Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Clin Exp Immunol ; 216(1): 45-54, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38133636

ABSTRACT

Cold agglutinin disease (CAD) is a rare B-cell lymphoproliferative disorder of the bone marrow, manifested by autoimmune hemolytic anemia caused by binding of monoclonal IgM autoantibodies to the I antigen. Underlying genetic changes have previously been reported, but their impact on gene expression profile has been unknown. Here, we define differentially expressed genes in CAD B cells. To unravel downstream alteration in cellular pathways, gene expression by RNA sequencing was undertaken. Clonal B-cell samples from 12 CAD patients and IgM-expressing memory B cells from 4 healthy individuals were analyzed. Differential expression analysis and filtering resulted in 93 genes with significant differential expression. Top upregulated genes included SLC4A1, SPTA1, YBX3, TESC, HBD, AHSP, TRAF1, HBA2, RHAG, CA1, SPTB, IL10, UBASH3B, ALAS2, HBA1, CRYM, RGCC, KANK2, and IGHV4-34. They were upregulated at least 8-fold, while complement receptor 1 (CR1/CD35) was downregulated 11-fold in clonal CAD B cells compared to control B cells. Flow cytometry analyses further confirmed reduced CR1 (CD35) protein expression by clonal CAD IgM+ B cells compared to IgM+ memory B cells in controls. CR1 (CD35) is an important negative regulator of B-cell activation and differentiation. Therefore, reduced CR1 (CD35) expression may increase activation, proliferation, and antibody production in CAD-associated clonal B cells.


Subject(s)
Anemia, Hemolytic, Autoimmune , Humans , Anemia, Hemolytic, Autoimmune/genetics , Anemia, Hemolytic, Autoimmune/metabolism , Down-Regulation , Receptors, Complement 3b/genetics , B-Lymphocytes , Immunoglobulin M , Gene Expression Profiling , Blood Proteins/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism
2.
Cell Rep Med ; 4(11): 101265, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37944527

ABSTRACT

Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.


Subject(s)
Chronic Pain , Humans , Mice , Animals , Chronic Pain/chemically induced , Chronic Pain/metabolism , Sensory Receptor Cells/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Inflammation/metabolism , Mitochondria/metabolism
3.
PLoS One ; 18(6): e0287558, 2023.
Article in English | MEDLINE | ID: mdl-37347777

ABSTRACT

The methyltransferase-like protein 13 (METTL13) methylates the eukaryotic elongation factor 1 alpha (eEF1A) on two locations: the N-terminal amino group and lysine 55. The absence of this methylation leads to reduced protein synthesis and cell proliferation in human cancer cells. Previous studies showed that METTL13 is dispensable in non-transformed cells, making it potentially interesting for cancer therapy. However, METTL13 has not been examined yet in whole animals. Here, we used the nematode Caenorhabditis elegans as a simple model to assess the functions of METTL13. Using methyltransferase assays and mass spectrometry, we show that the C. elegans METTL13 (METL-13) methylates eEF1A (EEF-1A) in the same way as the human protein. Crucially, the cancer-promoting role of METL-13 is also conserved and depends on the methylation of EEF-1A, like in human cells. At the same time, METL-13 appears dispensable for animal growth, development, and stress responses. This makes C. elegans a convenient whole-animal model for studying METL13-dependent carcinogenesis without the complications of interfering with essential wild-type functions.


Subject(s)
Neoplasms , Protein Methyltransferases , Animals , Humans , Caenorhabditis elegans/genetics , Methyltransferases/genetics , Carcinogenesis , Peptide Elongation Factor 1/genetics
4.
J Biol Chem ; 299(6): 104661, 2023 06.
Article in English | MEDLINE | ID: mdl-36997089

ABSTRACT

Lysine methylation is an abundant posttranslational modification, which has been most intensively studied in the context of histone proteins, where it represents an important epigenetic mark. Lysine methylation of histone proteins is primarily catalyzed by SET-domain methyltransferases (MTases). However, it has recently become evident that also another MTase family, the so-called seven-ß-strand (7BS) MTases, often denoted METTLs (methyltransferase-like), contains several lysine (K)-specific MTases (KMTs). These enzymes catalyze the attachment of up to three methyl groups to lysine residues in specific substrate proteins, using S-adenosylmethionine (AdoMet) as methyl donor. About a decade ago, only a single human 7BS KMT was known, namely the histone-specific DOT1L, but 15 additional 7BS KMTs have now been discovered and characterized. These KMTs typically target a single nonhistone substrate that, in most cases, belongs to one of the following three protein groups: components of the cellular protein synthesis machinery, mitochondrial proteins, and molecular chaperones. This article provides an extensive overview and discussion of the human 7BS KMTs and their biochemical and biological roles.


Subject(s)
Lysine , Methyltransferases , Humans , Methyltransferases/metabolism , Methylation , Lysine/metabolism , Protein Conformation, beta-Strand , Histones/metabolism , Protein Processing, Post-Translational , Protein Methyltransferases/metabolism
5.
Nat Commun ; 13(1): 4883, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35986016

ABSTRACT

How animals rewire cellular programs to survive cold is a fascinating problem with potential biomedical implications, ranging from emergency medicine to space travel. Studying a hibernation-like response in the free-living nematode Caenorhabditis elegans, we uncovered a regulatory axis that enhances the natural resistance of nematodes to severe cold. This axis involves conserved transcription factors, DAF-16/FoxO and PQM-1, which jointly promote cold survival by upregulating FTN-1, a protein related to mammalian ferritin heavy chain (FTH1). Moreover, we show that inducing expression of FTH1 also promotes cold survival of mammalian neurons, a cell type particularly sensitive to deterioration in hypothermia. Our findings in both animals and cells suggest that FTN-1/FTH1 facilitates cold survival by detoxifying ROS-generating iron species. We finally show that mimicking the effects of FTN-1/FTH1 with drugs protects neurons from cold-induced degeneration, opening a potential avenue to improved treatments of hypothermia.


Subject(s)
Caenorhabditis elegans Proteins , Hypothermia , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Ferritins/genetics , Ferritins/metabolism , Forkhead Transcription Factors/metabolism , Iron/metabolism , Mammals/metabolism , Mice , Neurons/metabolism
6.
J Biol Chem ; 298(4): 101791, 2022 04.
Article in English | MEDLINE | ID: mdl-35247388

ABSTRACT

Many proteins are modified by posttranslational methylation, introduced by a number of methyltransferases (MTases). Protein methylation plays important roles in modulating protein function and thus in optimizing and regulating cellular and physiological processes. Research has mainly focused on nuclear and cytosolic protein methylation, but it has been known for many years that also mitochondrial proteins are methylated. During the last decade, significant progress has been made on identifying the MTases responsible for mitochondrial protein methylation and addressing its functional significance. In particular, several novel human MTases have been uncovered that methylate lysine, arginine, histidine, and glutamine residues in various mitochondrial substrates. Several of these substrates are key components of the bioenergetics machinery, e.g., respiratory Complex I, citrate synthase, and the ATP synthase. In the present review, we report the status of the field of mitochondrial protein methylation, with a particular emphasis on recently discovered human MTases. We also discuss evolutionary aspects and functional significance of mitochondrial protein methylation and present an outlook for this emergent research field.


Subject(s)
Methyltransferases , Mitochondrial Proteins , Protein Processing, Post-Translational , Humans , Methylation , Methyltransferases/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
8.
Nucleic Acids Res ; 49(6): 3185-3203, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33693809

ABSTRACT

Protein methylation occurs primarily on lysine and arginine, but also on some other residues, such as histidine. METTL18 is the last uncharacterized member of a group of human methyltransferases (MTases) that mainly exert lysine methylation, and here we set out to elucidate its function. We found METTL18 to be a nuclear protein that contains a functional nuclear localization signal and accumulates in nucleoli. Recombinant METTL18 methylated a single protein in nuclear extracts and in isolated ribosomes from METTL18 knockout (KO) cells, identified as 60S ribosomal protein L3 (RPL3). We also performed an RPL3 interactomics screen and identified METTL18 as the most significantly enriched MTase. We found that His-245 in RPL3 carries a 3-methylhistidine (3MH; τ-methylhistidine) modification, which was absent in METTL18 KO cells. In addition, both recombinant and endogenous METTL18 were found to be automethylated at His-154, thus further corroborating METTL18 as a histidine-specific MTase. Finally, METTL18 KO cells displayed altered pre-rRNA processing, decreased polysome formation and codon-specific changes in mRNA translation, indicating that METTL18-mediated methylation of RPL3 is important for optimal ribosome biogenesis and function. In conclusion, we have here established METTL18 as the second human histidine-specific protein MTase, and demonstrated its functional relevance.


Subject(s)
Protein Biosynthesis , Protein Methyltransferases/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Amino Acid Motifs , Cell Nucleolus/enzymology , HEK293 Cells , HeLa Cells , Histidine/metabolism , Humans , Nuclear Localization Signals , Protein Methyltransferases/chemistry , RNA Processing, Post-Transcriptional , Ribosomal Protein L3 , Ribosomes/metabolism
9.
Nat Commun ; 12(1): 891, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33563959

ABSTRACT

Post-translational methylation plays a crucial role in regulating and optimizing protein function. Protein histidine methylation, occurring as the two isomers 1- and 3-methylhistidine (1MH and 3MH), was first reported five decades ago, but remains largely unexplored. Here we report that METTL9 is a broad-specificity methyltransferase that mediates the formation of the majority of 1MH present in mouse and human proteomes. METTL9-catalyzed methylation requires a His-x-His (HxH) motif, where "x" is preferably a small amino acid, allowing METTL9 to methylate a number of HxH-containing proteins, including the immunomodulatory protein S100A9 and the NDUFB3 subunit of mitochondrial respiratory Complex I. Notably, METTL9-mediated methylation enhances respiration via Complex I, and the presence of 1MH in an HxH-containing peptide reduced its zinc binding affinity. Our results establish METTL9-mediated 1MH as a pervasive protein modification, thus setting the stage for further functional studies on protein histidine methylation.


Subject(s)
Methylhistidines/metabolism , Methyltransferases/metabolism , Proteome/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Histidine/metabolism , Humans , Mammals/classification , Mammals/genetics , Mammals/metabolism , Methylation , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mutation , Protein Processing, Post-Translational , Proteome/chemistry , Substrate Specificity , Zinc/metabolism
10.
Nucleic Acids Res ; 48(2): 830-846, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31799605

ABSTRACT

RNA methylations are essential both for RNA structure and function, and are introduced by a number of distinct methyltransferases (MTases). In recent years, N6-methyladenosine (m6A) modification of eukaryotic mRNA has been subject to intense studies, and it has been demonstrated that m6A is a reversible modification that regulates several aspects of mRNA function. However, m6A is also found in other RNAs, such as mammalian 18S and 28S ribosomal RNAs (rRNAs), but the responsible MTases have remained elusive. 28S rRNA carries a single m6A modification, found at position A4220 (alternatively referred to as A4190) within a stem-loop structure, and here we show that the MTase ZCCHC4 is the enzyme responsible for introducing this modification. Accordingly, we found that ZCCHC4 localises to nucleoli, the site of ribosome assembly, and that proteins involved in RNA metabolism are overrepresented in the ZCCHC4 interactome. Interestingly, the absence of m6A4220 perturbs codon-specific translation dynamics and shifts gene expression at the translational level. In summary, we establish ZCCHC4 as the enzyme responsible for m6A modification of human 28S rRNA, and demonstrate its functional significance in mRNA translation.


Subject(s)
Adenosine/analogs & derivatives , Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Ribosomal, 28S/genetics , Adenosine/chemistry , Adenosine/genetics , Catalysis , Humans , Methylation , Methyltransferases/chemistry , Protein Binding/genetics , RNA, Ribosomal, 28S/chemistry
11.
J Biol Chem ; 294(31): 11654-11664, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31213526

ABSTRACT

Lysine methylation is a common posttranslational modification of nuclear and cytoplasmic proteins but is also present in mitochondria. The human protein denoted "family with sequence similarity 173 member B" (FAM173B) was recently uncovered as a mitochondrial lysine (K)-specific methyltransferase (KMT) targeting the c-subunit of mitochondrial ATP synthase (ATPSc), and was therefore renamed ATPSc-KMT. We here set out to investigate the biochemical function of its yet uncharacterized paralogue FAM173A. We demonstrate that FAM173A localizes to mitochondria, mediated by a noncanonical targeting sequence that is partially retained in the mature protein. Immunoblotting analysis using methyllysine-specific antibodies revealed that FAM173A knock-out (KO) abrogates lysine methylation of a single mitochondrial protein in human cells. Mass spectrometry analysis identified this protein as adenine nucleotide translocase (ANT), represented by two highly similar isoforms ANT2 and ANT3. We found that methylation occurs at Lys-52 of ANT, which was previously reported to be trimethylated. Complementation of KO cells with WT or enzyme-dead FAM173A indicated that the enzymatic activity of FAM173A is required for ANT methylation at Lys-52 to occur. Both in human cells and in rat organs, Lys-52 was exclusively trimethylated, indicating that this modification is constitutive, rather than regulatory and dynamic. Moreover, FAM173A-deficient cells displayed increased mitochondrial respiration compared with FAM173A-proficient cells. In summary, we demonstrate that FAM173A is the long-sought KMT responsible for ANT methylation at Lys-52, and point out the functional significance of Lys-52 methylation in ANT. Based on the established naming nomenclature for KMTs, we propose to rename FAM173A to ANT-KMT (gene name ANTKMT).


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Proteins/metabolism , Protein Methyltransferases/metabolism , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Liver/metabolism , Lysine/metabolism , Mass Spectrometry , Methylation , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Peptides/analysis , Protein Methyltransferases/genetics , Rats , Sequence Alignment
12.
J Biol Chem ; 294(4): 1128-1141, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30530489

ABSTRACT

Lysine methylation is an important post-translational modification that is also present on mitochondrial proteins, but the mitochondrial lysine-specific methyltransferases (KMTs) responsible for modification are in most cases unknown. Here, we set out to determine the function of human family with sequence similarity 173 member B (FAM173B), a mitochondrial methyltransferase (MTase) reported to promote chronic pain. Using bioinformatics analyses and biochemical assays, we found that FAM173B contains an atypical, noncleavable mitochondrial targeting sequence responsible for its localization to mitochondria. Interestingly, CRISPR/Cas9-mediated KO of FAM173B in mammalian cells abrogated trimethylation of Lys-43 in ATP synthase c-subunit (ATPSc), a modification previously reported as ubiquitous among metazoans. ATPSc methylation was restored by complementing the KO cells with enzymatically active human FAM173B or with a putative FAM173B orthologue from the nematode Caenorhabditis elegans Interestingly, lack of Lys-43 methylation caused aberrant incorporation of ATPSc into the ATP synthase complex and resulted in decreased ATP-generating ability of the complex, as well as decreased mitochondrial respiration. In summary, we have identified FAM173B as the long-sought KMT responsible for methylation of ATPSc, a key protein in cellular ATP production, and have demonstrated functional significance of ATPSc methylation. We suggest renaming FAM173B to ATPSc-KMT (gene name ATPSCKMT).


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Cell Line , Computational Biology , HeLa Cells , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mice , Mitochondria/metabolism
13.
Nat Commun ; 9(1): 3411, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143613

ABSTRACT

Eukaryotic elongation factor 1 alpha (eEF1A) delivers aminoacyl-tRNA to the ribosome and thereby plays a key role in protein synthesis. Human eEF1A is subject to extensive post-translational methylation, but several of the responsible enzymes remain unknown. Using a wide range of experimental approaches, we here show that human methyltransferase (MTase)-like protein 13 (METTL13) contains two distinct MTase domains targeting the N terminus and Lys55 of eEF1A, respectively. Our biochemical and structural analyses provide detailed mechanistic insights into recognition of the eEF1A N terminus by METTL13. Moreover, through ribosome profiling, we demonstrate that loss of METTL13 function alters translation dynamics and results in changed translation rates of specific codons. In summary, we here unravel the function of a human MTase, showing that it methylates eEF1A and modulates mRNA translation in a codon-specific manner.


Subject(s)
Codon/genetics , Methyltransferases/metabolism , Peptide Elongation Factor 1/metabolism , Humans , Methyltransferases/chemistry , Methyltransferases/genetics , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor 1/genetics , Protein Binding , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Ribosomes/metabolism
14.
RNA Biol ; 15(3): 314-319, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29447067

ABSTRACT

Lysine methylation is a frequent post-translational protein modification, which has been intensively studied in the case of histone proteins. Lysine methylations are also found on many non-histone proteins, and one prominent example is eukaryotic elongation factor 1 alpha (eEF1A). Besides its essential role in the protein synthesis machinery, a number of non-canonical functions have also been described for eEF1A, such as regulation of the actin cytoskeleton and the promotion of viral replication. The functional significance of the extensive lysine methylations on eEF1A, as well as the identity of the responsible lysine methyltransferases (KMTs), have until recently remained largely elusive. However, recent discoveries and characterizations of human eEF1A-specific KMTs indicate that lysine methylation of eEF1A can be dynamic and inducible, and modulates mRNA translation in a codon-specific fashion. Here, we give a general overview of eEF1A lysine methylation and discuss its possible functional and regulatory significance, with particular emphasis on newly discovered human KMTs.


Subject(s)
Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-1/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Lysine/chemistry , Actin Cytoskeleton/metabolism , Humans , Methylation , Models, Molecular , Protein Conformation , Protein Processing, Post-Translational , Virus Replication
15.
PLoS Biol ; 16(2): e2003452, 2018 02.
Article in English | MEDLINE | ID: mdl-29444090

ABSTRACT

Chronic pain is a debilitating problem, and insights in the neurobiology of chronic pain are needed for the development of novel pain therapies. A genome-wide association study implicated the 5p15.2 region in chronic widespread pain. This region includes the coding region for FAM173B, a functionally uncharacterized protein. We demonstrate here that FAM173B is a mitochondrial lysine methyltransferase that promotes chronic pain. Knockdown and sensory neuron overexpression strategies showed that FAM173B is involved in persistent inflammatory and neuropathic pain via a pathway dependent on its methyltransferase activity. FAM173B methyltransferase activity in sensory neurons hyperpolarized mitochondria and promoted macrophage/microglia activation through a reactive oxygen species-dependent pathway. In summary, we uncover a role for methyltransferase activity of FAM173B in the neurobiology of pain. These results also highlight FAM173B methyltransferase activity as a potential therapeutic target to treat debilitating chronic pain conditions.


Subject(s)
Chronic Pain/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Animals , Chromosomes, Human, Pair 5 , Chronic Pain/genetics , Female , Gene Knockdown Techniques , Genome-Wide Association Study , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Male , Mice, Inbred C57BL , Microglia/metabolism , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism
17.
J Biol Chem ; 292(43): 17950-17962, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28887308

ABSTRACT

Lysine methylation is an important and much-studied posttranslational modification of nuclear and cytosolic proteins but is present also in mitochondria. However, the responsible mitochondrial lysine-specific methyltransferases (KMTs) remain largely elusive. Here, we investigated METTL12, a mitochondrial human S-adenosylmethionine (AdoMet)-dependent methyltransferase and found it to methylate a single protein in mitochondrial extracts, identified as citrate synthase (CS). Using several in vitro and in vivo approaches, we demonstrated that METTL12 methylates CS on Lys-395, which is localized in the CS active site. Interestingly, the METTL12-mediated methylation inhibited CS activity and was blocked by the CS substrate oxaloacetate. Moreover, METTL12 was strongly inhibited by the reaction product S-adenosylhomocysteine (AdoHcy). In summary, we have uncovered a novel human mitochondrial KMT that introduces a methyl modification into a metabolic enzyme and whose activity can be modulated by metabolic cues. Based on the established naming nomenclature for similar enzymes, we suggest that METTL12 be renamed CS-KMT (gene name CSKMT).


Subject(s)
Citrate (si)-Synthase/metabolism , Methyltransferases/metabolism , Mitochondrial Proteins/metabolism , Oxaloacetic Acid/metabolism , S-Adenosylhomocysteine/metabolism , Citrate (si)-Synthase/genetics , HeLa Cells , Humans , Methylation , Methyltransferases/classification , Methyltransferases/genetics , Mitochondrial Proteins/classification , Mitochondrial Proteins/genetics
18.
Nucleic Acids Res ; 45(14): 8239-8254, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28520920

ABSTRACT

Many cellular proteins are methylated on lysine residues and this has been most intensively studied for histone proteins. Lysine methylations on non-histone proteins are also frequent, but in most cases the functional significance of the methylation event, as well as the identity of the responsible lysine (K) specific methyltransferase (KMT), remain unknown. Several recently discovered KMTs belong to the so-called seven-ß-strand (7BS) class of MTases and we have here investigated an uncharacterized human 7BS MTase currently annotated as part of the endothelin converting enzyme 2, but which should be considered a separate enzyme. Combining in vitro enzymology and analyzes of knockout cells, we demonstrate that this MTase efficiently methylates K36 in eukaryotic translation elongation factor 1 alpha (eEF1A) in vitro and in vivo. We suggest that this novel KMT is named eEF1A-KMT4 (gene name EEF1AKMT4), in agreement with the recently established nomenclature. Furthermore, by ribosome profiling we show that the absence of K36 methylation affects translation dynamics and changes translation speed of distinct codons. Finally, we show that eEF1A-KMT4 is part of a novel family of human KMTs, defined by a shared sequence motif in the active site and we demonstrate the importance of this motif for catalytic activity.


Subject(s)
Eukaryotic Initiation Factor-1/metabolism , Methyltransferases/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , Electrophoresis, Polyacrylamide Gel , Eukaryotic Initiation Factor-1/genetics , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase , Humans , Lysine/genetics , Lysine/metabolism , Methylation , Methyltransferases/genetics , Phylogeny , RNA, Messenger/genetics , Sequence Homology, Amino Acid
19.
Nucleic Acids Res ; 45(8): 4370-4389, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28108655

ABSTRACT

Lysine methylation is abundant on histone proteins, representing a dynamic regulator of chromatin state and gene activity, but is also frequent on many non-histone proteins, including eukaryotic elongation factor 1 alpha (eEF1A). However, the functional significance of eEF1A methylation remains obscure and it has remained unclear whether eEF1A methylation is dynamic and subject to active regulation. We here demonstrate, using a wide range of in vitro and in vivo approaches, that the previously uncharacterized human methyltransferase METTL21B specifically targets Lys-165 in eEF1A in an aminoacyl-tRNA- and GTP-dependent manner. Interestingly, METTL21B-mediated eEF1A methylation showed strong variation across different tissues and cell lines, and was induced by altering growth conditions or by treatment with certain ER-stress-inducing drugs, concomitant with an increase in METTL21B gene expression. Moreover, genetic ablation of METTL21B function in mammalian cells caused substantial alterations in mRNA translation, as measured by ribosomal profiling. A non-canonical function for eEF1A in organization of the cellular cytoskeleton has been reported, and interestingly, METTL21B accumulated in centrosomes, in addition to the expected cytosolic localization. In summary, the present study identifies METTL21B as the enzyme responsible for methylation of eEF1A on Lys-165 and shows that this modification is dynamic, inducible and likely of regulatory importance.


Subject(s)
Lysine/metabolism , Methyltransferases/genetics , Peptide Elongation Factor 1/genetics , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Transfer, Amino Acyl/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Gene Expression Regulation , Guanosine Triphosphate/metabolism , Humans , Methyltransferases/chemistry , Methyltransferases/metabolism , Organ Specificity , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor 1/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Messenger/metabolism , RNA, Transfer, Amino Acyl/metabolism , Rats , Sequence Alignment , Sequence Homology, Amino Acid
20.
Biochem J ; 473(14): 1995-2009, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27407169

ABSTRACT

Methylation of biomolecules is a frequent biochemical reaction within the cell, and a plethora of highly specific methyltransferases (MTases) catalyse the transfer of a methyl group from S-adenosylmethionine (AdoMet) to various substrates. The posttranslational methylation of lysine residues, catalysed by numerous lysine (K)-specific protein MTases (KMTs), is a very common and important protein modification, which recently has been subject to intense studies, particularly in the case of histone proteins. The majority of KMTs belong to a class of MTases that share a defining 'SET domain', and these enzymes mostly target lysines in the flexible tails of histones. However, the so-called seven-ß-strand (7BS) MTases, characterized by a twisted beta-sheet structure and certain conserved sequence motifs, represent the largest MTase class, and these enzymes methylate a wide range of substrates, including small metabolites, lipids, nucleic acids and proteins. Until recently, the histone-specific Dot1/DOT1L was the only identified eukaryotic 7BS KMT. However, a number of novel 7BS KMTs have now been discovered, and, in particular, several recently characterized human and yeast members of MTase family 16 (MTF16) have been found to methylate lysines in non-histone proteins. Here, we review the status and recent progress on the 7BS KMTs, and discuss these enzymes at the levels of sequence/structure, catalytic mechanism, substrate recognition and biological significance.


Subject(s)
Lysine/metabolism , Methyltransferases/metabolism , Animals , Histone-Lysine N-Methyltransferase , Humans , Methylation , Protein Processing, Post-Translational , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...