Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 311
Filter
1.
Proc Natl Acad Sci U S A ; 121(35): e2409628121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39163341

ABSTRACT

Protein kinase Gcn2 attenuates protein synthesis in response to amino acid starvation while stimulating translation of a transcriptional activator of amino acid biosynthesis. Gcn2 activation requires a domain related to histidyl-tRNA synthetase (HisRS), the enzyme that aminoacylates tRNAHis. While evidence suggests that deacylated tRNA binds the HisRS domain for kinase activation, ribosomal P-stalk proteins have been implicated as alternative activating ligands on stalled ribosomes. We report crystal structures of the HisRS domain of Chaetomium thermophilum Gcn2 that reveal structural mimicry of both catalytic (CD) and anticodon-binding (ABD) domains, which in authentic HisRS bind the acceptor stem and anticodon loop of tRNAHis. Elements for forming histidyl adenylate and aminoacylation are lacking, suggesting that Gcn2HisRS was repurposed for kinase activation, consistent with mutations in the CD that dysregulate yeast Gcn2 function. Substituting conserved ABD residues well positioned to contact the anticodon loop or that form a conserved ABD-CD interface impairs Gcn2 function in starved cells. Mimicry in Gcn2HisRS of two highly conserved structural domains for binding both ends of tRNA-each crucial for Gcn2 function-supports that deacylated tRNAs activate Gcn2 and exemplifies how a metabolic enzyme is repurposed to host new local structures and sequences that confer a novel regulatory function.


Subject(s)
Chaetomium , Histidine-tRNA Ligase , Protein Serine-Threonine Kinases , Chaetomium/enzymology , Chaetomium/genetics , Chaetomium/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Histidine-tRNA Ligase/metabolism , Histidine-tRNA Ligase/chemistry , Histidine-tRNA Ligase/genetics , Stress, Physiological , Fungal Proteins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Crystallography, X-Ray , Models, Molecular , Protein Domains , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry
2.
Front Immunol ; 15: 1399451, 2024.
Article in English | MEDLINE | ID: mdl-38895121

ABSTRACT

Introduction: Anti-SSA antibodies target two unrelated proteins, Ro52 (E3 ligase) and Ro60 (RNA binding protein). Previous studies indicate that anti-Ro52 antibodies are frequently associated with various myositis-specific autoantibodies (MSAs)-including anti-tRNA synthetase antibodies-and that the coexistence of MSAs and anti-Ro52 antibodies may portend worse clinical outcomes. Although not well-described in the setting of myositis, work from our animal model of HRS (histidyl-tRNA synthetase)-induced myositis suggests that anti-Ro60 antibodies may also be linked to specific MSAs such as anti-HRS/Jo-1. We therefore aimed to demonstrate the prevalence and clinical characteristics of Ro52 and Ro60 antibody positivity in patients possessing Jo-1 antibodies. Methods: To establish the immunological link between anti-synthetase, anti-Ro52, and anti-Ro60 antibodies, we evaluated the relative titers of these antibodies in blood and bronchoalveolar lavage fluid (BALF) of mice following immunization with HRS/Jo-1. In parallel, we used ELISA-based approaches to assess sera from 177 anti-Jo1 antibody-positive patients for the presence of anti-Ro52 and/or anti-Ro60 antibodies. We then determined statistical associations between co-existing anti-Jo-1, anti-Ro52, and/or anti-Ro60 antibodies and clinical manifestations associated with the anti-synthetase syndrome. Results: Mice immunized with HRS had higher levels of anti-Ro52 and anti-Ro60 antibodies in serum and BALF than PBS-immunized mice. In 177 anti-Jo-1 antibody-positive patients, the prevalence of anti-Ro52 and anti-Ro60 antibodies was 36% and 15%, respectively. The frequency of dry eye/dry mouth, interstitial pneumonia, and pulmonary events over time differed between patients with various combinations of anti-Ro52 and anti-Ro60 antibodies. While anti-Ro52 antibodies generally correlated with statistically significant increases in each of these clinical manifestations, the presence of Ro60 antibodies alone was associated with decreased frequency of ILD. Discussion: Anti-Ro52 and/or anti-Ro60 antibodies are often co-expressed with anti-Jo1 antibodies, defining clinical subsets with different disease course/outcomes.


Subject(s)
Myositis , Ribonucleoproteins , Animals , Humans , Ribonucleoproteins/immunology , Myositis/immunology , Female , Mice , Male , Middle Aged , Antibodies, Antinuclear/immunology , Antibodies, Antinuclear/blood , Autoantibodies/blood , Autoantibodies/immunology , Aged , Adult , Histidine-tRNA Ligase/immunology , Disease Models, Animal , Autoantigens/immunology , RNA, Small Cytoplasmic
3.
Front Immunol ; 14: 1238221, 2023.
Article in English | MEDLINE | ID: mdl-37809058

ABSTRACT

Introduction: Previous work in humans has demonstrated that both innate and adaptive immune signaling pathways contribute to the pathogenesis of idiopathic inflammatory myopathy (IIM), a systemic autoimmune disease targeting muscle as well as extra-muscular organs. To better define interactive signaling networks in IIM, we characterized the cellular phenotype and transcriptomic profiles of muscle-infiltrating cells in our established murine model of histidyl-tRNA synthetase (HRS)-induced myositis. Methods: Myositis was induced in wild type (WT) and various congenic/mutant strains of C57BL/6 mice through intramuscular immunization with recombinant HRS. Histopathological, immunohistochemical, flow cytometric, and transcriptomic assessments were used to characterize the functional relationship between muscle-infiltrating cell populations in these strains lacking different components of innate and/or adaptive immune signaling. Results: RAG1 KO mice developed markedly reduced muscle inflammation relative to WT mice, demonstrating a key requirement for T cells in driving HRS-induced myositis. While the reduction of mononuclear cell infiltrates in CD4-Cre.MyD88fl/fl conditional knockout mice and OT-II TCR transgenic mice highlighted roles for both innate and TCR-mediated/adaptive immune signaling in T cells, diminished inflammation in Lyz2-Cre.MyD88fl/fl conditional knockout mice underscored the importance of macrophage/myeloid cell populations in supporting T cell infiltration. Single cell RNA sequencing-based clustering of muscle-infiltrating subpopulations and associated pathway analyses showed that perturbations of T cell signaling/function alter the distribution and phenotype of macrophages, fibroblasts, and other non-lymphoid cell populations contributing to HRS-induced myositis. Discussion: Overall, HRS-induced myositis reflects the complex interplay between multiple cell types that collectively drive a TH1-predominant, pro-inflammatory tissue phenotype requiring antigen-mediated activation of both MyD88- and TCR-dependent T cell signaling pathways.


Subject(s)
Histidine-tRNA Ligase , Myositis , Humans , Mice , Animals , T-Lymphocytes , Mice, Inbred C57BL , Adaptive Immunity , Macrophages , Inflammation , Mice, Knockout , Receptors, Antigen, T-Cell
4.
Acta Trop ; 241: 106867, 2023 May.
Article in English | MEDLINE | ID: mdl-36878386

ABSTRACT

Visceral leishmaniasis is neglected tropical protozoan disease caused by Leishmania donovani and are associated with high fatality rate in developing countries since prophylactic vaccines are not available. In the present study, we evaluated the immunomodulatory potential of L. donovani histidyl-tRNA synthetase (LdHisRS) and predicted the epitopes using immunoinformatic tools. Histidyl-tRNA synthetase (HisRS) is a class IIa aminoacyl t-RNA synthetase enzyme (aaRS) required for histidine incorporation into proteins during protein synthesis. The recombinant LdHisRS protein (rLdHisRS) was expressed in E coli BL-21cells, and its immunomodulatory role was assessed in J774A.1 murine macrophage and in BALB/c mice, respectively. LdHisRS specifically stimulated and triggered enhance cell proliferation, nitric oxide release and IFN-γ (70%; P < 0.001), and IL-12 (55.37%; P < 0.05) cytokine release in vitro, whereas BALB/c mice immunized with rLdHisRS show higher NO release (80.95%; P<0.001), higher levels of Th1 cytokines IFN-γ (14%; P < 0.05), TNF-α (34.93%; P < 0.001), and IL-12 (28.49%; P < 0.001) and robust IgG (p<0.001) and IgG2a (p<0.001) production. We also identified 20 Helper T-lymphocytes (HTLs), 30 cytotoxic T lymphocytes (CTLs), and 18 B-cell epitopes from HisRS protein of L. donovani. All these epitopes can be further used to make a multiepitope vaccine against L. donovani.


Subject(s)
Leishmania donovani , Leishmaniasis Vaccines , Leishmaniasis, Visceral , Vaccines , Animals , Mice , Leishmania donovani/genetics , Leishmaniasis, Visceral/prevention & control , Histidine-tRNA Ligase , Escherichia coli , Th1 Cells , Cytokines , Interleukin-12 , Recombinant Proteins/genetics , Epitopes , Mice, Inbred BALB C , Protozoan Proteins/genetics , Antigens, Protozoan/genetics
5.
Genes (Basel) ; 14(2)2023 01 18.
Article in English | MEDLINE | ID: mdl-36833180

ABSTRACT

Histidyl-tRNA synthetase (HARS) ligates histidine to its cognate transfer RNA (tRNAHis). Mutations in HARS cause the human genetic disorders Usher syndrome type 3B (USH3B) and Charcot-Marie-Tooth syndrome type 2W (CMT2W). Treatment for these diseases remains symptomatic, and no disease specific treatments are currently available. Mutations in HARS can lead to destabilization of the enzyme, reduced aminoacylation, and decreased histidine incorporation into the proteome. Other mutations lead to a toxic gain-of-function and mistranslation of non-cognate amino acids in response to histidine codons, which can be rescued by histidine supplementation in vitro. We discuss recent advances in characterizing HARS mutations and potential applications of amino acid and tRNA therapy for future gene and allele specific therapy.


Subject(s)
Charcot-Marie-Tooth Disease , Histidine , Humans , Histidine/genetics , Histidine/metabolism , Mutation , Histidine-tRNA Ligase/genetics , Charcot-Marie-Tooth Disease/genetics , Aminoacylation
6.
Nucleic Acids Res ; 50(17): 10015-10025, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36107775

ABSTRACT

tRNAHis guanylyltransferase (Thg1) catalyzes the 3'-5' incorporation of guanosine into position -1 (G-1) of tRNAHis. G-1 is unique to tRNAHis and is crucial for recognition by histidyl-tRNA synthetase (HisRS). Yeast Thg1 requires ATP for G-1 addition to tRNAHis opposite A73, whereas archaeal Thg1 requires either ATP or GTP for G-1 addition to tRNAHis opposite C73. Paradoxically, human Thg1 (HsThg1) can add G-1 to tRNAsHis with A73 (cytoplasmic) and C73 (mitochondrial). As N73 is immediately followed by a CCA end (positions 74-76), how HsThg1 prevents successive 3'-5' incorporation of G-1/G-2/G-3 into mitochondrial tRNAHis (tRNAmHis) through a template-dependent mechanism remains a puzzle. We showed herein that mature native human tRNAmHis indeed contains only G-1. ATP was absolutely required for G-1 addition to tRNAmHis by HsThg1. Although HsThg1 could incorporate more than one GTP into tRNAmHisin vitro, a single-GTP incorporation prevailed when the relative GTP level was low. Surprisingly, HsThg1 possessed a tRNA-inducible GTPase activity, which could be inhibited by ATP. Similar activity was found in other high-eukaryotic dual-functional Thg1 enzymes, but not in yeast Thg1. This study suggests that HsThg1 may downregulate the level of GTP through its GTPase activity to prevent multiple-GTP incorporation into tRNAmHis.


Subject(s)
Nucleotidyltransferases/metabolism , RNA, Transfer, His , Adenosine Triphosphate , GTP Phosphohydrolases/genetics , Guanosine , Guanosine Triphosphate/metabolism , Histidine-tRNA Ligase , Humans , RNA, Transfer , RNA, Transfer, His/genetics , RNA, Transfer, His/metabolism , Saccharomyces cerevisiae/metabolism
7.
Arthritis Res Ther ; 24(1): 62, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35236390

ABSTRACT

BACKGROUND: To address the reactivity and affinity against histidyl-transfer RNA synthetase (HisRS) autoantigen of anti-Jo1 autoantibodies from serum and bronchoalveolar lavage fluid (BALF) in patients with idiopathic inflammatory myopathies/anti-synthetase syndrome (IIM/ASSD). To investigate the associations between the reactivity profile and clinical data over time. METHODS: Samples and clinical data were obtained from (i) 25 anti-Jo1+ patients (19 sera with 16 longitudinal samples and 6 BALF/matching sera at diagnosis), (ii) 29 anti-Jo1- patients (25 sera and 4 BALF/matching sera at diagnosis), and (iii) 27 age/gender-matched healthy controls (24 sera and 3 BALF/matching sera). Reactivity towards HisRS full-length (HisRS-FL), three HisRS domains (WHEP, antigen binding domain (ABD), and catalytic domain (CD)), and the HisRS splice variant (SV) was tested. Anti-Jo1 IgG reactivity was evaluated by ELISA and western blot using IgG purified from serum by affinity chromatography. In paired serum-BALF, anti-Jo1 IgG and IgA reactivity was analyzed by ELISA. Autoantibody affinity was measured by surface plasmon resonance using IgG purified from sera. Correlations between autoantibody reactivity and clinical data were evaluated at diagnosis and longitudinally. RESULTS: Anti-Jo1 IgG from serum and BALF bound HisRS-FL, WHEP, and SV with high reactivity at the time of diagnosis and recognized both conformation-dependent and conformation-independent HisRS epitopes. Anti-HisRS-FL IgG displayed high affinity early in the disease. At the time of IIM/ASSD diagnosis, the highest autoantibody levels against HisRS-FL were found in patients ever developing interstitial lung disease (ILD) and arthritis, but with less skin involvement. Moreover, the reactivity of anti-WHEP IgG in BALF correlated with poor pulmonary function. Levels of autoantibodies against HisRS-FL, HisRS domains, and HisRS splice variant generally decreased over time. With some exceptions, longitudinal anti-HisRS-FL antibody levels changed in line with ILD activity. CONCLUSION: High levels and high-affinity anti-Jo1 autoantibodies towards HisRS-FL were found early in disease in sera and BALF. In combination with the correlation of anti-HisRS-FL antibody levels with ILD and ILD activity in longitudinal samples as well as of anti-WHEP IgG in BALF with poor pulmonary function, this supports the previously raised hypothesis that the lung might have a role in the immune reaction in anti-Jo1-positive patients.


Subject(s)
Lung Diseases, Interstitial , Myositis , Autoantibodies , Histidine-tRNA Ligase , Humans , Ligases
8.
J Phys Chem B ; 125(42): 11606-11616, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34648705

ABSTRACT

Catalytic fields representing the topology of the optimal molecular environment charge distribution that reduces the activation barrier have been used to examine alternative reaction variants and to determine the role of conserved catalytic residues for two consecutive reactions catalyzed by the same enzyme. Until now, most experimental and conventional top-down theoretical studies employing QM/MM or ONIOM methods have focused on the role of enzyme electric fields acting on broken bonds of reactants. In contrast, our bottom-up approach dealing with a small reactant and transition-state model allows the analysis of the opposite effects: how the catalytic field resulting from the charge redistribution during the enzyme reaction acts on conserved amino acid residues and contributes to the reduction of the activation barrier. This approach has been applied to the family of histidyl tRNA synthetases involved in the translation of the genetic code into the protein amino acid sequence. Activation energy changes related to conserved charged amino acid residues for 12 histidyl tRNA synthetases from different biological species allowed to compare on equal footing the catalytic residues involved in ATP aminoacylation and tRNA charging reactions and to analyze different reaction mechanisms proposed in the literature. A scan of the library of atomic multipoles for amino acid side-chain rotamers within the catalytic field pointed out the change in the Glu83 conformation as the critical catalytic effect, providing, at low computational cost, insight into the electrostatic preorganization of the enzyme catalytic site at a level of detail that has not yet been accessible in conventional experimental or theoretical methods. This opens the way for rational reverse biocatalyst design at a very limited computational cost without resorting to empirical methods.


Subject(s)
Histidine-tRNA Ligase , Aminoacylation , Catalysis , Catalytic Domain , Histidine-tRNA Ligase/metabolism , Static Electricity
9.
FEBS J ; 288(1): 91-94, 2021 01.
Article in English | MEDLINE | ID: mdl-32940403

ABSTRACT

The pathogenic mechanism of neuropathy-associated aminoacyl-tRNA synthetase (ARS) gene variants is poorly defined. Mullen et al. generate new models of pathogenic, dominant HARS1 mutations and show that they increase eIF2α phosphorylation and decrease protein translation in neurons. These results are consistent with a dominant-negative mechanism of ARS-mediated peripheral neuropathy. Comment on: https://doi.org/10.1111/febs.15449.


Subject(s)
Histidine-tRNA Ligase , Peripheral Nervous System Diseases , Animals , Mutation , Neuronal Outgrowth , Peripheral Nervous System Diseases/genetics , Protein Biosynthesis , Zebrafish
10.
Cell Mol Immunol ; 18(6): 1463-1475, 2021 06.
Article in English | MEDLINE | ID: mdl-31797905

ABSTRACT

His-tRNA synthetase (HARS) is targeted by autoantibodies in chronic and acute inflammatory anti-Jo-1-positive antisynthetase syndrome. The extensive activation and migration of immune cells into lung and muscle are associated with interstitial lung disease, myositis, and morbidity. It is unknown whether the sequestration of HARS is an epiphenomenon or plays a causal role in the disease. Here, we show that HARS circulates in healthy individuals, but it is largely undetectable in the serum of anti-Jo-1-positive antisynthetase syndrome patients. In cultured primary human skeletal muscle myoblasts (HSkMC), HARS is released in increasing amounts during their differentiation into myotubes. We further show that HARS regulates immune cell engagement and inhibits CD4+ and CD8+ T-cell activation. In mouse and rodent models of acute inflammatory diseases, HARS administration downregulates immune activation. In contrast, neutralization of extracellular HARS by high-titer antibody responses during tissue injury increases susceptibility to immune attack, similar to what is seen in humans with anti-Jo-1-positive disease. Collectively, these data suggest that extracellular HARS is homeostatic in normal subjects, and its sequestration contributes to the morbidity of the anti-Jo-1-positive antisynthetase syndrome.


Subject(s)
Histidine-tRNA Ligase/blood , Immunity , Organ Specificity , Animals , Autoantibodies/blood , Case-Control Studies , Cell Differentiation/drug effects , Disease Models, Animal , Female , Histidine-tRNA Ligase/immunology , Humans , Immunity/drug effects , Immunomodulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Lung/drug effects , Lung/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Middle Aged , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscles/drug effects , Muscles/pathology , Myositis/blood , Myositis/diagnostic imaging , Myositis/immunology , Organ Specificity/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tomography, X-Ray Computed
11.
FEBS J ; 288(1): 142-159, 2021 01.
Article in English | MEDLINE | ID: mdl-32543048

ABSTRACT

Charcot-Marie-Tooth disease (CMT) encompasses a set of genetically and clinically heterogeneous neuropathies characterized by length-dependent dysfunction of the peripheral nervous system. Mutations in over 80 diverse genes are associated with CMT, and aminoacyl-tRNA synthetases (ARS) constitute a large gene family implicated in the disease. Despite considerable efforts to elucidate the mechanistic link between ARS mutations and the CMT phenotype, the molecular basis of the pathology is unknown. In this work, we investigated the impact of three CMT-associated substitutions (V155G, Y330C, and R137Q) in the cytoplasmic histidyl-tRNA synthetase (HARS1) on neurite outgrowth and peripheral nervous system development. The model systems for this work included a nerve growth factor-stimulated neurite outgrowth model in rat pheochromocytoma cells (PC12), and a zebrafish line with GFP/red fluorescent protein reporters of sensory and motor neuron development. The expression of CMT-HARS1 mutations led to attenuation of protein synthesis and increased phosphorylation of eIF2α in PC12 cells and was accompanied by impaired neurite and axon outgrowth in both models. Notably, these effects were phenocopied by histidinol, a HARS1 inhibitor, and cycloheximide, a protein synthesis inhibitor. The mutant proteins also formed heterodimers with wild-type HARS1, raising the possibility that CMT-HARS1 mutations cause disease through a dominant-negative mechanism. Overall, these findings support the hypothesis that CMT-HARS1 alleles exert their toxic effect in a neuronal context, and lead to dysregulated protein synthesis. These studies demonstrate the value of zebrafish as a model for studying mutant alleles associated with CMT, and for characterizing the processes that lead to peripheral nervous system dysfunction.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Histidine-tRNA Ligase/genetics , Neuronal Outgrowth/genetics , Neurons/metabolism , Peripheral Nervous System/metabolism , Protein Biosynthesis , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cycloheximide/pharmacology , Disease Models, Animal , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histidine-tRNA Ligase/antagonists & inhibitors , Histidine-tRNA Ligase/metabolism , Histidinol/pharmacology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mutation , Neuronal Outgrowth/drug effects , Neurons/drug effects , Neurons/pathology , PC12 Cells , Peripheral Nervous System/pathology , Protein Multimerization , Rats , Zebrafish , Red Fluorescent Protein
12.
Adv Rheumatol ; 61: 12, 2021. tab
Article in English | LILACS | ID: biblio-1152748

ABSTRACT

Abstract Background: To assess the prevalence and clinical relevance of anti-Jo-1 autoantibodies in a representative sample of patients with definite dermatomyositis (DM). Methods: This retrospective cohort study took place from 2005 to 2020 and assessed 118 adult patients from a tertiary center who were diagnosed with definite DM. A commercial kit was used to detect anti-Jo-1 autoantibodies. Results: The presence of anti-Jo-1 autoantibodies was observed in 10 out of 118 (8.5%) patients with definite DM. The following variables were comparable between individuals with and without anti-Jo-1 autoantibodies: age at diagnosis, sex, ethnicity, disease duration, follow-up period, recurrence rate, complete clinical response, death rate, and cancer incidence. There was no difference in clinical features between groups, except for an increased prevalence of "mechanic's hands," joint involvement, and lung disease, as well as a reduced occurrence of skin findings in patients positive for anti-Jo-1 autoantibodies. No anti-Jo-1-positive patients went into remission; they required greater use of glucocorticoids and immunosuppressive drugs. Conclusions: Anti-Jo-1 positivity was found in 8.5% of patients with definite DM. This autoantibody was associated with an antisynthetase syndrome phenotype and might predict clinical outcomes in patients with definite DM.(AU)


Subject(s)
Humans , Adult , Autoantibodies/analysis , Dermatomyositis/physiopathology , Histidine-tRNA Ligase/blood , Retrospective Studies , Cohort Studies , Muscular Diseases/physiopathology
13.
MAbs ; 12(1): 1836718, 2020.
Article in English | MEDLINE | ID: mdl-33131414

ABSTRACT

The autoimmune disease known as Jo-1 positive anti-synthetase syndrome (ASS) is characterized by circulating antibody titers to histidyl-tRNA synthetase (HARS), which may play a role in modulating the non-canonical functions of HARS. Monoclonal antibodies to HARS were isolated by single-cell screening and sequencing from three Jo-1 positive ASS patients and shown to be of high affinity, covering diverse epitope space. The immune response was further characterized by repertoire sequencing from the most productive of the donor samples. In line with previous studies of autoimmune repertoires, these antibodies tended to have long complementarity-determining region H3 sequences with more positive-charged residues than average. Clones of interest were clustered into groups with related sequences, allowing us to observe different somatic mutations in related clones. We postulated that these had found alternate structural solutions for high affinity binding, but that mutations might be transferable between clones to further enhance binding affinity. Transfer of somatic mutations between antibodies within the same clonal group was able to enhance binding affinity in a number of cases, including beneficial transfer of a mutation from a lower affinity clone into one of higher affinity. Affinity enhancement was seen with mutation transfer both between related single-cell clones, and directly from related repertoire sequences. To our knowledge, this is the first demonstration of somatic hypermutation transfer from repertoire sequences to further mature in vivo derived antibodies, and represents an additional tool to aid in affinity maturation for the development of antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Autoantibodies/immunology , Immunologic Techniques/methods , Myositis/immunology , Antibodies, Monoclonal/isolation & purification , Autoantibodies/isolation & purification , Histidine-tRNA Ligase/immunology , Humans , Somatic Hypermutation, Immunoglobulin/immunology
14.
Int J Mol Sci ; 21(20)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096801

ABSTRACT

Reprogramming of the genetic code system is limited by the difficulty in creating new tRNA structures. Here, I developed translationally active tRNA variants tagged with a small hairpin RNA aptamer, using Escherichia coli reporter assay systems. As the tRNA chassis for engineering, I employed amber suppressor variants of allo-tRNAs having the 9/3 composition of the 12-base pair amino-acid acceptor branch as well as a long variable arm (V-arm). Although their V-arm is a strong binding site for seryl-tRNA synthetase (SerRS), insertion of a bulge nucleotide in the V-arm stem region prevented allo-tRNA molecules from being charged by SerRS with serine. The SerRS-rejecting allo-tRNA chassis were engineered to have another amino-acid identity of either alanine, tyrosine, or histidine. The tip of the V-arms was replaced with diverse hairpin RNA aptamers, which were recognized by their cognate proteins expressed in E. coli. A high-affinity interaction led to the sequestration of allo-tRNA molecules, while a moderate-affinity aptamer moiety recruited histidyl-tRNA synthetase variants fused with the cognate protein domain. The new design principle for tRNA-aptamer fusions will enhance radical and dynamic manipulation of the genetic code.


Subject(s)
Aptamers, Nucleotide/genetics , Genetic Engineering/methods , RNA, Transfer/genetics , Anticodon , Aptamers, Nucleotide/chemistry , Escherichia coli/genetics , Genes, Suppressor , Histidine-tRNA Ligase/genetics , Point Mutation , RNA, Transfer/chemistry , Serine-tRNA Ligase/genetics , Serine-tRNA Ligase/metabolism
15.
Genes (Basel) ; 11(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32906706

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) play essential roles in protein translation. In addition, numerous aaRSs (mostly in vertebrates) have also been discovered to possess a range of non-canonical functions. Very few studies have been conducted to elucidate or characterize non-canonical functions of plant aaRSs. A genome-wide search for aaRS genes in Arabidopsis thaliana revealed a total of 59 aaRS genes. Among them, asparaginyl-tRNA synthetase (AsnRS) was found to possess a WHEP domain inserted into the catalytic domain in a plant-specific manner. This insertion was observed only in the cytosolic isoform. In addition, a long stretch of sequence that exhibited weak homology with histidine ammonia lyase (HAL) was found at the N-terminus of histidyl-tRNA synthetase (HisRS). This HAL-like domain has only been seen in plant HisRS, and only in cytosolic isoforms. Additionally, a number of genes lacking minor or major portions of the full-length aaRS sequence were found. These genes encode 14 aaRS fragments that lack key active site sequences and are likely catalytically null. These identified genes that encode plant-specific additional domains or aaRS fragment sequences are candidates for aaRSs possessing non-canonical functions.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Aspartate-tRNA Ligase/metabolism , Genome, Plant , Histidine-tRNA Ligase/metabolism , RNA, Transfer, Amino Acyl/metabolism , Amino Acyl-tRNA Synthetases/genetics , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Aspartate-tRNA Ligase/genetics , Catalytic Domain , Histidine-tRNA Ligase/genetics , Protein Biosynthesis , RNA, Transfer, Amino Acyl/genetics
16.
Hum Mutat ; 41(7): 1232-1237, 2020 07.
Article in English | MEDLINE | ID: mdl-32333447

ABSTRACT

Mutations in histidyl-tRNA synthetase (HARS1), an enzyme that charges transfer RNA with the amino acid histidine in the cytoplasm, have only been associated to date with autosomal recessive Usher syndrome type III and autosomal dominant Charcot-Marie-Tooth disease type 2W. Using massive parallel sequencing, we identified bi-allelic HARS1 variants in a child (c.616G>T, p.Asp206Tyr and c.730delG, p.Val244Cysfs*6) and in two sisters (c.1393A>C, p.Ile465Leu and c.910_912dupTTG, p.Leu305dup), all characterized by a multisystem ataxic syndrome. All mutations are rare, segregate with the disease, and are predicted to have a significant effect on protein function. Functional studies helped to substantiate their disease-related roles. Indeed, yeast complementation assays showing that one out of two mutations in each patient is loss-of-function, and the reduction of messenger RNA and protein levels and enzymatic activity in patient's skin-derived fibroblasts, together support the pathogenicity of the identified HARS1 variants in the patient phenotypes. Thus, our efforts expand the allelic and clinical spectrum of HARS1-related disease.


Subject(s)
Ataxia/genetics , Histidine-tRNA Ligase/genetics , Adult , Alleles , Child , Female , Humans , Male , Mutation, Missense
17.
J Clin Neurosci ; 74: 32-35, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31982271

ABSTRACT

Interstitial lung disease (ILD) is an extramuscular manifestation associated with increased mortality in idiopathic inflammatory myopathy (IIM). To identify risk factors for ILD in patients with IIM, this study retrospectively enrolled 117 eligible patients from a university medical center. After a comprehensive chart review, 56 patients were stratified into ILD (n = 28) and non-ILD (n = 28) groups. Clinical features, laboratory data, concomitant diseases, and serology profiles were compared. Patients with ILD had high prevalence of anti-Jo1 antibodies (p = 0.002), anti-Ro52 antibodies (p < 0.001), both anti-Jo1 and anti-Ro52 antibodies (p = 0.008), anti-Jo1 or anti-Ro52 antibodies (p < 0.001), and lower initial creatine kinase (CK) levels (p = 0.006). Moreover, patients with anti-Ro52 antibodies and either anti-Ro52 or anti-Jo1 antibodies had 9.17-fold (95% confidence interval [CI]: 2.858-33.487, p < 0.001) and 13.44-fold (95% CI: 4.008-52.757, p < 0.001) increased odds of developing ILD, respectively. By contrast, patients with higher CK levels had 0.99-fold (95% CI: 0.999-0.999, p = 0.011) increased odds of developing ILD. Both anti-Ro52 and anti-Jo1 antibodies were independent serological risk factors for IIM-associated ILD. Because these serology tests are commonly available, they can be used to guide pulmonary screening for patients with IIM to increase neurologist proactivity in recognizing and treating extramuscular conditions.


Subject(s)
Lung Diseases, Interstitial/diagnosis , Myositis/complications , Adult , Aged , Autoantibodies/blood , Creatine Kinase/blood , Female , Histidine-tRNA Ligase/immunology , Humans , Lung Diseases, Interstitial/blood , Lung Diseases, Interstitial/etiology , Male , Middle Aged , Myositis/diagnosis , Retrospective Studies , Ribonucleoproteins/immunology , Risk Factors
18.
J Hum Genet ; 65(3): 305-311, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31827252

ABSTRACT

HARS2 encodes mitochondrial histidyl-tRNA synthetase (HARS2), which links histidine to its cognate tRNA in the mitochondrial matrix. Biallelic variants in HARS2 are associated with Perrault syndrome, a rare recessive condition characterized by sensorineural hearing loss in both sexes and primary ovarian insufficiency in 46,XX females. Some individuals with Perrault syndrome have a broader phenotypic spectrum with neurological features, including ataxia and peripheral neuropathy. Here, we report a recurrent variant in HARS2 in association with sensorineural hearing loss. In affected individuals from three unrelated families, the variant HARS2 c.1439G>A p.(Arg480His) is present as a heterozygous variant in trans to a putative pathogenic variant. The low prevalence of the allele HARS2 c.1439G>A p.(Arg480His) in the general population and its presence in three families with hearing loss, confirm the pathogenicity of this variant and illustrate the presentation of Perrault syndrome as nonsyndromic hearing loss in males and prepubertal females.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Histidine-tRNA Ligase/genetics , Alleles , Child , Child, Preschool , Exome/genetics , Female , Gonadal Dysgenesis, 46,XX/genetics , Gonadal Dysgenesis, 46,XX/physiopathology , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Homozygote , Humans , Infant , Male , Mitochondria/genetics , Mutation, Missense/genetics , Pedigree , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/physiopathology
19.
Arthritis Rheumatol ; 72(1): 179-191, 2020 01.
Article in English | MEDLINE | ID: mdl-31403245

ABSTRACT

OBJECTIVE: Autoantibodies targeting histidyl-transfer RNA synthetase (HisRS; anti-Jo-1) are common in the idiopathic inflammatory myopathies (IIMs) and antisynthetase syndrome. This study was undertaken to investigate immunity against HisRS in the blood and lungs of patients with IIM/antisynthetase syndrome. METHODS: Bronchoalveolar lavage (BAL) fluid, BAL fluid cells, and peripheral blood mononuclear cells (PBMCs) from patients with IIM/antisynthetase syndrome (n = 24) were stimulated with full-length HisRS protein or a HisRS-derived peptide (HisRS11-23 ). BAL fluid and PBMCs from patients with sarcoidosis (n = 7) and healthy subjects (n = 12) were included as controls. The CD4+ T cell response was determined according to levels of CD40L up-regulation and cytokine expression using flow cytometry. Anti-Jo-1 autoantibody responses in the serum and BAL fluid were assessed by enzyme-linked immunosorbent assay. Lung biopsy samples from patients with IIM/antisynthetase syndrome (n = 14) were investigated by immunohistochemistry. RESULTS: In BAL fluid, CD4+ T cells from 3 of 4 patients with IIM/antisynthetase syndrome responded to stimulation with HisRS protein, as measured by the median fold change in CD40L expresssion in stimulated cells compared to unstimulated cells (median fold change 3.6, interquartile range [IQR] 2.7-14.7), and 2 of 3 patients with IIM/antisynthetase syndrome had the highest responses to HisRS11-23 (median fold change 88, IQR 27-149). In PBMCs, CD4+ T cells from 14 of 18 patients with IIM/antisynthetase syndrome responded to HisRS protein (median fold change 7.38, IQR 2.69-31.86; P < 0.001), whereas a HisRS11-23 response was present in 11 of 14 patients with IIM/antisynthetase syndrome (median fold change 3.4, IQR 1.87-10.9; P < 0.001). In the control group, there was a HisRS11-23 response in 3 of 7 patients with sarcoidosis (median fold change 2.09, IQR 1.45-3.29) and in 5 of 12 healthy controls (median fold change 2, IQR 1.89-2.42). CD4+ T cells from patients with IIM/antisynthetase syndrome displayed a pronounced Th1 phenotype in the BAL fluid when compared to the PBMCs (P < 0.001), producing high amounts of interferon-γ and interleukin-2 following stimulation. Anti-Jo-1 autoantibodies were detected in BAL fluid and germinal center (GC)-like structures were seen in the lung biopsy samples from patients with IIM/antisynthetase syndrome. CONCLUSION: The results of this study demonstrate a pronounced presence of HisRS-reactive CD4+ T cells in PBMCs and BAL fluid cells from patients with IIM/antisynthetase syndrome as compared to patients with sarcoidosis and healthy controls. These findings, combined with the presence of anti-Jo-1 autoantibodies in BAL fluid and GC-like structures in the lungs, suggest that immune activation against HisRS might take place within the lungs of patients with IIM/antisynthetase syndrome.


Subject(s)
Antibodies, Antinuclear/immunology , CD4-Positive T-Lymphocytes/immunology , Lung Diseases, Interstitial/immunology , Lung/immunology , Monocytes/immunology , Myositis/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Female , Histidine-tRNA Ligase/immunology , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Lung/cytology , Lung/pathology , Lung Diseases, Interstitial/pathology , Male , Middle Aged , Myositis/blood , Th1 Cells
20.
Proc Natl Acad Sci U S A ; 116(39): 19440-19448, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31501329

ABSTRACT

Aminoacyl-transfer RNA (tRNA) synthetases (aaRSs) are the largest protein family causatively linked to neurodegenerative Charcot-Marie-Tooth (CMT) disease. Dominant mutations cause the disease, and studies of CMT disease-causing mutant glycyl-tRNA synthetase (GlyRS) and tyrosyl-tRNA synthetase (TyrRS) showed their mutations create neomorphic structures consistent with a gain-of-function mechanism. In contrast, based on a haploid yeast model, loss of aminoacylation function was reported for CMT disease mutants in histidyl-tRNA synthetase (HisRS). However, neither that nor prior work of any CMT disease-causing aaRS investigated the aminoacylation status of tRNAs in the cellular milieu of actual patients. Using an assay that interrogated aminoacylation levels in patient cells, we investigated a HisRS-linked CMT disease family with the most severe disease phenotype. Strikingly, no difference in charged tRNA levels between normal and diseased family members was found. In confirmation, recombinant versions of 4 other HisRS CMT disease-causing mutants showed no correlation between activity loss in vitro and severity of phenotype in vivo. Indeed, a mutation having the most detrimental impact on activity was associated with a mild disease phenotype. In further work, using 3 independent biophysical analyses, structural opening (relaxation) of mutant HisRSs at the dimer interface best correlated with disease severity. In fact, the HisRS mutation in the severely afflicted patient family caused the largest degree of structural relaxation. These data suggest that HisRS-linked CMT disease arises from open conformation-induced mechanisms distinct from loss of aminoacylation.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/genetics , Histidine-tRNA Ligase/genetics , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation/genetics , Axons , Charcot-Marie-Tooth Disease/metabolism , Gain of Function Mutation/genetics , Histidine-tRNA Ligase/metabolism , Humans , Mutation , RNA, Transfer/genetics , RNA, Transfer/metabolism , Structure-Activity Relationship , Tyrosine-tRNA Ligase/genetics , Tyrosine-tRNA Ligase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL