Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2879, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38570506

RESUMO

Despite regulating overlapping gene enhancers and pathways, CREBBP and KMT2D mutations recurrently co-occur in germinal center (GC) B cell-derived lymphomas, suggesting potential oncogenic cooperation. Herein, we report that combined haploinsufficiency of Crebbp and Kmt2d induces a more severe mouse lymphoma phenotype (vs either allele alone) and unexpectedly confers an immune evasive microenvironment manifesting as CD8+ T-cell exhaustion and reduced infiltration. This is linked to profound repression of immune synapse genes that mediate crosstalk with T-cells, resulting in aberrant GC B cell fate decisions. From the epigenetic perspective, we observe interaction and mutually dependent binding and function of CREBBP and KMT2D on chromatin. Their combined deficiency preferentially impairs activation of immune synapse-responsive super-enhancers, pointing to a particular dependency for both co-activators at these specialized regulatory elements. Together, our data provide an example where chromatin modifier mutations cooperatively shape and induce an immune-evasive microenvironment to facilitate lymphomagenesis.


Assuntos
Linfoma Difuso de Grandes Células B , Animais , Camundongos , Linfócitos B/metabolismo , Cromatina/genética , Cromatina/metabolismo , Centro Germinativo/metabolismo , Linfoma Difuso de Grandes Células B/genética , Mutação , Microambiente Tumoral/genética
2.
Cancer Lett ; 575: 216404, 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37739210

RESUMO

Elevated expression and genetic aberration of IRTKS, also named as BAIAP2L1, have been observed in many tumors, especially in tumor progression. however, the molecular and cellular mechanisms involved in the IRTKS-enhanced tumor progression are obscure. Here we show that higher IRTKS level specifically increases histone H3 lysine 9 trimethylation (H3K9me3) by promoting accumulation of the histone methyltransferase SETDB1. Furthermore, we reveal that IRTKS recruits the deubiquitinase OTUD4 to remove Lys48-linked polyubiquitination at K182/K1050 sites of SETDB1, thus blocking SETDB1 degradation via the ubiquitin-proteasome pathway. Interestingly, the enhanced IRTKS-OTUD4-SETDB1-H3K9me3 axis leads to a general decrease in chromatin accessibility, which inhibits transcription of CDH1 encoding E-cadherin, a key molecule essential for maintaining epithelial cell phenotype, and therefore results in epithelial-mesenchymal transition (EMT) and malignant cell metastasis. Clinically, the elevated IRTKS levels in tumor specimens correlate with SETDB1 levels, but negatively associate with survival time. Our data reveal a novel mechanism for the IRTKS-enhanced tumor progression, where IRTKS cooperates with OTUD4 to enhance SETDB1-mediated H3K9 trimethylation that promotes tumor metastasis via suppressing E-cadherin expression. This study also provides a potential approach to reduce the activity and stability of the known therapeutic target SETDB1 possibly through regulating IRTKS or deubiquitinase OTUD4.

3.
Nature ; 574(7779): 575-580, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31645732

RESUMO

The Warburg effect, which originally described increased production of lactate in cancer, is associated with diverse cellular processes such as angiogenesis, hypoxia, polarization of macrophages and activation of T cells. This phenomenon is intimately linked to several diseases including neoplasia, sepsis and autoimmune diseases1,2. Lactate, which is converted from pyruvate in tumour cells, is widely known as an energy source and metabolic by-product. However, its non-metabolic functions in physiology and disease remain unknown. Here we show that lactate-derived lactylation of histone lysine residues serves as an epigenetic modification that directly stimulates gene transcription from chromatin. We identify 28 lactylation sites on core histones in human and mouse cells. Hypoxia and bacterial challenges induce the production of lactate by glycolysis, and this acts as a precursor that stimulates histone lactylation. Using M1 macrophages that have been exposed to bacteria as a model system, we show that histone lactylation has different temporal dynamics from acetylation. In the late phase of M1 macrophage polarization, increased histone lactylation induces homeostatic genes that are involved in wound healing, including Arg1. Collectively, our results suggest that an endogenous 'lactate clock' in bacterially challenged M1 macrophages turns on gene expression to promote homeostasis. Histone lactylation thus represents an opportunity to improve our understanding of the functions of lactate and its role in diverse pathophysiological conditions, including infection and cancer.


Assuntos
Epigênese Genética , Glicólise/genética , Histonas/química , Histonas/metabolismo , Ácido Láctico/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Homeostase , Humanos , Hipóxia/metabolismo , Lisina/química , Lisina/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reprodutibilidade dos Testes , Transcrição Gênica
4.
Mol Cell ; 62(2): 194-206, 2016 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-27105115

RESUMO

Here we report the identification and verification of a ß-hydroxybutyrate-derived protein modification, lysine ß-hydroxybutyrylation (Kbhb), as a new type of histone mark. Histone Kbhb marks are dramatically induced in response to elevated ß-hydroxybutyrate levels in cultured cells and in livers from mice subjected to prolonged fasting or streptozotocin-induced diabetic ketoacidosis. In total, we identified 44 histone Kbhb sites, a figure comparable to the known number of histone acetylation sites. By ChIP-seq and RNA-seq analysis, we demonstrate that histone Kbhb is a mark enriched in active gene promoters and that the increased H3K9bhb levels that occur during starvation are associated with genes upregulated in starvation-responsive metabolic pathways. Histone ß-hydroxybutyrylation thus represents a new epigenetic regulatory mark that couples metabolism to gene expression, offering a new avenue to study chromatin regulation and diverse functions of ß-hydroxybutyrate in the context of important human pathophysiological states, including diabetes, epilepsy, and neoplasia.


Assuntos
Cetoacidose Diabética/metabolismo , Metabolismo Energético , Regulação da Expressão Gênica , Histonas/metabolismo , Hidroxibutiratos/metabolismo , Fígado/metabolismo , Processamento de Proteína Pós-Traducional , Inanição/metabolismo , Animais , Sítios de Ligação , Montagem e Desmontagem da Cromatina , Cetoacidose Diabética/induzido quimicamente , Cetoacidose Diabética/genética , Modelos Animais de Doenças , Epigênese Genética , Ácidos Graxos/metabolismo , Glucose/metabolismo , Células HEK293 , Histonas/genética , Humanos , Lisina , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Inanição/genética , Estreptozocina
5.
Science ; 350(6266): 1383-6, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26659056

RESUMO

Release of promoter-proximal paused RNA polymerase II (Pol II) during early elongation is a critical step in transcriptional regulation in metazoan cells. Paused Pol II release is thought to require the kinase activity of cyclin-dependent kinase 9 (CDK9) for the phosphorylation of DRB sensitivity-inducing factor, negative elongation factor, and C-terminal domain (CTD) serine-2 of Pol II. We found that Pol II-associated factor 1 (PAF1) is a critical regulator of paused Pol II release, that positive transcription elongation factor b (P-TEFb) directly regulates the initial recruitment of PAF1 complex (PAF1C) to genes, and that the subsequent recruitment of CDK12 is dependent on PAF1C. These findings reveal cooperativity among P-TEFb, PAF1C, and CDK12 in pausing release and Pol II CTD phosphorylation.


Assuntos
Regulação da Expressão Gênica , Proteínas Nucleares/metabolismo , RNA Polimerase II/metabolismo , Elongação da Transcrição Genética , Linhagem Celular Tumoral , Quinase 9 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Humanos , Proteínas Nucleares/genética , Fosforilação , Fator B de Elongação Transcricional Positiva/metabolismo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , RNA Polimerase II/química , RNA Polimerase II/genética , Fatores de Transcrição/metabolismo
6.
Genes Dev ; 29(20): 2123-39, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26494788

RESUMO

RUNX1-RUNX1T1 (formerly AML1-ETO), a transcription factor generated by the t(8;21) translocation in acute myeloid leukemia (AML), dictates a leukemic program by increasing self-renewal and inhibiting differentiation. Here we demonstrate that the histone demethylase JMJD1C functions as a coactivator for RUNX1-RUNX1T1 and is required for its transcriptional program. JMJD1C is directly recruited by RUNX1-RUNX1T1 to its target genes and regulates their expression by maintaining low H3K9 dimethyl (H3K9me2) levels. Analyses in JMJD1C knockout mice also establish a JMJD1C requirement for RUNX1-RUNX1T1's ability to increase proliferation. We also show a critical role for JMJD1C in the survival of multiple human AML cell lines, suggesting that it is required for leukemic programs in different AML cell types through its association with key transcription factors.


Assuntos
Regulação Leucêmica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Leucemia Mieloide Aguda/fisiopatologia , Oxirredutases N-Desmetilantes/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Leucemia Mieloide Aguda/genética , Camundongos Knockout , Oxirredutases N-Desmetilantes/genética , Transporte Proteico/genética
7.
EMBO J ; 34(23): 2885-902, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26330467

RESUMO

The Mediator complex orchestrates multiple transcription factors with the Pol II apparatus for precise transcriptional control. However, its interplay with the surrounding chromatin remains poorly understood. Here, we analyze differential histone modifications between WT and MED23(-/-) (KO) cells and identify H2B mono-ubiquitination at lysine 120 (H2Bub) as a MED23-dependent histone modification. Using tandem affinity purification and mass spectrometry, we find that MED23 associates with the RNF20/40 complex, the enzyme for H2Bub, and show that this association is critical for the recruitment of RNF20/40 to chromatin. In a cell-free system, Mediator directly and substantially increases H2Bub on recombinant chromatin through its cooperation with RNF20/40 and the PAF complex. Integrative genome-wide analyses show that MED23 depletion specifically reduces H2Bub on a subset of MED23-controlled genes. Importantly, MED23-coupled H2Bub levels are oppositely regulated during myogenesis and lung carcinogenesis. In sum, these results establish a mechanistic link between the Mediator complex and a critical chromatin modification in coordinating transcription with cell growth and differentiation.


Assuntos
Histonas/metabolismo , Complexo Mediador/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Complexo Mediador/genética , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Ubiquitinação/genética , Ubiquitinação/fisiologia
8.
Mol Cell ; 58(2): 203-15, 2015 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-25818647

RESUMO

Acetylation of histones at DNA regulatory elements plays a critical role in transcriptional activation. Histones are also modified by other acyl moieties, including crotonyl, yet the mechanisms that govern acetylation versus crotonylation and the functional consequences of this "choice" remain unclear. We show that the coactivator p300 has both crotonyltransferase and acetyltransferase activities, and that p300-catalyzed histone crotonylation directly stimulates transcription to a greater degree than histone acetylation. Levels of histone crotonylation are regulated by the cellular concentration of crotonyl-CoA, which can be altered through genetic and environmental perturbations. In a cell-based model of transcriptional activation, increasing or decreasing the cellular concentration of crotonyl-CoA leads to enhanced or diminished gene expression, respectively, which correlates with the levels of histone crotonylation flanking the regulatory elements of activated genes. Our findings support a general principle wherein differential histone acylation (i.e., acetylation versus crotonylation) couples cellular metabolism to the regulation of gene expression.


Assuntos
Acil Coenzima A/metabolismo , Proteína p300 Associada a E1A/metabolismo , Histonas/metabolismo , Macrófagos/imunologia , RNA Mensageiro/metabolismo , Ativação Transcricional , Acetato-CoA Ligase/genética , Acetato-CoA Ligase/metabolismo , Acetilação , Acil Coenzima A/genética , Linhagem Celular , Sistema Livre de Células , Proteína p300 Associada a E1A/genética , Células HEK293 , Células HeLa , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Dados de Sequência Molecular
9.
Nat Struct Mol Biol ; 20(10): 1156-63, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23995757

RESUMO

Although histone H3 Lys4 (H3K4) methylation is widely associated with gene activation, direct evidence for its causal role in transcription, through specific MLL family members, is scarce. Here we have purified a human MLL2 (Kmt2b) complex that is highly active in H3K4 methylation and chromatin transcription in a cell-free system. This effect requires S-adenosyl methionine and intact H3K4, thus establishing a direct and causal role for MLL2-mediated H3K4 methylation in transcription. We also show that human AKAP95, a chromatin-associated protein, physically and functionally associates with MLL complexes and directly enhances their methyltransferase activity. Ectopic AKAP95 stimulates expression of a chromosomal reporter gene in synergy with MLL1 or MLL2, whereas AKAP95 depletion impairs retinoic acid-mediated gene induction in embryonic stem cells. These results demonstrate an important role for AKAP95 in regulating histone methylation and gene expression, particularly during cell-fate transitions.


Assuntos
Proteínas de Ancoragem à Quinase A/fisiologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Neoplasias/fisiologia , Transcrição Gênica/fisiologia , Diferenciação Celular , Sistema Livre de Células , Cromatina/genética , Metilação de DNA , Células-Tronco Embrionárias/citologia , Genes Reporter , Histonas/metabolismo , Humanos
10.
Nature ; 500(7460): 93-7, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23812588

RESUMO

Transcription factors are frequently altered in leukaemia through chromosomal translocation, mutation or aberrant expression. AML1-ETO, a fusion protein generated by the t(8;21) translocation in acute myeloid leukaemia, is a transcription factor implicated in both gene repression and activation. AML1-ETO oligomerization, mediated by the NHR2 domain, is critical for leukaemogenesis, making it important to identify co-regulatory factors that 'read' the NHR2 oligomerization and contribute to leukaemogenesis. Here we show that, in human leukaemic cells, AML1-ETO resides in and functions through a stable AML1-ETO-containing transcription factor complex (AETFC) that contains several haematopoietic transcription (co)factors. These AETFC components stabilize the complex through multivalent interactions, provide multiple DNA-binding domains for diverse target genes, co-localize genome wide, cooperatively regulate gene expression, and contribute to leukaemogenesis. Within the AETFC complex, AML1-ETO oligomerization is required for a specific interaction between the oligomerized NHR2 domain and a novel NHR2-binding (N2B) motif in E proteins. Crystallographic analysis of the NHR2-N2B complex reveals a unique interaction pattern in which an N2B peptide makes direct contact with side chains of two NHR2 domains as a dimer, providing a novel model of how dimeric/oligomeric transcription factors create a new protein-binding interface through dimerization/oligomerization. Intriguingly, disruption of this interaction by point mutations abrogates AML1-ETO-induced haematopoietic stem/progenitor cell self-renewal and leukaemogenesis. These results reveal new mechanisms of action of AML1-ETO, and provide a potential therapeutic target in t(8;21)-positive acute myeloid leukaemia.


Assuntos
Transformação Celular Neoplásica , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Complexos Multiproteicos/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Divisão Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/química , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/química , Proteínas de Fusão Oncogênica/química , Mutação Puntual , Ligação Proteica , Multimerização Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteína 1 Parceira de Translocação de RUNX1
11.
Cell ; 154(2): 297-310, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23870121

RESUMO

The H3K4me3 mark in chromatin is closely correlated with actively transcribed genes, although the mechanisms involved in its generation and function are not fully understood. In vitro studies with recombinant chromatin and purified human factors demonstrate a robust SET1 complex (SET1C)-mediated H3K4 trimethylation that is dependent upon p53- and p300-mediated H3 acetylation, a corresponding SET1C-mediated enhancement of p53- and p300-dependent transcription that reflects a primary effect of SET1C through H3K4 trimethylation, and direct SET1C-p53 and SET1C-p300 interactions indicative of a targeted recruitment mechanism. Complementary cell-based assays demonstrate a DNA-damage-induced p53-SET1C interaction, a corresponding enrichment of SET1C and H3K4me3 on a p53 target gene (p21/WAF1), and a corresponding codependency of H3K4 trimethylation and transcription upon p300 and SET1C. These results establish a mechanism in which SET1C and p300 act cooperatively, through direct interactions and coupled histone modifications, to facilitate the function of p53.


Assuntos
Proteína p300 Associada a E1A/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Sequência de Aminoácidos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Células HCT116 , Código das Histonas , Histonas/metabolismo , Humanos , Metilação , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Transcrição Gênica
12.
PLoS Genet ; 9(6): e1003524, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23754954

RESUMO

The interplay between polycomb and trithorax complexes has been implicated in embryonic stem cell (ESC) self-renewal and differentiation. It has been shown recently that WRD5 and Dpy-30, specific components of the SET1/MLL protein complexes, play important roles during ESC self-renewal and differentiation of neural lineages. However, not much is known about how and where specific trithorax complexes are targeted to genes involved in self-renewal or lineage-specification. Here, we report that the recruitment of the hSET1A histone H3K4 methyltransferase (HMT) complex by transcription factor USF1 is required for mesoderm specification and lineage differentiation. In undifferentiated ESCs, USF1 maintains hematopoietic stem/progenitor cell (HS/PC) associated bivalent chromatin domains and differentiation potential. Furthermore, USF1 directed recruitment of the hSET1A complex to the HoxB4 promoter governs the transcriptional activation of HoxB4 gene and regulates the formation of early hematopoietic cell populations. Disruption of USF or hSET1A function by overexpression of a dominant-negative AUSF1 mutant or by RNA-interference-mediated knockdown, respectively, led to reduced expression of mesoderm markers and inhibition of lineage differentiation. We show that USF1 and hSET1A together regulate H3K4me3 modifications and transcription preinitiation complex assembly at the hematopoietic-associated HoxB4 gene during differentiation. Finally, ectopic expression of USF1 in ESCs promotes mesoderm differentiation and enforces the endothelial-to-hematopoietic transition by inducing hematopoietic-associated transcription factors, HoxB4 and TAL1. Taken together, our findings reveal that the guided-recruitment of the hSET1A histone methyltransferase complex and its H3K4 methyltransferase activity by transcription regulator USF1 safeguards hematopoietic transcription programs and enhances mesoderm/hematopoietic differentiation.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Epigênese Genética , Histona-Lisina N-Metiltransferase/genética , Fatores Estimuladores Upstream/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Células K562 , Mesoderma/citologia , Mesoderma/metabolismo , Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/metabolismo , Ativação Transcricional , Fatores Estimuladores Upstream/metabolismo
13.
Cell ; 152(5): 1021-36, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23452851

RESUMO

Histone modifications regulate chromatin-dependent processes, yet the mechanisms by which they contribute to specific outcomes remain unclear. H3K4me3 is a prominent histone mark that is associated with active genes and promotes transcription through interactions with effector proteins that include initiation factor TFIID. We demonstrate that H3K4me3-TAF3 interactions direct global TFIID recruitment to active genes, some of which are p53 targets. Further analyses show that (1) H3K4me3 enhances p53-dependent transcription by stimulating preinitiation complex (PIC) formation; (2) H3K4me3, through TAF3 interactions, can act either independently or cooperatively with the TATA box to direct PIC formation and transcription; and (3) H3K4me3-TAF3/TFIID interactions regulate gene-selective functions of p53 in response to genotoxic stress. Our findings indicate a mechanism by which H3K4me3 directs PIC assembly for the rapid induction of specific p53 target genes.


Assuntos
Código das Histonas , Histonas/metabolismo , Fator de Transcrição TFIID/metabolismo , Iniciação da Transcrição Genética , Linhagem Celular Tumoral , Humanos , Lisina/metabolismo , Metilação , TATA Box , Fatores Associados à Proteína de Ligação a TATA , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
14.
Mol Cell ; 24(1): 39-50, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-17018291

RESUMO

The 26S proteasome degrades polyubiquitinated proteins by an energy-dependent mechanism. Here we define multiple roles for ATP in 26S proteasome function. ATP binding is necessary and sufficient for assembly of 26S proteasome from 20S proteasome and PA700/19S subcomplexes and for proteasome activation. Proteasome assembly and activation may require distinct ATP binding events. The 26S proteasome degrades nonubiquitylated, unstructured proteins without ATP hydrolysis, indicating that substrate translocation per se does not require the energy of hydrolysis. Nonubiquitylated folded proteins and certain polyubiquitylated folded proteins were refractory to proteolysis. The latter were deubiquitylated by an ATP-independent mechanism. Other folded as well as unstructured polyubiquitylated proteins required ATP hydrolysis for proteolysis and deubiquitylation. Thus, ATP hydrolysis is not used solely for substrate unfolding. These results indicate that 26S proteasome-catalyzed degradation of polyubiquitylated proteins involves mechanistic coupling of several processes and that such coupling imposes an energy requirement not apparent for any isolated process.


Assuntos
Trifosfato de Adenosina/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Bovinos , Estabilidade Enzimática , Humanos , Hidrólise , Modelos Biológicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Ubiquitina/metabolismo
15.
Mol Biol Cell ; 17(8): 3705-16, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16760428

RESUMO

Polo-like kinase 1 (Plk1) is required for the generation of the tension-sensing 3F3/2 kinetochore epitope and facilitates kinetochore localization of Mad2 and other spindle checkpoint proteins. Here, we investigate the mechanism by which Plk1 itself is recruited to kinetochores. We show that Plk1 binds to budding uninhibited by benzimidazole 1 (Bub1) in mitotic human cells. The Plk1-Bub1 interaction requires the polo-box domain (PBD) of Plk1 and is enhanced by cyclin-dependent kinase 1 (Cdk1)-mediated phosphorylation of Bub1 at T609. The PBD-dependent binding of Plk1 to Bub1 facilitates phosphorylation of Bub1 by Plk1 in vitro. Depletion of Bub1 in HeLa cells by RNA interference (RNAi) diminishes the kinetochore localization of Plk1. Ectopic expression of the wild-type Bub1, but not the Bub1-T609A mutant, in Bub1-RNAi cells restores the kinetochore localization of Plk1. Our results suggest that phosphorylation of Bub1 at T609 by Cdk1 creates a docking site for the PBD of Plk1 and facilitates the kinetochore recruitment of Plk1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sequência de Aminoácidos , Proteína Quinase CDC2/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Células HeLa , Humanos , Mitose , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Proteínas Quinases/química , Estrutura Terciária de Proteína , Transporte Proteico , Interferência de RNA , Quinase 1 Polo-Like
16.
Dev Cell ; 10(5): 575-85, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16580887

RESUMO

Loss of sister-chromatid cohesion triggers chromosome segregation in mitosis and occurs through two mechanisms in vertebrate cells: (1) phosphorylation and removal of cohesin from chromosome arms by mitotic kinases, including Plk1, during prophase, and (2) cleavage of centromeric cohesin by separase at the metaphase-anaphase transition. Bub1 and the MEI-S332/Shugoshin (Sgo1) family of proteins protect centromeric cohesin from mitotic kinases during prophase. We show that human Sgo1 binds to protein phosphatase 2A (PP2A). PP2A localizes to centromeres in a Bub1-dependent manner. The Sgo1-PP2A interaction is required for centromeric localization of Sgo1 and proper chromosome segregation in human cells. Depletion of Plk1 by RNA interference (RNAi) restores centromeric localization of Sgo1 and prevents chromosome missegregation in cells depleted of PP2A_Aalpha. Our findings suggest that Bub1 targets PP2A to centromeres, which in turn maintains Sgo1 at centromeres by counteracting Plk1-mediated chromosome removal of Sgo1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Segregação de Cromossomos , Fosfoproteínas Fosfatases/metabolismo , Expressão Gênica , Células HeLa , Humanos , Mitose , Mutação/genética , Ligação Proteica , Proteínas Quinases/metabolismo , Proteína Fosfatase 2 , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Subunidades Proteicas , Transporte Proteico , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Células Tumorais Cultivadas , Quinase 1 Polo-Like
17.
Proc Natl Acad Sci U S A ; 102(1): 63-8, 2005 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-15623561

RESUMO

Expression of the human T lymphotropic virus type I (HTLV-I) transactivator/oncoprotein, Tax, leads to faulty mitosis as reflected by chromosome aneuploidy, cytokinesis failure, and formation of micro- and multinucleated cells. Here we show that HTLV-I-transformed T cells progress through S/G(2)/M phases of the cell cycle with a delay. This delay is correlated with a decrease in the levels of cyclin A, cyclin B1, and securin. In tax-expressing cells, the Cdc20-associated anaphase promoting complex (APC(Cdc20)), an E3 ubiquitin ligase that controls metaphase to anaphase transition, becomes active before cellular entry into mitosis as evidenced by premature cyclin B1 polyubiquitination and degradation during S/G(2). Consistent with the notion that Tax activates APC(Cdc20) directly, Tax is found to coimmunoprecipitate with Cdc20 and Cdc27/APC3. The APC(Cdc20) activity prematurely activated by Tax remains sensitive to spindle checkpoint inhibition. Unscheduled activation of APC(Cdc20) by Tax provides an explanation for the mitotic abnormalities in HTLV-I-infected cells and is likely to play an important role in the development of adult T cell leukemia.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Produtos do Gene tax/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Proteínas Cdc20 , Ciclo Celular/genética , Ciclina B/genética , Ciclina B/metabolismo , Ciclina B1 , Ativação Enzimática , Produtos do Gene tax/genética , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Células Jurkat , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poliubiquitina/metabolismo , RNA Mensageiro/metabolismo , Securina
18.
J Biol Chem ; 278(45): 45027-33, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12947083

RESUMO

The presence of DNA damage activates a conserved cellular response known as the DNA damage checkpoint pathway. This pathway induces a cell cycle arrest that persists until the damage is repaired. Consequently, the failure to arrest in response to DNA damage is associated with genomic instability. In budding yeast, activation of the DNA damage checkpoint pathway leads to a mitotic cell cycle arrest. Following the detection of DNA damage, the checkpoint signal is transduced via the Mec1 kinase, which in turn activates two kinases, Rad53 and Chk1 that act in parallel pathways to bring about the cell cycle arrest. The downstream target of Rad53 is unknown. The target of Chk1 is Pds1, an inhibitor of anaphase initiation whose degradation is a prerequisite for mitotic progression. Pds1 degradation is dependent on its ubiquitination by the anaphase-promoting complex/cyclosome ubiquitin ligase, acting in conjunction with the Cdc20 protein (APC/CCdc20). Previous studies showed that the Rad53 and Chk1 pathways independently lead to Pds1 stabilization but the mechanism for this was unknown. In the present study we show that both the Chk1 and the Rad53 pathways inhibit the APC/CCdc20-dependent ubiquitination of Pds1 but they affect different steps of the process: the Rad53 pathway inhibits the Pds1-Cdc20 interaction whereas Chk1-dependent phosphorylation of Pds1 inhibits the ubiquitination reaction itself. Finally, we show that once the DNA damage is repaired, Pds1 dephosphorylation is involved in the recovery from the checkpoint induced cell cycle arrest.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/metabolismo , Proteínas Cdc20 , Proteínas de Ciclo Celular/antagonistas & inibidores , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Reparo do DNA , DNA Fúngico/metabolismo , DNA Fúngico/efeitos da radiação , Proteínas Fúngicas , Raios gama , Peptídeos e Proteínas de Sinalização Intracelular , Mitose , Proteínas Nucleares/antagonistas & inibidores , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Securina , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA