Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
EMBO J ; 42(5): e111372, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36514953

ABSTRACT

Mitophagy, the elimination of mitochondria via the autophagy-lysosome pathway, is essential for the maintenance of cellular homeostasis. The best characterised mitophagy pathway is mediated by stabilisation of the protein kinase PINK1 and recruitment of the ubiquitin ligase Parkin to damaged mitochondria. Ubiquitinated mitochondrial surface proteins are recognised by autophagy receptors including NDP52 which initiate the formation of an autophagic vesicle around the mitochondria. Damaged mitochondria also generate reactive oxygen species (ROS) which have been proposed to act as a signal for mitophagy, however the mechanism of ROS sensing is unknown. Here we found that oxidation of NDP52 is essential for the efficient PINK1/Parkin-dependent mitophagy. We identified redox-sensitive cysteine residues involved in disulphide bond formation and oligomerisation of NDP52 on damaged mitochondria. Oligomerisation of NDP52 facilitates the recruitment of autophagy machinery for rapid mitochondrial degradation. We propose that redox sensing by NDP52 allows mitophagy to function as a mechanism of oxidative stress response.


Subject(s)
Mitophagy , Nuclear Proteins , Protein Kinases , Humans , Autophagy , HeLa Cells , Mitophagy/physiology , Oxidation-Reduction , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Nuclear Proteins/metabolism
2.
Am J Hum Genet ; 108(11): 2195-2204, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34715011

ABSTRACT

Human mitochondrial RNase P (mt-RNase P) is responsible for 5' end processing of mitochondrial precursor tRNAs, a vital step in mitochondrial RNA maturation, and is comprised of three protein subunits: TRMT10C, SDR5C1 (HSD10), and PRORP. Pathogenic variants in TRMT10C and SDR5C1 are associated with distinct recessive or x-linked infantile onset disorders, resulting from defects in mitochondrial RNA processing. We report four unrelated families with multisystem disease associated with bi-allelic variants in PRORP, the metallonuclease subunit of mt-RNase P. Affected individuals presented with variable phenotypes comprising sensorineural hearing loss, primary ovarian insufficiency, developmental delay, and brain white matter changes. Fibroblasts from affected individuals in two families demonstrated decreased steady state levels of PRORP, an accumulation of unprocessed mitochondrial transcripts, and decreased steady state levels of mitochondrial-encoded proteins, which were rescued by introduction of the wild-type PRORP cDNA. In mt-tRNA processing assays performed with recombinant mt-RNase P proteins, the disease-associated variants resulted in diminished mitochondrial tRNA processing. Identification of disease-causing variants in PRORP indicates that pathogenic variants in all three subunits of mt-RNase P can cause mitochondrial dysfunction, each with distinct pleiotropic clinical presentations.


Subject(s)
Alleles , Genetic Pleiotropy , Mitochondria/enzymology , RNA, Mitochondrial/genetics , RNA, Transfer/genetics , Ribonuclease P/genetics , Adult , Female , Humans , Male , Pedigree
3.
Nature ; 563(7730): 235-240, 2018 11.
Article in English | MEDLINE | ID: mdl-30356213

ABSTRACT

Biosynthesis of glycogen, the essential glucose (and hence energy) storage molecule in humans, animals and fungi1, is initiated by the glycosyltransferase enzyme, glycogenin (GYG). Deficiencies in glycogen formation cause neurodegenerative and metabolic disease2-4, and mouse knockout5 and inherited human mutations6 of GYG impair glycogen synthesis. GYG acts as a 'seed core' for the formation of the glycogen particle by catalysing its own stepwise autoglucosylation to form a covalently bound gluco-oligosaccharide chain at initiation site Tyr 195. Precise mechanistic studies have so far been prevented by an inability to access homogeneous glycoforms of this protein, which unusually acts as both catalyst and substrate. Here we show that unprecedented direct access to different, homogeneously glucosylated states of GYG can be accomplished through a palladium-mediated enzyme activation 'shunt' process using on-protein C-C bond formation. Careful mimicry of GYG intermediates recapitulates catalytic activity at distinct stages, which in turn allows discovery of triphasic kinetics and substrate plasticity in GYG's use of sugar substrates. This reveals a tolerant but 'proof-read' mechanism that underlies the precision of this metabolic process. The present demonstration of direct, chemically controlled access to intermediate states of active enzymes suggests that such ligation-dependent activation could be a powerful tool in the study of mechanism.


Subject(s)
Glucose/biosynthesis , Palladium/metabolism , Biocatalysis , Enzyme Activation , Galactose/metabolism , Glucosyltransferases/metabolism , Glycoproteins/metabolism , Glycosylation , Humans , Kinetics , Uridine Diphosphate/metabolism
4.
Am J Hum Genet ; 106(2): 256-263, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004446

ABSTRACT

We report an inborn error of metabolism caused by TKFC deficiency in two unrelated families. Rapid trio genome sequencing in family 1 and exome sequencing in family 2 excluded known genetic etiologies, and further variant analysis identified rare homozygous variants in TKFC. TKFC encodes a bifunctional enzyme involved in fructose metabolism through its glyceraldehyde kinase activity and in the generation of riboflavin cyclic 4',5'-phosphate (cyclic FMN) through an FMN lyase domain. The TKFC homozygous variants reported here are located within the FMN lyase domain. Functional assays in yeast support the deleterious effect of these variants on protein function. Shared phenotypes between affected individuals with TKFC deficiency include cataracts and developmental delay, associated with cerebellar hypoplasia in one case. Further complications observed in two affected individuals included liver dysfunction and microcytic anemia, while one had fatal cardiomyopathy with lactic acidosis following a febrile illness. We postulate that deficiency of TKFC causes disruption of endogenous fructose metabolism leading to generation of by-products that can cause cataract. In line with this, an affected individual had mildly elevated urinary galactitol, which has been linked to cataract development in the galactosemias. Further, in light of a previously reported role of TKFC in regulating innate antiviral immunity through suppression of MDA5, we speculate that deficiency of TKFC leads to impaired innate immunity in response to viral illness, which may explain the fatal illness observed in the most severely affected individual.


Subject(s)
Cataract/etiology , Cerebellum/abnormalities , Developmental Disabilities/etiology , Mutation , Nervous System Malformations/etiology , Phosphorus-Oxygen Lyases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Alleles , Amino Acid Sequence , Cataract/pathology , Cerebellum/pathology , Child, Preschool , Developmental Disabilities/pathology , Female , Homozygote , Humans , Infant , Male , Nervous System Malformations/pathology , Pedigree , Phenotype , Phosphorylation , Sequence Homology , Exome Sequencing
5.
J Inherit Metab Dis ; 46(3): 406-420, 2023 05.
Article in English | MEDLINE | ID: mdl-36680553

ABSTRACT

Vitamin B12 (cobalamin, Cbl) is required as a cofactor by two human enzymes, 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR) and methylmalonyl-CoA mutase (MMUT). Within the body, a vast array of transporters, enzymes and chaperones are required for the generation and delivery of these cofactor forms. How they perform these functions is dictated by the structure and interactions of the proteins involved, the molecular bases of which are only now being elucidated. In this review, we highlight recent insights into human Cbl metabolism and address open questions in the field by employing a protein structure and interactome based perspective. We discuss how three very similar proteins-haptocorrin, intrinsic factor and transcobalamin-exploit slight structural differences and unique ligand receptor interactions to effect selective Cbl absorption and internalisation. We describe recent advances in the understanding of how endocytosed Cbl is transported across the lysosomal membrane and the implications of the recently solved ABCD4 structure. We detail how MMACHC and MMADHC cooperate to modify and target cytosolic Cbl to the client enzymes MTR and MMUT using ingenious modifications to an ancient nitroreductase fold, and how MTR and MMUT link with their accessory enzymes to sustainably harness the supernucleophilic potential of Cbl. Finally, we provide an outlook on how future studies may combine structural and interactome based approaches and incorporate knowledge of post-translational modifications to bring further insights.


Subject(s)
Methylmalonyl-CoA Mutase , Vitamin B 12 , Humans , Vitamin B 12/metabolism , Methylmalonyl-CoA Mutase/metabolism , Biological Transport , Molecular Chaperones , ATP-Binding Cassette Transporters/metabolism , Oxidoreductases/metabolism
6.
Am J Hum Genet ; 101(3): 466-477, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886345

ABSTRACT

RAC1 is a widely studied Rho GTPase, a class of molecules that modulate numerous cellular functions essential for normal development. RAC1 is highly conserved across species and is under strict mutational constraint. We report seven individuals with distinct de novo missense RAC1 mutations and varying degrees of developmental delay, brain malformations, and additional phenotypes. Four individuals, each harboring one of c.53G>A (p.Cys18Tyr), c.116A>G (p.Asn39Ser), c.218C>T (p.Pro73Leu), and c.470G>A (p.Cys157Tyr) variants, were microcephalic, with head circumferences between -2.5 to -5 SD. In contrast, two individuals with c.151G>A (p.Val51Met) and c.151G>C (p.Val51Leu) alleles were macrocephalic with head circumferences of +4.16 and +4.5 SD. One individual harboring a c.190T>G (p.Tyr64Asp) allele had head circumference in the normal range. Collectively, we observed an extraordinary spread of ∼10 SD of head circumferences orchestrated by distinct mutations in the same gene. In silico modeling, mouse fibroblasts spreading assays, and in vivo overexpression assays using zebrafish as a surrogate model demonstrated that the p.Cys18Tyr and p.Asn39Ser RAC1 variants function as dominant-negative alleles and result in microcephaly, reduced neuronal proliferation, and cerebellar abnormalities in vivo. Conversely, the p.Tyr64Asp substitution is constitutively active. The remaining mutations are probably weakly dominant negative or their effects are context dependent. These findings highlight the importance of RAC1 in neuronal development. Along with TRIO and HACE1, a sub-category of rare developmental disorders is emerging with RAC1 as the central player. We show that ultra-rare disorders caused by private, non-recurrent missense mutations that result in varying phenotypes are challenging to dissect, but can be delineated through focused international collaboration.


Subject(s)
Brain Diseases/genetics , Developmental Disabilities/genetics , Microcephaly/genetics , Mutation, Missense , rac1 GTP-Binding Protein/genetics , Adolescent , Amino Acid Sequence , Animals , Brain Diseases/pathology , Child , Child, Preschool , Developmental Disabilities/pathology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Female , Humans , Infant , Male , Mice , Microcephaly/pathology , Pedigree , Phenotype , Zebrafish/genetics , Zebrafish/growth & development
7.
Genet Med ; 22(1): 199-209, 2020 01.
Article in English | MEDLINE | ID: mdl-31462754

ABSTRACT

PURPOSE: Mitochondrial DNA (mtDNA) depletion syndrome (MDDS) encompasses a group of genetic disorders of mtDNA maintenance. Mutation of RRM2B is an uncommon cause of infantile-onset encephalomyopathic MDDS. Here we describe the natural history of this disease. METHODS: Multinational series of new genetically confirmed cases from six pediatric centers. RESULTS: Nine new cases of infantile-onset RRM2B deficiency, and 22 previously published cases comprised a total cohort of 31 patients. Infants presented at a mean of 1.95 months with truncal hypotonia, generalized weakness, and faltering growth. Seizures evolved in 39% at a mean of 3.1 months. Non-neurological manifestations included respiratory distress/failure (58%), renal tubulopathy (55%), sensorineural hearing loss (36%), gastrointestinal disturbance (32%), eye abnormalities (13%), and anemia (13%). Laboratory features included elevated lactate (blood, cerebrospinal fluid (CSF), urine, magnetic resonance (MR), spectroscopy), ragged-red and cytochrome c oxidase-deficient fibers, lipid myopathy, and multiple oxidative phosphorylation enzyme deficiencies in skeletal muscle. Eight new RRM2B variants were identified. Patients with biallelic truncating variants had the worst survival. Overall survival was 29% at 6 months and 16% at 1 year. CONCLUSIONS: Infantile-onset MDDS due to RRM2B deficiency is a severe disorder with characteristic clinical features and extremely poor prognosis. Presently management is supportive as there is no effective treatment. Novel treatments are urgently needed.


Subject(s)
Cell Cycle Proteins/genetics , Intestinal Pseudo-Obstruction/genetics , Muscular Dystrophy, Oculopharyngeal/genetics , Mutation, Missense , Ribonucleotide Reductases/genetics , Cell Cycle Proteins/chemistry , Female , Humans , Infant , Infant, Newborn , Intestinal Pseudo-Obstruction/mortality , Male , Models, Molecular , Muscular Dystrophy, Oculopharyngeal/mortality , Ophthalmoplegia/congenital , Prognosis , Protein Conformation , Ribonucleotide Reductases/chemistry , Survival Analysis
8.
Blood ; 131(9): 1000-1011, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29187380

ABSTRACT

Mutations in NBEAL2, the gene encoding the scaffolding protein Nbeal2, are causal of gray platelet syndrome (GPS), a rare recessive bleeding disorder characterized by platelets lacking α-granules and progressive marrow fibrosis. We present here the interactome of Nbeal2 with additional validation by reverse immunoprecipitation of Dock7, Sec16a, and Vac14 as interactors of Nbeal2. We show that GPS-causing mutations in its BEACH domain have profound and possible effects on the interaction with Dock7 and Vac14, respectively. Proximity ligation assays show that these 2 proteins are physically proximal to Nbeal2 in human megakaryocytes. In addition, we demonstrate that Nbeal2 is primarily localized in the cytoplasm and Dock7 on the membrane of or in α-granules. Interestingly, platelets from GPS cases and Nbeal2-/- mice are almost devoid of Dock7, resulting in a profound dysregulation of its signaling pathway, leading to defective actin polymerization, platelet activation, and shape change. This study shows for the first time proteins interacting with Nbeal2 and points to the dysregulation of the canonical signaling pathway of Dock7 as a possible cause of the aberrant formation of platelets in GPS cases and Nbeal2-deficient mice.


Subject(s)
Blood Platelets/metabolism , Blood Proteins/metabolism , GTPase-Activating Proteins/metabolism , Megakaryocytes/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Blood Platelets/cytology , Blood Proteins/genetics , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Megakaryocytes/cytology , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Protein Binding , Vesicular Transport Proteins/genetics
9.
Am J Med Genet A ; 182(11): 2685-2693, 2020 11.
Article in English | MEDLINE | ID: mdl-32808436

ABSTRACT

We report the case of an 11-year-old Syrian girl born to consanguineous parents, who presents an ataxic gait from early childhood. On clinical examination, she presented a severe static - kinetic cerebellar syndrome, walking without support is possible for short distances only. Strikingly, three consecutive MRIs did not show any sign of cerebellar abnormalities, but a brain positron emission tomography (PET) using [18F]-fluorodeoxyglucose (FDG) demonstrated a clear decrease in glucose metabolism in the cerebellum as well as the anterior and medial temporal lobe bilaterally. A clinical exome analysis identified a novel homozygous c.251A > G (p.Asn84Ser) likely pathogenic variant in the carbonic anhydrase 8 (CA8) gene. CA8 mutations cause cerebellar ataxia, mental retardation, and disequilibrium syndrome subtype 3 (CAMRQ3), a rare genetically autosomal recessive disorder, only described in four families, so far with the frequent observation of quadrupedal gait. The proband differed with other reported CA8 mutations by the absence of clear cerebellar signs on brain MRI and the presence of focal seizures. This report expands the clinical spectrum associated with mutations in CA8 and illustrates the possible discrepancy between (mild) neuro-radiological images (MRI) and (severe) clinical phenotype in young individuals. In contrast, the observation of clear cerebellar abnormal metabolic findings suggests that the FDG-PET scan may be used as an early marker for hereditary ataxia.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Ataxia/pathology , Homozygote , Intellectual Disability/pathology , Mutation , Phenotype , Cerebellar Ataxia/genetics , Child , Consanguinity , Female , Humans , Intellectual Disability/genetics , Male , Pedigree
10.
J Biol Chem ; 293(33): 12862-12876, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29880640

ABSTRACT

Mitochondrial tRNAs are transcribed as long polycistronic transcripts of precursor tRNAs and undergo posttranscriptional modifications such as endonucleolytic processing and methylation required for their correct structure and function. Among them, 5'-end processing and purine 9 N1-methylation of mitochondrial tRNA are catalyzed by two proteinaceous complexes with overlapping subunit composition. The Mg2+-dependent RNase P complex for 5'-end cleavage comprises the methyltransferase domain-containing protein tRNA methyltransferase 10C, mitochondrial RNase P subunit (TRMT10C/MRPP1), short-chain oxidoreductase hydroxysteroid 17ß-dehydrogenase 10 (HSD17B10/MRPP2), and metallonuclease KIAA0391/MRPP3. An MRPP1-MRPP2 subcomplex also catalyzes the formation of 1-methyladenosine/1-methylguanosine at position 9 using S-adenosyl-l-methionine as methyl donor. However, a lack of structural information has precluded insights into how these complexes methylate and process mitochondrial tRNA. Here, we used a combination of X-ray crystallography, interaction and activity assays, and small angle X-ray scattering (SAXS) to gain structural insight into the two tRNA modification complexes and their components. The MRPP1 N terminus is involved in tRNA binding and monomer-monomer self-interaction, whereas the C-terminal SPOUT fold contains key residues for S-adenosyl-l-methionine binding and N1-methylation. The entirety of MRPP1 interacts with MRPP2 to form the N1-methylation complex, whereas the MRPP1-MRPP2-MRPP3 RNase P complex only assembles in the presence of precursor tRNA. This study proposes low-resolution models of the MRPP1-MRPP2 and MRPP1-MRPP2-MRPP3 complexes that suggest the overall architecture, stoichiometry, and orientation of subunits and tRNA substrates.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/chemistry , Methyltransferases/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , RNA, Mitochondrial/chemistry , RNA, Transfer/chemistry , Ribonuclease P/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Crystallography, X-Ray , Humans , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , RNA, Mitochondrial/metabolism , RNA, Transfer/metabolism , Ribonuclease P/metabolism , Scattering, Small Angle
11.
Kidney Int ; 95(5): 1138-1152, 2019 05.
Article in English | MEDLINE | ID: mdl-30885509

ABSTRACT

Mutations in leucine-rich-repeats and immunoglobulin-like-domains 2 (LRIG2) or in heparanase 2 (HPSE2) cause urofacial syndrome, a devastating autosomal recessive disease of functional bladder outlet obstruction. It has been speculated that urofacial syndrome has a neural basis, but it is unknown whether defects in urinary bladder innervation are present. We hypothesized that urofacial syndrome features a peripheral neuropathy of the bladder. Mice with homozygous targeted Lrig2 mutations had urinary defects resembling those found in urofacial syndrome. There was no anatomical blockage of the outflow tract, consistent with a functional bladder outlet obstruction. Transcriptome analysis revealed differential expression of 12 known transcripts in addition to Lrig2, including 8 with established roles in neurobiology. Mice with homozygous mutations in either Lrig2 or Hpse2 had increased nerve density within the body of the urinary bladder and decreased nerve density around the urinary outflow tract. In a sample of 155 children with chronic kidney disease and urinary symptoms, we discovered novel homozygous missense LRIG2 variants that were predicted to be pathogenic in 2 individuals with non-syndromic bladder outlet obstruction. These observations provide evidence that a peripheral neuropathy is central to the pathobiology of functional bladder outlet obstruction in urofacial syndrome, and emphasize the importance of LRIG2 and heparanase 2 for nerve patterning in the urinary tract.


Subject(s)
Glucuronidase/genetics , Membrane Glycoproteins/genetics , Peripheral Nervous System Diseases/genetics , Urinary Bladder Neck Obstruction/genetics , Urinary Bladder/innervation , Urologic Diseases/genetics , Animals , Child , DNA Mutational Analysis , Facies , Female , Gene Expression Profiling , Homozygote , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Peripheral Nervous System Diseases/pathology , Urinary Bladder/pathology , Urinary Bladder Neck Obstruction/pathology , Urologic Diseases/pathology
12.
J Inherit Metab Dis ; 42(3): 565-574, 2019 05.
Article in English | MEDLINE | ID: mdl-30663059

ABSTRACT

Pyridoxine-dependent epilepsy (PDE) is often characterized as an early onset epileptic encephalopathy with dramatic clinical improvement following pyridoxine supplementation. Unfortunately, not all patients present with classic neonatal seizures or respond to an initial pyridoxine trial, which can result in the under diagnosis of this treatable disorder. Restriction of lysine intake and transport is associated with improved neurologic outcomes, although treatment should be started in the first year of life to be effective. Because of the documented diagnostic delay and benefit of early treatment, we aimed to develop a newborn screening method for PDE. Previous studies have demonstrated the accumulation of Δ1 -piperideine-6-carboxylate and α-aminoadipic semialdehyde in individuals with PDE, although these metabolites are unstable at room temperature (RT) limiting their utility for newborn screening. As a result, we sought to identify a biomarker that could be applied to current newborn screening paradigms. We identified a novel metabolite, 6-oxo-pipecolate (6-oxo-PIP), which accumulates in substantial amounts in blood, plasma, urine, and cerebral spinal fluid of individuals with PDE. Using a stable isotope-labeled internal standard, we developed a nonderivatized liquid chromatography tandem mass spectrometry-based method to quantify 6-oxo-PIP. This method replicates the analytical techniques used in many laboratories and could be used with few modifications in newborn screening programs. Furthermore, 6-oxo-PIP was measurable in urine for 4 months even when stored at RT. Herein, we report a novel biomarker for PDE that is stable at RT and can be quantified using current newborn screening techniques.


Subject(s)
Epilepsy/diagnosis , Neonatal Screening/methods , Pipecolic Acids/analysis , Biomarkers , Chromatography, Liquid , Female , Humans , Infant, Newborn , Male
13.
J Inherit Metab Dis ; 42(5): 809-817, 2019 09.
Article in English | MEDLINE | ID: mdl-31177572

ABSTRACT

The first step in branched-chain amino acid (BCAA) catabolism is catalyzed by the two BCAA transferase isoenzymes, cytoplasmic branched-chain amino acid transferase (BCAT) 1, and mitochondrial BCAT2. Defects in the second step of BCAA catabolism cause maple syrup urine disease (MSUD), a condition which has been far more extensively investigated. Here, we studied the consequences of BCAT2 deficiency, an ultra-rare condition in humans. We present genetic, clinical, and functional data in five individuals from four different families with homozygous or compound heterozygous BCAT2 mutations which were all detected following abnormal biochemical profile results or familial mutation segregation studies. We demonstrate that BCAT2 deficiency has a recognizable biochemical profile with raised plasma BCAAs and, in contrast with MSUD, low-normal branched-chain keto acids (BCKAs) with undetectable l-allo-isoleucine. Interestingly, unlike in MSUD, none of the individuals with BCAT2 deficiency developed acute encephalopathy even with exceptionally high BCAA levels. We observed wide-ranging clinical phenotypes in individuals with BCAT2 deficiency. While one adult was apparently asymptomatic, three individuals had presented with developmental delay and autistic features. We show that the biochemical characteristics of BCAT2 deficiency may be amenable to protein-restricted diet and that early treatment may improve outcome in affected individuals. BCAT2 deficiency is an inborn error of BCAA catabolism. At present, it is unclear whether developmental delay and autism are parts of the variable phenotypic spectrum of this condition or coincidental. Further studies will be required to explore this.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acids, Branched-Chain/blood , Brain/pathology , Mitochondria/pathology , Pregnancy Proteins/deficiency , Transaminases/deficiency , Adolescent , Adult , Brain/diagnostic imaging , Child , Child, Preschool , Female , Homozygote , Humans , Magnetic Resonance Imaging , Male , Minor Histocompatibility Antigens/genetics , Mutation , Phenotype , Pregnancy Proteins/genetics , Transaminases/genetics
14.
Hum Mol Genet ; 25(11): 2234-2244, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27005423

ABSTRACT

Classic galactosemia is a potentially lethal disease caused by the dysfunction of galactose 1-phosphate uridylyltransferase (GALT). Over 300 disease-associated GALT mutations have been reported, with the majority being missense changes, although a better understanding of their underlying molecular effects has been hindered by the lack of structural information for the human enzyme. Here, we present the 1.9 Å resolution crystal structure of human GALT (hGALT) ternary complex, revealing a homodimer arrangement that contains a covalent uridylylated intermediate and glucose-1-phosphate in the active site, as well as a structural zinc-binding site, per monomer. hGALT reveals significant structural differences from bacterial GALT homologues in metal ligation and dimer interactions, and therefore is a zbetter model for understanding the molecular consequences of disease mutations. Both uridylylation and zinc binding influence the stability and aggregation tendency of hGALT. This has implications for disease-associated variants where p.Gln188Arg, the most commonly detected, increases the rate of aggregation in the absence of zinc likely due to its reduced ability to form the uridylylated intermediate. As such our structure serves as a template in the future design of pharmacological chaperone therapies and opens new concepts about the roles of metal binding and activity in protein misfolding by disease-associated mutants.


Subject(s)
Galactosemias/genetics , Structure-Activity Relationship , Ternary Complex Factors/chemistry , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics , Binding Sites/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , Galactose/chemistry , Galactose/metabolism , Galactosemias/metabolism , Galactosemias/pathology , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Ternary Complex Factors/genetics , UTP-Hexose-1-Phosphate Uridylyltransferase/chemistry
15.
Biochem J ; 474(20): 3403-3420, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28827282

ABSTRACT

Glycogen storage disorders (GSDs) are caused by excessive accumulation of glycogen. Some GSDs [adult polyglucosan (PG) body disease (APBD), and Tarui and Lafora diseases] are caused by intracellular accumulation of insoluble inclusions, called PG bodies (PBs), which are chiefly composed of malconstructed glycogen. We developed an APBD patient skin fibroblast cell-based assay for PB identification, where the bodies are identified as amylase-resistant periodic acid-Schiff's-stained structures, and quantified. We screened the DIVERSet CL 10 084 compound library using this assay in high-throughput format and discovered 11 dose-dependent and 8 non-dose-dependent PB-reducing hits. Approximately 70% of the hits appear to act through reducing glycogen synthase (GS) activity, which can elongate glycogen chains and presumably promote PB generation. Some of these GS inhibiting hits were also computationally predicted to be similar to drugs interacting with the GS activator protein phosphatase 1. Our work paves the way to discovering medications for the treatment of PB-involving GSD, which are extremely severe or fatal disorders.


Subject(s)
Fibroblasts/enzymology , Glycogen Storage Disease , Glycogen Synthase/metabolism , Nervous System Diseases , Adult , Drug Evaluation, Preclinical/methods , Female , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/drug therapy , Glycogen Storage Disease/enzymology , Humans , Male , Nervous System Diseases/diagnosis , Nervous System Diseases/drug therapy , Nervous System Diseases/enzymology
16.
Hum Mutat ; 38(8): 988-1001, 2017 08.
Article in English | MEDLINE | ID: mdl-28497574

ABSTRACT

Mutations in the human MMAA gene cause the metabolic disorder cblA-type methylmalonic aciduria (MMA), although knowledge of the mechanism of dysfunction remains lacking. MMAA regulates the incorporation of the cofactor adenosylcobalamin (AdoCbl), generated from the MMAB adenosyltransferase, into the destination enzyme methylmalonyl-CoA mutase (MUT). This function of MMAA depends on its GTPase activity, which is stimulated by an interaction with MUT. Here, we present 67 new patients with cblA-type MMA, identifying 19 novel mutations. We biochemically investigated how missense mutations in MMAA in 22 patients lead to disease. About a third confer instability to the recombinant protein in bacterial and human expression systems. All 15 purified mutant proteins demonstrated wild-type like intrinsic GTPase activity and only one (p.Asp292Val), where the mutation is in the GTP binding domain, revealed decreased GTP binding. However, all mutations strongly decreased functional association with MUT by reducing GTPase activity stimulation upon incubation with MUT, while nine mutant proteins additionally lost the ability to physically bind MUT. Finally, all mutations interfered with gating the transfer of AdoCbl from MMAB to MUT. This work suggests loss of functional interaction between MMAA and MUT as a disease-causing mechanism that impacts processing and assembly of a cofactor to its destination enzyme.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Mitochondrial Proteins/metabolism , Amino Acid Metabolism, Inborn Errors/genetics , Child , Child, Preschool , Cobamides/metabolism , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Membrane Transport Proteins/metabolism , Methylmalonyl-CoA Mutase/metabolism , Mitochondrial Proteins/genetics , Mutation , Mutation, Missense/genetics , Protein Binding
17.
Hum Mutat ; 38(11): 1555-1568, 2017 11.
Article in English | MEDLINE | ID: mdl-28752568

ABSTRACT

Mucopolysaccharidosis Type I (MPS I) is a lysosomal storage disorder with varying degrees of phenotypic severity caused by mutations in IDUA. Over 200 disease-causing variants in IDUA have been reported. We describe the profile of disease-causing variants in 291 individuals with MPS I for whom IDUA sequencing was performed, focusing on the UK subset of the cohort. A total of 63 variants were identified, of which 20 were novel, and the functional significance of the novel variants is explored. The severe form of MPS I is treated with hematopoietic stem cell transplantation, known to have improved outcomes with earlier age at treatment. Developing genotype-phenotype relationships would therefore have considerable clinical utility, especially in the light of the development of newborn screening programs for MPS I. Associations between genotype and phenotype are examined in this cohort, particularly in the context of the profile of variants identified in UK individuals. Relevant associations can be made for the majority of UK individuals based on the presence of nonsense or truncating variants as well as other associations described in this report.


Subject(s)
Genetic Association Studies , Iduronidase/genetics , Mucopolysaccharidosis I/diagnosis , Mucopolysaccharidosis I/genetics , Mutation , Alleles , Enzyme Activation , Genotype , Humans , Iduronidase/metabolism , Mucopolysaccharidosis I/epidemiology , Phenotype , Sequence Analysis, DNA , Severity of Illness Index , United Kingdom/epidemiology
18.
Hum Mol Genet ; 24(20): 5667-76, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26199317

ABSTRACT

Glycogen branching enzyme 1 (GBE1) plays an essential role in glycogen biosynthesis by generating α-1,6-glucosidic branches from α-1,4-linked glucose chains, to increase solubility of the glycogen polymer. Mutations in the GBE1 gene lead to the heterogeneous early-onset glycogen storage disorder type IV (GSDIV) or the late-onset adult polyglucosan body disease (APBD). To better understand this essential enzyme, we crystallized human GBE1 in the apo form, and in complex with a tetra- or hepta-saccharide. The GBE1 structure reveals a conserved amylase core that houses the active centre for the branching reaction and harbours almost all GSDIV and APBD mutations. A non-catalytic binding cleft, proximal to the site of the common APBD mutation p.Y329S, was found to bind the tetra- and hepta-saccharides and may represent a higher-affinity site employed to anchor the complex glycogen substrate for the branching reaction. Expression of recombinant GBE1-p.Y329S resulted in drastically reduced protein yield and solubility compared with wild type, suggesting this disease allele causes protein misfolding and may be amenable to small molecule stabilization. To explore this, we generated a structural model of GBE1-p.Y329S and designed peptides ab initio to stabilize the mutation. As proof-of-principle, we evaluated treatment of one tetra-peptide, Leu-Thr-Lys-Glu, in APBD patient cells. We demonstrate intracellular transport of this peptide, its binding and stabilization of GBE1-p.Y329S, and 2-fold increased mutant enzymatic activity compared with untreated patient cells. Together, our data provide the rationale and starting point for the screening of small molecule chaperones, which could become novel therapies for this disease.


Subject(s)
Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type IV/enzymology , Glycogen Storage Disease/enzymology , Mutation, Missense , Nervous System Diseases/enzymology , Peptides/therapeutic use , Amino Acid Sequence , Computational Biology , Glycogen Debranching Enzyme System/drug effects , Glycogen Debranching Enzyme System/metabolism , Glycogen Storage Disease/drug therapy , Glycogen Storage Disease/genetics , Glycogen Storage Disease Type IV/genetics , Humans , Molecular Sequence Data , Nervous System Diseases/drug therapy , Nervous System Diseases/genetics , Protein Structure, Tertiary , Sequence Alignment
19.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3294-3302, 2017 12.
Article in English | MEDLINE | ID: mdl-28888424

ABSTRACT

MRPP2 (also known as HSD10/SDR5C1) is a multifunctional protein that harbours both catalytic and non-catalytic functions. The protein belongs to the short-chain dehydrogenase/reductases (SDR) family and is involved in the catabolism of isoleucine in vivo and steroid metabolism in vitro. MRPP2 also moonlights in a complex with the MRPP1 (also known as TRMT10C) protein for N1-methylation of purines at position 9 of mitochondrial tRNA, and in a complex with MRPP1 and MRPP3 (also known as PRORP) proteins for 5'-end processing of mitochondrial precursor tRNA. Inherited mutations in the HSD17B10 gene encoding MRPP2 protein lead to a childhood disorder characterised by progressive neurodegeneration, cardiomyopathy or both. Here we report two patients with novel missense mutations in the HSD17B10 gene (c.34G>C and c.526G>A), resulting in the p.V12L and p.V176M substitutions. Val12 and Val176 are highly conserved residues located at different regions of the MRPP2 structure. Recombinant mutant proteins were expressed and characterised biochemically to investigate their effects towards the functions of MRPP2 and associated complexes in vitro. Both mutant proteins showed significant reduction in the dehydrogenase, methyltransferase and tRNA processing activities compared to wildtype, associated with reduced stability for protein with p.V12L, whereas the protein carrying p.V176M showed impaired kinetics and complex formation. This study therefore identified two distinctive molecular mechanisms to explain the biochemical defects for the novel missense patient mutations.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Mitochondria/metabolism , RNA, Transfer/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/chemistry , Female , Gene Expression , Humans , Infant , Male , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Mutation, Missense , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonuclease P/genetics , Ribonuclease P/metabolism
20.
Brain ; 139(11): 2844-2854, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27604308

ABSTRACT

Neurometabolic disorders are markedly heterogeneous, both clinically and genetically, and are characterized by variable neurological dysfunction accompanied by suggestive neuroimaging or biochemical abnormalities. Despite early specialist input, delays in diagnosis and appropriate treatment initiation are common. Next-generation sequencing approaches still have limitations but are already enabling earlier and more efficient diagnoses in these patients. We designed a gene panel targeting 614 genes causing inborn errors of metabolism and tested its diagnostic efficacy in a paediatric cohort of 30 undiagnosed patients presenting with variable neurometabolic phenotypes. Genetic defects that could, at least partially, explain observed phenotypes were identified in 53% of cases. Where biochemical abnormalities pointing towards a particular gene defect were present, our panel identified diagnoses in 89% of patients. Phenotypes attributable to defects in more than one gene were seen in 13% of cases. The ability of in silico tools, including structure-guided prediction programmes to characterize novel missense variants were also interrogated. Our study expands the genetic, clinical and biochemical phenotypes of well-characterized (POMGNT1, TPP1) and recently identified disorders (PGAP2, ACSF3, SERAC1, AFG3L2, DPYS). Overall, our panel was accurate and efficient, demonstrating good potential for applying similar approaches to clinically and biochemically diverse neurometabolic disease cohorts.


Subject(s)
Brain Diseases, Metabolic/genetics , Genetic Predisposition to Disease , Metabolism, Inborn Errors/genetics , Adolescent , Brain Diseases, Metabolic/diagnostic imaging , Child , Child, Preschool , Cohort Studies , Female , Genetic Testing , Genotype , Humans , Imaging, Three-Dimensional , Infant , Magnetic Resonance Imaging , Male , Metabolism, Inborn Errors/diagnostic imaging , Phenotype , Tripeptidyl-Peptidase 1 , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL