• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 1
  • Tagged with
  • 7
  • 7
  • 7
  • 6
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Kinetic Mechanism and Inhibitory Study of Protein Arginine Methyltransferase 1

Feng, You 28 July 2012 (has links)
Protein arginine methyltransferase 1 (PRMT1) is a key posttranslational modification enzyme that catalyzes the methylation of specific arginine residues in histone and nonhistone protein substrates, regulating diverse cellular processes such as transcriptional initiation, RNA splicing, DNA repair, and signal transduction. Recently the essential roles of PRMT1 in cancer and cardiovascular complications have intrigued much attention. Developing effective PRMT inhibitors therefore is of significant therapeutic value. The research on PRMT inhibitor development however is greatly hindered by poor understanding of the biochemical basis of protein arginine methylation and lack of effective assays for PRMT1 inhibitor screening. Herein, we report our effort in the kinetic mechanism study as well as the fluorescent probe and inhibitor development for PRMT1. New fluorescent reporters were designed and applied to perform single-step analysis of substrate binding and methylation of PRMT1. Using these reporters, we performed transient-state fluorescence measurements to dissect the rate constants along the PRMT1 catalytic coordinate. The data give evidence that the chemistry of methyl transfer is the major rate-limiting step, and that binding of the cofactor SAM or SAH affects the association and dissociation of H4 with PRMT1. Importantly, we identified a critical kinetic step suggesting a precatalytic conformational transition induced by substrate binding. On the other hand, we discovered a type of naphthyl-sulfo (NS) compounds that block PRMT1- mediated arginine methylation at micromolar potency through a unique mechanism: they directly target the substrates but not PRMT enzymes for the observed inhibition. We also found that suramin, an anti-parasite and anti-cancer drug bearing similar functional groups, effectively inhibited PRMT1 mediated methylation. These findings about novel PRMT inhibitors and their unique inhibition mechanism provide a new way for chemical regulation of protein arginine methylation. Addionally, to dissect the interplaying relationship between different histone modification marks, we investigated how individual lysine acetylations and their different combinations at the H4 tail affect Arg-3 methylation in cis. Our data reveal that the effect of lysine acetylation on arginine methylation depends on the site of acetylation and the type of methylation. While certain acetylations present a repressive impact on PRMT-1 mediated methylation (type I methylation), lysine acetylation generally is correlated with enhanced methylation by PRMT5 (type II dimethylation). In particular, Lys-5 acetylation decreases activity of PRMT1 but increases that of PRMT5. Furthermore, hyperacetylation increases the content of ordered secondary structures of H4 tail. These findings provide new insights into the regulatory mechanism of Arg-3 methylation by H4 acetylation, and unravel that complex intercommunications exist between different posttranslational marks in cis.
2

Development of Novel Methods and their Utilization in the Analysis of the Effect of the N-terminus of Human Protein Arginine Methyltransferase 1 Variant 1 on Enzymatic Activity, Protein-protein Interactions, and Substrate Specificity

Suh-Lailam, Brenda Bienka 01 May 2010 (has links)
Protein arginine methyltransferases (PRMTs) are enzymes that catalyze the methylation of protein arginine residues, resulting in the formation of monomethylarginine, and/or asymmetric or symmetric dimethylarginines. Although understanding of the PRMTs has grown rapidly over the last few years, several challenges still remain in the PRMT field. Here, we describe the development of two techniques that will be very useful in investigating PRMT regulation, small molecule inhibition, oligomerization, protein-protein interaction, and substrate specificity, which will ultimately lead to the advancement of the PRMT field. Studies have shown that having an N-terminal tag can influence enzyme activity and substrate specificity. The first protocol tackles this problem by developing a way to obtain active untagged recombinant PRMT proteins. The second protocol describes a fast and efficient method for quantitative measurement of AdoMet-dependent methyltranseferase activity with protein substrates. In addition to being very sensitive, this method decreases the processing time for the analysis of PRMT activity to a few minutes compared to weeks by traditional methods, and generates 3000-fold less radioactive waste. We then used these methods to investigate the effect of truncating the NT of human PRMT1 variant 1 (hPRMT1-V1) on enzyme activity, protein-protein interactions, and substrate specificity. Our studies show that the NT of hPRMT1-V1 influences enzymatic activity and protein-protein interactions. In particular, methylation of a variety of protein substrates was more efficient when the first 10 amino acids of hPRMT1v1 were removed, suggesting an autoinhibitory role for this small section of the N-terminus. Likewise, as portions of the NT were removed, the altered hPRMT1v1 constructs were able to interact with more proteins. Overall, my studies suggest the the sequence and length of the NT of hPRMT1v1 is capable of enforcing specific protein interactions.
3

New Roles for Arginine Methylation in RNA Metabolism and Cancer

Goulet, Isabelle 05 October 2011 (has links)
Because it can expand the range of a protein’s interactions or modulate its activity, post-translational methylation of arginine residues in proteins must be duly coordinated and ‘decoded’ to ensure appropriate cellular interpretation of this biological cue. This can be achieved through modulation of the enzymatic activity/specificity of the protein arginine methyltransferases (PRMTs) and proper recognition of the methylation ‘mark’ by a subset of proteins containing ‘methyl-sensing’ protein modules known as ‘Tudor’ domains. In order to gain a better understanding of these regulatory mechanisms, we undertook a detailed biochemical characterization of the predominant member of the PRMT family, PRMT1, and of the novel Tudor domain-containing protein 3 (TDRD3). First, we found that PRMT1 function can be modulated by 1) the expression of up to seven PRMT1 isoforms (v1-7), each with a unique N-terminal region that confers distinct substrate specificity, and by 2) differential subcellular localization, as revealed by the presence of a nuclear export sequence unique to PRMT1v2. Second, our findings suggest that TDRD3 is recruited to cytoplasmic stress granules (SGs) in response to environmental stress potentially by engaging in methyl-dependent protein-protein interactions with proteins involved in the control of gene expression. We also found that arginine methylation may serve as a general regulator of overall SG dynamics. Finally, we uncovered that alteration of PRMT1, TDRD3, and global arginine methylation levels in breast cancer cells may be closely associated with disease progression and poor prognosis. Therefore, further studies into the pathophysiological consequences ensuing from misregulation of arginine methylation will likely lead to the development of novel strategies for the prevention and treatment of breast cancer.
4

New Roles for Arginine Methylation in RNA Metabolism and Cancer

Goulet, Isabelle 05 October 2011 (has links)
Because it can expand the range of a protein’s interactions or modulate its activity, post-translational methylation of arginine residues in proteins must be duly coordinated and ‘decoded’ to ensure appropriate cellular interpretation of this biological cue. This can be achieved through modulation of the enzymatic activity/specificity of the protein arginine methyltransferases (PRMTs) and proper recognition of the methylation ‘mark’ by a subset of proteins containing ‘methyl-sensing’ protein modules known as ‘Tudor’ domains. In order to gain a better understanding of these regulatory mechanisms, we undertook a detailed biochemical characterization of the predominant member of the PRMT family, PRMT1, and of the novel Tudor domain-containing protein 3 (TDRD3). First, we found that PRMT1 function can be modulated by 1) the expression of up to seven PRMT1 isoforms (v1-7), each with a unique N-terminal region that confers distinct substrate specificity, and by 2) differential subcellular localization, as revealed by the presence of a nuclear export sequence unique to PRMT1v2. Second, our findings suggest that TDRD3 is recruited to cytoplasmic stress granules (SGs) in response to environmental stress potentially by engaging in methyl-dependent protein-protein interactions with proteins involved in the control of gene expression. We also found that arginine methylation may serve as a general regulator of overall SG dynamics. Finally, we uncovered that alteration of PRMT1, TDRD3, and global arginine methylation levels in breast cancer cells may be closely associated with disease progression and poor prognosis. Therefore, further studies into the pathophysiological consequences ensuing from misregulation of arginine methylation will likely lead to the development of novel strategies for the prevention and treatment of breast cancer.
5

New Roles for Arginine Methylation in RNA Metabolism and Cancer

Goulet, Isabelle 05 October 2011 (has links)
Because it can expand the range of a protein’s interactions or modulate its activity, post-translational methylation of arginine residues in proteins must be duly coordinated and ‘decoded’ to ensure appropriate cellular interpretation of this biological cue. This can be achieved through modulation of the enzymatic activity/specificity of the protein arginine methyltransferases (PRMTs) and proper recognition of the methylation ‘mark’ by a subset of proteins containing ‘methyl-sensing’ protein modules known as ‘Tudor’ domains. In order to gain a better understanding of these regulatory mechanisms, we undertook a detailed biochemical characterization of the predominant member of the PRMT family, PRMT1, and of the novel Tudor domain-containing protein 3 (TDRD3). First, we found that PRMT1 function can be modulated by 1) the expression of up to seven PRMT1 isoforms (v1-7), each with a unique N-terminal region that confers distinct substrate specificity, and by 2) differential subcellular localization, as revealed by the presence of a nuclear export sequence unique to PRMT1v2. Second, our findings suggest that TDRD3 is recruited to cytoplasmic stress granules (SGs) in response to environmental stress potentially by engaging in methyl-dependent protein-protein interactions with proteins involved in the control of gene expression. We also found that arginine methylation may serve as a general regulator of overall SG dynamics. Finally, we uncovered that alteration of PRMT1, TDRD3, and global arginine methylation levels in breast cancer cells may be closely associated with disease progression and poor prognosis. Therefore, further studies into the pathophysiological consequences ensuing from misregulation of arginine methylation will likely lead to the development of novel strategies for the prevention and treatment of breast cancer.
6

New Roles for Arginine Methylation in RNA Metabolism and Cancer

Goulet, Isabelle January 2011 (has links)
Because it can expand the range of a protein’s interactions or modulate its activity, post-translational methylation of arginine residues in proteins must be duly coordinated and ‘decoded’ to ensure appropriate cellular interpretation of this biological cue. This can be achieved through modulation of the enzymatic activity/specificity of the protein arginine methyltransferases (PRMTs) and proper recognition of the methylation ‘mark’ by a subset of proteins containing ‘methyl-sensing’ protein modules known as ‘Tudor’ domains. In order to gain a better understanding of these regulatory mechanisms, we undertook a detailed biochemical characterization of the predominant member of the PRMT family, PRMT1, and of the novel Tudor domain-containing protein 3 (TDRD3). First, we found that PRMT1 function can be modulated by 1) the expression of up to seven PRMT1 isoforms (v1-7), each with a unique N-terminal region that confers distinct substrate specificity, and by 2) differential subcellular localization, as revealed by the presence of a nuclear export sequence unique to PRMT1v2. Second, our findings suggest that TDRD3 is recruited to cytoplasmic stress granules (SGs) in response to environmental stress potentially by engaging in methyl-dependent protein-protein interactions with proteins involved in the control of gene expression. We also found that arginine methylation may serve as a general regulator of overall SG dynamics. Finally, we uncovered that alteration of PRMT1, TDRD3, and global arginine methylation levels in breast cancer cells may be closely associated with disease progression and poor prognosis. Therefore, further studies into the pathophysiological consequences ensuing from misregulation of arginine methylation will likely lead to the development of novel strategies for the prevention and treatment of breast cancer.
7

Synthèse de ligands à la proteine CARM1 pour l'étude de son activité enzymatique et la synthèse d'inhibiteurs sélectifs / Insights into CARM1 methylation : design of selective inhibitors and peptide mimics : a structure based approach

Ajebbar, Samira 11 May 2012 (has links)
Les protéines arginine méthyl transférases ("PRMTs") sont impliquées dans de nombreux processus cellulaires essentiels. La protéine CARMI ("Coactivator-associated arginine methyltransferase 1", appelée aussi "PRMT4") a été initialement identifiée par sa fonction co-activatrice de la transcription impliquantplusieurs récepteurs nucléaires des hormones. CARMI est une enzyme qui catalyse la réaction de méthylation sur les histones via un donneur de méthyl naturel, la S-adénosY-L -méthionine (SAM). De nombreux travaux ont montré que CARMI est surexprimée dans les cancers du sein et de la prostate. L' objectif de ce travail est la compréhension à l'échelle moléculaire du mode d'action de CARMI et l'étude du mécanisme de reconnaissances moléculaires et de transferts d' informations gouvernés par la protéine CARMI. La structure cristallographique obtenue de cette enzyme en présence de cofacteur, la S-AdénosyhHomocystéine ou la Sinefungine a eu un effet stabilisant. Ainsi, notre stratégie a été de créer des molécules hameçons basées sur le motif de la SAM capables d' ancrer un peptide mimant la séquence de l' histone H3, pour ensuite les tester en co-cristallisation avec CARMI. Ainsi, grâce à la diffraction aux rayons X, les interactions mises en jeu dans le complexe CARMlImolécules hameçons/peptide pourront être déterminées. Cette stratégie s'est effectuée en trois étapes : la première étape, décrite dans le chapitre 2, a consisté en la synthèse d'analogues de la SAM obtenus grâce à des modifications réalisées autour de l'atome de soufre. Ces composés nous ont permis d' explorer la « poche du sulfonium ». Puis la seconde étape, décrite dans le chapitre 3, a été la synthèse • d'analogues de bisubstrats nécessaires pour l'exploration de la « poche de l'arginine ». Dans une dernière étape, décrite dans les chapitres 4 et 5, nous avons abordé la synthèse d'adduits SAM-peptide pouf pouvoir étudier le « domaine de fixation du peptide ». Dans le quatrième chapitre, la méthode de choix est la création d'un lien covalent entre une molécule hameçon électrophile etun peptide par chimie de click in-situ : par réaction de cycloaddition de Huisgen; par réaction entre des molécules hameçons électrophiles capables de piéger un peptide cystéine ou un peptide arginine. Ces essais se sont révélés infructueux et une nouvelle stratégie a été employée en utilisant des molécules ancres. Dansle cinquième chapitre des molécules ancres ont donc été préformés pour ensuite être testés en cocristallisation dans CARMl. / Protein aginine methyltransferases (PRMTs) have been implicated in a variety of biological processes. Coactivator-associated arginine methyltransferase 1 (CARM1 , also known as PRMT4) was identified as an enhancer of the transcriptional activation by several nuclear hormone receptors CARM1 is an enzyme which methylates the arginines of histones via a natural methyl donor, the SAdenosyh-Methionine (SAM). Recent studies have shown that CARM-1 is over-expressed in breastumors and in hormone dependent prostate tumors. The goal of this work is to understand at the molecular level the mode of binding of substrate/product arginine-containing peptides, reflectingstates prior and subsequent to methylation and the detailed mechanism of action of this protein. Several crystal structures of the catalytic domain of CARM1 have shown that cofactor-binding, such as S-Adenosyl-L -Homocysteine or sinefungin, produces large conformational changes in the catalytic domain. These crystal structures clearly illustrate that SAM binding is a prerequisite for peptide binding and build up the productive peptide binding site. Our strategy was to design fishhook molecules derivatives of the SAM capable of anchoring a mimic peptide of the histone H3 in order to test the co-crystallization in CARM1. Consequently, thanks to X-Ray structure, interactions involvement in the complexe CARM1/fishhook molecule/peptide could be determined. This strategy was do ne in three steps: the first one, described in the chapter 2, consisted in synthesizing SAManalogues with several modifications around sulfur atom. These compounds permitted to explore the "sulfonium pocket". The second step, described in chapter 3, consisted in synthesizing analogues of bisubstrats to explore "arginine binding pocket". Finally, the last step, described in the chapter 4 and 5, consisted in synthesizing SAM-peptide adducts to study "peptide binding domain". ln the chapter 4, the chosen method is the creation of covalent link between this molecule and a peptide by click chemistry in-situ: by reaction of Huisgen's cycloaddition; by reaction between electrophilic fishhook molecules capable of capturing with a cystein or arginine peptides. Unfortunately, ail of these trials have been unsuccessful. Consequently SAM-peptide adducts were performed to be co-crystallized in CARM1. This part was described in the last chapter.

Page generated in 0.1438 seconds