cause mitochondrial dysfunction of energy production and mitochondrial disorders. More than 250 genes that cause mitochondrial disorders have been reported to date. However exact genetic diagnosis for patients still remained largely unknown. To reveal this heterogeneity, we performed comprehensive genomic analyses for 142 patients with childhood-onset mitochondrial respiratory chain complex deficiencies. The approach includes whole mtDNA and exome analyses using high-throughput sequencing, and chromosomal aberration analyses using high-density oligonucleotide arrays. We identified 37 novel mutations in known mitochondrial disease genes and 3 mitochondria-related genes (MRPS23, QRSL1, and PNPLA4) as novel causative genes. We also identified 2 genes known to cause monogenic diseases (MECP2 and TNNI3) and 3 chromosomal aberrations (6q24.3-q25.1, 17p12, and 22q11.21) as causes in this cohort. Our approaches enhance the ability to identify pathogenic gene mutations in patients with biochemically defined mitochondrial respiratory chain complex deficiencies in clinical settings. They also underscore clinical and genetic heterogeneity and will improve patient care of this complex disorder.


Subject(s)
Exome/genetics , Genetic Heterogeneity , Mitochondria/genetics , Mitochondrial Diseases/genetics , Adolescent , Child , Child, Preschool , Chromosome Aberrations , DNA, Mitochondrial/genetics , Female , Fibroblasts , High-Throughput Nucleotide Sequencing , Humans , INDEL Mutation/genetics , Infant , Infant, Newborn , Male , Mitochondria/pathology , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/pathology , Polymorphism, Single Nucleotide/genetics
10.
Am J Hum Genet ; 97(5): 761-8, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26522469

ABSTRACT

S-adenosylmethionine (SAM) is the predominant methyl group donor and has a large spectrum of target substrates. As such, it is essential for nearly all biological methylation reactions. SAM is synthesized by methionine adenosyltransferase from methionine and ATP in the cytoplasm and subsequently distributed throughout the different cellular compartments, including mitochondria, where methylation is mostly required for nucleic-acid modifications and respiratory-chain function. We report a syndrome in three families affected by reduced intra-mitochondrial methylation caused by recessive mutations in the gene encoding the only known mitochondrial SAM transporter, SLC25A26. Clinical findings ranged from neonatal mortality resulting from respiratory insufficiency and hydrops to childhood acute episodes of cardiopulmonary failure and slowly progressive muscle weakness. We show that SLC25A26 mutations cause various mitochondrial defects, including those affecting RNA stability, protein modification, mitochondrial translation, and the biosynthesis of CoQ10 and lipoic acid.


Subject(s)
Amino Acid Transport Systems/genetics , Calcium-Binding Proteins/genetics , DNA Methylation , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Muscle Weakness/genetics , Mutation/genetics , S-Adenosylmethionine/metabolism , Amino Acid Sequence , Child, Preschool , Female , Humans , Male , Molecular Sequence Data , Muscle Weakness/pathology , Pedigree , Prognosis , RNA Stability , Sequence Homology, Amino Acid , Thioctic Acid/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
11.
Am J Hum Genet ; 96(2): 309-17, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25658047

ABSTRACT

Primary coenzyme Q10 (CoQ10) deficiencies are rare, clinically heterogeneous disorders caused by mutations in several genes encoding proteins involved in CoQ10 biosynthesis. CoQ10 is an essential component of the electron transport chain (ETC), where it shuttles electrons from complex I or II to complex III. By whole-exome sequencing, we identified five individuals carrying biallelic mutations in COQ4. The precise function of human COQ4 is not known, but it seems to play a structural role in stabilizing a multiheteromeric complex that contains most of the CoQ10 biosynthetic enzymes. The clinical phenotypes of the five subjects varied widely, but four had a prenatal or perinatal onset with early fatal outcome. Two unrelated individuals presented with severe hypotonia, bradycardia, respiratory insufficiency, and heart failure; two sisters showed antenatal cerebellar hypoplasia, neonatal respiratory-distress syndrome, and epileptic encephalopathy. The fifth subject had an early-onset but slowly progressive clinical course dominated by neurological deterioration with hardly any involvement of other organs. All available specimens from affected subjects showed reduced amounts of CoQ10 and often displayed a decrease in CoQ10-dependent ETC complex activities. The pathogenic role of all identified mutations was experimentally validated in a recombinant yeast model; oxidative growth, strongly impaired in strains lacking COQ4, was corrected by expression of human wild-type COQ4 cDNA but failed to be corrected by expression of COQ4 cDNAs with any of the mutations identified in affected subjects. COQ4 mutations are responsible for early-onset mitochondrial diseases with heterogeneous clinical presentations and associated with CoQ10 deficiency.


Subject(s)
Ataxia/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Phenotype , Ubiquinone/deficiency , Amino Acid Sequence , Ataxia/pathology , Base Sequence , Exome/genetics , Fatal Outcome , Female , Gene Components , Humans , Male , Mitochondrial Diseases/pathology , Molecular Sequence Data , Muscle Weakness/pathology , Mutation/genetics , Pedigree , Saccharomyces cerevisiae , Sequence Analysis, DNA , Ubiquinone/genetics
12.
J Pediatr ; 193: 256-260, 2018 02.
Article in English | MEDLINE | ID: mdl-29249525

ABSTRACT

The diagnosis of Barth syndrome is challenging owing to the wide phenotypic spectrum with allelic heterogeneity. Here we report 3 cases of Barth syndrome with phenotypic and allelic heterogeneity that were diagnosed by different approaches, including whole exome sequencing and final confirmation by reverse-transcription polymease chain reaction.


Subject(s)
Barth Syndrome/diagnosis , Transcription Factors/genetics , Acyltransferases , Barth Syndrome/genetics , Humans , Infant , Infant, Newborn , Male , Mutation , Phenotype , Reverse Transcriptase Polymerase Chain Reaction/methods , Exome Sequencing/methods
13.
Brain ; 140(6): 1595-1610, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28549128

ABSTRACT

Although mitochondrial disorders are clinically heterogeneous, they frequently involve the central nervous system and are among the most common neurogenetic disorders. Identifying the causal genes has benefited enormously from advances in high-throughput sequencing technologies; however, once the defect is known, researchers face the challenge of deciphering the underlying disease mechanism. Here we characterize large biallelic deletions in the region encoding the ATAD3C, ATAD3B and ATAD3A genes. Although high homology complicates genomic analysis of the ATAD3 defects, they can be identified by targeted analysis of standard single nucleotide polymorphism array and whole exome sequencing data. We report deletions that generate chimeric ATAD3B/ATAD3A fusion genes in individuals from four unrelated families with fatal congenital pontocerebellar hypoplasia, whereas a case with genomic rearrangements affecting the ATAD3C/ATAD3B genes on one allele and ATAD3B/ATAD3A genes on the other displays later-onset encephalopathy with cerebellar atrophy, ataxia and dystonia. Fibroblasts from affected individuals display mitochondrial DNA abnormalities, associated with multiple indicators of altered cholesterol metabolism. Moreover, drug-induced perturbations of cholesterol homeostasis cause mitochondrial DNA disorganization in control cells, while mitochondrial DNA aggregation in the genetic cholesterol trafficking disorder Niemann-Pick type C disease further corroborates the interdependence of mitochondrial DNA organization and cholesterol. These data demonstrate the integration of mitochondria in cellular cholesterol homeostasis, in which ATAD3 plays a critical role. The dual problem of perturbed cholesterol metabolism and mitochondrial dysfunction could be widespread in neurological and neurodegenerative diseases.


Subject(s)
Adenosine Triphosphatases/genetics , Cerebellum/abnormalities , DNA, Mitochondrial/genetics , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Nervous System Malformations/genetics , ATPases Associated with Diverse Cellular Activities , Adult , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Consanguinity , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Humans , Infant , Infant, Newborn , Male , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Diseases/physiopathology , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/physiopathology
14.
Int J Clin Oncol ; 23(3): 497-503, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29330641

ABSTRACT

BACKGROUND: The base excision repair gene MUTYH is the causative gene of colorectal polyposis syndrome, which is an autosomal recessive disorder associated with a high risk of colorectal cancer. Since few studies have investigated the genotype-phenotype association in Japanese patients with MUTYH variants, the aim of this study was to clarify the clinicopathological findings in Japanese patients with MUTYH gene variants who were detected by screening causative genes associated with hereditary colorectal polyposis. METHODS: After obtaining informed consent, genetic testing was performed using target enrichment sequencing of 26 genes, including MUTYH. RESULTS: Of the 31 Japanese patients with suspected hereditary colorectal polyposis, eight MUTYH variants were detected in five patients. MUTYH hotspot variants known for Caucasians, namely p.G396D and p.Y179D, were not among the detected variants.Of five patients, two with biallelic MUTYH variants were diagnosed with MUTYH-associated polyposis, while two others had monoallelic MUTYH variants. One patient had the p.P18L and p.G25D variants on the same allele; however, supportive data for considering these two variants 'pathogenic' were lacking. CONCLUSIONS: Two patients with biallelic MUTYH variants and two others with monoallelic MUTYH variants were identified among Japanese colorectal polyposis patients. Hotspot variants of the MUTYH gene for Caucasians were not hotspots for Japanese patients.


Subject(s)
Adenomatous Polyposis Coli/etiology , DNA Glycosylases/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adolescent , Adult , Aged , Alleles , Asian People/genetics , Cohort Studies , Female , Genetic Variation , Humans , Male , Middle Aged , Mutation , White People/genetics
15.
Hum Mutat ; 38(12): 1796-1800, 2017 12.
Article in English | MEDLINE | ID: mdl-28722338

ABSTRACT

High-density oligonucleotide arrays have widely been used to detect pathogenic chromosomal deletions. In addition to high-density oligonucleotide arrays, programs using whole-exome sequencing have become available for estimating copy-number variations using depth of coverage. Here, we propose a new statistical method, HDR-del, to prioritize pathogenic chromosomal deletions based on Hamming distance in exome sequencing. In vcf (variant call format) files generated from exome sequencing, hemizygous chromosomal deletion regions lack heterozygous variants and lead to apparent long runs of homozygosity (ROH). In our Hamming distance ratio (HDR)-del approach, we calculate the "difference" in heterozygous status between an affected individual and control individuals using the HDR over all candidate chromosomal deletion regions defined as ROH longer than 1Mbp. Using a suitable test statistic, which is expected to be large for a true pathogenic deletion region, we prioritize candidate chromosomal deletion regions based on this statistic. In our approach, we were able to considerably narrow down true pathogenic chromosomal deletion regions, which were confirmed by high-density oligonucleotide arrays in four mitochondrial disease patients. Our HDR-del approach represents an easy method for detecting chromosomal deletions.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , DNA Copy Number Variations , Exome Sequencing/methods , Exome/genetics , Mitochondrial Diseases/genetics , Child , Datasets as Topic , Homozygote , Humans , Oligonucleotide Array Sequence Analysis , Statistics as Topic
16.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25434004

ABSTRACT

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Subject(s)
Acidosis, Lactic/genetics , Brain Diseases/genetics , Cardiomyopathy, Hypertrophic/genetics , GTP-Binding Proteins/genetics , Protein Processing, Post-Translational , Acidosis, Lactic/physiopathology , Amino Acid Sequence , Brain/pathology , Brain Diseases/physiopathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Line , Child , Child, Preschool , Consanguinity , Female , Fibroblasts , GTP-Binding Proteins/metabolism , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , Mutation , Pedigree , Protein Biosynthesis , RNA Interference , RNA, Transfer/genetics , RNA, Transfer/metabolism , Sequence Alignment
17.
J Hum Genet ; 62(5): 539-547, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28123175

ABSTRACT

Tafazzin, encoded by the TAZ gene, is a mitochondrial membrane-associated protein that remodels cardiolipin (CL), an important mitochondrial phospholipid. TAZ mutations are associated with Barth syndrome (BTHS). BTHS is an X-linked multisystemic disorder affecting usually male patients. Through sequence analysis of TAZ, we found one novel mutation c.39_60del p.(Pro14Alafs*19) by whole-exome sequencing and a reported missense mutation c.280C>T p.(Arg94Cys) by Sanger sequencing in two male patients (Pt1 and Pt2). Patient with c.280C>T mutation had dilated cardiomyopathy, while another patient with c.39_60del mutation had no feature of cardiomyopathy. A reported m.1555A>G homoplasmic variant was also identified in the patient having mutation c.39_60del by whole mitochondrial DNA sequencing method. This variant was not considered to be the main cause of mitochondrial dysfunction based on a cytoplasmic hybrid (cybrid) assay. Tafazzin expression was absent in both patient-derived fibroblast cells. Complementation of TAZ expression in fibroblasts from the patient with the novel mutation c.39_60del restored mitochondrial respiratory complex assembly. High-performance liquid chromatography-tandem mass spectrometry-based metabolic analysis revealed the decline of CL and the accumulation of monolysocardiolipin, indicating the loss of tafazzin activity. Owing to phenotypic variability, it is difficult to diagnose BTHS based on clinical features only. We conclude that genetic analysis should be performed to avoid underdiagnosis of this potentially life-threatening inborn error of metabolism.


Subject(s)
Cardiomyopathies/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , Transcription Factors/genetics , Acyltransferases , Base Sequence , Child , Child, Preschool , Electron Transport/genetics , Female , Genotype , Humans , Infant, Newborn , Male , Phenotype , Pregnancy , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism
18.
J Inherit Metab Dis ; 40(5): 685-693, 2017 09.
Article in English | MEDLINE | ID: mdl-28429146

ABSTRACT

Leigh syndrome (LS) is a progressive neurodegenerative disorder of infancy and early childhood. It is clinically diagnosed by typical manifestations and characteristic computed tomography (CT) or magnetic resonance imaging (MRI) studies. Unravelling mitochondrial respiratory chain (MRC) dysfunction behind LS is essential for deeper understanding of the disease, which may lead to the development of new therapies and cure. The aim of this study was to evaluate the clinical validity of various diagnostic tools in confirming MRC disorder in LS and Leigh-like syndrome (LL). The results of enzyme assays, molecular analysis, and cellular oxygen consumption rate (OCR) measurements were examined. Of 106 patients, 41 were biochemically and genetically verified, and 34 had reduced MRC activity but no causative mutations. Seven patients with normal MRC complex activities had mutations in the MT-ATP6 gene. Five further patients with normal activity in MRC were identified with causative mutations. Conversely, 12 out of 60 enzyme assays performed for genetically verified patients returned normal results. No biochemical or genetic background was confirmed for 19 patients. OCR was reduced in ten out of 19 patients with negative enzyme assay results. Inconsistent enzyme assay results between fibroblast and skeletal muscle biopsy samples were observed in 33% of 37 simultaneously analyzed cases. These data suggest that highest diagnostic rate is reached using a combined enzymatic and genetic approach, analyzing more than one type of biological materials where suitable. Microscale oxygraphy detected MRC impairment in 50% cases with no defect in MRC complex activities.


Subject(s)
Leigh Disease/diagnosis , Adolescent , Adult , Asian People , Child , Electron Transport/genetics , Female , Fibroblasts/physiology , Humans , Leigh Disease/genetics , Male , Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Muscle, Skeletal/physiology , Mutation/genetics , Oxygen Consumption/genetics , Young Adult
20.
J Hum Genet ; 61(11): 959-963, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27357426

ABSTRACT

In the search for sequence variants underlying disease, commonly applied filtering steps usually result in a number of candidate variants that cannot further be narrowed down. In autosomal recessive families, disease usually occurs only in one generation so that genetic linkage analysis is unlikely to help. Because homozygous recessive mutations tend to be inherited together with flanking homozygous variants, we developed a statistical method to detect pathogenic variants in autosomal recessive families: We look for differences in patterns of homozygosity around candidate variants between patients and control individuals and expect that such differences are greater for pathogenic variants than random candidate variants. In six autosomal recessive mitochondrial disease families, in which pathogenic homozygous variants have already been identified, our approach succeeded in prioritizing pathogenic mutations. Our method is applicable to single patients from recessive families with at least a few dozen control individuals from the same population; it is easy to use and is highly effective for detecting causative mutations in autosomal recessive families.


Subject(s)
Computational Biology/methods , Genes, Recessive , Genetic Association Studies/methods , Genetic Linkage , Software , Case-Control Studies , Genetic Predisposition to Disease , Genotype , Humans , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL