Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Mol Cell ; 75(4): 823-834.e5, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31302001

RESUMO

Sirt3, as a major mitochondrial nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, is required for mitochondrial metabolic adaption to various stresses. However, how to regulate Sirt3 activity responding to metabolic stress remains largely unknown. Here, we report Sirt3 as a SUMOylated protein in mitochondria. SUMOylation suppresses Sirt3 catalytic activity. SUMOylation-deficient Sirt3 shows elevated deacetylation on mitochondrial proteins and increased fatty acid oxidation. During fasting, SUMO-specific protease SENP1 is accumulated in mitochondria and quickly de-SUMOylates and activates Sirt3. SENP1 deficiency results in hyper-SUMOylation of Sirt3 and hyper-acetylation of mitochondrial proteins, which reduces mitochondrial metabolic adaption responding to fasting. Furthermore, we find that fasting induces SENP1 translocation into mitochondria to activate Sirt3. The studies on mice show that Sirt3 SUMOylation mutation reduces fat mass and antagonizes high-fat diet (HFD)-induced obesity via increasing oxidative phosphorylation and energy expenditure. Our results reveal that SENP1-Sirt3 signaling modulates Sirt3 activation and mitochondrial metabolism during metabolic stress.


Assuntos
Cisteína Endopeptidases/metabolismo , Mitocôndrias/metabolismo , Mutação , Obesidade/metabolismo , Transdução de Sinais , Sirtuína 3/metabolismo , Sumoilação , Acetilação , Animais , Cisteína Endopeptidases/genética , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Mutantes , Mitocôndrias/genética , Mitocôndrias/patologia , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Sirtuína 3/genética
2.
Mol Cell ; 65(2): 296-309, 2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28065600

RESUMO

In mammalian cells, histone deacetylase (HDAC) and Sirtuin (SIRT) are two families responsible for removing acetyl groups from acetylated proteins. Here, we describe protein deacetylation coupled with deacetylimination as a function of lysyl oxidase (LOX) family members. LOX-like 3 (Loxl3) associates with Stat3 in the nucleus to deacetylate and deacetyliminate Stat3 on multiple acetyl-lysine sites. Surprisingly, Loxl3 N-terminal scavenger receptor cysteine-rich (SRCR) repeats, rather than the C-terminal oxidase catalytic domain, represent the major deacetylase/deacetyliminase activity. Loxl3-mediated deacetylation/deacetylimination disrupts Stat3 dimerization, abolishes Stat3 transcription activity, and restricts cell proliferation. In Loxl3-/- mice, Stat3 is constitutively acetylated and naive CD4+ T cells are potentiated in Th17/Treg cell differentiation. When overexpressed, the SRCR repeats from other LOX family members can catalyze protein deacetylation/deacetylimination. Thus, our findings delineate a hitherto-unknown mechanism of protein deacetylation and deacetylimination catalyzed by lysyl oxidases.


Assuntos
Aminoácido Oxirredutases/metabolismo , Linfócitos T CD4-Positivos/enzimologia , Colite/enzimologia , Processamento de Proteína Pós-Traducional , Fator de Transcrição STAT3/metabolismo , Acetilação , Aminoácido Oxirredutases/deficiência , Aminoácido Oxirredutases/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Catálise , Diferenciação Celular , Núcleo Celular/enzimologia , Proliferação de Células , Colite/genética , Colite/imunologia , Modelos Animais de Doenças , Genótipo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Domínios Proteicos , Multimerização Proteica , Interferência de RNA , Fator de Transcrição STAT3/genética , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia , Células Th17/enzimologia , Células Th17/imunologia , Transcrição Gênica , Transfecção
3.
EMBO J ; 39(10): e103111, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32187724

RESUMO

The homeostatic link between oxidative stress and autophagy plays an important role in cellular responses to a wide variety of physiological and pathological conditions. However, the regulatory pathway and outcomes remain incompletely understood. Here, we show that reactive oxygen species (ROS) function as signaling molecules that regulate autophagy through ataxia-telangiectasia mutated (ATM) and cell cycle checkpoint kinase 2 (CHK2), a DNA damage response (DDR) pathway activated during metabolic and hypoxic stress. We report that CHK2 binds to and phosphorylates Beclin 1 at Ser90/Ser93, thereby impairing Beclin 1-Bcl-2 autophagy-regulatory complex formation in a ROS-dependent fashion. We further demonstrate that CHK2-mediated autophagy has an unexpected role in reducing ROS levels via the removal of damaged mitochondria, which is required for cell survival under stress conditions. Finally, CHK2-/- mice display aggravated infarct phenotypes and reduced Beclin 1 p-Ser90/Ser93 in a cerebral stroke model, suggesting an in vivo role of CHK2-induced autophagy in cell survival. Taken together, these results indicate that the ROS-ATM-CHK2-Beclin 1-autophagy axis serves as a physiological adaptation pathway that protects cells exposed to pathological conditions from stress-induced tissue damage.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína Beclina-1/metabolismo , Quinase do Ponto de Checagem 2/metabolismo , AVC Isquêmico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Autofagia , Linhagem Celular , Modelos Animais de Doenças , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Camundongos , Estresse Oxidativo , Fosforilação
4.
Mol Cell ; 55(1): 31-46, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24882211

RESUMO

MutS protein homolog 2 (MSH2) is a key DNA mismatch repair protein. It forms the MSH2-MSH6 (MutSα) and MSH2-MSH3 (MutSß) heterodimers, which help to ensure genomic integrity. MutSα not only recognizes and repairs mismatched nucleotides but also recognizes DNA adducts induced by DNA-damaging agents, and triggers cell-cycle arrest and apoptosis. Loss or depletion of MutSα from cells leads to microsatellite instability (MSI) and resistance to DNA damage. Although the level of MutSα can be reduced by the ubiquitin-proteasome pathway, the detailed mechanisms of this regulation remain elusive. Here we report that histone deacetylase 6 (HDAC6) sequentially deacetylates and ubiquitinates MSH2, leading to MSH2 degradation. In addition, HDAC6 significantly reduces cellular sensitivity to DNA-damaging agents and decreases cellular DNA mismatch repair activities by downregulation of MSH2. Overall, these findings reveal a mechanism by which proper levels of MutSα are maintained.


Assuntos
Histona Desacetilases/fisiologia , Proteína 2 Homóloga a MutS/metabolismo , Acetilação , Animais , Células Cultivadas , Células HEK293 , Células HeLa , Desacetilase 6 de Histona , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Camundongos , Estabilidade Proteica , Ubiquitinação
5.
J Biol Chem ; 288(23): 16567-16578, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23612972

RESUMO

The proteasome activator REGγ has been reported to promote degradation of steroid receptor coactivator-3 and cyclin-dependent kinase inhibitors p21, p16, and p19 in a ubiquitin- and ATP-independent manner. A recent comparative analysis of REGγ expression in mouse and human tissues reveals a unique pattern of REGγ in specific cell types, suggesting undisclosed functions and biological importance of this molecule. Despite the emerging progress made in REGγ-related studies, how REGγ function is regulated remains to be explored. In this study, we report for the first time that REGγ can be acetylated mostly on its lysine 195 (Lys-195) residue by CREB binding protein (CBP), which can be reversed by sirtuin 1 (SIRT1) in mammalian cells. Site-directed mutagenesis abrogated acetylation at Lys-195 and significantly attenuated the capability of REGγ to degrade its target substrates, p21 and hepatitis C virus core protein. Mechanistically, acetylation at Lys-195 is important for the interactions between REGγ monomers and ultimately influences REGγ heptamerization. Biological analysis of cells containing REGγ-WT or REGγ-K195R mutant indicates an impact of acetylation on REGγ-mediated regulation of cell proliferation and cell cycle progression. These findings reveal a previously unknown mechanism in the regulation of REGγ assembly and activity, suggesting a potential venue for the intervention of the ubiquitin-independent REGγ proteasome activity.


Assuntos
Autoantígenos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Multimerização Proteica/fisiologia , Proteólise , Acetilação , Substituição de Aminoácidos , Animais , Autoantígenos/genética , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Ciclo Celular/fisiologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Complexo de Endopeptidases do Proteassoma/genética , Ubiquitina/genética , Ubiquitina/metabolismo
6.
J Biol Chem ; 288(24): 17532-43, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23629655

RESUMO

Trithorax group proteins methylate lysine 4 of histone 3 (H3K4) at active gene promoters. MLL5 protein, a member of the Trithorax protein family, has been implicated in the control of the cell cycle progression; however, the underlying molecular mechanism(s) have not been fully determined. In this study, we found that the MLL5 protein can associate with the cell cycle regulator "host cell factor" (HCF-1). The interaction between MLL5 and HCF-1 is mediated by the "HCF-1 binding motif" (HBM) of the MLL5 protein and the Kelch domain of the HCF-1 protein. Confocal microscopy showed that the MLL5 protein largely colocalized with HCF-1 in the nucleus. Knockdown of MLL5 resulted in reduced cell proliferation and cell cycle arrest in the G1 phase. Moreover, down-regulation of E2F1 target gene expression and decreased H3K4me3 levels at E2F1-responsive promoters were observed in MLL5 knockdown cells. Additionally, the core subunits, including ASH2L, RBBP5, and WDR5, that are necessary for effective H3K4 methyltransferase activities of the Trithorax protein complexes, were absent in the MLL5 complex, suggesting that a distinct mechanism may be used by MLL5 for exerting its H3K4 methyltransferase activity. Together, our findings demonstrate that MLL5 could associate with HCF-1 and then be recruited to E2F1-responsive promoters to stimulate H3K4 trimethylation and transcriptional activation, thereby facilitating the cell cycle G1 to S phase transition.


Assuntos
Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição E2F1/metabolismo , Regulação da Expressão Gênica , Fator C1 de Célula Hospedeira/metabolismo , Sequência de Aminoácidos , Núcleo Celular , Proliferação de Células , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Fator C1 de Célula Hospedeira/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Espectrometria de Massas , Metilação , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Mapeamento de Peptídeos , Regiões Promotoras Genéticas , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/genética , Fatores de Transcrição/metabolismo
7.
J Biol Chem ; 288(46): 33156-70, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24089523

RESUMO

Histone deacetylase 6 (HDAC6) is well known for its ability to promote cell migration through deacetylation of its cytoplasmic substrates such as α-tubulin. However, how HDAC6 itself is regulated to control cell motility remains elusive. Previous studies have shown that one third of extracellular signal-regulated kinase (ERK) is associated with the microtubule cytoskeleton in cells. Yet, no connection between HDAC6 and ERK has been discovered. Here, for the first time, we reveal that ERK binds to and phosphorylates HDAC6 to promote cell migration via deacetylation of α-tubulin. We have identified two novel ERK-mediated phosphorylation sites: threonine 1031 and serine 1035 in HDAC6. Both sites were phosphorylated by ERK1 in vitro, whereas Ser-1035 was phosphorylated in response to the activation of EGFR-Ras-Raf-MEK-ERK signaling pathway in vivo. HDAC6-null mouse embryonic fibroblasts rescued by the nonphosphorylation mimicking mutant displayed significantly reduced cell migration compared with those rescued by the wild type. Consistently, the nonphosphorylation mimicking mutant exerted lower tubulin deacetylase activity in vivo compared with the wild type. These data indicate that ERK/HDAC6-mediated cell motility is through deacetylation of α-tubulin. Overall, our results suggest that HDAC6-mediated cell migration could be governed by EGFR-Ras-Raf-MEK-ERK signaling.


Assuntos
Movimento Celular/fisiologia , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Tubulina (Proteína)/metabolismo , Acetilação , Animais , Células CHO , Cricetinae , Cricetulus , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Desacetilase 6 de Histona , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Mutantes , Proteína Quinase 3 Ativada por Mitógeno/genética , Tubulina (Proteína)/genética
8.
Polymers (Basel) ; 16(6)2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38543402

RESUMO

Bamboo scrimber is acknowledged for its eco-friendly potential as a structural material. Its properties are significantly affected by both its density and resin content, but the effect of resin content on the properties under high density is not yet known. In this study, the microstructure, water resistance, mechanical properties, and thermal stability of bamboo scrimbers with varying resin content at a density of 1.30 g/cm3 were investigated. The results unearthed that phenolic resin assisted in the densification of bamboo cells during hot pressing, and a higher resin content could effectively reduce the cracks in the scrimber. The inherent cellulose I structure remained unaffected, but an increase in resin content led to a noticeable decline in crystallinity. Additionally, an increase in resin content pronouncedly improved the water resistance and dimensional stability of bamboo scrimbers. The water absorption and thickness swelling were as low as 9.67% and 7.62%, respectively. The modulus of rupture (MOR) exhibited a marginal increase with the amount of resin, whereas the compressive strength and short-beam shearing strength first increased and then decreased. Their peak strengths were 327.87 MPa at a resin content of 15 wt.%, and 168.85 MPa and 25.96 MPa at 11 wt.%, respectively. However, phenolic resin accelerated the thermal decomposition of bamboo scrimbers, and more resin worsened the thermal stability. These research outcomes offer a dual advantage, providing both a theoretical foundation and concrete data that can inform the production and practical application of high-density bamboo scrimbers.

9.
J Biol Chem ; 287(23): 18937-52, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22496453

RESUMO

The Kruppel-associated box (KRAB)-associated co-repressor KAP1 is an essential nuclear co-repressor for the KRAB zinc finger protein superfamily of transcriptional factors. Ataxia telangiectasia mutated (ATM)-Chk2 and ATM- and Rad3-related (ATR)-Chk1 are two primary kinase signaling cascades activated in response to DNA damage. A growing body of evidence suggests that ATM and ATR phosphorylate KAP1 at Ser-824 in response to DNA damage and regulate KAP1-dependent chromatin condensation, DNA repair, and gene expression. Here, we show that, depending on the type of DNA damage that occurs, KAP1 Ser-473 can be phosphorylated by ATM-Chk2 or ATR-Chk1 kinases. Phosphorylation of KAP1 at Ser-473 attenuated its binding to the heterochromatin protein 1 family proteins and inhibited its transcriptional repression of KRAB-zinc finger protein (KRAB-ZFP) target genes. Moreover, KAP1 Ser-473 phosphorylation induced by DNA damage stimulated KAP1-E2F1 binding. Overexpression of heterochromatin protein 1 significantly inhibited E2F1-KAP1 binding. Elimination of KAP1 Ser-473 phosphorylation increased E2F1-targeted proapoptotic gene expression and E2F1-induced apoptosis in response to DNA damage. Furthermore, loss of phosphorylation of KAP1 Ser-473 led to less BRCA1 focus formation and slower kinetics of loss of γH2AX foci after DNA damage. KAP1 Ser-473 phosphorylation was required for efficient DNA repair and cell survival in response to DNA damage. Our studies reveal novel functions of KAP1 Ser-473 phosphorylation under stress.


Assuntos
Apoptose/fisiologia , Montagem e Desmontagem da Cromatina , Dano ao DNA , Proteínas Repressoras/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Células HEK293 , Células HeLa , Humanos , Fosforilação/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/genética , Serina/genética , Serina/metabolismo , Proteína 28 com Motivo Tripartido , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Nat Cell Biol ; 8(9): 1025-31, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16892051

RESUMO

The nicotinamide adenine dinucleotide (NAD)-dependent deacetylase Sir2 (silent information regulator 2) regulates gene silencing in yeast and promotes lifespan extension during caloric restriction. The mammalian homologue of Sir2 (SirT1) regulates p53, NF-kappaB and Forkhead transcription factors, and is implicated in stress response. This report shows that the cell-cycle and apoptosis regulator E2F1 induces SirT1 expression at the transcriptional level. Furthermore, SirT1 binds to E2F1 and inhibits E2F1 activities, forming a negative feedback loop. Knockdown of SirT1 by small interference RNA (siRNA) increases E2F1 transcriptional and apoptotic functions. DNA damage by etoposide causes E2F1-dependent induction of SirT1 expression and knockdown of SirT1 increases sensitivity to etoposide. These results reveal a mutual regulation between E2F1 and SirT1 that affects cellular sensitivity to DNA damage.


Assuntos
Apoptose , Dano ao DNA , Fator de Transcrição E2F1/metabolismo , Sirtuínas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Etoposídeo/toxicidade , Retroalimentação Fisiológica , Humanos , Mutação , Ligação Proteica , RNA Interferente Pequeno/genética , Sirtuína 1 , Sirtuínas/genética
11.
Analyst ; 138(22): 7016-22, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24098881

RESUMO

Cell surface glycans are a class of sophisticated biomolecules related to cancer development and progression, and their analysis is of great significance for early cancer diagnosis and treatment. In this paper, we proposed a fluorescence assay to evaluate glycan expression on living cancer cells based on a competitive strategy coupled with dual-functionalized nanobiocomposites. The competitive assay was conducted between living cancer cells and thiomannosyl derivatives using concanavalin A (Con A)-modified electrode as the interaction platform. To impart fluorescence signaling ability to competitive derivatives, quantum dots (QDs) were anchored on BSA-protected Au nanoparticles, and thiomannosyl derivatives were further immobilized on the nanoparticle surface through Au-S binding. Due to the spacing between QDs and Au nanoparticles by BSA, the {QDs-Au-BSA-mannose} nanobiocomposites maintained the fluorescence of QDs and showed binding ability with the Con A-modified electrode. Au nanorods (AuNRs)-modified electrode was used as an effective substrate to immobilize Con A. This assay was successfully applied to the analysis of two cancer cells lines (A549 and QGY-7701). The method is simple and shows promise for the study of glycan expression on living cancer cells.


Assuntos
Bioensaio/métodos , Nanotecnologia , Polissacarídeos/genética , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Fluorescência , Ouro/química , Humanos , Microscopia Eletrônica de Transmissão , Polissacarídeos/metabolismo , Receptores de Concanavalina A/química
12.
J Cell Sci ; 123(Pt 23): 4076-84, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21084564

RESUMO

The proteasome activator REGγ mediates a shortcut for the destruction of intact mammalian proteins. The biological roles of REGγ and the underlying mechanisms are not fully understood. Here we provide evidence that REGγ regulates cellular distribution of p53 by facilitating its multiple monoubiquitylation and subsequent nuclear export and degradation. We also show that inhibition of p53 tetramerization by REGγ might further enhance cytoplasmic relocation of p53 and reduce active p53 in the nucleus. Furthermore, multiple monoubiquitylation of p53 enhances its physical interaction with HDM2 and probably facilitates subsequent polyubiquitylation of p53, suggesting that monoubiquitylation can act as a signal for p53 degradation. Depletion of REGγ sensitizes cells to stress-induced apoptosis, validating its crucial role in the control of apoptosis, probably through regulation of p53 function. Using a mouse xenograft model, we show that REGγ knockdown results in a significant reduction of tumor growth, suggesting an important role for REGγ in tumor development. Our study therefore demonstrates that REGγ-mediated inactivation of p53 is one of the mechanisms involved in cancer progression.


Assuntos
Autoantígenos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Autoantígenos/genética , Linhagem Celular Tumoral , Núcleo Celular/química , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Multimerização Proteica , Transporte Proteico , Distribuição Aleatória , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Ubiquitinação
13.
Biochem J ; 434(2): 275-85, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21133853

RESUMO

The tumour suppressor ARF (alternative reading frame) is one of the most important oncogenic stress sensors. ARF provides an 'oncogenic checkpoint' function through both p53-dependent and p53-independent mechanisms. In the present study, we demonstrate a novel p53-independent interaction between p14(ARF) and the adenovirus oncoprotein E1A. p14(ARF) inhibits E1A transcriptional function and promotes ubiquitination-dependent degradation of E1A. p14(ARF) overexpression relocalizes E1A into the nucleolus and inhibits E1A-induced cellular DNA replication independent of p53. Knockdown of endogenous p14(ARF) increases E1A transactivation. In addition, E1A can competitively inhibit ARF-Mdm2 (murine double minute 2) complex formation. These results identify a novel binding partner of p14(ARF) and reveal a mutually inhibitory interaction between p14(ARF) and E1A. We speculate that the ARF-E1A interaction may represent an additional host defence mechanism to limit viral replication. Alternatively, the interaction may allow adenovirus to sense the functional state of p53 in host cells, and fine-tune its own replication activity to prevent the triggering of a detrimental host response.


Assuntos
Proteínas E1A de Adenovirus/antagonistas & inibidores , Proteínas E1A de Adenovirus/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Sítios de Ligação , Linhagem Celular , Replicação do DNA , Inativação Gênica , Células HeLa , Humanos , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Replicação Viral/fisiologia
14.
Nat Commun ; 13(1): 6548, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36319643

RESUMO

Aberrant expression of the Forkhead box transcription factor, FOXQ1, is a prevalent mechanism of epithelial-mesenchymal transition (EMT) and metastasis in multiple carcinoma types. However, it remains unknown how FOXQ1 regulates gene expression. Here, we report that FOXQ1 initiates EMT by recruiting the MLL/KMT2 histone methyltransferase complex as a transcriptional coactivator. We first establish that FOXQ1 promoter recognition precedes MLL complex assembly and histone-3 lysine-4 trimethylation within the promoter regions of critical genes in the EMT program. Mechanistically, we identify that the Forkhead box in FOXQ1 functions as a transactivation domain directly binding the MLL core complex subunit RbBP5 without interrupting FOXQ1 DNA binding activity. Moreover, genetic disruption of the FOXQ1-RbBP5 interaction or pharmacologic targeting of KMT2/MLL recruitment inhibits FOXQ1-dependent gene expression, EMT, and in vivo tumor progression. Our study suggests that targeting the FOXQ1-MLL epigenetic axis could be a promising strategy to combat triple-negative breast cancer metastatic progression.


Assuntos
Neoplasias da Mama , Segunda Neoplasia Primária , Feminino , Humanos , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Segunda Neoplasia Primária/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Melanoma Maligno Cutâneo
15.
Nat Commun ; 13(1): 6350, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36289222

RESUMO

The methyltransferase like 3 (METTL3) has been generally recognized as a nuclear protein bearing oncogenic properties. We find predominantly cytoplasmic METTL3 expression inversely correlates with node metastasis in human cancers. It remains unclear if nuclear METTL3 is functionally distinct from cytosolic METTL3 in driving tumorigenesis and, if any, how tumor cells sense oncogenic insults to coordinate METTL3 functions within these intracellular compartments. Here, we report an acetylation-dependent regulation of METTL3 localization that impacts on metastatic dissemination. We identify an IL-6-dependent positive feedback axis to facilitate nuclear METTL3 functions, eliciting breast cancer metastasis. IL-6, whose mRNA transcript is subjected to METTL3-mediated m6A modification, promotes METTL3 deacetylation and nuclear translocation, thereby inducing global m6A abundance. This deacetylation-mediated nuclear shift of METTL3 can be counterbalanced by SIRT1 inhibition, a process that is further enforced by aspirin treatment, leading to ablated lung metastasis via impaired m6A methylation. Intriguingly, acetylation-mimetic METTL3 mutant reconstitution results in enhanced translation and compromised metastatic potential. Our study identifies an acetylation-dependent regulatory mechanism determining the subcellular localization of METTL3, which may provide mechanistic clues for developing therapeutic strategies to combat breast cancer metastasis.


Assuntos
Neoplasias da Mama , Metiltransferases , Humanos , Feminino , Metiltransferases/metabolismo , Acetilação , Sirtuína 1/metabolismo , Interleucina-6/metabolismo , RNA Mensageiro/metabolismo , Carcinogênese , Neoplasias da Mama/genética , Proteínas Nucleares/metabolismo , Aspirina
16.
Cell Rep ; 40(2): 111062, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35830807

RESUMO

Aging is a primary risk factor for neurodegenerative diseases, such as Alzheimer's disease (AD). SIRT2, an NAD+(nicotinamide adenine dinucleotide)-dependent deacetylase, accumulates in the aging brain. Here, we report that, in the amyloid precursor protein (APP)/PS1 transgenic mouse model of AD, genetic deletion of SIRT2 or pharmacological inhibition of SIRT2 ameliorates cognitive impairment. We find that suppression of SIRT2 enhances acetylation of APP, which promotes non-amyloidogenic processing of APP at the cell surface, leading to increased soluble APP-α (sAPPα). We discover that lysines 132 and 134 of the major pathogenic protein ß-amyloid (Aß) precursor are acetylated and that these residues are deacetylated by SIRT2. Strikingly, exogenous expression of wild-type or an acetylation-mimic APP mutant protects cultured primary neurons from Aß42 challenge. Our study identifies SIRT2-mediated deacetylation of APP on K132 and K134 as a regulated post-translational modification (PTM) and suggests inhibition of SIRT2 as a potential therapeutic strategy for AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Acetilação , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Presenilina-1/metabolismo , Processamento de Proteína Pós-Traducional , Sirtuína 2/genética , Sirtuína 2/metabolismo
17.
J Biol Chem ; 285(43): 32988-32998, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20729194

RESUMO

Mps1 is a dual specificity protein kinase with key roles in regulating the spindle assembly checkpoint and chromosome-microtubule attachments. Consistent with these mitotic functions, Mps1 protein levels fluctuate during the cell cycle, peaking at early mitosis and abruptly declining during mitotic exit and progression into the G(1) phase. Although evidence in budding yeast indicates that Mps1 is targeted for degradation at anaphase by the anaphase-promoting complex (APC)-c(Cdc20) complex, little is known about the regulatory mechanisms that govern Mps1 protein levels in human cells. Here, we provide evidence for the ubiquitin ligase/proteosome pathway in regulating human Mps1 levels during late mitosis through G(1) phase. First, we showed that treatment of HEK 293T cells with the proteosome inhibitor MG132 resulted in an increase in both the polyubiquitination and the accumulation of Mps1 protein levels. Next, Mps1 was shown to co-precipitate with APC and its activators Cdc20 and Cdh1 in a cell cycle-dependent manner. Consistent with this, overexpression of Cdc20 or Cdh1 led to a marked reduction of endogenous Mps1 levels during anaphase or G(1) phase, respectively. In contrast, depletion of Cdc20 or Cdh1 by RNAi treatment both led to the stabilization of Mps1 protein during mitosis or G(1) phase, respectively. Finally, we identified a single D-box motif in human Mps1 that is required for its ubiquitination and degradation. Failure to appropriately degrade Mps1 is sufficient to trigger centrosome amplification and mitotic abnormalities in human cells. Thus, our results suggest that the sequential actions of the APC-c(Cdc20) and APC-c(Cdh1) ubiquitin ligases regulate the clearance of Mps1 levels and are critical for Mps1 functions during the cell cycle in human cells.


Assuntos
Caderinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Antígenos CD , Caderinas/genética , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Linhagem Celular , Centrossomo/enzimologia , Estabilidade Enzimática/genética , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases , Ubiquitina-Proteína Ligases/genética
18.
Nat Commun ; 12(1): 20, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33397932

RESUMO

Drug resistance and tumor recurrence are major challenges in cancer treatment. Cancer cells often display centrosome amplification. To maintain survival, cancer cells achieve bipolar division by clustering supernumerary centrosomes. Targeting centrosome clustering is therefore considered a promising therapeutic strategy. However, the regulatory mechanisms of centrosome clustering remain unclear. Here we report that KIFC1, a centrosome clustering regulator, is positively associated with tumor recurrence. Under DNA damaging treatments, the ATM and ATR kinases phosphorylate KIFC1 at Ser26 to selectively maintain the survival of cancer cells with amplified centrosomes via centrosome clustering, leading to drug resistance and tumor recurrence. Inhibition of KIFC1 phosphorylation represses centrosome clustering and tumor recurrence. This study identified KIFC1 as a prognostic tumor recurrence marker, and revealed that tumors can acquire therapeutic resistance and recurrence via triggering centrosome clustering under DNA damage stresses, suggesting that blocking KIFC1 phosphorylation may open a new vista for cancer therapy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Centrossomo/metabolismo , Cinesinas/metabolismo , Recidiva Local de Neoplasia/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Instabilidade Cromossômica , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Cinesinas/química , Camundongos , Recidiva Local de Neoplasia/patologia , Fosforilação , Fosfosserina/metabolismo
19.
Sci Adv ; 7(9)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33627431

RESUMO

Improper distribution of chromosomes during mitosis can contribute to malignant transformation. Higher eukaryotes have evolved a mitotic catastrophe mechanism for eliminating mitosis-incompetent cells; however, the signaling cascade and its epigenetic regulation are poorly understood. Our analyses of human cancerous tissue revealed that the NAD-dependent deacetylase SIRT2 is up-regulated in early-stage carcinomas of various organs. Mass spectrometry analysis revealed that SIRT2 interacts with and deacetylates the structural maintenance of chromosomes protein 1 (SMC1A), which then promotes SMC1A phosphorylation to properly drive mitosis. We have further demonstrated that inhibition of SIRT2 activity or continuously increasing SMC1A-K579 acetylation causes abnormal chromosome segregation, which, in turn, induces mitotic catastrophe in cancer cells and enhances their vulnerability to chemotherapeutic agents. These findings suggest that regulation of the SIRT2-SMC1A axis through deacetylation-phosphorylation permits escape from mitotic catastrophe, thus allowing early precursor lesions to overcome oncogenic stress.


Assuntos
Antimitóticos , Sirtuína 2 , Acetilação , Carcinogênese/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética , Humanos , Fosforilação , Sirtuína 2/genética , Sirtuína 2/metabolismo
20.
J Biol Chem ; 284(27): 18210-7, 2009 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-19433578

RESUMO

The NAD-dependent deacetylase SirT1 regulates factors involved in stress response and cell survival and is a potential drug target of activators and inhibitors. Determination of SirT1 function in tumor cells is important for its targeting in cancer therapy. We found that SirT1 knockdown by short hairpin RNA accelerates tumor xenograft formation by HCT116 cells, whereas SirT1 overexpression inhibits tumor formation. Furthermore, pharmacological inhibition of SirT1 stimulates cell proliferation under conditions of growth factor deprivation. Paradoxically, SirT1 inhibition also sensitizes cells to apoptosis by chemotherapy drugs. Immunohistochemical staining revealed high level SirT1 in normal colon mucosa and benign adenomas. SirT1 overexpression was observed in approximately 25% of stage I/II/III colorectal adenocarcinomas but rarely found in advanced stage IV tumors. Furthermore, approximately 30% of carcinomas showed lower than normal SirT1 expression. This pattern is consistent with SirT1 having pleiotropic effects during cancer development (anti-proliferation and anti-apoptotic). These results suggest a rationale for the use of SirT1 activators and inhibitors in the prevention and treatment of colon cancer.


Assuntos
Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Sirtuínas/genética , Sirtuínas/metabolismo , Adenocarcinoma/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Divisão Celular/fisiologia , Colo/patologia , Colo/fisiologia , Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Imuno-Histoquímica , Camundongos , Transplante de Neoplasias , RNA Interferente Pequeno , Sirtuína 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA