Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Exp Eye Res ; 233: 109542, 2023 08.
Article in English | MEDLINE | ID: mdl-37331647

ABSTRACT

Retinoblastoma (Rb) is a rare malignant disorder affecting the developing retina of children under the age of five. Chemotherapeutic agents used for treating Rb have been associated with defects of the retinal pigment epithelium (RPE), such as hyperplasia, gliosis, and mottling. Herein, we have developed two pluripotent stem cell (PSC)-RPE models to assess the cytotoxicity of known Rb chemotherapeutics such as Melphalan, Topotecan and TW-37. Our findings demonstrate that these drugs alter the RPE by decreasing the monolayer barrier's trans-epithelial resistance and affecting the cells' phagocytic activity. Transcriptional analyses demonstrate an altered expression of genes involved in melanin and retinol processing, tight junction and apical-basal polarity pathways in both models. When applied within the clinical range, none of the drug treatments caused significant cytotoxic effects, changes to the apical-basal polarity, tight junction network or cell cycle. Together, our results demonstrate that although the most commonly used Rb chemotherapeutic drugs do not cause cytotoxicity in RPE, their application in vitro leads to compromised phagocytosis and strength of the barrier function, in addition to changes in gene expression that could alter the visual cycle in vivo. Our data demonstrate that widely used Rb chemotherapeutic drugs can have a deleterious impact on RPE cells and thus great care has to be exercised with regard to their delivery so the adjacent healthy RPE is not damaged during the course of tumor eradication.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Child , Humans , Retinal Pigment Epithelium/metabolism , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retina , Retinal Neoplasms/drug therapy , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Gene Expression , Cell Differentiation
2.
J Biol Chem ; 292(11): 4519-4532, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28082677

ABSTRACT

Mitochondrial gene expression is a fundamental process that is largely dependent on nuclear-encoded proteins. Several steps of mitochondrial RNA processing and maturation, including RNA post-transcriptional modification, appear to be spatially organized into distinct foci, which we have previously termed mitochondrial RNA granules (MRGs). Although an increasing number of proteins have been localized to MRGs, a comprehensive analysis of the proteome of these structures is still lacking. Here, we have applied a microscopy-based approach that has allowed us to identify novel components of the MRG proteome. Among these, we have focused our attention on RPUSD4, an uncharacterized mitochondrial putative pseudouridine synthase. We show that RPUSD4 depletion leads to a severe reduction of the steady-state level of the 16S mitochondrial (mt) rRNA with defects in the biogenesis of the mitoribosome large subunit and consequently in mitochondrial translation. We report that RPUSD4 binds 16S mt-rRNA, mt-tRNAMet, and mt-tRNAPhe, and we demonstrate that it is responsible for pseudouridylation of the latter. These data provide new insights into the relevance of RNA pseudouridylation in mitochondrial gene expression.


Subject(s)
Intramolecular Transferases/metabolism , RNA/metabolism , Cell Line , Humans , Intramolecular Transferases/analysis , Intramolecular Transferases/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Transport , RNA Interference , RNA, Mitochondrial , RNA, Ribosomal, 16S/metabolism , RNA, Small Interfering/genetics , RNA, Transfer, Met/metabolism , RNA, Transfer, Phe/metabolism
3.
Biochem J ; 474(13): 2145-2158, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28512204

ABSTRACT

Accurate assembly and maturation of human mitochondrial ribosomes is essential for synthesis of the 13 polypeptides encoded by the mitochondrial genome. This process requires the correct integration of 80 proteins, 1 mt (mitochondrial)-tRNA and 2 mt-rRNA species, the latter being post-transcriptionally modified at many sites. Here, we report that human ribosome-binding factor A (RBFA) is a mitochondrial RNA-binding protein that exerts crucial roles in mitoribosome biogenesis. Unlike its bacterial orthologue, RBFA associates mainly with helices 44 and 45 of the 12S rRNA in the mitoribosomal small subunit to promote dimethylation of two highly conserved consecutive adenines. Characterization of RBFA-depleted cells indicates that this dimethylation is not a prerequisite for assembly of the small ribosomal subunit. However, the RBFA-facilitated modification is necessary for completing mt-rRNA maturation and regulating association of the small and large subunits to form a functional monosome implicating RBFA in the quality control of mitoribosome formation.


Subject(s)
Escherichia coli Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/metabolism , RNA, Ribosomal/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Amino Acid Sequence , Escherichia coli Proteins/genetics , HEK293 Cells , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , RNA, Ribosomal/genetics , RNA-Binding Proteins/genetics , Ribosomal Proteins/genetics , Sequence Homology, Amino Acid
4.
Nucleic Acids Res ; 44(14): 6868-82, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27353330

ABSTRACT

LRPPRC is a protein that has attracted interest both for its role in post-transcriptional regulation of mitochondrial gene expression and more recently because numerous mutated variants have been characterized as causing severe infantile mitochondrial neurodegeneration. LRPPRC belongs to the pentatricopeptide repeat (PPR) protein family, originally defined by their RNA binding capacity, and forms a complex with SLIRP that harbours an RNA recognition motif (RRM) domain. We show here that LRPPRC displays a broad and strong RNA binding capacity in vitro in contrast to SLIRP that associates only weakly with RNA. The LRPPRC-SLIRP complex comprises a hetero-dimer via interactions by polar amino acids in the single RRM domain of SLIRP and three neighbouring PPR motifs in the second quarter of LRPPRC, which critically contribute to the LRPPRC-SLIRP binding interface to enhance its stability. Unexpectedly, specific amino acids at this interface are located within the PPRs of LRPPRC at positions predicted to interact with RNA and within the RNP1 motif of SLIRP's RRM domain. Our findings thus unexpectedly establish that despite the prediction that these residues in LRPPRC and SLIRP should bind RNA, they are instead used to facilitate protein-protein interactions, enabling the formation of a stable complex between these two proteins.


Subject(s)
Neoplasm Proteins/metabolism , RNA Recognition Motif , RNA-Binding Proteins/metabolism , Repetitive Sequences, Amino Acid , Amino Acid Sequence , Amino Acids/genetics , Conserved Sequence , Cross-Linking Reagents/metabolism , HEK293 Cells , Humans , Models, Biological , Mutation/genetics , Neoplasm Proteins/chemistry , Protein Binding , Protein Multimerization , Protein Stability , RNA/metabolism , RNA-Binding Proteins/chemistry
5.
Biochim Biophys Acta ; 1832(8): 1304-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23603806

ABSTRACT

Multiple respiratory chain deficiencies represent a common cause of mitochondrial diseases and are associated with a wide range of clinical symptoms. We report a subject, born to consanguineous parents, with growth retardation and neurological deterioration. Multiple respiratory chain deficiency was found in muscle and fibroblasts of the subject as well as abnormal assembly of complexes I and IV. A microsatellite genotyping of the family members detected only one region of homozygosity on chromosome 17q24.2-q25.3 in which we focused our attention to genes involved in mitochondrial translation. We sequenced MRPL12, encoding the mitochondrial ribosomal protein L12 and identified a c.542C>T transition in exon 5 changing a highly conserved alanine into a valine (p.Ala181Val). This mutation resulted in a decreased steady-state level of MRPL12 protein, with altered integration into the large ribosomal subunit. Moreover, an overall mitochondrial translation defect was observed in the subject's fibroblasts with a significant reduction of synthesis of COXI, COXII and COXIII subunits. Modeling of MRPL12 shows Ala181 positioned in a helix potentially involved in an interface of interaction suggesting that the p.Ala181Val change might be predicted to alter interactions with the elongation factors. These results contrast with the eubacterial orthologues of human MRPL12, where L7/L12 proteins do not appear to have a selective effect on translation. Therefore, analysis of the mutated version found in the subject presented here suggests that the mammalian protein does not function in an entirely analogous manner to the eubacterial L7/L12 equivalent.


Subject(s)
Cell Cycle Proteins/genetics , Growth Disorders/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mutation , Nervous System Diseases/genetics , Nuclear Proteins/genetics , Ribosomal Proteins/genetics , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cells, Cultured , Child, Preschool , Fibroblasts/metabolism , Genotype , Growth Disorders/metabolism , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nervous System Diseases/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptide Chain Elongation, Translational , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Sequence Alignment
6.
iScience ; 27(4): 109397, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38510120

ABSTRACT

Molecular information on the early stages of human retinal development remains scarce due to limitations in obtaining early human eye samples. Pluripotent stem cell-derived retinal organoids (ROs) provide an unprecedented opportunity for studying early retinogenesis. Using a combination of single cell RNA-seq and spatial transcriptomics we present for the first-time a single cell spatiotemporal transcriptome of RO development. Our data demonstrate that ROs recapitulate key events of retinogenesis including optic vesicle/cup formation, presence of a putative ciliary margin zone, emergence of retinal progenitor cells and their orderly differentiation to retinal neurons. Combining the scRNA- with scATAC-seq data, we were able to reveal cell-type specific transcription factor binding motifs on accessible chromatin at each stage of organoid development, and to show that chromatin accessibility is highly correlated to the developing human retina, but with some differences in the temporal emergence and abundance of some of the retinal neurons.

7.
Nat Commun ; 15(1): 3567, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670973

ABSTRACT

The emergence of retinal progenitor cells and differentiation to various retinal cell types represent fundamental processes during retinal development. Herein, we provide a comprehensive single cell characterisation of transcriptional and chromatin accessibility changes that underline retinal progenitor cell specification and differentiation over the course of human retinal development up to midgestation. Our lineage trajectory data demonstrate the presence of early retinal progenitors, which transit to late, and further to transient neurogenic progenitors, that give rise to all the retinal neurons. Combining single cell RNA-Seq with spatial transcriptomics of early eye samples, we demonstrate the transient presence of early retinal progenitors in the ciliary margin zone with decreasing occurrence from 8 post-conception week of human development. In retinal progenitor cells, we identified a significant enrichment for transcriptional enhanced associate domain transcription factor binding motifs, which when inhibited led to loss of cycling progenitors and retinal identity in pluripotent stem cell derived organoids.


Subject(s)
Cell Differentiation , Retina , Single-Cell Analysis , Stem Cells , Humans , Single-Cell Analysis/methods , Retina/cytology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Organoids/metabolism , Organoids/cytology , Gene Expression Regulation, Developmental , Chromatin/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA-Seq , Cell Lineage , Transcriptome
8.
Nucleic Acids Res ; 39(18): 8092-104, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21724598

ABSTRACT

Tra2ß regulates a number of splicing switches including activation of the human testis-specific exon HIPK3-T in the Homeodomain Interacting Protein Kinase 3 gene. By testing HIPK3-T exons of different intrinsic strengths, we found Tra2ß most efficiently activated splicing inclusion of intrinsically weak exons, although these were spliced at a lower overall level. Both the RRM and N-terminal RS-rich region of Tra2ß were required for splicing activation. Bioinformatic searches for splicing enhancers and repressors mapped four physically distinct exonic splicing enhancers (ESEs) within HIPK3-T, each containing the known Tra2ß AGAA-rich binding site. Surprisingly disruption of each single ESE prevented Tra2ß-mediated activation, although single mutated exons could still bind Tra2ß protein by gel shifts and functional splicing analyses. Titration experiments indicate an additive model of HIPK3-T splicing activation, requiring availability of an array of four distinct ESEs to enable splicing activation. To enable this efficient Tra2ß-mediated splicing switch to operate, a closely adjacent downstream and potentially competitive stronger 5'-splice site is actively repressed. Our data indicate that a novel arrangement of multiple mono-specific AGAA-rich ESEs coupled to a weak 5'-splice site functions as a responsive gauge. This gauge monitors changes in the specific nuclear concentration of the RNA binding protein Tra2ß, and co-ordinately regulates HIPK3-T exon splicing inclusion.


Subject(s)
Alternative Splicing , Intracellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Exons , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA Splice Sites , RNA-Binding Proteins/chemistry , Serine-Arginine Splicing Factors
9.
Stem Cells Transl Med ; 11(4): 415-433, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35325233

ABSTRACT

Retinoblastoma (Rb) is a childhood cancer of the developing retina, accounting for up to 17% of all tumors in infancy. To gain insights into the transcriptional events of cell state transitions during Rb development, we established 2 disease models via retinal organoid differentiation of a pRB (retinoblastoma protein)-depleted human embryonic stem cell line (RB1-null hESCs) and a pRB patient-specific induced pluripotent (iPSC) line harboring a RB1 biallelic mutation (c.2082delC). Both models were characterized by pRB depletion and accumulation of retinal progenitor cells at the expense of amacrine, horizontal and retinal ganglion cells, which suggests an important role for pRB in differentiation of these cell lineages. Importantly, a significant increase in the fraction of proliferating cone precursors (RXRγ+Ki67+) was observed in both pRB-depleted organoid models, which were defined as Rb-like clusters by single-cell RNA-Seq analysis. The pRB-depleted retinal organoids displayed similar features to Rb tumors, including mitochondrial cristae aberrations and rosette-like structures, and were able to undergo cell growth in an anchorage-independent manner, indicative of cell transformation in vitro. In both models, the Rb cones expressed retinal ganglion and horizontal cell markers, a novel finding, which could help to better characterize these tumors with possible therapeutic implications. Application of Melphalan, Topotecan, and TW-37 led to a significant reduction in the fraction of Rb proliferating cone precursors, validating the suitability of these in vitro models for testing novel therapeutics for Rb.


Subject(s)
Pluripotent Stem Cells , Retinal Neoplasms , Retinoblastoma , Cell Differentiation , Child , Humans , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Retina/metabolism , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma/pathology , Retinoblastoma Protein/genetics
10.
Biochem J ; 430(3): 551-8, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20604745

ABSTRACT

The bacterial Ras-like protein Era has been reported previously to bind 16S rRNA within the 30S ribosomal subunit and to play a crucial role in ribosome assembly. An orthologue of this essential GTPase ERAL1 (Era G-protein-like 1) exists in higher eukaryotes and although its exact molecular function and cellular localization is unknown, its absence has been linked to apoptosis. In the present study we show that human ERAL1 is a mitochondrial protein important for the formation of the 28S small mitoribosomal subunit. We also show that ERAL1 binds in vivo to the rRNA component of the small subunit [12S mt (mitochondrial)-rRNA]. Bacterial Era associates with a 3' unstructured nonanucleotide immediately downstream of the terminal stem-loop (helix 45) of 16S rRNA. This site contains an AUCA sequence highly conserved across all domains of life, immediately upstream of the anti-Shine-Dalgarno sequence, which is conserved in bacteria. Strikingly, this entire region is absent from 12S mt-rRNA. We have mapped the ERAL1-binding site to a 33 nucleotide section delineating the 3' terminal stem-loop region of 12S mt-rRNA. This loop contains two adenine residues that are reported to be dimethylated on mitoribosome maturation. Furthermore, and also in contrast with the bacterial orthologue, loss of ERAL1 leads to rapid decay of nascent 12S mt-rRNA, consistent with a role as a mitochondrial RNA chaperone. Finally, whereas depletion of ERAL1 leads to apoptosis, cell death occurs prior to any appreciable loss of mitochondrial protein synthesis or reduction in the stability of mitochondrial mRNA.


Subject(s)
GTP-Binding Proteins/metabolism , RNA, Ribosomal, 28S/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Blotting, Northern , Cell Line , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Mitochondrial Proteins/biosynthesis , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , RNA/genetics , RNA Interference , RNA Stability , RNA, Mitochondrial , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal, 28S/genetics , RNA-Binding Proteins/genetics
11.
Biochem Soc Trans ; 38(6): 1523-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118119

ABSTRACT

Mitochondria are ubiquitous and essential organelles for all nucleated cells of higher eukaryotes. They contain their own genome [mtDNA (mitochondrial DNA)], and this autosomally replicating extranuclear DNA encodes a complement of genes whose products are required to couple oxidative phosphorylation. Sequencing of this human mtDNA more than 20 years ago revealed unusual features that included a modified codon usage. Specific deviations from the standard genetic code include recoding of the conventional UGA stop to tryptophan, and, strikingly, the apparent recoding of two arginine triplets (AGA and AGG) to termination signals. This latter reassignment was made because of the absence of cognate mtDNA-encoded tRNAs, and a lack of tRNAs imported from the cytosol. Each of these codons only occurs once and, in both cases, at the very end of an open reading frame. The presence of both AGA and AGG is rarely found in other mammals, and the molecular mechanism that has driven the change from encoding arginine to dictating a translational stop has posed a challenging conundrum. Mitochondria from the majority of other organisms studied use only UAA and UAG, leaving the intriguing question of why human organelles appear to have added the complication of a further two stop codons, AGA and AGG, or have they? In the present review, we report recent data to show that mammalian mitochondria can utilize a -1 frameshift such that only the standard UAA and UAG stop codons are required to terminate the synthesis of all 13 polypeptides.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Peptide Chain Termination, Translational , Protein Biosynthesis , Ribosomes/metabolism , Codon , DNA, Mitochondrial/genetics , Genetic Code , Humans , Ribosomes/genetics
12.
Clin Cancer Res ; 15(4): 1241-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19174487

ABSTRACT

PURPOSE: High-risk neuroblastoma is characterized by poor survival rates, and the development of improved therapeutic approaches is a priority. Temozolomide and topotecan show promising clinical activity against neuroblastoma. Poly(ADP-ribose) polymerase-1 (PARP-1) promotes DNA repair and cell survival following genotoxic insult; we postulated that its inhibition may enhance the efficacy of these DNA-damaging drugs in pediatric cancers. EXPERIMENTAL DESIGN: We evaluated the chemosensitizing properties of the PARP inhibitor AG014699 (Pfizer, Inc.) in combination with temozolomide and topotecan, against human neuroblastoma cells and xenografts, alongside associated pharmacologic and toxicologic indices. RESULTS: Addition of PARP-inhibitory concentrations of AG014699 significantly potentiated growth inhibition by both topotecan (1.5- to 2.3-fold) and temozolomide (3- to 10-fold) in vitro, with equivalent effects confirmed in clonogenic assays. In two independent in vivo models (NB1691 and SHSY5Y xenografts), temozolomide caused a xenograft growth delay, which was enhanced by co-administration of AG014699, and resulted in complete and sustained tumor regression in the majority (6 of 10; 60%) of cases. Evidence of enhanced growth delay by topotecan/AG014699 co-administration was observed in NB1691 xenografts. AG014699 metabolites distributed rapidly into the plasma (Cmax, 1.2-1.9 nmol/L at 30 min) and accumulated in xenograft tissues (Cmax, 1-2 micromol/L at 120 min), associated with a sustained suppression of PARP-1 enzyme activity. Doses of AG014699 required for potentiation were not toxic per se. CONCLUSIONS: These data show enhancement of temozolomide and topotecan efficacy by PARP inhibition in neuroblastoma. Coupled with the acceptable pharmacokinetic, pharmacodynamic, and toxicity profiles of AG014699, our findings provide strong rationale for investigation of PARP inhibitors in pediatric early clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Neuroblastoma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Topotecan/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Drug Synergism , Humans , Mice , Neuroblastoma/pathology , Poly (ADP-Ribose) Polymerase-1 , Temozolomide , Xenograft Model Antitumor Assays
13.
FEBS Lett ; 588(15): 2496-503, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24911204

ABSTRACT

Mitochondrial protein synthesis is essential for all mammals, being responsible for providing key components of the oxidative phosphorylation complexes. Although only thirteen different polypeptides are made, the molecular details of this deceptively simple process remain incomplete. Central to this process is a non-canonical ribosome, the mitoribosome, which has evolved to address its unique mandate. In this review, we integrate the current understanding of the molecular aspects of mitochondrial translation with recent advances in structural biology. We identify numerous key questions that we will need to answer if we are to increase our knowledge of the molecular mechanisms underlying mitochondrial protein synthesis.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Animals , Humans , Mitochondrial Proteins/genetics , RNA/genetics , RNA/metabolism , RNA, Mitochondrial , Ribosomal Proteins/genetics , Ribosomes/metabolism
14.
EMBO Mol Med ; 6(2): 183-93, 2014 02.
Article in English | MEDLINE | ID: mdl-24413189

ABSTRACT

Disorders of the mitochondrial genome cause a wide spectrum of disease, these present mainly as neurological and/or muscle related pathologies. Due to the intractability of the human mitochondrial genome there are currently no effective treatments for these disorders. The majority of the pathogenic mutations lie in the genes encoding mitochondrial tRNAs. Consequently, the biochemical deficiency is due to mitochondrial protein synthesis defects, which manifest as aberrant cellular respiration and ATP synthesis. It has previously been reported that overexpression of mitochondrial aminoacyl tRNA synthetases has been effective, in cell lines, at partially suppressing the defects resulting from mutations in their cognate mt-tRNAs. We now show that leucyl tRNA synthetase is able to partially rescue defects caused by mutations in non-cognate mt-tRNAs. Further, a C terminal peptide alone can enter mitochondria and interact with the same spectrum of mt-tRNAs as the entire synthetase, in intact cells. These data support the possibility that a small peptide could correct at least the biochemical defect associated with many mt-tRNA mutations, inferring a novel therapy for these disorders.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Mitochondria/genetics , Mutation/genetics , RNA, Transfer, Leu/genetics , Suppression, Genetic , Amino Acyl-tRNA Synthetases/chemistry , Cell Proliferation , Humans , Mitochondria/enzymology , Oxidative Phosphorylation , Phenotype , Protein Binding , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL