Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Structure ; 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-39029460

RESUMO

Complex associating with SET1 (COMPASS) is a histone H3K4 tri-methyltransferase controlled by several regulatory subunits including CXXC zinc finger protein 1 (Cfp1). Prior studies established the structural underpinnings controlling H3K4me3 recognition by the PHD domain of Cfp1's yeast homolog (Spp1). However, metazoans Cfp1PHD lacks structural elements important for H3K4me3 stabilization in Spp1, suggesting that in metazoans, Cfp1PHD domain binds H3K4me3 differently. The structure of Cfp1PHD in complex with H3K4me3 shows unique features such as non-canonical coordination of the first zinc atom and a disulfide bond forcing the reorientation of Cfp1PHD N-terminus, thereby leading to an atypical H3K4me3 binding pocket. This configuration minimizes Cfp1PHD reliance on canonical residues important for histone binding functions of other PHD domains. Cancer-related mutations in Cfp1PHD impair H3K4me3 binding, implying a potential impact on epigenetic signaling. Our work highlights a potential diversification of PHD histone binding modes and the impact of cancer mutations on Cfp1 functions.

2.
FEBS Lett ; 596(7): 898-909, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35122247

RESUMO

Crohn's disease (CD) is characterized by the chronic inflammation of the gastrointestinal tract. A dysbiotic microbiome and a defective immune system are linked to CD, where hydrogen sulfide (H2 S) microbial producers positively correlate with the severity of the disease. Atopobium parvulum is a key H2 S producer from the microbiome of CD patients. In this study, the biochemical characterization of two Atopobium parvulum cysteine desulfurases, ApSufS and ApCsdB, shows that the enzymes are allosterically regulated. Structural analyses reveal that ApSufS forms a dimer with conserved characteristics observed in type II cysteine desulfurases. Four residues surrounding the active site are essential to catalyse cysteine desulfurylation, and a segment of short-chain residues grant access for substrate binding. A better understanding of ApSufS will help future avenues for CD treatment.


Assuntos
Doença de Crohn , Cisteína , Actinobacteria , Liases de Carbono-Enxofre/química , Cisteína/metabolismo , Humanos
3.
FASEB J ; 35(8): e21790, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34320252

RESUMO

CXXC Zinc finger protein 1 (CFP1) is a multitasking protein playing essential roles during various developmental processes. Its ability to interact with several proteins contribute to several epigenetic events. Here, we review CFP1's functions and its impact on DNA methylation and the post-translational modification of histone proteins such as lysine acetylation and methylation. We will also discuss the potential role of CFP1 in carcinogenesis and the impact of the mutations identified in patients suffering from various cancers.


Assuntos
Epigênese Genética , Mutação , Neoplasias/metabolismo , Transativadores/metabolismo , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Transativadores/genética
4.
Nat Commun ; 11(1): 4120, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807798

RESUMO

Lysine acetylation (Kac), an abundant post-translational modification (PTM) in prokaryotes, regulates various microbial metabolic pathways. However, no studies have examined protein Kac at the microbiome level, and it remains unknown whether Kac level is altered in patient microbiomes. Herein, we use a peptide immuno-affinity enrichment strategy coupled with mass spectrometry to characterize protein Kac in the microbiome, which successfully identifies 35,200 Kac peptides from microbial or human proteins in gut microbiome samples. We demonstrate that Kac is widely distributed in gut microbial metabolic pathways, including anaerobic fermentation to generate short-chain fatty acids. Applying to the analyses of microbiomes of patients with Crohn's disease identifies 52 host and 136 microbial protein Kac sites that are differentially abundant in disease versus controls. This microbiome-wide acetylomic approach aids in advancing functional microbiome research.


Assuntos
Doença de Crohn/metabolismo , Microbioma Gastrointestinal/fisiologia , Lisina/metabolismo , Acetilação , Voluntários Saudáveis , Humanos , Análise Multivariada , Proteômica , Espectrometria de Massas em Tandem
5.
Biochemistry ; 59(6): 755-765, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31909602

RESUMO

Ribonuclease 6 (RNase 6) is one of eight catalytically active human pancreatic-type RNases that belong to a superfamily of rapidly evolving enzymes. Like some of its human homologues, RNase 6 exhibits host defense properties such as antiviral and antibacterial activities. Recently solved crystal structures of this enzyme in its nucleotide-free form show the conservation of the prototypical kidney-shaped fold preserved among vertebrate RNases, in addition to revealing the presence of a unique secondary active site. In this study, we determine the structural and conformational properties experienced by RNase 6 upon binding to substrate and product analogues. We present the first crystal structures of RNase 6 bound to a nucleotide ligand (adenosine 5'-monophosphate), in addition to RNase 6 bound to phosphate ions. While the enzyme preserves B2 subsite ligand preferences, our results show a lack of typical B2 subsite interactions normally observed in homologous ligand-bound RNases. A comparison of the dynamical properties of RNase 6 in its apo-, substrate-, and product-bound states highlight the unique dynamical properties experienced on time scales ranging from nano- to milliseconds. Overall, our results confirm the specific evolutionary adaptation of RNase 6 relative to its unique catalytic and biological activities.


Assuntos
Ressonância Magnética Nuclear Biomolecular/métodos , Ribonucleases/química , Ribonucleases/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Sítios de Ligação/fisiologia , Humanos , Ligantes , Estrutura Secundária de Proteína
6.
ACS Appl Mater Interfaces ; 11(19): 17697-17705, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-31013043

RESUMO

The effect of accounting for the total surface in the association of thiol-containing molecules to nanosilver was assessed using isothermal titration calorimetry, along with a new open access algorithm that calculates the total surface area for samples of different polydispersity. Further, we used advanced molecular dynamic calculations to explore the underlying mechanisms for the interaction of the studied molecules in the presence of a nanosilver surface in the form of flat surfaces or as three-dimensional pseudospherical nanostructures. Our data indicate that not only is the total surface area available for binding but also the supramolecular arrangements of the molecules in the near proximity of the nanosilver surface strongly affects the affinity of thiol-containing molecules to nanosilver surfaces.

7.
Nat Commun ; 9(1): 2794, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-30022074

RESUMO

Mutations in proteins like FUS which cause Amyotrophic Lateral Sclerosis (ALS) result in the aberrant formation of stress granules while ALS-linked mutations in other proteins impede elimination of stress granules. Repeat expansions in C9ORF72, the major cause of ALS, reduce C9ORF72 levels but how this impacts stress granules is uncertain. Here, we demonstrate that C9ORF72 associates with the autophagy receptor p62 and controls elimination of stress granules by autophagy. This requires p62 to associate via the Tudor protein SMN with proteins, including FUS, that are symmetrically methylated on arginines. Mice lacking p62 accumulate arginine-methylated proteins and alterations in FUS-dependent splicing. Patients with C9ORF72 repeat expansions accumulate symmetric arginine dimethylated proteins which co-localize with p62. This suggests that C9ORF72 initiates a cascade of ALS-linked proteins (C9ORF72, p62, SMN, FUS) to recognize stress granules for degradation by autophagy and hallmarks of a defect in this process are observable in ALS patients.


Assuntos
Esclerose Lateral Amiotrófica/genética , Autofagia/genética , Proteína C9orf72/genética , Proteína FUS de Ligação a RNA/genética , Proteína Sequestossoma-1/genética , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Arginina/metabolismo , Proteína C9orf72/metabolismo , Linhagem Celular Tumoral , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , Embrião de Mamíferos , Células HeLa , Humanos , Metilação , Camundongos , Camundongos Knockout , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Cultura Primária de Células , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteína Sequestossoma-1/metabolismo , Estresse Fisiológico , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo
8.
Chem Commun (Camb) ; 52(31): 5474-7, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27021271

RESUMO

Methylation of arginine and lysine (RK) residues play essential roles in epigenetics and the regulation of gene expression. However, research in this area is often hindered by the lack of effective tools for probing the protein methylation. Here, we present an antibody-free strategy to capture protein methylation on RK residues by using chemical reactions to eliminate the charges on un-modified RK residues and peptide N-termini. Peptides containing methylated RK residues remain positively charged and are then enriched by strong cation exchange chromatography, followed by high-resolution mass spectrometry identification.


Assuntos
Arginina/análise , Lisina/análise , Peptídeos/química , Proteínas/química , Sequência de Aminoácidos , Cromatografia por Troca Iônica , Espectrometria de Massas , Metilação
9.
Genes Dev ; 30(5): 508-21, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944678

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous group of hematological tumors composed of distinct subtypes that vary in their genetic abnormalities, gene expression signatures, and prognoses. However, it remains unclear whether T-ALL subtypes differ at the functional level, and, as such, T-ALL treatments are uniformly applied across subtypes, leading to variable responses between patients. Here we reveal the existence of a subtype-specific epigenetic vulnerability in T-ALL by which a particular subgroup of T-ALL characterized by expression of the oncogenic transcription factor TAL1 is uniquely sensitive to variations in the dosage and activity of the histone 3 Lys27 (H3K27) demethylase UTX/KDM6A. Specifically, we identify UTX as a coactivator of TAL1 and show that it acts as a major regulator of the TAL1 leukemic gene expression program. Furthermore, we demonstrate that UTX, previously described as a tumor suppressor in T-ALL, is in fact a pro-oncogenic cofactor essential for leukemia maintenance in TAL1-positive (but not TAL1-negative) T-ALL. Exploiting this subtype-specific epigenetic vulnerability, we propose a novel therapeutic approach based on UTX inhibition through in vivo administration of an H3K27 demethylase inhibitor that efficiently kills TAL1-positive primary human leukemia. These findings provide the first opportunity to develop personalized epigenetic therapy for T-ALL patients.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/genética , Terapia Genética , Histona Desmetilases/genética , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas Proto-Oncogênicas/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Histona Desmetilases/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T
10.
Stem Cell Reports ; 5(5): 741-752, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26455415

RESUMO

The molecules and mechanisms pertinent to the low immunogenicity of undifferentiated embryonic stem cells (ESCs) remain poorly understood. Here, we provide evidence that milk fat globule epidermal growth factor 8 (MFG-E8) is a vital mediator in this phenomenon and directly suppresses T cell immune responses. MFG-E8 is enriched in undifferentiated ESCs but diminished in differentiated ESCs. Upregulation of MFG-E8 in ESCs increases the successful engraftment of both undifferentiated and differentiated ESCs across major histocompatibility complex barriers. MFG-E8 suppresses T cell activation/proliferation and inhibits Th1, Th2, and Th17 subpopulations while increasing regulatory T cell subsets. Neutralizing MFG-E8 substantially abrogates these effects, whereas addition of recombinant MFG-E8 to differentiated ESCs restores immunosuppression. Furthermore, we provide the evidence that MFG-E8 suppresses T cell activation and regulates T cell polarization by inhibiting PKCθ phosphorylation through the α3/5ßV integrin receptor. Our findings offer an approach to facilitate transplantation acceptance.


Assuntos
Antígenos de Superfície/metabolismo , Células-Tronco Embrionárias/imunologia , Histocompatibilidade , Ativação Linfocitária , Proteínas do Leite/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Antígenos de Superfície/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Leite/genética
11.
Genes Dev ; 29(2): 123-8, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25593305

RESUMO

The methyltransferase activity of the trithorax group (TrxG) protein MLL1 found within its COMPASS (complex associated with SET1)-like complex is allosterically regulated by a four-subunit complex composed of WDR5, RbBP5, Ash2L, and DPY30 (also referred to as WRAD). We report structural evidence showing that in WRAD, a concave surface of the Ash2L SPIa and ryanodine receptor (SPRY) domain binds to a cluster of acidic residues, referred to as the D/E box, in RbBP5. Mutational analysis shows that residues forming the Ash2L/RbBP5 interface are important for heterodimer formation, stimulation of MLL1 catalytic activity, and erythroid cell terminal differentiation. We also demonstrate that a phosphorylation switch on RbBP5 stimulates WRAD complex formation and significantly increases KMT2 (lysine [K] methyltransferase 2) enzyme methylation rates. Overall, our findings provide structural insights into the assembly of the WRAD complex and point to a novel regulatory mechanism controlling the activity of the KMT2/COMPASS family of lysine methyltransferases.


Assuntos
Histonas/metabolismo , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Cristalização , Análise Mutacional de DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/genética , Células Eritroides/citologia , Células Eritroides/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Metilação/efeitos dos fármacos , Metiltransferases/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
12.
Nucleic Acids Res ; 41(15): 7438-52, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23761443

RESUMO

APOBEC3G (A3G) is a host-encoded protein that potently restricts the infectivity of a broad range of retroviruses. This can occur by mechanisms dependent on catalytic activity, resulting in the mutagenic deamination of nascent viral cDNA, and/or by other means that are independent of its catalytic activity. It is not yet known to what extent deamination-independent processes contribute to the overall restriction, how they exactly work or how they are regulated. Here, we show that alanine substitution of either tryptophan 94 (W94A) or 127 (W127A) in the non-catalytic N-terminal domain of A3G severely impedes RNA binding and alleviates deamination-independent restriction while still maintaining DNA mutator activity. Substitution of both tryptophans (W94A/W127A) produces a more severe phenotype in which RNA binding and RNA-dependent protein oligomerization are completely abrogated. We further demonstrate that RNA binding is specifically required for crippling late reverse transcript accumulation, preventing proviral DNA integration and, consequently, restricting viral particle release. We did not find that deaminase activity made a significant contribution to the restriction of any of these processes. In summary, this work reveals that there is a direct correlation between A3G's capacity to bind RNA and its ability to inhibit retroviral infectivity in a deamination-independent manner.


Assuntos
Domínio Catalítico , Citosina Desaminase/metabolismo , Vírus da Leucemia Murina de Moloney/fisiologia , RNA Viral/genética , Desaminases APOBEC , Alanina/genética , Alanina/metabolismo , Substituição de Aminoácidos , Animais , Citidina Desaminase , Citosina Desaminase/genética , Desaminação , Ativação Enzimática , Células HEK293 , HIV-1/fisiologia , Humanos , Camundongos , Células NIH 3T3 , Multimerização Proteica , Transcrição Reversa , Triptofano/genética , Triptofano/metabolismo , Montagem de Vírus , Integração Viral , Liberação de Vírus
13.
Biochem J ; 449(1): 151-9, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22989411

RESUMO

WDR5 (WD40 repeat protein 5) is an essential component of the human trithorax-like family of SET1 [Su(var)3-9 enhancer-of-zeste trithorax 1] methyltransferase complexes that carry out trimethylation of histone 3 Lys4 (H3K4me3), play key roles in development and are abnormally expressed in many cancers. In the present study, we show that the interaction between WDR5 and peptides from the catalytic domain of MLL (mixed-lineage leukaemia protein) (KMT2) can be antagonized with a small molecule. Structural and biophysical analysis show that this antagonist binds in the WDR5 peptide-binding pocket with a Kd of 450 nM and inhibits the catalytic activity of the MLL core complex in vitro. The degree of inhibition was enhanced at lower protein concentrations consistent with a role for WDR5 in directly stabilizing the MLL multiprotein complex. Our data demonstrate inhibition of an important protein-protein interaction and form the basis for further development of inhibitors of WDR5-dependent enzymes implicated in MLL-rearranged leukaemias or other cancers.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/metabolismo , Domínio Catalítico/fisiologia , Cristalografia por Raios X , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia
14.
Biopolymers ; 99(2): 136-45, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175388

RESUMO

In the last 20 years, we have witnessed an exponential number of evidences linking the human mixed lineage leukemia-1 (MLL1) gene to several acute and myelogenous leukemias. MLL1 is one of the founding members of the SET1 family of lysine methyltransferases and is key for the proper control of developmentally regulated gene expression. MLL1 is a structurally complex protein composed of several functional domains. These domains play pivotal roles for the recruitment of regulatory proteins. These MLL1 regulatory proteins (MRPs) dynamically interact with MLL1 and consequently control gene expression. In this review, we summarize recent structural and functional studies of MRPs and discuss emergent structural paradigms for the control of MLL1 activity.


Assuntos
Redes Reguladoras de Genes , Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Leucemia Mieloide/fisiopatologia , Proteína de Leucina Linfoide-Mieloide/metabolismo
15.
Sports Health ; 4(1): 47-50, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23016068

RESUMO

BACKGROUND: Meniscal tears have been associated with meniscal cysts and fullness of the knee joint line on physical examination. HYPOTHESIS: Joint line fullness is an accurate, sensitive, and specific test to detect meniscal tears. STUDY DESIGN: Prospective cohort study. METHODS: One hundred consecutive patients undergoing knee arthroscopy were included. All had physical examinations documenting the presence of joint line fullness, joint line tenderness, and the McMurray sign. Arthroscopy was the gold standard for tears. Accuracy, sensitivity, and specificity were calculated and correlated with type of tear. Sixty-one patients had a magnetic resonance imaging preoperatively (the gold standard for determining the presence of a cyst). RESULTS: Meniscal tears were found in 67 patients at arthroscopy. The accuracy, sensitivity, and specificity of joint line fullness were, respectively, 73%, 70%, and 82% in detecting meniscal tears; 68%, 87%, and 30% for joint line tenderness; and 47%, 32%, and 78% for the McMurray sign. The highest positive predictive value for detecting a tear was 88% for joint line fullness, compared with 77% for joint line tenderness and 76% for the McMurray sign. However, joint line fullness did not correlate well with the presence of a cyst, with a low positive predictive value (29%). Of those patients with joint line fullness on physical examination, 89% had a horizontal cleavage component of their tear at arthroscopy. CONCLUSION: Joint line fullness is an accurate, sensitive, and specific test to detect meniscal tears. CLINICAL RELEVANCE: The findings support the routine use of joint line fullness during physical examination along with other common tests to improve the accuracy of clinically diagnosing meniscal tears.

16.
Nucleic Acids Res ; 40(9): 4237-46, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22266653

RESUMO

In mammals, the SET1 family of lysine methyltransferases (KMTs), which includes MLL1-5, SET1A and SET1B, catalyzes the methylation of lysine-4 (Lys-4) on histone H3. Recent reports have demonstrated that a three-subunit complex composed of WD-repeat protein-5 (WDR5), retinoblastoma-binding protein-5 (RbBP5) and absent, small, homeotic disks-2-like (ASH2L) stimulates the methyltransferase activity of MLL1. On the basis of studies showing that this stimulation is in part controlled by an interaction between WDR5 and a small region located in close proximity of the MLL1 catalytic domain [referred to as the WDR5-interacting motif (Win)], it has been suggested that WDR5 might play an analogous role in scaffolding the other SET1 complexes. We herein provide biochemical and structural evidence showing that WDR5 binds the Win motifs of MLL2-4, SET1A and SET1B. Comparative analysis of WDR5-Win complexes reveals that binding of the Win motifs is achieved by the plasticity of WDR5 peptidyl-arginine-binding cleft allowing the C-terminal ends of the Win motifs to be maintained in structurally divergent conformations. Consistently, enzymatic assays reveal that WDR5 plays an important role in the optimal stimulation of MLL2-4, SET1A and SET1B methyltransferase activity by the RbBP5-ASH2L heterodimer. Overall, our findings illustrate the function of WDR5 in scaffolding the SET1 family of KMTs and further emphasize on the important role of WDR5 in regulating global histone H3 Lys-4 methylation.


Assuntos
Histona-Lisina N-Metiltransferase/química , Subunidades Proteicas/química , Motivos de Aminoácidos , Sítios de Ligação , Cristalografia , Histona-Lisina N-Metiltransferase/metabolismo , Modelos Moleculares , Peptídeos/química , Ligação Proteica , Subunidades Proteicas/metabolismo
17.
J Mol Cell Biol ; 3(5): 301-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028380

RESUMO

The SMYD (SET and MYND domain) family of lysine methyltransferases (KMTs) plays pivotal roles in various cellular processes, including gene expression regulation and DNA damage response. Initially identified as genuine histone methyltransferases, specific members of this family have recently been shown to methylate non-histone proteins such as p53, VEGFR, and the retinoblastoma tumor suppressor (pRb). To gain further functional insights into this family of KMTs, we generated the protein interaction network for three different human SMYD proteins (SMYD2, SMYD3, and SMYD5). Characterization of each SMYD protein network revealed that they associate with both shared and unique sets of proteins. Among those, we found that HSP90 and several of its co-chaperones interact specifically with the tetratrico peptide repeat (TPR)-containing SMYD2 and SMYD3. Moreover, using proteomic and biochemical techniques, we provide evidence that SMYD2 methylates K209 and K615 on HSP90 nucleotide-binding and dimerization domains, respectively. In addition, we found that each methylation site displays unique reactivity in regard to the presence of HSP90 co-chaperones, pH, and demethylation by the lysine amine oxidase LSD1, suggesting that alternative mechanisms control HSP90 methylation by SMYD2. Altogether, this study highlights the ability of SMYD proteins to form unique protein complexes that may underlie their various biological functions and the SMYD2-mediated methylation of the key molecular chaperone HSP90.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteoma/análise , Células HEK293 , Proteínas de Choque Térmico HSP90/genética , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Lisina/metabolismo , Metilação , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Conformação Proteica , Mapas de Interação de Proteínas
18.
Structure ; 19(1): 101-8, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21220120

RESUMO

Histone H3 Lys-4 methylation is predominantly catalyzed by a family of methyltransferases whose enzymatic activity depends on their interaction with a three-subunit complex composed of WDR5, RbBP5, and Ash2L. Here, we report that a segment of 50 residues of RbBP5 bridges the Ash2L C-terminal domain to WDR5. The crystal structure of WDR5 in ternary complex with RbBP5 and MLL1 reveals that both proteins binds peptide-binding clefts located on opposite sides of WDR5's ß-propeller domain. RbBP5 engages in several hydrogen bonds and van der Waals contacts within a V-shaped cleft formed by the junction of two blades on WDR5. Mutational analyses of both the WDR5 V-shaped cleft and RbBP5 residues reveal that the interactions between RbBP5 and WDR5 are important for the stimulation of MLL1 methyltransferase activity. Overall, this study provides the structural basis underlying the formation of the WDR5-RbBP5 subcomplex and further highlight the crucial role of WDR5 in scaffolding the MLL1 core complex.


Assuntos
Histona-Lisina N-Metiltransferase/química , Proteína de Leucina Linfoide-Mieloide/química , Proteínas Nucleares/química , Proteínas Recombinantes de Fusão/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas de Ligação a DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência
19.
FASEB J ; 25(3): 960-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21135039

RESUMO

The SET1 family of methyltransferases carries out the bulk of histone H3 Lys-4 methylation in vivo. One of the common features of this family is the regulation of their methyltransferase activity by a tripartite complex composed of WDR5, RbBP5, and Ash2L. To selectively probe the role of the SET1 family of methyltransferases, we have developed a library of histone H3 peptide mimetics and report herein the characterization of an Nα acetylated form of histone H3 peptide (NαH3). Binding and inhibition studies reveal that the addition of an acetyl moiety to the N terminus of histone H3 significantly enhances its binding to WDR5 and prevents the stimulation of MLL1 methyltransferase activity by the WDR5-RbBP5-Ash2L complex. The crystal structure of NαH3 in complex with WDR5 reveals that a high-affinity hydrophobic pocket accommodates the binding of the acetyl moiety. These results provide the structural basis to control WDR5-RbBP5-Ash2L-MLL1 activity and a tool to manipulate stem cell differentiation programs.


Assuntos
Metilação de DNA/fisiologia , Epigenômica , Histonas/metabolismo , Proteína de Leucina Linfoide-Mieloide , Células-Tronco/enzimologia , Acetilação , Diferenciação Celular/fisiologia , Células Cultivadas , Cristalografia , Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Histonas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Químicos , Mimetismo Molecular , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Nucleares/metabolismo , Nucleossomos/fisiologia , Biblioteca de Peptídeos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
20.
J Biol Chem ; 285(41): 31849-58, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20675860

RESUMO

SET domain lysine methyltransferases (KMTs) methylate specific lysine residues in histone and non-histone substrates. These enzymes also display product specificity by catalyzing distinct degrees of methylation of the lysine ε-amino group. To elucidate the molecular mechanism underlying this specificity, we have characterized the Y245A and Y305F mutants of the human KMT SET7/9 (also known as KMT7) that alter its product specificity from a monomethyltransferase to a di- and a trimethyltransferase, respectively. Crystal structures of these mutants in complex with peptides bearing unmodified, mono-, di-, and trimethylated lysines illustrate the roles of active site water molecules in aligning the lysine ε-amino group for methyl transfer with S-adenosylmethionine. Displacement or dissociation of these solvent molecules enlarges the diameter of the active site, accommodating the increasing size of the methylated ε-amino group during successive methyl transfer reactions. Together, these results furnish new insights into the roles of active site water molecules in modulating lysine multiple methylation by SET domain KMTs and provide the first molecular snapshots of the mono-, di-, and trimethyl transfer reactions catalyzed by these enzymes.


Assuntos
Substituição de Aminoácidos , Histona-Lisina N-Metiltransferase/química , Lisina/química , Mutação de Sentido Incorreto , Água/química , Catálise , Domínio Catalítico , Cristalografia por Raios X , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Lisina/metabolismo , Metilação , Água/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA