Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell Rep ; 43(8): 114570, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39093697

ABSTRACT

A wide variety of electrophilic derivatives of itaconate, the Kreb's cycle-derived metabolite, are immunomodulatory, yet these derivatives have overlapping and sometimes contradictory activities. Therefore, we generated a genetic system to interrogate the immunomodulatory functions of endogenously produced itaconate in human macrophages. Endogenous itaconate is driven by multiple innate signals restraining inflammatory cytokine production. Endogenous itaconate directly targets cysteine 13 in IRAK4 (disrupting IRAK4 autophosphorylation and activation), drives the degradation of nuclear factor κB, and modulates global ubiquitination patterns. As a result, cells unable to make itaconate overproduce inflammatory cytokines such as tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and IL-1ß in response to these innate activators. In contrast, the production of interferon (IFN)ß, downstream of LPS, requires the production of itaconate. These data demonstrate that itaconate is a critical arbiter of inflammatory cytokine production downstream of multiple innate signaling pathways, laying the groundwork for the development of itaconate mimetics for the treatment of autoimmunity.


Subject(s)
Cytokines , Immunity, Innate , Macrophages , Succinates , Ubiquitination , Humans , Succinates/pharmacology , Succinates/metabolism , Ubiquitination/drug effects , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Cytokines/metabolism , Immunity, Innate/drug effects , NF-kappa B/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Signal Transduction/drug effects , Lipopolysaccharides/pharmacology , HEK293 Cells
2.
Nat Med ; 8(9): 1004-10, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12145646

ABSTRACT

Adult bone marrow (BM) contains cells capable of differentiating along hematopoietic (Lin(+)) or non-hematopoietic (Lin(-)) lineages. Lin(-) hematopoietic stem cells (HSCs) have recently been shown to contain a population of endothelial precursor cells (EPCs) capable of forming blood vessels. Here we show that intravitreally injected Lin(-) BM cells selectively target retinal astrocytes, cells that serve as a template for both developmental and injury-associated retinal angiogenesis. When Lin(-) BM cells were injected into neonatal mouse eyes, they extensively and stably incorporated into forming retinal vasculature. When EPC-enriched HSCs were injected into the eyes of neonatal rd/rd mice, whose vasculature ordinarily degenerates with age, they rescued and maintained a normal vasculature. In contrast, normal retinal angiogenesis was inhibited when EPCs expressing a potent angiostatic protein were injected. We have demonstrated that Lin(-) BM cells and astrocytes specifically interact with one another during normal angiogenesis and pathological vascular degeneration in the retina. Selective targeting with Lin(-) HSC may be a useful therapeutic approach for the treatment of many ocular diseases.


Subject(s)
Astrocytes/pathology , Bone Marrow Cells , Neovascularization, Pathologic , Retinal Vessels/pathology , Animals , Animals, Newborn , Bone Marrow Transplantation , Endothelium, Vascular/pathology , Genetic Therapy/methods , Mice , Mice, Inbred Strains , Retina , Retinal Degeneration/pathology , Retinal Degeneration/therapy , Transfection
3.
Structure ; 15(7): 793-805, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17637340

ABSTRACT

Higher eukaryote tRNA synthetases have expanded functions that come from enlarged, more differentiated structures that were adapted to fit aminoacylation function. How those adaptations affect catalytic mechanisms is not known. Presented here is the structure of a catalytically active natural splice variant of human tryptophanyl-tRNA synthetase (TrpRS) that is a potent angiostatic factor. This and related structures suggest that a eukaryote-specific N-terminal extension of the core enzyme changed substrate recognition by forming an active site cap. At the junction of the extension and core catalytic unit, an arginine is recruited to replace a missing landmark lysine almost 200 residues away. Mutagenesis, rapid kinetic, and substrate binding studies support the functional significance of the cap and arginine recruitment. Thus, the enzyme function of human TrpRS has switched more to the N terminus of the sequence. This switch has the effect of creating selective pressure to retain the N-terminal extension for functional expansion.


Subject(s)
Angiogenesis Inhibitors/chemistry , Models, Molecular , Protein Folding , Tryptophan-tRNA Ligase/chemistry , Acetylation , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Geobacillus stearothermophilus/metabolism , Humans , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , RNA Splicing , Substrate Specificity , Tryptophan-tRNA Ligase/genetics
4.
Chem Biol ; 14(12): 1323-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18096501

ABSTRACT

Disease-causing mutations occur in genes for aminoacyl tRNA synthetases. That some mutations are dominant suggests a gain of function. Native tRNA synthetases, such as tyrosyl-tRNA synthetase (TyrRS) and tryptophanyl-tRNA synthetase, catalyze aminoacylation and are also procytokines that are activated by natural fragmentation. In principle, however, gain-of-function phenotypes could arise from mutational activation of synthetase procytokines. From crystal structure analysis, we hypothesized that a steric block of a critical Glu-Leu-Arg (ELR) motif in full-length TyrRS suppresses the cytokine activity of a natural fragment. To test this hypothesis, we attempted to uncover ELR in the procytokine by mutating a conserved tyrosine (Y341) that tethers ELR. Site-specific proteolytic cleavage and small-angle X-ray scattering established subtle opening of the structure by the mutation. Strikingly, four different assays demonstrated mutational activation of cytokine functions. The results prove the possibilities for constitutive gain-of-function mutations in tRNA synthetases.


Subject(s)
Cytokines/metabolism , Mutation , Tyrosine-tRNA Ligase/metabolism , Amino Acid Motifs/genetics , Amino Acid Substitution , Animals , Cattle , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemotaxis, Leukocyte/drug effects , Chick Embryo , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Mice , Mice, Nude , Models, Molecular , Neovascularization, Physiologic/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Conformation , Protein Structure, Tertiary , Scattering, Small Angle , Tyrosine-tRNA Ligase/chemistry , Tyrosine-tRNA Ligase/genetics , X-Ray Diffraction
5.
J Exp Med ; 210(1): 23-30, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23267013

ABSTRACT

B cell development requires tight regulation to allow for the generation of a diverse repertoire while preventing the development of autoreactive cells. We report, using N-ethyl-N-nitrosourea (ENU)-induced mutagenesis, the identification of a mutant mouse (chompB) with a block in early B cell development. The blockade occurs after the transitional 1 (T1) stage and leads to a decrease in mature B cell subsets and deficits in T cell-dependent antibody responses. Additionally, chompB mice have decreases in myeloid dendritic cells (DCs). The mutation was mapped to the intramembrane protease signal peptide peptidase-like 2a (Sppl2a), a gene not previously implicated in immune cell development. Proteomic analysis identified the invariant chain (CD74) as a key substrate of Sppl2a and suggests that regulated intramembrane proteolysis of CD74 by Sppl2a contributes to B cell and DC survival. Moreover, these data suggest that modulation of Sppl2a may be a useful therapeutic strategy for treatment of B cell dependent autoimmune disorders.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , B-Lymphocytes/physiology , Dendritic Cells/pathology , Membrane Proteins/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Aspartic Acid Endopeptidases/genetics , B-Lymphocytes/pathology , Cell Survival , Dendritic Cells/physiology , Ethylnitrosourea/pharmacology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Immunoglobulins/metabolism , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mutagenesis/drug effects , Mutation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Chem Biol ; 16(5): 531-9, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19477417

ABSTRACT

Aminoacyl tRNA synthetases are known for catalysis of aminoacylation. Significantly, some mammalian synthetases developed cytokine functions possibly linked to disease-causing mutations in tRNA synthetases. Not understood is how epitopes for cytokine signaling were introduced into catalytic scaffolds without disturbing aminoacylation. Here we investigate human tyrosyl-tRNA synthetase, where a catalytic-domain surface helix, next to the active site, was recruited for interleukin-8-like cytokine signaling. Taking advantage of our high resolution structure, the reciprocal impact of rational mutations designed to disrupt aminoacylation or cytokine signaling was investigated with multiple assays. The collective analysis demonstrated a protective fine-structure separation of aminoacylation from cytokine activities within the conserved catalytic domain. As a consequence, disease-causing mutations affecting cell signaling can arise without disturbing aminoacylation. These results with TyrRS also predict the previously unknown binding conformation of interleukin-8-like CXC cytokines.


Subject(s)
Cytokines/chemistry , Mutation , Tyrosine-tRNA Ligase/chemistry , Amino Acid Substitution , Aminoacylation , Biocatalysis , Catalytic Domain , Computer Simulation , Humans , Mutant Proteins/metabolism , Protein Binding , Tyrosine-tRNA Ligase/metabolism
7.
J Biol Chem ; 284(18): 12328-38, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19286662

ABSTRACT

A growing number of orphan G-protein-coupled receptors (GPCRs) have been reported to be activated by lipid ligands, such as lysophosphatidic acid, sphingosine 1-phosphate (S1P), and cannabinoids, for which there are already well established receptors. These new ligand claims are controversial due to either lack of independent confirmations or conflicting reports. We used the beta-arrestin PathHunter assay system, a newly developed, generic GPCR assay format that measures beta-arrestin binding to GPCRs, to evaluate lipid receptor and ligand pairing. This assay eliminates interference from endogenous receptors on the parental cells because it measures a signal that is specifically generated by the tagged receptor and is immediately downstream of receptor activation. We screened a large number of newly "deorphaned" receptors (GPR23, GPR92, GPR55, G2A, GPR18, GPR3, GPR6, GPR12, and GPR63) and control receptors against a collection of approximately 400 lipid molecules to try to identify the receptor ligand in an unbiased fashion. GPR92 was confirmed to be a lysophosphatidic acid receptor with weaker responses to farnesyl pyrophosphate and geranylgeranyl diphosphate. The putative cannabinoid receptor GPR55 responded strongly to AM251, rimonabant, and lysophosphatidylinositol but only very weakly to endocannabinoids. G2A receptor was confirmed to be an oxidized free fatty acid receptor. In addition, we discovered that 3,3'-diindolylmethane, a dietary molecule from cruciferous vegetables, which has known anti-cancer properties, to be a CB(2) receptor partial agonist, with binding affinity around 1 microm. The anti-inflammatory effect of 3,3'-diindolylmethane in RAW264.7 cells was shown to be partially mediated by CB(2).


Subject(s)
Arrestins/metabolism , Cannabinoids/metabolism , Lysophospholipids/metabolism , Receptors, G-Protein-Coupled/metabolism , Sphingosine/analogs & derivatives , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Sphingosine/metabolism , beta-Arrestins
8.
J Biol Chem ; 283(4): 2070-7, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-17999956

ABSTRACT

In mammalian cells, specific aminoacyl-transfer RNA (tRNA) synthetases have cytokine functions that require interactions with partners outside of the translation apparatus. Little is known about these interactions and how they facilitate expanded functions that link protein translation to other cellular pathways. For example, an alternative splice fragment of tryptophanyl-tRNA synthetase (TrpRS) and a similar natural proteolytic fragment are potent angiostatic factors that act through the vascular endothelial-cadherin receptor and Akt signaling pathway. Here we demonstrate mobilization of TrpRS for exocytosis from endothelial cells and the potential for plasmin to activate the cytokine function of the extracellular synthetase. Direct physical evidence showed that the annexin II-S100A10 complex, which regulates exocytosis, forms a ternary complex with TrpRS. Functional studies demonstrate that both annexin II and S100A10 regulate trafficking of TrpRS. Thus, complexes of mammalian tRNA synthetases with seemingly disparate proteins may in general be relevant to understanding how their expanded functions are implemented.


Subject(s)
Angiostatic Proteins/metabolism , Annexin A2/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Fibrinolysin/metabolism , S100 Proteins/metabolism , Tryptophan-tRNA Ligase/metabolism , Alternative Splicing/physiology , Angiostatic Proteins/genetics , Annexin A2/genetics , Cells, Cultured , Cytokines/genetics , Endothelial Cells/cytology , Exocytosis/physiology , Fibrinolysin/genetics , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Biosynthesis/physiology , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , S100 Proteins/genetics , Signal Transduction/physiology , Tryptophan-tRNA Ligase/genetics
9.
EMBO J ; 25(12): 2919-29, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16724112

ABSTRACT

Aminoacylation of tRNA is the first step of protein synthesis. Here, we report the co-crystal structure of human tryptophanyl-tRNA synthetase and tRNATrp. This enzyme is reported to interact directly with elongation factor 1alpha, which carries charged tRNA to the ribosome. Crystals were generated from a 50/50% mixture of charged and uncharged tRNATrp. These crystals captured two conformations of the complex, which are nearly identical with respect to the protein and a bound tryptophan. They are distinguished by the way tRNA is bound. In one, uncharged tRNA is bound across the dimer, with anticodon and acceptor stem interacting with separate subunits. In this cross-dimer tRNA complex, the class I enzyme has a class II-like tRNA binding mode. This structure accounts for biochemical investigations of human TrpRS, including species-specific charging. In the other conformation, presumptive aminoacylated tRNA is bound only by the anticodon, the acceptor stem being free and having space to interact precisely with EF-1alpha, suggesting that the product of aminoacylation can be directly handed off to EF-1alpha for the next step of protein synthesis.


Subject(s)
Nucleic Acid Conformation , Protein Biosynthesis , RNA, Transfer, Trp/chemistry , RNA, Transfer, Trp/metabolism , Tryptophan-tRNA Ligase/chemistry , Tryptophan-tRNA Ligase/metabolism , Amino Acid Sequence , Anticodon/genetics , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Peptide Elongation Factor 1/metabolism , Protein Binding , Protein Conformation , Sequence Alignment , Tryptophan/genetics
10.
Biochemistry ; 44(11): 4216-21, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15766249

ABSTRACT

In cellular environments, coupled hydrolytic reactions are used to force efficient product formation in enzyme-catalyzed reactions. In the first step of protein synthesis, aminoacyl-tRNA synthetases react with amino acid and ATP to form an enzyme-bound adenylate that, in the next step, reacts with tRNA to form aminoacyl-tRNA. The reaction liberates pyrophosphate (PP(i)) which, in turn, can be hydrolyzed by pyrophosphatase to drive efficient aminoacylation. A potential polymorphic variant of human tryptophanyl-tRNA synthetase is shown here to sequester tryptophanyl adenylate. The bound adenylate does not react efficiently with the liberated PP(i) that normally competes with tRNA to resynthesize ATP and free amino acid. Structural analysis of this variant showed that residues needed for binding ATP phosphates and thus PP(i) were reoriented from their conformations in the structure of the more common sequence variant. Significantly, the reorientation does not affect reaction with tRNA, so that efficient aminoacylation is achieved.


Subject(s)
Genetic Variation , Mutagenesis, Site-Directed , Tryptophan-tRNA Ligase/chemistry , Tryptophan-tRNA Ligase/metabolism , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Catalysis , Diphosphates/chemistry , Diphosphates/metabolism , Enzyme Activation/genetics , Humans , Kinetics , Models, Chemical , Molecular Sequence Data , Point Mutation , Polymorphism, Genetic , Tryptophan/chemistry , Tryptophan/metabolism
11.
Proc Natl Acad Sci U S A ; 100(26): 15376-80, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14671330

ABSTRACT

Early forms of the genetic code likely generated "statistical" proteins, with similar side chains occupying the same sequence positions at different ratios. In this scenario, groups of related side chains were treated by aminoacyl-tRNA synthetases as a single molecular species until a discrimination mechanism developed that could separate them. The aromatic amino acids tryptophan, tyrosine, and phenylalanine likely constituted one of these groups. A crystal structure of human tryptophanyl-tRNA synthetase was solved at 2.1 A with a tryptophanyl-adenylate bound at the active site. A cocrystal structure of an active fragment of human tyrosyl-tRNA synthetase with its cognate amino acid analog was also solved at 1.6 A. The two structures enabled active site identifications and provided the information for structure-based sequence alignments of approximately 45 orthologs of each enzyme. Two critical positions shared by all tyrosyl-tRNA synthetases and tryptophanyl-tRNA synthetases for amino acid discrimination were identified. The variations at these two positions and phylogenetic analyses based on the structural information suggest that, in contrast to many other amino acids, discrimination of tyrosine from tryptophan occurred late in the development of the genetic code.


Subject(s)
Genetic Code , Tryptophan-tRNA Ligase/chemistry , Tryptophan-tRNA Ligase/genetics , Adenosine Monophosphate , Amino Acid Sequence , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Geobacillus stearothermophilus/enzymology , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Sensitivity and Specificity , Sequence Alignment , Sequence Homology, Amino Acid , Tryptophan
12.
Mol Cell ; 13(6): 829-41, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-15053876

ABSTRACT

Early work on aminoacylation of alanine-specific tRNA (tRNA(Ala)) by alanyl-tRNA synthetase (AlaRS) gave rise to the concept of an early "second genetic code" imbedded in the acceptor stems of tRNAs. A single conserved and position-specific G:U base pair in the tRNA acceptor stem is the key identity determinant. Further understanding has been limited due to lack of a crystal structure of the enzyme. We determined a 2.14 A crystal structure of the 453 amino acid catalytic fragment of Aquifex aeolicus AlaRS. It contains the catalytic domain characteristic of class II synthetases, a helical domain with a hairpin motif critical for acceptor-stem recognition, and a C-terminal domain of a mixed alpha/beta fold. Docking of tRNA(Ala) on AlaRS shows critical contacts with the three domains, consistent with previous mutagenesis and functional data. It also suggests conformational flexibility within the C domain, which might allow for the positional variation of the key G:U base pair seen in some tRNA(Ala)s.


Subject(s)
Alanine-tRNA Ligase/chemistry , Bacterial Proteins/chemistry , Alanine-tRNA Ligase/genetics , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/genetics , Base Pairing , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Bacterial/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL