Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 37(5-6): 204-217, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36921996

RESUMO

Although it is well established that Huntington's disease (HD) is mainly caused by polyglutamine-expanded mutant huntingtin (mHTT), the molecular mechanism of mHTT-mediated actions is not fully understood. Here, we showed that expression of the N-terminal fragment containing the expanded polyglutamine (HTTQ94) of mHTT is able to promote both the ACSL4-dependent and the ACSL4-independent ferroptosis. Surprisingly, inactivation of the ACSL4-dependent ferroptosis fails to show any effect on the life span of Huntington's disease mice. Moreover, by using RNAi-mediated screening, we identified ALOX5 as a major factor required for the ACSL4-independent ferroptosis induced by HTTQ94. Although ALOX5 is not required for the ferroptotic responses triggered by common ferroptosis inducers such as erastin, loss of ALOX5 expression abolishes HTTQ94-mediated ferroptosis upon reactive oxygen species (ROS)-induced stress. Interestingly, ALOX5 is also required for HTTQ94-mediated ferroptosis in neuronal cells upon high levels of glutamate. Mechanistically, HTTQ94 activates ALOX5-mediated ferroptosis by stabilizing FLAP, an essential cofactor of ALOX5-mediated lipoxygenase activity. Notably, inactivation of the Alox5 gene abrogates the ferroptosis activity in the striatal neurons from the HD mice; more importantly, loss of ALOX5 significantly ameliorates the pathological phenotypes and extends the life spans of these HD mice. Taken together, these results demonstrate that ALOX5 is critical for mHTT-mediated ferroptosis and suggest that ALOX5 is a potential new target for Huntington's disease.


Assuntos
Ferroptose , Doença de Huntington , Animais , Camundongos , Modelos Animais de Doenças , Ferroptose/genética , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Neurônios/metabolismo , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio/metabolismo
2.
Mol Cell ; 75(6): 1299-1314.e6, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31353207

RESUMO

MRE11 nuclease forms a trimeric complex (MRN) with RAD50 and NBS1 and plays a central role in preventing genomic instability. When DNA double-strand breaks (DSBs) occur, MRN is quickly recruited to the damage site and initiates DNA end resection; accordingly, MRE11 must be tightly regulated to avoid inefficient repair or nonspecific resection. Here, we show that MRE11 and RAD50 form a complex (MRC) with C1QBP, which stabilizes MRE11/RAD50, while inhibiting MRE11 nuclease activity by preventing its binding to DNA or chromatin. Upon DNA damage, ATM phosphorylates MRE11-S676/S678 to quickly dissociate the MRC complex. Either excess or insufficient C1QBP impedes the recruitment of MRE11 to DSBs and impairs the DNA damage response. C1QBP is highly expressed in breast cancer and positively correlates with MRE11 expression, and the inhibition of C1QBP enhances tumor regression with chemotherapy. By influencing MRE11 at multiple levels, C1QBP is, thus, an important player in the DNA damage response.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga , Proteína Homóloga a MRE11/metabolismo , Proteínas Mitocondriais/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Hidrolases Anidrido Ácido/genética , Animais , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos , Proteína Homóloga a MRE11/genética , Proteínas Mitocondriais/genética , Complexos Multiproteicos/genética , Proteínas Nucleares/genética , Estabilidade Proteica , Células Sf9 , Spodoptera
3.
Mol Cell ; 74(6): 1250-1263.e6, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31054974

RESUMO

Alternative pre-mRNA-splicing-induced post-transcriptional gene expression regulation is one of the pathways for tumors maintaining proliferation rates accompanying the malignant phenotype under stress. Here, we uncover a list of hyperacetylated proteins in the context of acutely reduced Acetyl-CoA levels under nutrient starvation. PHF5A, a component of U2 snRNPs, can be acetylated at lysine 29 in response to multiple cellular stresses, which is dependent on p300. PHF5A acetylation strengthens the interaction among U2 snRNPs and affects global pre-mRNA splicing pattern and extensive gene expression. PHF5A hyperacetylation-induced alternative splicing stabilizes KDM3A mRNA and promotes its protein expression. Pathologically, PHF5A K29 hyperacetylation and KDM3A upregulation axis are correlated with poor prognosis of colon cancer. Our findings uncover a mechanism of an anti-stress pathway through which acetylation on PHF5A promotes the cancer cells' capacity for stress resistance and consequently contributes to colon carcinogenesis.


Assuntos
Processamento Alternativo , Carcinogênese/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas de Ligação a RNA/genética , Transativadores/genética , Acetilcoenzima A/deficiência , Acetilação , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/metabolismo , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Transdução de Sinais , Análise de Sobrevida , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
4.
EMBO Rep ; 24(12): e57500, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37870259

RESUMO

SIRT2, a cytoplasmic member of the Sirtuin family, has important roles in immunity and inflammation. However, its function in regulating the response to DNA virus infection remains elusive. Here, we find that SIRT2 is a unique regulator among the Sirtuin family that negatively modulates the cGAS-STING-signaling pathway. SIRT2 is down-regulated after Herpes simplex virus-1 (HSV-1) infection, and SIRT2 deficiency markedly elevates the expression levels of type I interferon (IFN). SIRT2 inhibits the DNA binding ability and droplet formation of cGAS by interacting with and deacetylating G3BP1 at K257, K276, and K376, leading to the disassembly of the cGAS-G3BP1 complex, which is critical for cGAS activation. Administration of AGK2, a selective SIRT2 inhibitor, protects mice from HSV-1 infection and increases the expression of IFN and IFN-stimulated genes. Our study shows that SIRT2 negatively regulates cGAS activation through G3BP1 deacetylation, suggesting a potential antiviral strategy by modulating SIRT2 activity.


Assuntos
DNA Helicases , Imunidade Inata , Animais , Camundongos , DNA Helicases/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Helicases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Transdução de Sinais , Sirtuína 2/genética , Sirtuína 2/metabolismo
5.
Am J Pathol ; 193(7): 899-912, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37068638

RESUMO

The accuracy and timeliness of the pathologic diagnosis of soft tissue tumors (STTs) critically affect treatment decision and patient prognosis. Thus, it is crucial to make a preliminary judgement on whether the tumor is benign or malignant with hematoxylin and eosin-stained images. A deep learning-based system, Soft Tissue Tumor Box (STT-BOX), is presented herein, with only hematoxylin and eosin images for malignant STT identification from benign STTs with histopathologic similarity. STT-BOX assumed gastrointestinal stromal tumor as a baseline for malignant STT evaluation, and distinguished gastrointestinal stromal tumor from leiomyoma and schwannoma with 100% area under the curve in patients from three hospitals, which achieved higher accuracy than the interpretation of experienced pathologists. Particularly, this system performed well on six common types of malignant STTs from The Cancer Genome Atlas data set, accurately highlighting the malignant mass lesion. STT-BOX was able to distinguish ovarian malignant sex-cord stromal tumors without any fine-tuning. This study included mesenchymal tumors that originated from the digestive system, bone and soft tissues, and reproductive system, where the high accuracy of migration verification may reveal the morphologic similarity of the nine types of malignant tumors. Further evaluation in a pan-STT setting would be potential and prospective, obviating the overuse of immunohistochemistry and molecular tests, and providing a practical basis for clinical treatment selection in a timely manner.


Assuntos
Aprendizado Profundo , Tumores do Estroma Gastrointestinal , Neoplasias Ovarianas , Neoplasias de Tecidos Moles , Feminino , Humanos , Tumores do Estroma Gastrointestinal/diagnóstico , Tumores do Estroma Gastrointestinal/patologia , Amarelo de Eosina-(YS) , Hematoxilina , Estudos Prospectivos , Neoplasias de Tecidos Moles/diagnóstico
6.
Nucleic Acids Res ; 50(7): 3817-3834, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35349706

RESUMO

Reactive oxygen species (ROS) are constantly produced in cells, an excess of which causes oxidative stress. ROS has been linked to regulation of the Hippo pathway; however, the underlying detailed mechanisms remain unclear. Here, we report that MOB1, a substrate of MST1/2 and co-activator of LATS1/2 in the canonical Hippo pathway, interacts with and is acetylated at lysine 11 by acetyltransferase CBP and deacetylated by HDAC6. MOB1-K11 acetylation stabilizes itself by reducing its binding capacity with E3 ligase Praja2 and subsequent ubiquitination. MOB1-K11 acetylation increases its phosphorylation and activates LATS1. Importantly, upstream oxidative stress signals promote MOB1 acetylation by suppressing CBP degradation, independent of MST1/2 kinase activity and HDAC6 deacetylation effect, thereby linking oxidative stress to activation of the Hippo pathway. Functionally, the acetylation-deficient mutant MOB1-K11R promotes lung cancer cell proliferation, migration and invasion in vitro and accelerates tumor growth in vivo, compared to the wild-type MOB1. Clinically, acetylated MOB1 corresponds to better prediction of overall survival in patients with non-small cell lung cancer. Therefore, as demonstrated, an oxidative stress-CBP regulatory axis controls MOB1-K11 acetylation and activates LATS1, thereby activating the Hippo pathway and suppressing YAP/TAZ nuclear translocation and tumor progression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Quimiocina CXCL10/metabolismo , Via de Sinalização Hippo , Neoplasias Pulmonares , Acetilação , Humanos , Neoplasias Pulmonares/genética , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio
7.
Biol Chem ; 404(6): 585-599, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-36420535

RESUMO

The dysregulation of the translation elongation factor families which are responsible for reprogramming of mRNA translation has been shown to contribute to tumor progression. Here, we report that the acetylation of eukaryotic Elongation Factor 1 Alpha 1 (eEF1A1/EF1A1) is required for genotoxic stress response and maintaining the malignancy of colorectal cancer (CRC) cells. The evolutionarily conserved site K439 is identified as the key acetylation site. Tissue expression analysis demonstrates that the acetylation level of eEF1A1 K439 is higher than paired normal tissues. Most importantly, hyperacetylation of eEF1A1 at K439 negatively correlates with CRC patient survival. Mechanistically, CBP and SIRT1 are the major acetyltransferase and deacetylase of eEF1A1. Hyperacetylation of eEF1A1 at K439 shows a significant tumor-promoting effect by increasing the capacity of proliferation, migration, and invasion of CRC cells. Our findings identify the altered post-translational modification at the translation machines as a critical factor in stress response and susceptibility to colorectal carcinogenesis.


Assuntos
Neoplasias Colorretais , Fator 1 de Elongação de Peptídeos , Humanos , Fator 1 de Elongação de Peptídeos/genética , Fator 1 de Elongação de Peptídeos/metabolismo , Acetilação , Processamento de Proteína Pós-Traducional , Carcinogênese
8.
FASEB J ; 36(8): e22409, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35792897

RESUMO

Interferon regulatory factor 7 (IRF7), as the interferon-stimulated gene, maximally drives type I interferon (IFN) production. However, the mechanisms by which the biological function of IRF7 is regulated remain elusive. In this study, we found that IRF7 selectively interacted with the neuralized E3 ubiquitin-protein ligase 3 (NEURL3). In concomitant with IRF7 induction, NEURL3 is upregulated by NF-κB signaling in the late phase of viral infection. Moreover, NEURL3 augmented the host antiviral immune response through ubiquitinating IRF7. A mechanistic study revealed that NEURL3 triggered K63-linked poly-ubiquitination on IRF7 lysine 375, which in turn epigenetically enhanced the transcription of interferon-stimulated genes (ISGs) through disruption of the association of IRF7 with Histone Deacetylase 1 (HDAC1), consequently augmenting host antiviral immune response. Accordingly, Neurl3-/- mice produced less type I IFNs and exhibited increased susceptibility to viral infection. Taken together, our findings identify NEURL3 as an E3 ubiquitin ligase of IRF7 and shed new light on the positive regulation of IRF7 in host antiviral immune signaling.


Assuntos
Interferon Tipo I , Ubiquitina-Proteína Ligases/metabolismo , Viroses , Animais , Antivirais/farmacologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Camundongos , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
9.
EMBO Rep ; 21(4): e48183, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32141187

RESUMO

Protein lysine acetylation affects colorectal cancer (CRC) distant metastasis through multiple pathways. In a previous proteomics screen, we found that isocitrate dehydrogenase 1 (IDH1) is hyperacetylated in CRC primary tumors and liver metastases. Here, we further investigate the function of IDH1 hyperacetylation at lysine 224 in CRC progression. We find that IDH1 K224 deacetylation promotes its enzymatic activity and the production of α-KG, and we identify sirtuin-2 (SIRT2) as a major deacetylase for IDH1. SIRT2 overexpression significantly inhibits CRC cell proliferation, migration, and invasion. IDH1 acetylation is modulated in response to intracellular metabolite concentration and regulates cellular redox hemostasis. Moreover, IDH1 acetylation reversely regulates HIF1α-dependent SRC transcription which in turn controls CRC progression. Physiologically, our data indicate that IDH1 deacetylation represses CRC cell invasion and migration in vitro and in vivo, while the hyperacetylation of IDH1 on K224 is significantly correlated to distant metastasis and poor survival of colorectal cancer patients. In summary, our study uncovers a novel mechanism through which SIRT2-dependent IDH1 deacetylation regulates cellular metabolism and inhibits liver metastasis of colorectal cancer.


Assuntos
Neoplasias Colorretais , Neoplasias Hepáticas , Acetilação , Neoplasias Colorretais/genética , Humanos , Isocitrato Desidrogenase/genética , Neoplasias Hepáticas/genética , Processamento de Proteína Pós-Traducional , Sirtuína 2/genética , Sirtuína 2/metabolismo
10.
Br J Cancer ; 125(3): 351-357, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33953345

RESUMO

BACKGROUND: Oesophageal cancer (EC) ranks high in both morbidity and mortality. A non-invasive and high-sensitivity diagnostic approach is necessary to improve the prognosis of EC patients. METHODS: A total of 525 serum samples were subjected to lipidomic analysis. We combined serum lipidomics and machine-learning algorithms to select important metabolite features for the detection of oesophageal squamous cell carcinoma (ESCC), the major subtype of EC in developing countries. A diagnostic model using a panel of selected features was developed and evaluated. Integrative analyses of tissue transcriptome and serum lipidome were conducted to reveal the underlying mechanism of lipid dysregulation. RESULTS: Our optimised diagnostic model with a panel of 12 lipid biomarkers together with age and gender reaches a sensitivity of 90.7%, 91.3% and 90.7% and an area under receiver-operating characteristic curve of 0.958, 0.966 and 0.818 in detecting ESCC for the training cohort, validation cohort and independent validation cohort, respectively. Integrative analysis revealed matched variation trend of genes encoding key enzymes in lipid metabolism. CONCLUSIONS: We have identified a panel of 12 lipid biomarkers for diagnostic modelling and potential mechanisms of lipid dysregulation in the serum of ESCC patients. This is a reliable, rapid and non-invasive tumour-diagnostic approach for clinical application.


Assuntos
Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Neoplasias Esofágicas/diagnóstico , Carcinoma de Células Escamosas do Esôfago/diagnóstico , Perfilação da Expressão Gênica/métodos , Lipidômica/métodos , Idoso , Área Sob a Curva , Estudos de Casos e Controles , Detecção Precoce de Câncer , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/sangue , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Aprendizado de Máquina , Masculino , Pessoa de Meia-Idade , Prognóstico , Curva ROC , Sensibilidade e Especificidade , Máquina de Vetores de Suporte
11.
Clin Proteomics ; 18(1): 16, 2021 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-34022816

RESUMO

BACKGROUND: Gastrointestinal stromal tumor (GIST) is a common digestive tract tumor with high rate of metastasis and recurrence. Currently, we understand the genome, transcriptome and proteome in GIST. However, posttranscriptional modification features in GIST remain unclear. In the present study, we aimed to construct a complete profile of acetylome in GIST. METHODS: Five common protein modifications, including acetylation, succinylation, crotonylation, 2-hydroxyisobutyrylation, and malonylation were tested among GIST subgroups and significantly differentially- expressed lysine acetylation was found. The acetylated peptides labeled with Tandem Mass Tag (TMT)under high sensitive mass spectrometry, and some proteins with acetylation sites were identified. Subsequently, these proteins and peptides were classified into high/moderate (H/M) risk and low (L) risk groups according to the modified NIH classification standard. Furthermore, cell components, molecular function, biological processes, KEGG pathways and protein interaction networks were analyzed. RESULTS: A total of 2904 acetylation sites from 1319 proteins were identified, of which quantitative information of 2548 sites from 1169 proteins was obtained. Finally, the differentially-expressed lysine acetylation sites were assessed and we found that 42 acetylated sites of 38 proteins were upregulated in the H/M risk group compared with the L risk group, while 48 acetylated sites of 44 proteins were downregulated, of which Ki67 K1063Ac and FCHSD2 K24Ac were the two acetylated proteins that were most changed. CONCLUSIONS: Our novel findings provide further understanding of acetylome in GIST and might demonstrate the possibility in the acetylation targeted diagnosis and therapy of GIST.

12.
FASEB J ; 34(9): 11488-11497, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32652764

RESUMO

Werner syndrome protein (WRN) plays critical roles in DNA replication, recombination, and repair, as well as transcription and cellular senescence. Ubiquitination and degradation of WRN have been reported, however, the E3 ubiquitin ligase of WRN is little known. Here, we identify mindbomb E3 ubiquitin protein ligase 1 (MIB1) as a novel E3 ubiquitin ligase for WRN protein. MIB1 physically interacts with WRN in vitro and in vivo and induces ubiquitination and degradation of WRN in the ubiquitin-proteasome pathway. Camptothecin (CPT) enhances the interaction between MIB1 and WRN, and promotes WRN degradation in a MIB1-dependent manner. In addition, CPT-induced cellular senescence is facilitated by the expression of MIB1 and attenuated by WRN expression. Our results show that MIB1-mediated degradation of WRN promotes cellular senescence and reveal a novel model executed by MIB1 and WRN to regulate cellular senescence.


Assuntos
Camptotecina/farmacologia , Senescência Celular/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Helicase da Síndrome de Werner/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Senescência Celular/genética , Técnicas de Inativação de Genes , Células HCT116 , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica , Proteólise/efeitos dos fármacos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos dos fármacos , Helicase da Síndrome de Werner/genética
13.
Amino Acids ; 53(12): 1841-1850, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34273023

RESUMO

Pyrroline-5-carboxylate reductase (PYCR), the last enzyme in proline synthesis that converts P5C into proline, was found promoting cancer growth and inhibiting apoptosis through multiple approaches, including regulating cell cycle and redox homeostasis, and promoting growth signaling pathways. Proline is abnormally up-regulated in multiple cancers and becomes one of the critical players in the reprogramming of cancer metabolism. As the last key enzymes in proline generation, PYCRs have been the subject of many investigations, and have been demonstrated to play an indispensable role in promoting tumorigenesis and cancer progression. In this article, we will thoroughly review the recent investigations on PYCRs in cancer development.


Assuntos
Carcinogênese/metabolismo , Neoplasias/metabolismo , Prolina/metabolismo , Pirrolina Carboxilato Redutases/metabolismo , Animais , Ciclo Celular/fisiologia , Humanos , Oxirredução
14.
Biol Chem ; 401(9): 1031-1039, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32284438

RESUMO

Citrate synthase (CS), the rate-limiting enzyme in the tricarboxylic acid (TCA) cycle catalyzes the first step of the cycle, namely, the condensation of oxaloacetate and acetyl-CoA to produce citrate. The expression and enzymatic activity of CS are altered in cancers, but posttranslational modification (PTM) of CS and its regulation in tumorigenesis remain largely obscure. SIRT5 belongs to the nicotinamide adenine dinucleotide (NAD)+-dependent deacetylase sirtuin family and plays vital roles in multiple biological processes via modulating various substrates. Here, we show that SIRT5 interacts with CS and that SIRT5 desuccinylates CS at the evolutionarily conserved residues K393 and K395. Moreover, hypersuccinylation of CS at K393 and K395 dramatically reduces its enzymatic activity and suppresses colon cancer cell proliferation and migration. These results provide experimental evidence in support of a potential therapeutic approach for colon cancer.


Assuntos
Citrato (si)-Sintase/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Sirtuínas/metabolismo , Movimento Celular/fisiologia , Proliferação de Células , Neoplasias do Colo/enzimologia , Humanos
15.
FASEB J ; 33(4): 5535-5547, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30649986

RESUMO

Tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN) plays a critical role in regulating cell survival, cell growth, and proliferation by antagonizing the PI3K-AKT-mTOR pathway. The regulatory mechanism of PTEN protein is still not completely understood. Here, we found that Sirtuin 4 (SIRT4) interacts with PTEN and regulates its stability. Overexpression of SIRT4 in cells causes down-regulation of PTEN. This regulation is independent of PTEN acetylation and ubiquitination. We further found that SIRT4 degrades PTEN through lysosome pathway mediated by insulin degrading enzyme (IDE). SIRT4 bridges PTEN and IDE for degradation in response to nutritional starvation stresses. Our results suggest that when cells were exposed to nutritional starvation, SIRT4 was induced and cooperated with IDE to degrade PTEN; low levels of PTEN promote cells to survive from cellular stress. Our findings provide a new regulation of PTEN in response to cellular stresses.-Liu, M., Wang, Z., Ren, M., Yang, X., Liu, B., Qi, H., Yu, M., Song, S., Chen, S., Liu, L., Zhang, Y., Zou, J., Zhu, W.-G., Yin, Y., Luo, J. SIRT4 regulates PTEN stability through IDE in response to cellular stresses.


Assuntos
Insulisina/metabolismo , Proteínas Mitocondriais/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Sirtuínas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Regulação para Baixo/fisiologia , Células HEK293 , Humanos , Células MCF-7 , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
16.
Nucleic Acids Res ; 46(18): 9601-9616, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30165671

RESUMO

Anabolism and catabolism are tightly regulated according to the cellular energy supply. Upon energy stress, ribosomal RNA (rRNA) biogenesis is inhibited, and autophagy is induced. However, the mechanism linking rRNA biogenesis and autophagy is unclear. Here, we demonstrate that the nucleolar protein NAT10 plays a role in the transition between rRNA biogenesis and autophagy. Under normal conditions, NAT10 is acetylated to activate rRNA biogenesis and inhibit autophagy induction. Mechanistic studies demonstrate that NAT10 binds to and acetylates the autophagy regulator Che-1 at K228 to suppress the Che-1-mediated transcriptional activation of downstream genes Redd1 and Deptor under adequate energy supply conditions. Upon energy stress, NAT10 is deacetylated by Sirt1, leading to suppression of NAT10-activated rRNA biogenesis. In addition, deacetylation of NAT10 abolishes the NAT10-mediated transcriptional repression of Che-1, leading to the release of autophagy inhibition. Collectively, we demonstrate that the acetylation status of NAT10 is important for the anabolism-catabolism transition in response to energy stress, providing a novel mechanism by which nucleolar proteins control rRNA synthesis and autophagy in response to the cellular energy supply.


Assuntos
Autofagia/genética , Acetiltransferase N-Terminal E/genética , RNA Ribossômico/genética , Estresse Fisiológico/genética , Acetilação , Proteínas Reguladoras de Apoptose/genética , Nucléolo Celular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metabolismo/genética , Acetiltransferase N-Terminal E/metabolismo , Acetiltransferases N-Terminal , RNA Ribossômico/biossíntese , RNA Ribossômico/metabolismo , Proteínas Repressoras/genética , Sirtuína 1/genética , Fatores de Transcrição/genética
17.
Int J Mol Sci ; 21(11)2020 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-32527012

RESUMO

Serine-threonine kinase receptor-associated protein (STRAP) functions as a regulator of both TGF-ß and p53 signaling that participates in the regulation of cell proliferation and cell death in response to various stresses. Here, we demonstrate that STRAP acetylation plays an important role in p53-mediated cell cycle arrest and apoptosis. STRAP is acetylated at lysines 147, 148, and 156 by the acetyltransferases CREB-binding protein (CBP) and that the acetylation is reversed by the deacetylase sirtuin7 (SIRT7). Hypo- or hyperacetylation mutations of STRAP at lysines 147, 148, and 156 (3KR or 3KQ) influence its activation and stabilization of p53. Moreover, following 5-fluorouracil (5-FU) treatment, STRAP is mobilized from the cytoplasm to the nucleus and promotes STRAP acetylation. Our finding on the regulation of STRAP links p53 with SIRT7 influencing p53 activity and stability.


Assuntos
Proteínas de Ligação a RNA/metabolismo , Sirtuínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Fluoruracila/farmacologia , Células HCT116 , Humanos , Lisina/metabolismo , Estabilidade Proteica , Proteínas de Ligação a RNA/genética , Sirtuínas/genética , Proteína Supressora de Tumor p53/genética
18.
J Biol Chem ; 293(46): 17769-17779, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30282801

RESUMO

The histone transmethylase complex comprising WD repeat domain 77 (WDR77) and protein arginine methyltransferase 5 (PRMT5) catalyzes dimethylation of H4R3 (H4R3me2) and drives cancer cell proliferation and migration, but its regulation is not fully understood. Here, we report that sirtuin 7 (SIRT7) directly deacetylates WDR77 and that this deacetylation interferes with the WDR77-PRMT5 interaction and suppresses proliferation of human colon cancer HCT116 cells. Using co-expression in HEK293T cells and co-immunoprecipitation assays, we observed that SIRT7 deacetylates WDR77 at Lys-3 and Lys-243, which reduced of WDR77's interaction with PRMT5. More importantly, this reduction suppressed the transmethylase activity of the WDR77/PRMT5 complex, resulting in a reduction of the H4R3me2 modification. Rescue of the WDR77-KO HCT116 cells with a WDR77-2KR (K3R and K243R) variant yielded cell migration and proliferation rates that were significantly lower than those of WDR77-KO HCT116 cells rescued with WT WDR77. In summary, SIRT7 is a major deacetylase for WDR77, and SIRT7-mediated deacetylation of WDR77 at Lys-3 and Lys-243 weakens the WDR77-PRMT5 interaction and activity and thereby suppresses growth of cancer cells.


Assuntos
Neoplasias do Colo/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Sirtuínas/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Proteína de Ligação a CREB/metabolismo , Proliferação de Células , Neoplasias do Colo/patologia , Células HCT116 , Células HEK293 , Humanos , Lisina/química , Multimerização Proteica
19.
EMBO Rep ; 17(3): 349-66, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26882543

RESUMO

As a genome guardian, p53 maintains genome stability by arresting cells for damage repair or inducing cell apoptosis to eliminate the damaged cells in stress response. Several nucleolar proteins stabilize p53 by interfering Mdm2-p53 interaction upon cellular stress, while other mechanisms by which nucleolar proteins activate p53 remain to be determined. Here, we identify NAT10 as a novel regulator for p53 activation. NAT10 acetylates p53 at K120 and stabilizes p53 by counteracting Mdm2 action. In addition, NAT10 promotes Mdm2 degradation with its intrinsic E3 ligase activity. After DNA damage, NAT10 translocates to nucleoplasm and activates p53-mediated cell cycle control and apoptosis. Finally, NAT10 inhibits cell proliferation and expression of NAT10 decreases in human colorectal carcinomas. Thus, our data demonstrate that NAT10 plays a critical role in p53 activation via acetylating p53 and counteracting Mdm2 action, providing a novel pathway by which nucleolar protein activates p53 as a cellular stress sensor.


Assuntos
Neoplasias Colorretais/metabolismo , Acetiltransferase N-Terminal E/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação , Acetilação , Transporte Ativo do Núcleo Celular , Apoptose , Núcleo Celular/metabolismo , Dano ao DNA , Células HCT116 , Células HEK293 , Humanos , Acetiltransferase N-Terminal E/genética , Acetiltransferases N-Terminal , Ligação Proteica , Estabilidade Proteica , Proteólise
20.
Mol Cell ; 39(2): 247-58, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20670893

RESUMO

SIRT1, a NAD(+)-dependent histone deacetylase, plays crucial roles in multiple biological processes including gene transcription, cellular metabolism, stress response, and tumorigenesis. Xeroderma pigmentosum group A (XPA) is a core nucleotide excision repair (NER) factor essential for NER process. Here we show that SIRT1 plays an important role in the regulation of NER pathway. Downregulation of SIRT1 significantly sensitizes cells to UV irradiation. SIRT1 interacts with XPA, and the interaction is enhanced after UV irradiation. XPA can be acetylated at lysines 63 and 67. SIRT1 deacetylates XPA both in vitro and in cells. Importantly, SIRT1-mediated deacetylation of XPA is required for optimal NER pathway since XPA-deficient cells complemented with XPA-K6367Q, which mimics hyperacetylated XPA, display significantly higher UV sensitivity compared with the XPA cells complemented with wild-type XPA. Furthermore, SIRT1-mediated XPA deacetylation enhances its interaction with RPA32. Our results demonstrate that SIRT1 regulates NER pathway through modulation of XPA acetylation status.


Assuntos
Reparo do DNA/efeitos da radiação , Processamento de Proteína Pós-Traducional/efeitos da radiação , Sirtuína 1/metabolismo , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo , Acetilação , Reparo do DNA/genética , Teste de Complementação Genética , Células HeLa , Humanos , Mutação de Sentido Incorreto , Processamento de Proteína Pós-Traducional/genética , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Sirtuína 1/genética , Proteína de Xeroderma Pigmentoso Grupo A/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA