Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Trends Biochem Sci ; 49(3): 257-276, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38233282

RESUMEN

Histone lysine demethylases (KDMs) regulate eukaryotic gene transcription by catalysing the removal of methyl groups from histone proteins. These enzymes are intricately regulated by the kinase signalling system in response to internal and external stimuli. Here, we review the mechanisms by which kinase-mediated phosphorylation influence human histone KDM function. These include the changing of histone KDM subcellular localisation or chromatin binding, the altering of protein half-life, changes to histone KDM complex formation that result in histone demethylation, non-histone demethylation or demethylase-independent effects, and effects on histone KDM complex dissociation. We also explore the structural context of phospho-sites on histone KDMs and evaluate how this relates to function.


Asunto(s)
Histona Demetilasas , Histonas , Humanos , Histona Demetilasas/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Fosforilación , Desmetilación
2.
J Biol Chem ; 300(2): 105639, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199565

RESUMEN

Translation elongation factor 1A (eEF1A) is an essential and highly conserved protein required for protein synthesis in eukaryotes. In both Saccharomyces cerevisiae and human, five different methyltransferases methylate specific residues on eEF1A, making eEF1A the eukaryotic protein targeted by the highest number of dedicated methyltransferases after histone H3. eEF1A methyltransferases are highly selective enzymes, only targeting eEF1A and each targeting just one or two specific residues in eEF1A. However, the mechanism of this selectivity remains poorly understood. To reveal how S. cerevisiae elongation factor methyltransferase 4 (Efm4) specifically methylates eEF1A at K316, we have used AlphaFold-Multimer modeling in combination with crosslinking mass spectrometry (XL-MS) and enzyme mutagenesis. We find that a unique beta-hairpin motif, which extends out from the core methyltransferase fold, is important for the methylation of eEF1A K316 in vitro. An alanine mutation of a single residue on this beta-hairpin, F212, significantly reduces Efm4 activity in vitro and in yeast cells. We show that the equivalent residue in human eEF1A-KMT2 (METTL10), F220, is also important for its activity towards eEF1A in vitro. We further show that the eEF1A guanine nucleotide exchange factor, eEF1Bα, inhibits Efm4 methylation of eEF1A in vitro, likely due to competitive binding. Lastly, we find that phosphorylation of eEF1A at S314 negatively crosstalks with Efm4-mediated methylation of K316. Our findings demonstrate how protein methyltransferases can be highly selective towards a single residue on a single protein in the cell.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Humanos , Metilación , Metiltransferasas/metabolismo , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/química , Factor 1 de Elongación Peptídica/metabolismo , Fosforilación , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína , Estructura Cuaternaria de Proteína
3.
FEBS J ; 291(12): 2590-2614, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38270553

RESUMEN

Disruptor of telomeric silencing 1 (Dot1p) is an exquisitely conserved histone methyltransferase and is the sole enzyme responsible for H3K79 methylation in the budding yeast Saccharomyces cerevisiae. It has been shown to be highly phosphorylated in vivo; however, the upstream kinases that act on Dot1p are almost entirely unknown in yeast and all other eukaryotes. Here, we used in vitro and in vivo kinase discovery approaches to show that mitogen-activated protein kinase HOG1 (Hog1p) is a bona fide kinase of the Dot1p methyltransferase. In vitro kinase assays showed that Hog1p phosphorylates Dot1p at multiple sites, including at several proline-adjacent sites that are consistent with known Hog1p substrate preferences. The activity of Hog1p was specifically enhanced at these proline-adjacent sites on Dot1p upon Hog1p activation by the osmostress-responsive MAP kinase kinase PBS2 (Pbs2p). Genomic deletion of HOG1 reduced phosphorylation at specific sites on Dot1p in vivo, providing further evidence for Hog1p kinase activity on Dot1p in budding yeast cells. Phenotypic analysis of knockout and phosphosite mutant yeast strains revealed the importance of Hog1p-catalysed phosphorylation of Dot1p for cellular responses to ultraviolet-induced DNA damage. In mammalian systems, this kinase-substrate relationship was found to be conserved: human DOT1L (the ortholog of yeast Dot1p) can be phosphorylated by the proline-directed kinase p38ß (also known as MAPK11; the ortholog of yeast Hog1p) at multiple sites in vitro. Taken together, our findings establish Hog1p and p38ß as newly identified upstream kinases of the Dot1p/DOT1L H3K79 methyltransferase enzymes in eukaryotes.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Proteínas Quinasas Activadas por Mitógenos , Prolina , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Fosforilación , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Prolina/metabolismo , Metiltransferasas/metabolismo , Metiltransferasas/genética , Histonas/metabolismo , Histonas/genética , Especificidad por Sustrato , Proteínas Nucleares , Quinasas de Proteína Quinasa Activadas por Mitógenos
4.
Proc Natl Acad Sci U S A ; 120(23): e2215431120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252976

RESUMEN

Defining all sites for a post-translational modification in the cell, and identifying their upstream modifying enzymes, is essential for a complete understanding of a modification's function. However, the complete mapping of a modification in the proteome and definition of its associated enzyme-substrate network is rarely achieved. Here, we present the protein methylation network for Saccharomyces cerevisiae. Through a formal process of defining and quantifying all potential sources of incompleteness, for both the methylation sites in the proteome and also protein methyltransferases, we prove that this protein methylation network is now near-complete. It contains 33 methylated proteins and 28 methyltransferases, comprising 44 enzyme-substrate relationships, and a predicted further three enzymes. While the precise molecular function of most methylation sites is unknown, and it remains possible that other sites and enzymes remain undiscovered, the completeness of this protein modification network is unprecedented and allows us to holistically explore the role and evolution of protein methylation in the eukaryotic cell. We show that while no single protein methylation event is essential in yeast, the vast majority of methylated proteins are themselves essential, being primarily involved in the core cellular processes of transcription, RNA processing, and translation. This suggests that protein methylation in lower eukaryotes exists to fine-tune proteins whose sequences are evolutionarily constrained, providing an improvement in the efficiency of their cognate processes. The approach described here, for the construction and evaluation of post-translational modification networks and their constituent enzymes and substrates, defines a formal process of utility for other post-translational modifications.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Metilación , Proteínas de Saccharomyces cerevisiae/metabolismo , Células Eucariotas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Procesamiento Proteico-Postraduccional
5.
J Proteome Res ; 21(7): 1628-1639, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35612954

RESUMEN

Alternative splicing can lead to distinct protein isoforms. These can have different functions in specific cells and tissues or in different developmental stages. In this study, we explored whether transcripts assembled from long read, nanopore-based, direct RNA-sequencing (RNA-seq) could improve the identification of protein isoforms in human K562 cells. By comparing with Illumina-based short read RNA-seq, we showed that a large proportion of Ensembl transcripts (5949/14,326) and genes expressing alternatively spliced transcripts (486/2981) identified with long direct reads were missed by short paired-end reads. By co-analyzing proteomic and transcriptomic data, we also showed that some peptides (826/35,976), proteins (262/3215), and protein isoforms arising from distinct transcript variants (574/1212) identified with isoform-specific peptides via custom long-read-based databases were missed in Illumina-derived databases. Finally, we generated unequivocal peptide evidence for a set of protein isoforms and showed that long read, direct RNA-seq allows the discovery of novel protein isoforms not already in reference databases or custom databases built from short read RNA-seq data. Our analysis highlights the benefits of long read RNA-seq data in the generation of reference databases to increase tandem mass spectrometry (MS/MS) identification of protein isoforms.


Asunto(s)
Proteómica , Espectrometría de Masas en Tándem , Empalme Alternativo , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Péptidos/genética , Péptidos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN/metabolismo , Análisis de Secuencia de ARN , Espectrometría de Masas en Tándem/métodos , Transcriptoma
6.
Mol Cell Proteomics ; 21(7): 100249, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35609787

RESUMEN

The methylation of histidine is a post-translational modification whose function is poorly understood. Methyltransferase histidine protein methyltransferase 1 (Hpm1p) monomethylates H243 in the ribosomal protein Rpl3p and represents the only known histidine methyltransferase in Saccharomyces cerevisiae. Interestingly, the hpm1 deletion strain is highly pleiotropic, with many extraribosomal phenotypes including improved growth rates in alternative carbon sources. Here, we investigate how the loss of histidine methyltransferase Hpm1p results in diverse phenotypes, through use of targeted mass spectrometry (MS), growth assays, quantitative proteomics, and differential crosslinking MS. We confirmed the localization and stoichiometry of the H243 methylation site, found unreported sensitivities of Δhpm1 yeast to nonribosomal stressors, and identified differentially abundant proteins upon hpm1 knockout with clear links to the coordination of sugar metabolism. We adapted the emerging technique of quantitative large-scale stable isotope labeling of amino acids in cell culture crosslinking MS for yeast, which resulted in the identification of 1267 unique in vivo lysine-lysine crosslinks. By reproducibly monitoring over 350 of these in WT and Δhpm1, we detected changes to protein structure or protein-protein interactions in the ribosome, membrane proteins, chromatin, and mitochondria. Importantly, these occurred independently of changes in protein abundance and could explain a number of phenotypes of Δhpm1, not addressed by expression analysis. Further to this, some phenotypes were predicted solely from changes in protein structure or interactions and could be validated by orthogonal techniques. Taken together, these studies reveal a broad role for Hpm1p in yeast and illustrate how crosslinking MS will be an essential tool for understanding complex phenotypes.


Asunto(s)
Metiltransferasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Histidina/metabolismo , Lisina/metabolismo , Metiltransferasas/metabolismo , Proteoma/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
J Mol Biol ; 434(7): 167500, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35183557

RESUMEN

Histone lysine methylation is a key epigenetic modification that regulates eukaryotic transcription. In Saccharomyces cerevisiae, it is controlled by a reduced but evolutionarily conserved suite of methyltransferase (Set1p, Set2p, Dot1p, and Set5p) and demethylase (Jhd1p, Jhd2p, Rph1p, and Gis1p) enzymes. Many of these enzymes are extensively phosphorylated in vivo; however, the functions of almost all phosphosites remain unknown. Here, we comprehensively analyse the phosphoregulation of the yeast histone methylation network by functionally investigating 40 phosphosites on six enzymes. A total of 82 genomically-edited S. cerevisiae strains were generated through mutagenesis of sites to aspartate as a phosphomimetic or alanine as a phosphonull. These phosphosite mutants were screened for changes in native H3K4, H3K36, and H3K79 methylation levels, and for sensitivity to environmental stress conditions. For methyltransferase Set2p, we found that phosphorylation at threonine 127 significantly decreased H3K36 methylation in vivo, and that an N-terminal phosphorylation cluster at serine residues 6, 8, and 10 is required for the diamide stress response. Proteomic analysis of Set2p phosphosite mutants revealed a specific downregulation of membrane-associated proteins and processes, consistent with changes brought about by SET2 deletion and the sensitivity of mutants to diamide. For demethylase Jhd1p, we found that its sole phosphorylation site at serine 44 is required for the cold stress response. This study represents the first systematic investigation into the phosphoregulation of the epigenetic network in any eukaryote, and shows that phosphosites on histone methylation enzymes are required for a normal cellular response to stress in S.cerevisiae.


Asunto(s)
Histona Metiltransferasas , Histona Demetilasas con Dominio de Jumonji , Metiltransferasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Estrés Fisiológico , Diamida/farmacología , Histona Metiltransferasas/genética , Histona Metiltransferasas/fisiología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Metiltransferasas/genética , Metiltransferasas/fisiología , Fosforilación , Proteómica , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Serina/metabolismo
8.
J Proteome Res ; 20(5): 2420-2434, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33856219

RESUMEN

The formation of condensates in membraneless organelles is thought to be driven by protein phase separation. Arginine methylation and serine/threonine phosphorylation are important in the phase separation process; however, these post-translational modifications are often present in intrinsically disordered regions that are difficult to analyze with standard proteomic techniques. To understand their presence and co-occurrence in condensate-associated proteins, here, we use a multiprotease and multi-tandem mass spectrometry (MS/MS) fragmentation approach, coupled with heavy methyl stable isotope labeling of amino acids in cell culture (SILAC) and phospho- or methyl-peptide enrichment. For Saccharomyces cerevisiae, we report a 50% increase in the known arginine methylproteome, involving 15 proteins that are all condensate-associated. Importantly, some of these proteins have arginine methylation on all predicted sites-providing evidence that this modification can be pervasive. We explored whether arginine-methylated, condensate-associated proteins are also phosphorylated and found 12 such proteins to carry phosphorylated serine or threonine. In Npl3, Ded1, and Sbp1, single peptides were found to carry both modifications, indicating a co-occurrence in close proximity and on the same protein molecule. These co-modifications occur in regions of disorder, whereas arginine methylation is typically on regions of disorder that are also basic. For phosphorylation, its association with charged regions of condensate-associated proteins was less consistent, although some regions with multisite phosphorylation sites were strongly acidic. We conclude that arginine-methylated proteins associated with condensates are typically also modified with protein phosphorylation.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Arginina/metabolismo , ARN Helicasas DEAD-box , Metilación , Fosforilación , Procesamiento Proteico-Postraduccional , Proteómica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Espectrometría de Masas en Tándem
9.
FEBS J ; 288(19): 5668-5691, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33764612

RESUMEN

Protein arginine methyltransferase 6 (PRMT6) catalyses the asymmetric dimethylation of arginines on numerous substrate proteins within the human cell. In particular, PRMT6 methylates histone H3 arginine 2 (H3R2) which affects both gene repression and activation. However, the substrate specificity of PRMT6 has not been comprehensively analysed. Here, we systematically characterise the substrate recognition motif of PRMT6, finding that it has broad specificity and recognises the RG motif. Working with a H3 tail peptide as a template, on which we made 204 amino acid substitutions, we use targeted mass spectrometry to measure their effect on PRMT6 in vitro activity. We first show that PRMT6 methylates R2 and R8 in the H3 peptide, although H3R8 is methylated with lower efficiency and is not an in vivo PRMT6 substrate. We then quantify the effect of 194 of these amino acid substitutions on methylation at both H3R2 and H3R8. In both cases, we find that PRMT6 tolerates essentially any amino acid substitution in the H3 peptide, but that positively charged and bulky residues are preferred near the target arginine. We show that PRMT6 also has preference for glycine, but only in the position immediately following the target arginine. This indicates that PRMT6 recognises the RG motif rather than the RGG motif. We further confirm this preference for the RG motif on another PRMT6 substrate, histone H4R3. This broad specificity and recognition of RG rather than RGG are distinctive among the PRMT family and has implications for the development of drugs to selectively target PRMT6. DATABASES: Panorama Public (https://panoramaweb.org/PRMT6motif.url); ProteomeXchange (PXD016711).


Asunto(s)
Secuencias de Aminoácidos/genética , Sustitución de Aminoácidos/genética , Proteínas Nucleares/genética , Péptidos/genética , Proteína-Arginina N-Metiltransferasas/genética , Arginina/genética , Histonas/genética , Humanos , Metilación , Procesamiento Proteico-Postraduccional , Especificidad por Sustrato/genética
10.
J Biol Chem ; 296: 100192, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33334889

RESUMEN

Histone methylation is central to the regulation of eukaryotic transcription. In Saccharomyces cerevisiae, it is controlled by a system of four methyltransferases (Set1p, Set2p, Set5p, and Dot1p) and four demethylases (Jhd1p, Jhd2p, Rph1p, and Gis1p). While the histone targets for these enzymes are well characterized, the connection of the enzymes with the intracellular signaling network and thus their regulation is poorly understood; this also applies to all other eukaryotes. Here we report the detailed characterization of the eight S. cerevisiae enzymes and show that they carry a total of 75 phosphorylation sites, 92 acetylation sites, and two ubiquitination sites. All enzymes are subject to phosphorylation, although demethylases Jhd1p and Jhd2p contained one and five sites respectively, whereas other enzymes carried 14 to 36 sites. Phosphorylation was absent or underrepresented on catalytic and other domains but strongly enriched for regions of disorder on methyltransferases, suggesting a role in the modulation of protein-protein interactions. Through mutagenesis studies, we show that phosphosites within the acidic and disordered N-terminus of Set2p affect H3K36 methylation levels in vivo, illustrating the functional importance of such sites. While most kinases upstream of the yeast histone methylation enzymes remain unknown, we model the possible connections between the cellular signaling network and the histone-based gene regulatory system and propose an integrated regulatory structure. Our results provide a foundation for future, detailed exploration of the role of specific kinases and phosphosites in the regulation of histone methylation.


Asunto(s)
Histona Metiltransferasas/metabolismo , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Metilación , Fosforilación , Procesamiento Proteico-Postraduccional
11.
Int J Parasitol ; 51(4): 225-239, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33275945

RESUMEN

Diarrheal disease caused by Giardia duodenalis is highly prevalent, causing over 200 million cases globally each year. The processes that drive parasite virulence, host immune evasion and transmission involve coordinated gene expression and have been linked to epigenetic regulation. Epigenetic regulatory systems are eukaryote-conserved, including in deep branching excavates such as Giardia, with several studies already implicating histone post-translational modifications in regulation of its pathogenesis and life cycle. However, further insights into Giardia chromatin dynamics have been hindered by a lack of site-specific knowledge of histone modifications. Using mass spectrometry, we have provided the first known molecular map of histone methylation, acetylation and phosphorylation modifications in Giardia core histones. We have identified over 50 previously unreported histone modifications including sites with established roles in epigenetic regulation, and co-occurring modifications indicative of post-translational modification crosstalk. These demonstrate conserved histone modifications in Giardia which are equivalent to many other eukaryotes, and suggest that similar epigenetic mechanisms are in place in this parasite. Further, we used sequence, domain and structural homology to annotate putative histone enzyme networks in Giardia, highlighting representative chromatin modifiers which appear sufficient for identified sites, particularly those from H3 and H4 variants. This study is to our knowledge the first and most comprehensive, complete and accurate view of Giardia histone post-translational modifications to date, and a substantial step towards understanding their associations in parasite development and virulence.


Asunto(s)
Giardia lamblia , Histonas , Epigénesis Genética , Eucariontes/metabolismo , Giardia lamblia/genética , Giardia lamblia/metabolismo , Histonas/genética , Histonas/metabolismo , Espectrometría de Masas , Procesamiento Proteico-Postraduccional
12.
Mol Cell Proteomics ; 19(11): 1876-1895, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32817346

RESUMEN

Co-fractionation MS (CF-MS) is a technique with potential to characterize endogenous and unmanipulated protein complexes on an unprecedented scale. However this potential has been offset by a lack of guidelines for best-practice CF-MS data collection and analysis. To obtain such guidelines, this study thoroughly evaluates novel and published Saccharomyces cerevisiae CF-MS data sets using very high proteome coverage libraries of yeast gold standard complexes. A new method for identifying gold standard complexes in CF-MS data, Reference Complex Profiling, and the Extending 'Guilt-by-Association' by Degree (EGAD) R package are used for these evaluations, which are verified with concurrent analyses of published human data. By evaluating data collection designs, which involve fractionation of cell lysates, it is found that near-maximum recall of complexes can be achieved with fewer samples than published studies. Distributing sample collection across orthogonal fractionation methods, rather than a single high resolution data set, leads to particularly efficient recall. By evaluating 17 different similarity scoring metrics, which are central to CF-MS data analysis, it is found that two metrics rarely used in past CF-MS studies - Spearman and Kendall correlations - and the recently introduced Co-apex metric frequently maximize recall, whereas a popular metric-Euclidean distance-delivers poor recall. The common practice of integrating external genomic data into CF-MS data analysis is also evaluated, revealing that this practice may improve the precision and recall of known complexes but is generally unsuitable for predicting novel complexes in model organisms. If studying nonmodel organisms using orthologous genomic data, it is found that particular subsets of fractionation profiles (e.g. the lowest abundance quartile) should be excluded to minimize false discovery. These assessments are summarized in a series of universally applicable guidelines for precise, sensitive and efficient CF-MS studies of known complexes, and effective predictions of novel complexes for orthogonal experimental validation.


Asunto(s)
Fraccionamiento Químico/métodos , Espectrometría de Masas/métodos , Proteoma/metabolismo , Proteómica/métodos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Cromatografía en Gel , Cromatografía Liquida/métodos , Ontología de Genes , Humanos , Estándares de Referencia
13.
Mol Biol Evol ; 37(12): 3525-3549, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-32702104

RESUMEN

Methylation is a common posttranslational modification of arginine and lysine in eukaryotic proteins. Methylproteomes are best characterized for higher eukaryotes, where they are functionally expanded and evolved complex regulation. However, this is not the case for protist species evolved from the earliest eukaryotic lineages. Here, we integrated bioinformatic, proteomic, and drug-screening data sets to comprehensively explore the methylproteome of Giardia duodenalis-a deeply branching parasitic protist. We demonstrate that Giardia and related diplomonads lack arginine-methyltransferases and have remodeled conserved RGG/RG motifs targeted by these enzymes. We also provide experimental evidence for methylarginine absence in proteomes of Giardia but readily detect methyllysine. We bioinformatically infer 11 lysine-methyltransferases in Giardia, including highly diverged Su(var)3-9, Enhancer-of-zeste and Trithorax proteins with reduced domain architectures, and novel annotations demonstrating conserved methyllysine regulation of eukaryotic elongation factor 1 alpha. Using mass spectrometry, we identify more than 200 methyllysine sites in Giardia, including in species-specific gene families involved in cytoskeletal regulation, enriched in coiled-coil features. Finally, we use known methylation inhibitors to show that methylation plays key roles in replication and cyst formation in this parasite. This study highlights reduced methylation enzymes, sites, and functions early in eukaryote evolution, including absent methylarginine networks in the Diplomonadida. These results challenge the view that arginine methylation is eukaryote conserved and demonstrate that functional compensation of methylarginine was possible preceding expansion and diversification of these key networks in higher eukaryotes.


Asunto(s)
Giardia/enzimología , Proteína Metiltransferasas/metabolismo , Proteoma , Evolución Biológica , Proteínas del Citoesqueleto/metabolismo , Metilación , Trofozoítos/crecimiento & desarrollo
14.
Anal Chem ; 92(2): 1874-1882, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31851481

RESUMEN

Saccharomyces cerevisiae has the most comprehensively characterized protein-protein interaction network, or interactome, of any eukaryote. This has predominantly been generated through multiple, systematic studies of protein-protein interactions by two-hybrid techniques and of affinity-purified protein complexes. A pressing question is to understand how large-scale cross-linking mass spectrometry (XL-MS) can confirm and extend this interactome. Here, intact yeast nuclei were subject to cross-linking with disuccinimidyl sulfoxide (DSSO) and analyzed using hybrid MS2-MS3 methods. XlinkX identified a total of 2,052 unique residue pair cross-links at 1% FDR. Intraprotein cross-links were found to provide extensive structural constraint data, with almost all intralinks that mapped to known structures and slightly fewer of those mapping to homology models being within 30 Å. Intralinks provided structural information for a further 366 proteins. A method for optimizing interprotein cross-link score cut-offs was developed, through use of extensive known yeast interactions. Its application led to a high confidence, yeast nuclear interactome. Strikingly, almost half of the interactions were not previously detected by two-hybrid or AP-MS techniques. Multiple lines of evidence existed for many such interactions, whether through literature or ortholog interaction data, through multiple unique interlinks between proteins, and/or through replicates. We conclude that XL-MS is a powerful means to measure interactions, that complements two-hybrid and affinity-purification techniques.


Asunto(s)
Núcleo Celular/química , Reactivos de Enlaces Cruzados/química , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/química , Espectrometría de Masas/métodos , Proteínas Nucleares/química , Péptidos/química , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , Multimerización de Proteína , Proteínas de Saccharomyces cerevisiae/química , Succinimidas/química , Sulfóxidos/química
15.
J Mol Biol ; 432(2): 448-466, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31756331

RESUMEN

Crosstalk exists when two or more post-translational modifications, nearby in sequence or 3D space, affect each other or a protein's interactions. Saccharomyces cerevisiae protein Npl3p has six repeats of sequence SRGG, in a disordered domain, which can carry arginine methylation and serine phosphorylation. Crosstalk of the modifications controls Npl3p interactions with nuclear import, export, and other proteins. Here, we asked whether repeated SRGG motifs existed in other S. cerevisiae proteins and whether they serve a related function. Two other proteins had multiple SRGG motifs: Nop1p (fibrillarin) and Gar1p, both nucleolar proteins, which had nine and four motifs, respectively. For Nop1p, we first showed it to be extensively methylated in vivo. We then showed that the Nop1p SRGG motif is subjected to methylation by Hmt1p, phosphorylation by Sky1p, and Glc7p dephosphorylation and that there is crosstalk whereby phosphorylation blocks methylation. This is consistent with our recent motif analysis of Hmt1p, which revealed a negative specificity for acidic residues at -1 and -2 positions. On knockout of HMT1, Nop1p-GFP localization was not typically nucleolar. Conditional two-hybrid analysis, of Nop1p with C/D box small ribonuclear proteins Nop56p and Nop58p, suggested this may be associated with decreased protein-protein interactions on loss of arginine methylation. The effect of SRGG phosphorylation on the interactions of Nop1p remains unknown yet was predicted to cause a structural disorder-to-order transition in the Nop1p N-terminal domain. The SRGG motif is one of very few examples of modification crosstalk that has related functions in multiple proteins from the same species.


Asunto(s)
Secuencias de Aminoácidos/genética , Núcleo Celular/genética , Proteínas Cromosómicas no Histona/genética , Secuencias Repetitivas de Aminoácido/genética , Transporte Activo de Núcleo Celular/genética , Arginina/genética , Núcleo Celular/ultraestructura , Proteínas Cromosómicas no Histona/química , Metilación , Proteínas Nucleares/genética , Fosforilación/genética , Proteína Fosfatasa 1/genética , Proteínas Serina-Treonina Quinasas/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Ribonucleoproteínas Nucleolares Pequeñas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Serina/genética
16.
Proteomics ; 19(17): e1800444, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31328383

RESUMEN

High-resolution MS/MS spectra of peptides can be deisotoped to identify monoisotopic masses of peptide fragments. The use of such masses should improve protein identification rates. However, deisotoping is not universally used and its benefits have not been fully explored. Here, MS2-Deisotoper, a tool for use prior to database search, is used to identify monoisotopic peaks in centroided MS/MS spectra. MS2-Deisotoper works by comparing the mass and relative intensity of each peptide fragment peak to every other peak of greater mass, and by applying a set of rules concerning mass and intensity differences. After comprehensive parameter optimization, it is shown that MS2-Deisotoper can improve the number of peptide spectrum matches (PSMs) identified by up to 8.2% and proteins by up to 2.8%. It is effective with SILAC and non-SILAC MS/MS data. The identification of unique peptide sequences is also improved, increasing the number of human proteoforms by 3.7%. Detailed investigation of results shows that deisotoping increases Mascot ion scores, improves FDR estimation for PSMs, and leads to greater protein sequence coverage. At a peptide level, it is found that the efficacy of deisotoping is affected by peptide mass and charge. MS2-Deisotoper can be used via a user interface or as a command-line tool.


Asunto(s)
Isótopos de Carbono/análisis , Marcaje Isotópico/métodos , Isótopos de Nitrógeno/análisis , Fragmentos de Péptidos/análisis , Proteínas/análisis , Programas Informáticos , Espectrometría de Masas en Tándem/estadística & datos numéricos , Algoritmos , Isótopos de Carbono/química , Bases de Datos de Proteínas , Humanos , Isótopos de Nitrógeno/química , Fragmentos de Péptidos/química , Proteínas/química , Espectrometría de Masas en Tándem/métodos
17.
Cancers (Basel) ; 11(5)2019 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-31086012

RESUMEN

In this review, we describe the attributes of histone H3 mutants identified in cancer. H3 mutants were first identified in genes encoding H3.3, in pediatric high-grade glioma, and subsequently in chondrosarcomas and giant cell tumors of bone (GCTB) in adolescents. The most heavily studied are the lysine to methionine mutants K27M and K36M, which perturb the target site for specific lysine methyltransferases and dominantly perturb methylation of corresponding lysines in other histone H3 proteins. We discuss recent progress in defining the consequences of these mutations on chromatin, including a newly emerging view of the central importance of the disruption of H3K36 modification in many distinct K to M histone mutant cancers. We also review new work exploring the role of H3.3 G34 mutants identified in pediatric glioma and GCTB. G34 is not itself post-translationally modified, but G34 mutation impinges on the modification of H3K36. Here, we ask if G34R mutation generates a new site for methylation on the histone tail. Finally, we consider evidence indicating that histone mutations might be more widespread in cancer than previously thought, and if the perceived bias towards mutation of H3.3 is real or reflects the biology of tumors in which the histone mutants were first identified.

18.
Mol Cell Proteomics ; 17(12): 2462-2479, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30206180

RESUMEN

Hmt1p is the predominant arginine methyltransferase in Saccharomyces cerevisiae Its substrate proteins are involved in transcription, transcriptional regulation, nucleocytoplasmic transport and RNA splicing. Hmt1p-catalyzed methylation can also modulate protein-protein interactions. Hmt1p is conserved from unicellular eukaryotes through to mammals where its ortholog, PRMT1, is lethal upon knockout. In yeast, however, the effect of knockout on the transcriptome and proteome has not been described. Transcriptome analysis revealed downregulation of phosphate-responsive genes in hmt1Δ, including acid phosphatases PHO5, PHO11, and PHO12, phosphate transporters PHO84 and PHO89 and the vacuolar transporter chaperone VTC3 Analysis of the hmt1Δ proteome revealed decreased abundance of phosphate-associated proteins including phosphate transporter Pho84p, vacuolar alkaline phosphatase Pho8p, acid phosphatase Pho3p and subunits of the vacuolar transporter chaperone complex Vtc1p, Vtc3p and Vtc4p. Consistent with this, phosphate homeostasis was dysregulated in hmt1Δ cells, showing decreased extracellular phosphatase levels and decreased total Pi in phosphate-depleted medium. In vitro, we showed that transcription factor Pho4p can be methylated at Arg-241, which could explain phosphate dysregulation in hmt1Δ if interplay exists with phosphorylation at Ser-242 or Ser-243, or if Arg-241 methylation affects the capacity of Pho4p to homodimerize or interact with Pho2p. However, the Arg-241 methylation site was not validated in vivo and the localization of a Pho4p-GFP fusion in hmt1Δ was not different from wild type. To our knowledge, this is the first study to reveal an association between Hmt1p and phosphate homeostasis and one which suggests a regulatory link between S-adenosyl methionine and intracellular phosphate.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fosfatos/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Fosfatasa Ácida/genética , Arginina/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Homeostasis/genética , Metilación , Microscopía Fluorescente , Proteoma/genética , Espectrometría de Masas en Tándem , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
19.
J Proteome Res ; 17(10): 3485-3491, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30156103

RESUMEN

Protein methyltransferases often recognize their substrates through linear sequence motifs. The determination of these motifs is critical to understand methyltransferase mechanism, function, and drug targeting. Here we describe MT-MAMS (methyltransferase motif analysis by mass spectrometry), a quantitative approach to characterize methyltransferase substrate recognition motifs. In MT-MAMS, peptide sets are synthesized which contain all amino acid substitutions at single positions within a template sequence. These are then incubated with the methyltransferase of interest in the presence of deuterated S-adenosyl methionine (D3-AdoMet). The use of this heavy methyl donor gives unique mass shifts to methylated peptides, allowing their unambiguous quantification by mass spectrometry. The stoichiometry of methylation resulting from each substitution is then derived, and finally the methyltransferase substrate recognition motif is generated. We validated MT-MAMS by application to lysine methyltransferase G9a, generating the substrate recognition motif (TKRN)-(A > RS > G)-(R ≫ K)-K-(STRCKMAQHG)-Φ; this is highly similar to that previously determined by peptide arrays. We then determined the recognition motif of yeast lysine elongation factor methyltransferase 1 (Efm1) to be (Y > FW)-K-^P-G-G-Φ. This is a new type of lysine methyltransferase recognition motif that only contains noncharged residues, excluding the target lysine. We further determined recognition motifs of major yeast and human arginine methyltransferases Hmt1 and PRMT1, revealing them to be ^(DE)-^(DE)-R-(G ≫ A)-(GN > RAW)-(FYW > ILKHM) and ^(DE)-^(DE)-R-(G ≫ N)-(GR > ANK)-(K > YHMFILW), respectively. These motifs expand significantly on the canonical RGG recognition motif and include the negative specificity of these enzymes, a feature unique to MT-MAMS. Finally, we show that MT-MAMS can be used to generate insights into the processivity of protein methyltransferases.


Asunto(s)
Secuencias de Aminoácidos , Espectrometría de Masas/métodos , Péptidos/metabolismo , Proteína Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Humanos , Metilación , Proteína Metiltransferasas/genética , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
20.
Trends Biochem Sci ; 43(3): 211-223, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29398204

RESUMEN

Eukaryotic elongation factor 1A (eEF1A) is an essential and highly conserved protein involved in diverse cellular processes, including translation, cytoskeleton organisation, nuclear export, and proteasomal degradation. Recently, nine novel and site-specific methyltransferases were discovered that target eEF1A, five in yeast and four in human, making it the eukaryotic protein with the highest number of independent methyltransferases. Some of these methyltransferases show striking evolutionary conservation. Yet, they come from diverse methyltransferase families, indicating they confer competitive advantage through independent origins. As might be expected, the first functional studies of specific methylation sites found them to have distinct effects, notably on eEF1A-related processes of translation and tRNA aminoacylation. Further functional studies of sites will likely reveal other unique roles for this interesting modification.


Asunto(s)
Factor 1 de Elongación Peptídica/química , Factor 1 de Elongación Peptídica/metabolismo , Humanos , Metilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA