Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Biochemistry (Mosc) ; 86(Suppl 1): S12-S23, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33827397

ABSTRACT

Aminoacyl-RNA synthetases (aaRSs) are among the key enzymes of protein biosynthesis. They are responsible for conducting the first step in the protein biosynthesis, namely attaching amino acids to the corresponding tRNA molecules both in cytoplasm and mitochondria. More and more research demonstrates that mutations in the genes encoding aaRSs lead to the development of various neurodegenerative diseases, such as incurable Charcot-Marie-Tooth disease (CMT) and distal spinal muscular atrophy. Some mutations result in the loss of tRNA aminoacylation activity, while other mutants retain their classical enzyme activity. In the latter case, disease manifestations are associated with additional neuron-specific functions of aaRSs. At present, seven aaRSs (GlyRS, TyrRS, AlaRS, HisRS, TrpRS, MetRS, and LysRS) are known to be involved in the CMT etiology with glycyl-tRNA synthetase (GlyRS) being the most studied of them.


Subject(s)
Glycine-tRNA Ligase/genetics , Mutation , Nervous System Diseases/enzymology , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Female , Humans , Male , Muscular Atrophy, Spinal/enzymology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Neurons/enzymology , Neurons/physiology
2.
J Neurochem ; 157(3): 351-369, 2021 05.
Article in English | MEDLINE | ID: mdl-33236345

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is one of the most common inherited neurodegenerative disorders with an increasing number of CMT-associated variants identified as causative factors, however, there has been no effective therapy for CMT to date. Aminoacyl-tRNA synthetases (aaRS) are essential enzymes in translation by charging amino acids onto their cognate tRNAs during protein synthesis. Dominant monoallelic variants of aaRSs have been largely implicated in CMT. Some aaRSs variants affect enzymatic activity, demonstrating a loss-of-function property. In contrast, loss of aminoacylation activity is neither necessary nor sufficient for some aaRSs variants to cause CMT. Instead, accumulating evidence from CMT patient samples, animal genetic studies or protein conformational analysis has pinpointed toxic gain-of-function of aaRSs variants in CMT, suggesting complicated mechanisms underlying the pathogenesis of CMT. In this review, we summarize the latest advances in studies on CMT-linked aaRSs, with a particular focus on their functions. The current challenges, future direction and the promising candidates for potential treatment of CMT are also discussed.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Amino Acyl-tRNA Synthetases/drug effects , Animals , Charcot-Marie-Tooth Disease/drug therapy , Disease Models, Animal , Humans , Mutation
3.
Int J Mol Sci ; 21(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33049996

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is one of the most common inherited peripheral neuropathies. CMT patients typically show slowly progressive muscle weakness and sensory loss in a distal dominant pattern in childhood. The diagnosis of CMT is based on clinical symptoms, electrophysiological examinations, and genetic testing. Advances in genetic testing technology have revealed the genetic heterogeneity of CMT; more than 100 genes containing the disease causative mutations have been identified. Because a single genetic alteration in CMT leads to progressive neurodegeneration, studies of CMT patients and their respective models revealed the genotype-phenotype relationships of targeted genes. Conventionally, rodents and cell lines have often been used to study the pathogenesis of CMT. Recently, Drosophila has also attracted attention as a CMT model. In this review, we outline the clinical characteristics of CMT, describe the advantages and disadvantages of using Drosophila in CMT studies, and introduce recent advances in CMT research that successfully applied the use of Drosophila, in areas such as molecules associated with mitochondria, endosomes/lysosomes, transfer RNA, axonal transport, and glucose metabolism.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Peripheral Nervous System Diseases/genetics , Amino Acyl-tRNA Synthetases/genetics , Animals , Axonal Transport/genetics , Charcot-Marie-Tooth Disease/classification , Charcot-Marie-Tooth Disease/enzymology , Child , Humans , Intracellular Membranes/metabolism , L-Iditol 2-Dehydrogenase/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Peripheral Nervous System Diseases/classification , Peripheral Nervous System Diseases/enzymology
4.
ACS Chem Neurosci ; 11(3): 258-267, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31845794

ABSTRACT

Disruption of axonal transport causes a number of rare, inherited axonopathies and is heavily implicated in a wide range of more common neurodegenerative disorders, many of them age-related. Acetylation of α-tubulin is one important regulatory mechanism, influencing microtubule stability and motor protein attachment. Of several strategies so far used to enhance axonal transport, increasing microtubule acetylation through inhibition of the deacetylase enzyme histone deacetylase 6 (HDAC6) has been one of the most effective. Several inhibitors have been developed and tested in animal and cellular models, but better drug candidates are still needed. Here we report the development and characterization of two highly potent HDAC6 inhibitors, which show low toxicity, promising pharmacokinetic properties, and enhance microtubule acetylation in the nanomolar range. We demonstrate their capacity to rescue axonal transport of mitochondria in a primary neuronal culture model of the inherited axonopathy Charcot-Marie-Tooth Type 2F, caused by a dominantly acting mutation in heat shock protein beta 1.


Subject(s)
Histone Deacetylase 6/antagonists & inhibitors , Mitochondria/metabolism , Neurons/drug effects , Tubulin/drug effects , Acetylation/drug effects , Animals , Charcot-Marie-Tooth Disease/enzymology , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Mice, Inbred C57BL , Microtubules/metabolism , Mitochondria/drug effects , Neurons/metabolism , Tubulin/metabolism
5.
Nat Commun ; 10(1): 4914, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664033

ABSTRACT

Mitofusin-2 (MFN2) is a dynamin-like GTPase that plays a central role in regulating mitochondrial fusion and cell metabolism. Mutations in MFN2 cause the neurodegenerative disease Charcot-Marie-Tooth type 2A (CMT2A). The molecular basis underlying the physiological and pathological relevance of MFN2 is unclear. Here, we present crystal structures of truncated human MFN2 in different nucleotide-loading states. Unlike other dynamin superfamily members including MFN1, MFN2 forms sustained dimers even after GTP hydrolysis via the GTPase domain (G) interface, which accounts for its high membrane-tethering efficiency. The biochemical discrepancy between human MFN2 and MFN1 largely derives from a primate-only single amino acid variance. MFN2 and MFN1 can form heterodimers via the G interface in a nucleotide-dependent manner. CMT2A-related mutations, mapping to different functional zones of MFN2, lead to changes in GTP hydrolysis and homo/hetero-association ability. Our study provides fundamental insight into how mitofusins mediate mitochondrial fusion and the ways their disruptions cause disease.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Dimerization , GTP Phosphohydrolases/genetics , Guanosine Triphosphate/metabolism , Humans , Mitochondria/chemistry , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mutation , Protein Domains
6.
Mol Neurobiol ; 56(12): 8656-8667, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31313076

ABSTRACT

Charcot-Marie-Tooth disease type-4J (CMT4J), an autosomal recessively inherited peripheral neuropathy characterized by neuronal degeneration, segmental demyelination, and limb muscle weakness, is caused by compound heterozygous mutations in the SAC3/FIG4 gene, resulting in SAC3/FIG4 protein deficiency. SAC3/FIG4 is a phosphatase that not only turns over PtdIns(3,5)P2 to PtdIns3P but also promotes PtdIns(3,5)P2 synthesis by activating the PIKFYVE kinase that also makes PtdIns5P. Whether CMT4J patients have alterations in PtdIns(3,5)P2, PtdIns5P or in other phosphoinositides (PIs), and if yes, in what direction these changes might be, has never been examined. We performed PI profiling in primary fibroblasts from a cohort of CMT4J patients. Subsequent to myo-[2-3H]inositol cell labeling to equilibrium, steady-state levels of PIs were quantified by HPLC under conditions concurrently detecting PtdIns5P, PtdIns(3,5)P2, and the other PIs. Immunoblotting verified SAC3/FIG4 depletion in CMT4J fibroblasts. Compared to normal human controls (n = 9), both PtdIns(3,5)P2 and PtdIns5P levels were significantly decreased in CMT4J fibroblasts (n = 13) by 36.4 ± 3.6% and 43.1 ± 4.4%, respectively (p < 0.0001). These reductions were independent of patients' gender or disease onset. Although mean values for PtdIns3P in the CMT4J cohort remained unchanged, there were high variations in PtdIns3P among individual patients. Aberrant endolysosomal vacuoles, typically seen under PtdIns(3,5)P2 reduction, were apparent but not in fibroblasts from all patients. The subset of patients without aberrant vacuoles exhibited especially low PtdIns3P levels. Concomitant decreases in PtdIns5P and PtdIns(3,5)P2 and the link between PtdIns3P levels and cellular vacuolization are novel insights shedding further light into the molecular determinants in CMT4J polyneuropathy.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/deficiency , Adolescent , Adult , Age of Onset , Child , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Flavoproteins/metabolism , Humans , Male , Middle Aged , Models, Biological , Phosphatidylinositols/chemistry , Phosphoric Monoester Hydrolases/metabolism , Vacuoles/metabolism
7.
Am J Hum Genet ; 104(3): 520-529, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30824121

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes responsible for charging tRNA molecules with cognate amino acids. Consistent with the essential function and ubiquitous expression of ARSs, mutations in 32 of the 37 ARS-encoding loci cause severe, early-onset recessive phenotypes. Previous genetic and functional data suggest a loss-of-function mechanism; however, our understanding of the allelic and locus heterogeneity of ARS-related disease is incomplete. Cysteinyl-tRNA synthetase (CARS) encodes the enzyme that charges tRNACys with cysteine in the cytoplasm. To date, CARS variants have not been implicated in any human disease phenotype. Here, we report on four subjects from three families with complex syndromes that include microcephaly, developmental delay, and brittle hair and nails. Each affected person carries bi-allelic CARS variants: one individual is compound heterozygous for c.1138C>T (p.Gln380∗) and c.1022G>A (p.Arg341His), two related individuals are compound heterozygous for c.1076C>T (p.Ser359Leu) and c.1199T>A (p.Leu400Gln), and one individual is homozygous for c.2061dup (p.Ser688Glnfs∗2). Measurement of protein abundance, yeast complementation assays, and assessments of tRNA charging indicate that each CARS variant causes a loss-of-function effect. Compared to subjects with previously reported ARS-related diseases, individuals with bi-allelic CARS variants are unique in presenting with a brittle-hair-and-nail phenotype, which most likely reflects the high cysteine content in human keratins. In sum, our efforts implicate CARS variants in human inherited disease, expand the locus and clinical heterogeneity of ARS-related clinical phenotypes, and further support impaired tRNA charging as the primary mechanism of recessive ARS-related disease.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/etiology , Developmental Disabilities/etiology , Hair Diseases/etiology , Microcephaly/etiology , Mutation , Nail Diseases/etiology , Adult , Amino Acid Sequence , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/pathology , Developmental Disabilities/enzymology , Developmental Disabilities/pathology , Female , Genes, Recessive , Genetic Predisposition to Disease , Hair Diseases/enzymology , Hair Diseases/pathology , Humans , Male , Microcephaly/enzymology , Microcephaly/pathology , Nail Diseases/enzymology , Nail Diseases/pathology , Pedigree , Phenotype , Prognosis , Sequence Homology , Young Adult
8.
J Biol Chem ; 294(14): 5321-5339, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30643024

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are essential enzymes that catalyze the first reaction in protein biosynthesis, namely the charging of transfer RNAs (tRNAs) with their cognate amino acids. aaRSs have been increasingly implicated in dominantly and recessively inherited human diseases. The most common aaRS-associated monogenic disorder is the incurable neurodegenerative disease Charcot-Marie-Tooth neuropathy (CMT), caused by dominant mono-allelic mutations in aaRSs. With six currently known members (GlyRS, TyrRS, AlaRS, HisRS, TrpRS, and MetRS), aaRSs represent the largest protein family implicated in CMT etiology. After the initial discovery linking aaRSs to CMT, the field has progressed from understanding whether impaired tRNA charging is a critical component of this disease to elucidating the specific pathways affected by CMT-causing mutations in aaRSs. Although many aaRS CMT mutants result in loss of tRNA aminoacylation function, animal genetics studies demonstrated that dominant mutations in GlyRS cause CMT through toxic gain-of-function effects, which also may apply to other aaRS-linked CMT subtypes. The CMT-causing mechanism is likely to be multifactorial and involves multiple cellular compartments, including the nucleus and the extracellular space, where the normal WT enzymes also appear. Thus, the association of aaRSs with neuropathy is relevant to discoveries indicating that aaRSs also have nonenzymatic regulatory functions that coordinate protein synthesis with other biological processes. Through genetic, functional, and structural analyses, commonalities among different mutations and different aaRS-linked CMT subtypes have begun to emerge, providing insights into the nonenzymatic functions of aaRSs and the pathogenesis of aaRS-linked CMT to guide therapeutic development to treat this disease.


Subject(s)
Amino Acyl-tRNA Synthetases , Cell Nucleus , Charcot-Marie-Tooth Disease , Protein Biosynthesis , RNA, Transfer , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Gain of Function Mutation , Humans , RNA, Transfer/genetics , RNA, Transfer/metabolism
9.
Neurosci Res ; 139: 69-78, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30261202

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is composed of a heterogeneous group of hereditary peripheral neuropathies. The peripheral nervous system primarily comprises two types of cells: neuronal cells and myelinating glial Schwann cells. CMT2 N is an autosomal dominant disease and its responsible gene encodes alanyl-tRNA synthetase (AARS), which is a family of cytoplasmic aminoacyl-tRNA synthetases. CMT2 N is associated with the mutation, including a missense mutation, which is known to decrease the enzymatic activity of AARS, but whether and how its mutation affects AARS localization and neuronal process formation remains to be understood. First, we show that the AARS mutant harboring Asn71-to-Tyr (N71Y) is not localized in cytoplasm. The expression of AARS mutant proteins in COS-7 cells mainly leads to localization into lysosome, whereas the wild type is indeed localized in cytoplasm. Second, in N1E-115 cells as the neuronal cell model, cells expressing the N71Y mutant do not have the ability to grow processes. Third, pretreatment with antiepileptic valproic acid reverses the inhibitory effect of the N71Y mutant on process growth. Taken together, the N71Y mutation of AARS leads to abnormal intracellular localization, inhibiting process growth, yet this inhibition is reversed by valproic acid.


Subject(s)
Alanine-tRNA Ligase/metabolism , Axons/metabolism , Charcot-Marie-Tooth Disease/enzymology , Neurites/metabolism , Valproic Acid/pharmacology , Alanine-tRNA Ligase/genetics , Charcot-Marie-Tooth Disease/metabolism , Humans , Mutation/genetics , Neurites/drug effects , Phenotype
10.
FEBS Lett ; 592(5): 703-717, 2018 03.
Article in English | MEDLINE | ID: mdl-29288497

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed enzymes responsible for charging tRNAs with their cognate amino acids, therefore essential for the first step in protein synthesis. Although the majority of protein synthesis happens in the cytosol, an additional translation apparatus is required to translate the 13 mitochondrial DNA-encoded proteins important for oxidative phosphorylation. Most ARS genes in these cellular compartments are distinct, but two genes are common, encoding aminoacyl-tRNA synthetases of glycine (GARS) and lysine (KARS) in both mitochondria and the cytosol. Mutations in the majority of the 37 nuclear-encoded human ARS genes have been linked to a variety of recessive and dominant tissue-specific disorders. Current data indicate that impaired enzyme function could explain the pathogenicity, however not all pathogenic ARSs mutations result in deficient catalytic function; thus, the consequences of mutations may arise from other molecular mechanisms. The peripheral nerves are frequently affected, as illustrated by the high number of mutations in cytosolic and bifunctional tRNA synthetases causing Charcot-Marie-Tooth disease (CMT). Here we provide insights on the pathomechanisms of CMT-causing tRNA synthetases with specific focus on the two bifunctional tRNA synthetases (GARS, KARS).


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Charcot-Marie-Tooth Disease , Cytosol , Mitochondria , Mitochondrial Proteins , Neuromuscular Diseases , Amino Acyl-tRNA Synthetases/genetics , Animals , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Cytosol/enzymology , Cytosol/pathology , Humans , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neuromuscular Diseases/enzymology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/pathology , Oxidative Phosphorylation
11.
Nucleic Acids Res ; 45(13): 8091-8104, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28531329

ABSTRACT

While having multiple aminoacyl-tRNA synthetases implicated in Charcot-Marie-Tooth (CMT) disease suggests a common mechanism, a defect in enzymatic activity is not shared among the CMT-causing mutants. Protein misfolding is a common hypothesis underlying the development of many neurological diseases. Its process usually involves an initial reduction in protein stability and then the subsequent oligomerization and aggregation. Here, we study the structural effect of three CMT-causing mutations in tyrosyl-tRNA synthetase (TyrRS or YARS). Through various approaches, we found that the mutations do not induce changes in protein secondary structures, or shared effects on oligomerization state and stability. However, all mutations provide access to a surface masked in the wild-type enzyme, and that access correlates with protein misinteraction. With recent data on another CMT-linked tRNA synthetase, we suggest that an inherent plasticity, engendering the formation of alternative stable conformations capable of aberrant interactions, links the tRNA synthetase family to CMT.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Tyrosine-tRNA Ligase/chemistry , Tyrosine-tRNA Ligase/metabolism , Amino Acid Substitution , Crystallography, X-Ray , Deuterium Exchange Measurement , Enzyme Stability/genetics , Humans , Kinetics , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Conformation , Protein Folding , Protein Multimerization/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Scattering, Small Angle , Tripartite Motif-Containing Protein 28 , Tyrosine-tRNA Ligase/genetics , X-Ray Diffraction
12.
Neurotherapeutics ; 14(2): 417-428, 2017 04.
Article in English | MEDLINE | ID: mdl-27957719

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is the most common inherited peripheral neuropathy, with an estimated prevalence of 1 in 2500. The degeneration of motor and sensory nerve axons leads to motor and sensory symptoms that progress over time and have an important impact on the daily life of these patients. Currently, there is no curative treatment available. Recently, we identified histone deacetylase 6 (HDAC6), which deacetylates α-tubulin, as a potential therapeutic target in axonal CMT (CMT2). Pharmacological inhibition of the deacetylating function of HDAC6 reversed the motor and sensory deficits in a mouse model for mutant "small heat shock protein B1" (HSPB1)-induced CMT2 at the behavioral and electrophysiological level. In order to translate this potential therapeutic strategy into a clinical application, small drug-like molecules that are potent and selective HDAC6 inhibitors are essential. To screen for these, we developed a method that consisted of 3 distinct phases and that was based on the pathological findings in the mutant HSPB1-induced CMT2 mouse model. Three different inhibitors (ACY-738, ACY-775, and ACY-1215) were tested and demonstrated to be both potent and selective HDAC6 inhibitors. Moreover, these inhibitors increased the innervation of the neuromuscular junctions in the gastrocnemius muscle and improved the motor and sensory nerve conduction, confirming that HDAC6 inhibition is a potential therapeutic strategy in CMT2. Furthermore, ACY-1215 is an interesting lead molecule as it is currently tested in clinical trials for cancer. Taken together, these results may speed up the translation of pharmacological inhibition of HDAC6 into a therapy against CMT2.


Subject(s)
Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/enzymology , Drug Evaluation, Preclinical , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Pyrimidines/pharmacology , Animals , Axonal Transport/drug effects , Ganglia, Spinal/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/innervation , Neuromuscular Junction/drug effects , Neurons/drug effects , Pyrimidines/therapeutic use , Tumor Cells, Cultured
13.
J Biol Chem ; 291(11): 5740-5752, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26797133

ABSTRACT

Glycyl-tRNA synthetase (GlyRS) is the enzyme that covalently links glycine to cognate tRNA for translation. It is of great research interest because of its nonconserved quaternary structures, unique species-specific aminoacylation properties, and noncanonical functions in neurological diseases, but none of these is fully understood. We report two crystal structures of human GlyRS variants, in the free form and in complex with tRNA(Gly) respectively, and reveal new aspects of the glycylation mechanism. We discover that insertion 3 differs considerably in conformation in catalysis and that it acts like a "switch" and fully opens to allow tRNA to bind in a cross-subunit fashion. The flexibility of the protein is supported by molecular dynamics simulation, as well as enzymatic activity assays. The biophysical and biochemical studies suggest that human GlyRS may utilize its flexibility for both the traditional function (regulate tRNA binding) and alternative functions (roles in diseases).


Subject(s)
Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/metabolism , RNA, Transfer, Gly/metabolism , Amino Acid Sequence , Aminoacylation , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Crystallography, X-Ray , Glycine-tRNA Ligase/genetics , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Protein Conformation
14.
ACS Chem Neurosci ; 7(2): 240-58, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26599234

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a disorder of the peripheral nervous system where progressive degeneration of motor and sensory nerves leads to motor problems and sensory loss and for which no pharmacological treatment is available. Recently, it has been shown in a model for the axonal form of CMT that histone deacetylase 6 (HDAC6) can serve as a target for the development of a pharmacological therapy. Therefore, we aimed at developing new selective and activity-specific HDAC6 inhibitors with improved biochemical properties. By utilizing a bicyclic cap as the structural scaffold from which to build upon, we developed several analogues that showed improved potency compared to tubastatin A while maintaining excellent selectivity compared to HDAC1. Further screening in N2a cells examining both the acetylation of α-tubulin and histones narrowed down the library of compounds to three potent and selective HDAC6 inhibitors. In mutant HSPB1-expressing DRG neurons, serving as an in vitro model for CMT2, these inhibitors were able to restore the mitochondrial axonal transport deficits. Combining structure-based development of HDAC6 inhibitors, screening in N2a cells and in a neuronal model for CMT2F, and preliminary ADMET and pharmacokinetic profiles, resulted in the selection of compound 23d that possesses improved biochemical, functional, and druglike properties compared to tubastatin A.


Subject(s)
Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/enzymology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Acetylation/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Ganglia, Spinal/cytology , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Interleukin-2/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neuroblastoma/pathology , Neurons/drug effects , Neurons/enzymology , Tubulin/genetics , Tubulin/metabolism
15.
Hum Mol Genet ; 25(4): 681-92, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26662798

ABSTRACT

FIG4 is a phosphoinositide phosphatase that is mutated in several diseases including Charcot-Marie-Tooth Disease 4J (CMT4J) and Yunis-Varon syndrome (YVS). To investigate the mechanism of disease pathogenesis, we generated Drosophila models of FIG4-related diseases. Fig4 null mutant animals are viable but exhibit marked enlargement of the lysosomal compartment in muscle cells and neurons, accompanied by an age-related decline in flight ability. Transgenic animals expressing Drosophila Fig4 missense mutations corresponding to human pathogenic mutations can partially rescue lysosomal expansion phenotypes, consistent with these mutations causing decreased FIG4 function. Interestingly, Fig4 mutations predicted to inactivate FIG4 phosphatase activity rescue lysosome expansion phenotypes, and mutations in the phosphoinositide (3) phosphate kinase Fab1 that performs the reverse enzymatic reaction also causes a lysosome expansion phenotype. Since FIG4 and FAB1 are present together in the same biochemical complex, these data are consistent with a model in which FIG4 serves a phosphatase-independent biosynthetic function that is essential for lysosomal membrane homeostasis. Lysosomal phenotypes are suppressed by genetic inhibition of Rab7 or the HOPS complex, demonstrating that FIG4 functions after endosome-to-lysosome fusion. Furthermore, disruption of the retromer complex, implicated in recycling from the lysosome to Golgi, does not lead to similar phenotypes as Fig4, suggesting that the lysosomal defects are not due to compromised retromer-mediated recycling of endolysosomal membranes. These data show that FIG4 plays a critical noncatalytic function in maintaining lysosomal membrane homeostasis, and that this function is disrupted by mutations that cause CMT4J and YVS.


Subject(s)
Flavoproteins/genetics , Lysosomes/pathology , Phosphoric Monoester Hydrolases/genetics , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Drosophila , Drosophila Proteins/metabolism , Endosomes/enzymology , Endosomes/genetics , Flavoproteins/metabolism , Homeostasis , Lysosomes/enzymology , Lysosomes/genetics , Mutation , Neurons/enzymology , Phenotype , Phosphoric Monoester Hydrolases/metabolism
16.
PLoS Biol ; 13(9): e1002258, 2015.
Article in English | MEDLINE | ID: mdl-26406915

ABSTRACT

The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)-axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease.


Subject(s)
Cell Adhesion Molecules/metabolism , Charcot-Marie-Tooth Disease/genetics , Myelin P0 Protein/genetics , Myelin Sheath/physiology , Nerve Growth Factors/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Charcot-Marie-Tooth Disease/enzymology , Gene Knockout Techniques , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice
17.
PLoS Biol ; 13(9): e1002259, 2015.
Article in English | MEDLINE | ID: mdl-26407016

ABSTRACT

The protein P0 has long been known to play a crucial role in holding together the myelin sheath that insulates peripheral nerves. A new study reveals that P0 is also important for organizing the nodes of Ranvier that occupy the gaps in the insulation. Read the Research Article.


Subject(s)
Cell Adhesion Molecules/metabolism , Charcot-Marie-Tooth Disease/genetics , Myelin P0 Protein/genetics , Myelin Sheath/physiology , Nerve Growth Factors/metabolism , Animals , Charcot-Marie-Tooth Disease/enzymology , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice
18.
Ukr Biochem J ; 87(6): 142-53, 2015.
Article in Ukrainian | MEDLINE | ID: mdl-27025069

ABSTRACT

The computational structural models of human tyrosyl-tRNA synthetase and its mutant form G41R (Charcot-Marie-Tooth associated) were constructed, while their whole structural coordinates are still unknown. Grid-services of MolDynGrid Virtual Laboratory and Ukrainian National Grid-infrastructure were used for molecular dynamics (MD) simulations. The analyses of trajectories of MD simulations have shown the ß-sheet formation in region Lys147 - Glu157 between H9 and H10 helices (CP1 insertion of Rossman fold) for G41R mutant.


Subject(s)
Molecular Dynamics Simulation , Mutation , Tyrosine-tRNA Ligase/chemistry , Amino Acid Sequence , Charcot-Marie-Tooth Disease/enzymology , Humans , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary
19.
J Biol Chem ; 289(29): 20359-69, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24898252

ABSTRACT

Aminoacyl-tRNA synthetases are an ancient enzyme family that specifically charges tRNA molecules with cognate amino acids for protein synthesis. Glycyl-tRNA synthetase (GlyRS) is one of the most intriguing aminoacyl-tRNA synthetases due to its divergent quaternary structure and abnormal charging properties. In the past decade, mutations of human GlyRS (hGlyRS) were also found to be associated with Charcot-Marie-Tooth disease. However, the mechanisms of traditional and alternative functions of hGlyRS are poorly understood due to a lack of studies at the molecular basis. In this study we report crystal structures of wild type and mutant hGlyRS in complex with tRNA and with small substrates and describe the molecular details of enzymatic recognition of the key tRNA identity elements in the acceptor stem and the anticodon loop. The cocrystal structures suggest that insertions 1 and 3 work together with the active site in a cooperative manner to facilitate efficient substrate binding. Both the enzyme and tRNA molecules undergo significant conformational changes during glycylation. A working model of multiple conformations for hGlyRS catalysis is proposed based on the crystallographic and biochemical studies. This study provides insights into the catalytic pathway of hGlyRS and may also contribute to our understanding of Charcot-Marie-Tooth disease.


Subject(s)
Glycine-tRNA Ligase/chemistry , RNA, Transfer, Gly/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain/genetics , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Crystallography, X-Ray , Glycine-tRNA Ligase/genetics , Glycine-tRNA Ligase/metabolism , Glycosylation , HeLa Cells , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Models, Molecular , Mutagenesis, Insertional , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleic Acid Conformation , Protein Conformation , RNA, Transfer, Gly/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...