Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(10): e2309957121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38422022

ABSTRACT

Hypoxia signaling influences tumor development through both cell-intrinsic and -extrinsic pathways. Inhibiting hypoxia-inducible factor (HIF) function has recently been approved as a cancer treatment strategy. Hence, it is important to understand how regulators of HIF may affect tumor growth under physiological conditions. Here we report that in aging mice factor-inhibiting HIF (FIH), one of the most studied negative regulators of HIF, is a haploinsufficient suppressor of spontaneous B cell lymphomas, particular pulmonary B cell lymphomas. FIH deficiency alters immune composition in aged mice and creates a tumor-supportive immune environment demonstrated in syngeneic mouse tumor models. Mechanistically, FIH-defective myeloid cells acquire tumor-supportive properties in response to signals secreted by cancer cells or produced in the tumor microenvironment with enhanced arginase expression and cytokine-directed migration. Together, these data demonstrate that under physiological conditions, FIH plays a key role in maintaining immune homeostasis and can suppress tumorigenesis through a cell-extrinsic pathway.


Subject(s)
Lymphoma, B-Cell , Repressor Proteins , Animals , Mice , Hypoxia/metabolism , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , Tumor Microenvironment
2.
J Med Chem ; 66(15): 10849-10865, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37527664

ABSTRACT

Jumonji-C domain-containing protein 5 (JMJD5) is a 2-oxoglutarate (2OG)-dependent oxygenase that plays important roles in development, circadian rhythm, and cancer through unclear mechanisms. JMJD5 has been reported to have activity as a histone protease, as an Nε-methyl lysine demethylase, and as an arginine residue hydroxylase. Small-molecule JMJD5-selective inhibitors will be useful for investigating its (patho)physiological roles. Following the observation that the broad-spectrum 2OG oxygenase inhibitor pyridine-2,4-dicarboxylic acid (2,4-PDCA) is a 2OG-competing JMJD5 inhibitor, we report that 5-aminoalkyl-substituted 2,4-PDCA derivatives are potent JMJD5 inhibitors manifesting selectivity for JMJD5 over other human 2OG oxygenases. Crystallographic analyses with five inhibitors imply induced fit binding and reveal that the 2,4-PDCA C5 substituent orients into the JMJD5 substrate-binding pocket. Cellular studies indicate that the lead compounds display similar phenotypes as reported for clinically observed JMJD5 variants, which have a reduced catalytic activity compared to wild-type JMJD5.


Subject(s)
Histones , Neoplasms , Humans , Circadian Rhythm , Pyridines/pharmacology , Oxygenases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism
3.
Genet Med ; 25(9): 100893, 2023 09.
Article in English | MEDLINE | ID: mdl-37179472

ABSTRACT

PURPOSE: Developmentally regulated Guanosine-5'-triphosphate-binding protein 1 (DRG1) is a highly conserved member of a class of GTPases implicated in translation. Although the expression of mammalian DRG1 is elevated in the central nervous system during development, and its function has been implicated in fundamental cellular processes, no pathogenic germline variants have yet been identified. Here, we characterize the clinical and biochemical consequences of DRG1 variants. METHODS: We collate clinical information of 4 individuals with germline DRG1 variants and use in silico, in vitro, and cell-based studies to study the pathogenicity of these alleles. RESULTS: We identified private germline DRG1 variants, including 3 stop-gained p.Gly54∗, p.Arg140∗, p.Lys263∗, and a p.Asn248Phe missense variant. These alleles are recessively inherited in 4 affected individuals from 3 distinct families and cause a neurodevelopmental disorder with global developmental delay, primary microcephaly, short stature, and craniofacial anomalies. We show that these loss-of-function variants (1) severely disrupt DRG1 messenger RNA/protein stability in patient-derived fibroblasts, (2) impair its GTPase activity, and (3) compromise its binding to partner protein ZC3H15. Consistent with the importance of DRG1 in humans, targeted inactivation of mouse Drg1 resulted in preweaning lethality. CONCLUSION: Our work defines a new Mendelian disorder of DRG1 deficiency. This study highlights DRG1's importance for normal mammalian development and underscores the significance of translation factor GTPases in human physiology and homeostasis.


Subject(s)
GTP-Binding Proteins , Neurodevelopmental Disorders , Animals , Humans , Mice , Carrier Proteins , GTP Phosphohydrolases/genetics , Mammals/metabolism , Neurodevelopmental Disorders/genetics , RNA, Messenger
4.
J Clin Invest ; 133(7)2023 04 03.
Article in English | MEDLINE | ID: mdl-36795492

ABSTRACT

Although protein hydroxylation is a relatively poorly characterized posttranslational modification, it has received significant recent attention following seminal work uncovering its role in oxygen sensing and hypoxia biology. Although the fundamental importance of protein hydroxylases in biology is becoming clear, the biochemical targets and cellular functions often remain enigmatic. JMJD5 is a "JmjC-only" protein hydroxylase that is essential for murine embryonic development and viability. However, no germline variants in JmjC-only hydroxylases, including JMJD5, have yet been described that are associated with any human pathology. Here we demonstrate that biallelic germline JMJD5 pathogenic variants are deleterious to JMJD5 mRNA splicing, protein stability, and hydroxylase activity, resulting in a human developmental disorder characterized by severe failure to thrive, intellectual disability, and facial dysmorphism. We show that the underlying cellular phenotype is associated with increased DNA replication stress and that this is critically dependent on the protein hydroxylase activity of JMJD5. This work contributes to our growing understanding of the role and importance of protein hydroxylases in human development and disease.


Subject(s)
Histone Demethylases , Mixed Function Oxygenases , Humans , Animals , Mice , Histone Demethylases/genetics , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Protein Processing, Post-Translational
5.
J Biol Chem ; 298(6): 102020, 2022 06.
Article in English | MEDLINE | ID: mdl-35537551

ABSTRACT

The aspariginyl hydroxylase human factor inhibiting hypoxia-inducible factor (FIH) is an important regulator of the transcriptional activity of hypoxia-inducible factor. FIH also catalyzes the hydroxylation of asparaginyl and other residues in ankyrin repeat domain-containing proteins, including apoptosis stimulating of p53 protein (ASPP) family members. ASPP2 is reported to undergo a single FIH-catalyzed hydroxylation at Asn-986. We report biochemical and crystallographic evidence showing that FIH catalyzes the unprecedented post-translational hydroxylation of both asparaginyl residues in "VNVN" and related motifs of ankyrin repeat domains in ASPPs (i.e., ASPP1, ASPP2, and iASPP) and the related ASB11 and p18-INK4C proteins. Our biochemical results extend the substrate scope of FIH catalysis and may have implications for its biological roles, including in the hypoxic response and ASPP family function.


Subject(s)
Ankyrin Repeat , Mixed Function Oxygenases , Repressor Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Apoptosis Regulatory Proteins , Catalysis , Humans , Hydroxylation , Hypoxia , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism
6.
J Med Chem ; 64(23): 17031-17050, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34843649

ABSTRACT

MINA53 is a JmjC domain 2-oxoglutarate-dependent oxygenase that catalyzes ribosomal hydroxylation and is a target of the oncogenic transcription factor c-MYC. Despite its anticancer target potential, no small-molecule MINA53 inhibitors are reported. Using ribosomal substrate fragments, we developed mass spectrometry assays for MINA53 and the related oxygenase NO66. These assays enabled the identification of 2-(aryl)alkylthio-3,4-dihydro-4-oxoypyrimidine-5-carboxylic acids as potent MINA53 inhibitors, with selectivity over NO66 and other JmjC oxygenases. Crystallographic studies with the JmjC demethylase KDM5B revealed active site binding but without direct metal chelation; however, molecular modeling investigations indicated that the inhibitors bind to MINA53 by directly interacting with the iron cofactor. The MINA53 inhibitors manifest evidence for target engagement and selectivity for MINA53 over KDM4-6. The MINA53 inhibitors show antiproliferative activity with solid cancer lines and sensitize cancer cells to conventional chemotherapy, suggesting that further work investigating their potential in combination therapies is warranted.


Subject(s)
Dioxygenases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Ribosomes/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallization , Dioxygenases/chemistry , Dioxygenases/metabolism , Enzyme Inhibitors/metabolism , Histone Demethylases/chemistry , Histone Demethylases/metabolism , Humans , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Conformation , Substrate Specificity
7.
Cell Mol Life Sci ; 78(23): 7219-7235, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34664086

ABSTRACT

GTPases are a large superfamily of evolutionarily conserved proteins involved in a variety of fundamental cellular processes. The developmentally regulated GTP-binding protein (DRG) subfamily of GTPases consists of two highly conserved paralogs, DRG1 and DRG2, both of which have been implicated in the regulation of cell proliferation, translation and microtubules. Furthermore, DRG1 and 2 proteins both have a conserved binding partner, DRG family regulatory protein 1 and 2 (DFRP1 and DFRP2), respectively, that prevents them from being degraded. Similar to DRGs, the DFRP proteins have also been studied in the context of cell growth control and translation. Despite these proteins having been implicated in several fundamental cellular processes they remain relatively poorly characterized, however. In this review, we provide an overview of the structural biology and biochemistry of DRG GTPases and discuss current understanding of DRGs and DFRPs in normal physiology, as well as their emerging roles in diseases such as cancer.


Subject(s)
Cell Proliferation/physiology , GTP-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Neoplasms/pathology , Animals , GTP-Binding Proteins/genetics , Humans , Microtubules/metabolism , Protein Biosynthesis/physiology , Protein Domains/physiology , RNA-Binding Proteins/metabolism
9.
Biochem Soc Trans ; 48(5): 1843-1858, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32985654

ABSTRACT

Fe(II)/2-oxoglutarate (2OG)-dependent oxygenases are a conserved enzyme class that catalyse diverse oxidative reactions across nature. In humans, these enzymes hydroxylate a broad range of biological substrates including DNA, RNA, proteins and some metabolic intermediates. Correspondingly, members of the 2OG-dependent oxygenase superfamily have been linked to fundamental biological processes, and found dysregulated in numerous human diseases. Such findings have stimulated efforts to understand both the biochemical activities and cellular functions of these enzymes, as many have been poorly studied. In this review, we focus on human 2OG-dependent oxygenases catalysing the hydroxylation of protein and polynucleotide substrates. We discuss their modulation by changes in the cellular microenvironment, particularly with respect to oxygen, iron, 2OG and the effects of oncometabolites. We also describe emerging evidence that these enzymes are responsive to cellular stresses including hypoxia and DNA damage. Moreover, we examine how dysregulation of 2OG-dependent oxygenases is associated with human disease, and the apparent paradoxical role for some of these enzymes during cancer development. Finally, we discuss some of the challenges associated with assigning biochemical activities and cellular functions to 2OG-dependent oxygenases.


Subject(s)
DNA Damage , Ketoglutaric Acids/pharmacology , Oxygenases/metabolism , Ascorbic Acid/chemistry , Biological Phenomena , Catalysis , DNA/chemistry , Gene Expression Regulation , Humans , Hydroxylation , Hypoxia , Mixed Function Oxygenases/metabolism , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Oxygen/chemistry , Protein Processing, Post-Translational , RNA/chemistry
10.
J Biol Chem ; 295(13): 4065-4078, 2020 03 27.
Article in English | MEDLINE | ID: mdl-31690629

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) directly dictate the expression of multiple RNA species including novel and as yet uncharacterized long noncoding transcripts with unknown function. We used pan-genomic HIF-binding and transcriptomic data to identify a novel long noncoding RNA Noncoding Intergenic Co-Induced transcript (NICI) on chromosome 12p13.31 which is regulated by hypoxia via HIF-1 promoter-binding in multiple cell types. CRISPR/Cas9-mediated deletion of the hypoxia-response element revealed co-regulation of NICI and the neighboring protein-coding gene, solute carrier family 2 member 3 (SLC2A3) which encodes the high-affinity glucose transporter 3 (GLUT3). Knockdown or knockout of NICI attenuated hypoxic induction of SLC2A3, indicating a direct regulatory role of NICI in SLC2A3 expression, which was further evidenced by CRISPR/Cas9-VPR-mediated activation of NICI expression. We also demonstrate that regulation of SLC2A3 is mediated through transcriptional activation rather than posttranscriptional mechanisms because knockout of NICI leads to reduced recruitment of RNA polymerase 2 to the SLC2A3 promoter. Consistent with this we observe NICI-dependent regulation of glucose consumption and cell proliferation. Furthermore, NICI expression is regulated by the von Hippel-Lindau (VHL) tumor suppressor and is highly expressed in clear cell renal cell carcinoma (ccRCC), where SLC2A3 expression is associated with patient prognosis, implying an important role for the HIF/NICI/SLC2A3 axis in this malignancy.


Subject(s)
Carcinoma, Renal Cell/genetics , Glucose Transporter Type 3/genetics , RNA, Long Noncoding/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , CRISPR-Cas Systems/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , Transcriptional Activation/genetics , Tumor Hypoxia/genetics
11.
J Clin Invest ; 129(9): 3640-3656, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31162141

ABSTRACT

Physiological effects of cellular hypoxia are sensed by prolyl hydroxylase (PHD) enzymes which regulate HIFs. Genetic interventions on HIF/PHD pathways reveal multiple phenotypes that extend the known biology of hypoxia. Recent studies unexpectedly implicate HIF in aspects of multiple immune and inflammatory pathways. However such studies are often limited by systemic lethal effects and/or use tissue-specific recombination systems, which are inherently irreversible, un-physiologically restricted and difficult to time. To study these processes better we developed recombinant mice which express tetracycline-regulated shRNAs broadly targeting the main components of the HIF/PHD pathway, permitting timed bi-directional intervention. We have shown that stabilization of HIF levels in adult mice through PHD2 enzyme silencing by RNA interference, or inducible recombination of floxed alleles, results in multi-lineage leukocytosis and features of autoimmunity. This phenotype was rapidly normalized on re-establishment of the hypoxia-sensing machinery when shRNA expression was discontinued. In both situations these effects were mediated principally through the Hif2a isoform. Assessment of cells bearing regulatory T cell markers from these mice revealed defective function and pro-inflammatory effects in vivo. We believe our findings have shown a new role for the PHD2/Hif2a couple in the reversible regulation of T cell and immune activity.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , RNA Interference/immunology , Signal Transduction , T-Lymphocytes, Regulatory , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/immunology , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
12.
PLoS One ; 14(2): e0211244, 2019.
Article in English | MEDLINE | ID: mdl-30807587

ABSTRACT

Expulsion of parasitic gastrointestinal nematodes requires diverse effector mechanisms coordinated by a Th2-type response. The evolutionarily conserved JmjC protein; Myc Induced Nuclear Antigen (Mina) has been shown to repress IL4, a key Th2 cytokine, suggesting Mina may negatively regulate nematode expulsion. Here we report that expulsion of the parasitic nematode Trichuris muris was indeed accelerated in Mina deficient mice. Unexpectedly, this was associated not with an elevated Th2- but rather an impaired Th1-type response. Further reciprocal bone marrow chimera and conditional KO experiments demonstrated that retarded parasite expulsion and a normal Th1-type response both required Mina in intestinal epithelial cells (IECs). Transcriptional profiling experiments in IECs revealed anti-microbial α-defensin peptides to be the major target of Mina-dependent retention of worms in infected mice. In vitro exposure to recombinant α-defensin peptides caused cytotoxic damage to whipworms. These results identify a latent IEC-intrinsic anthelmintic pathway actively constrained by Mina and point to α-defensins as important effectors that together with Mina may be attractive therapeutic targets for the control of nematode infection.


Subject(s)
Epithelial Cells/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Trichuris/immunology , Animals , Cytokines/analysis , Epithelial Cells/cytology , Intestines/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/deficiency , Nuclear Proteins/deficiency , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Transcriptome , Trichuriasis/drug therapy , Trichuriasis/immunology , Trichuriasis/pathology , Trichuris/drug effects , Trichuris/pathogenicity , alpha-Defensins/genetics , alpha-Defensins/metabolism
13.
Cell Mol Life Sci ; 75(22): 4093-4105, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30151692

ABSTRACT

Hydroxylation is a novel protein modification catalyzed by a family of oxygenases that depend on fundamental nutrients and metabolites for activity. Protein hydroxylases have been implicated in a variety of key cellular processes that play important roles in both normal homeostasis and pathogenesis. Here, in this review, we summarize the current literature on a highly conserved sub-family of oxygenases that catalyze protein histidyl hydroxylation. We discuss the evidence supporting the biochemical assignment of these emerging enzymes as ribosomal protein hydroxylases, and provide an overview of their role in immunology, bone development, and cancer.


Subject(s)
Mixed Function Oxygenases/metabolism , Ribosomes/enzymology , Animals , Histone Demethylases/metabolism , Humans , Mixed Function Oxygenases/chemistry , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism
14.
Nat Chem Biol ; 14(10): 988, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29950663

ABSTRACT

In the version of this article initially published, authors Sarah E. Wilkins, Charlotte D. Eaton, Martine I. Abboud and Maximiliano J. Katz were incorrectly included in the equal contributions footnote in the affiliations list. Footnote number seven linking to the equal contributions statement should be present only for Suzana Markolovic and Qinqin Zhuang, and the statement should read "These authors contributed equally: Suzana Markolovic, Qinqin Zhuang." The error has been corrected in the HTML and PDF versions of the article.

15.
Nat Chem Biol ; 14(7): 688-695, 2018 07.
Article in English | MEDLINE | ID: mdl-29915238

ABSTRACT

Biochemical, structural and cellular studies reveal Jumonji-C (JmjC) domain-containing 7 (JMJD7) to be a 2-oxoglutarate (2OG)-dependent oxygenase that catalyzes (3S)-lysyl hydroxylation. Crystallographic analyses reveal JMJD7 to be more closely related to the JmjC hydroxylases than to the JmjC demethylases. Biophysical and mutation studies show that JMJD7 has a unique dimerization mode, with interactions between monomers involving both N- and C-terminal regions and disulfide bond formation. A proteomic approach identifies two related members of the translation factor (TRAFAC) family of GTPases, developmentally regulated GTP-binding proteins 1 and 2 (DRG1/2), as activity-dependent JMJD7 interactors. Mass spectrometric analyses demonstrate that JMJD7 catalyzes Fe(II)- and 2OG-dependent hydroxylation of a highly conserved lysine residue in DRG1/2; amino-acid analyses reveal that JMJD7 catalyzes (3S)-lysyl hydroxylation. The functional assignment of JMJD7 will enable future studies to define the role of DRG hydroxylation in cell growth and disease.


Subject(s)
Biocatalysis , GTP Phosphohydrolases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , GTP Phosphohydrolases/chemistry , Humans , Hydroxylation , Jumonji Domain-Containing Histone Demethylases/chemistry , Models, Molecular
16.
Mol Cell ; 58(5): 729-41, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046647

ABSTRACT

Hydroxylation is an emerging modification generally catalyzed by a family of ∼70 enzymes that are dependent on oxygen, Fe(II), ascorbate, and the Kreb's cycle intermediate 2-oxoglutarate (2OG). These "2OG oxygenases" sit at the intersection of nutrient availability and metabolism where they have the potential to regulate gene expression and growth in response to changes in co-factor abundance. Characterized 2OG oxygenases regulate fundamental cellular processes by catalyzing the hydroxylation or demethylation (via hydroxylation) of DNA, RNA, or protein. As such they have been implicated in various syndromes and diseases, but particularly cancer. In this review we discuss the emerging role of 2OG oxygenases in gene expression control, examine the regulation of these unique enzymes by nutrient availability and metabolic intermediates, and describe these properties in relation to the expanding role of these enzymes in cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Animals , DNA Methylation , Gene Expression , Humans , Hydroxylation , Mixed Function Oxygenases/physiology , Neoplasms/metabolism , Protein Processing, Post-Translational
17.
Translation (Austin) ; 3(1): e1009331, 2015.
Article in English | MEDLINE | ID: mdl-26779412

ABSTRACT

The complexity of the eukaryotic protein synthesis machinery is partly driven by extensive and diverse modifications to associated proteins and RNAs. These modifications can have important roles in regulating translation factor activity and ribosome biogenesis and function. Further investigation of 'translational modifications' is warranted considering the growing evidence implicating protein synthesis as a critical point of gene expression control that is commonly deregulated in disease. New evidence suggests that translation is a major new target for oxidative modifications, specifically hydroxylations and demethylations, which generally are catalyzed by a family of emerging oxygenase enzymes that act at the interface of nutrient availability and metabolism. This review summarizes what is currently known about the role or these enzymes in targeting rRNA synthesis, protein translation and associated cellular processes.

18.
Proc Natl Acad Sci U S A ; 111(11): 4031-6, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24550447

ABSTRACT

2-Oxoglutarate (2OG) and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) is predicted to be a conserved 2OG oxygenase, the catalytic domain of which is related to hypoxia-inducible factor prolyl hydroxylases. OGFOD1 homologs in yeast are implicated in diverse cellular functions ranging from oxygen-dependent regulation of sterol response genes (Ofd1, Schizosaccharomyces pombe) to translation termination/mRNA polyadenylation (Tpa1p, Saccharomyces cerevisiae). However, neither the biochemical activity of OGFOD1 nor the identity of its substrate has been defined. Here we show that OGFOD1 is a prolyl hydroxylase that catalyzes the posttranslational hydroxylation of a highly conserved residue (Pro-62) in the small ribosomal protein S23 (RPS23). Unusually OGFOD1 retained a high affinity for, and forms a stable complex with, the hydroxylated RPS23 substrate. Knockdown or inactivation of OGFOD1 caused a cell type-dependent induction of stress granules, translational arrest, and growth impairment in a manner complemented by wild-type but not inactive OGFOD1. The work identifies a human prolyl hydroxylase with a role in translational regulation.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Prolyl Hydroxylases/metabolism , Protein Biosynthesis/physiology , Protein Processing, Post-Translational/physiology , Ribosomal Proteins/metabolism , Analysis of Variance , Carrier Proteins/genetics , Computational Biology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Hydroxylation , Immunoblotting , Immunoprecipitation , Ketoglutaric Acids/metabolism , Luciferases , Nuclear Proteins/genetics , Proline/metabolism , Protein Biosynthesis/genetics , Yeasts
19.
Mol Cell ; 53(4): 645-54, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24486019

ABSTRACT

Efficient stop codon recognition and peptidyl-tRNA hydrolysis are essential in order to terminate translational elongation and maintain protein sequence fidelity. Eukaryotic translational termination is mediated by a release factor complex that includes eukaryotic release factor 1 (eRF1) and eRF3. The N terminus of eRF1 contains highly conserved sequence motifs that couple stop codon recognition at the ribosomal A site to peptidyl-tRNA hydrolysis. We reveal that Jumonji domain-containing 4 (Jmjd4), a 2-oxoglutarate- and Fe(II)-dependent oxygenase, catalyzes carbon 4 (C4) lysyl hydroxylation of eRF1. This posttranslational modification takes place at an invariant lysine within the eRF1 NIKS motif and is required for optimal translational termination efficiency. These findings further highlight the role of 2-oxoglutarate/Fe(II) oxygenases in fundamental cellular processes and provide additional evidence that ensuring fidelity of protein translation is a major role of hydroxylation.


Subject(s)
Gene Expression Regulation , Histone Demethylases/metabolism , Mixed Function Oxygenases/chemistry , Peptide Chain Termination, Translational/genetics , Peptide Termination Factors/chemistry , Protein Biosynthesis , Amino Acid Sequence , Animals , Catalysis , Cell Line, Tumor , Codon, Terminator , HeLa Cells , Humans , Hydrolysis , Hydroxylation , Jumonji Domain-Containing Histone Demethylases , Models, Molecular , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Homology, Amino Acid
20.
Nature ; 496(7446): 461-8, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23467089

ABSTRACT

Despite their importance, the molecular circuits that control the differentiation of naive T cells remain largely unknown. Recent studies that reconstructed regulatory networks in mammalian cells have focused on short-term responses and relied on perturbation-based approaches that cannot be readily applied to primary T cells. Here we combine transcriptional profiling at high temporal resolution, novel computational algorithms, and innovative nanowire-based perturbation tools to systematically derive and experimentally validate a model of the dynamic regulatory network that controls the differentiation of mouse TH17 cells, a proinflammatory T-cell subset that has been implicated in the pathogenesis of multiple autoimmune diseases. The TH17 transcriptional network consists of two self-reinforcing, but mutually antagonistic, modules, with 12 novel regulators, the coupled action of which may be essential for maintaining the balance between TH17 and other CD4(+) T-cell subsets. Our study identifies and validates 39 regulatory factors, embeds them within a comprehensive temporal network and reveals its organizational principles; it also highlights novel drug targets for controlling TH17 cell differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Cells, Cultured , DNA/genetics , DNA/metabolism , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Genome/genetics , Interferon-gamma/biosynthesis , Interleukin-2/genetics , Mice , Mice, Inbred C57BL , Nanowires , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Silicon , Th17 Cells/immunology , Time Factors , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...