Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
1.
PeerJ ; 12: e17234, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38666079

RESUMO

Background: Post-translational modification by Small Ubiquitin-like MOdifier (SUMO) is an important mechanism to regulate protein activity, protein stability, and localization of substrates. Zbtb21 is a zinc finger and BTB (Broad-complex, Tram-track and Bric à brac) domain-containing transcription factor. Bioinformatic prediction suggests several putative SUMOylated sites in Zbtb21 protein. Methods: Two evolutionarily conserved lysine residues in Zbtb21 protein were mutated alone or in combination to disrupt the binding with SUMO molecules. Western blot and co-immunoprecipitation analyses were performed to detect the SUMOylation state of wild type and mutant Zbtb21 proteins, respectively. Luciferase reporter assays were conducted to evaluate their transcription activities. Meanwhile, immunofluorescence staining was carried out to show their sub-nuclear localizations. Finally, co-immunoprecipitation was performed to detect the interaction between Zbtb21 and its partners. Results: Phylogenetically conserved lysines 419 and 845 of zebrafish Zbtb21 protein can be conjugated with SUMO molecules. SUMOylation does not affect the subcellular localization and protein stability of Zbtb21, as well as the interaction with Zbtb14 or Zbtb21. Nevertheless, luciferase reporter assays revealed that Zbtb21 is a dual-function transcription factor which exerts activation or repression effect on different promoters, and SUMOylation can modulate the transcriptional activity of Zbtb21 in regulating downstream target genes. Hence, Zbtb21 is identified as a novel substrate of SUMOylation, which would be important for its function. Conclusions: Zebrafish Zbtb21 protein can be SUMOylated on lysines 419 and 845, which is evolutionary conserved. SUMOylation affects the dual role of Zbtb21 on transcription.


Assuntos
Sumoilação , Proteínas de Peixe-Zebra , Peixe-Zebra , Sumoilação/genética , Animais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/genética , Humanos
2.
J Cell Physiol ; 239(3): e31080, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37450667

RESUMO

SUMOylation plays an essential role in diverse physiological and pathological processes. Identification of wild-type SUMO1-modification sites by mass spectrometry is still challenging. In this study, we produced a monoclonal SUMO1C-K antibody recognizing SUMOylated peptides and proposed an efficient streamline for identification of SUMOylation sites. We identified 471 SUMOylation sites in 325 proteins from five raw data. These identified sites exhibit a high positive rate when evaluated by mutation-verified SUMOylation sites. We identified many SUMOylated proteins involved in mitochondrial metabolism and non-membrane-bounded organelles formation. We proposed a SUMOylation motif, ΨKXD/EP, where proline is required for efficient SUMOylation. We further revealed SUMOylation of TFII-I was stimulated by growth signals and was required for nucleus-localization of p-ERK1/2. Mutation of SUMOylation sites of TFII-I suppressed tumor cell growth in vitro and in vivo. Taken together, we provided a strategy for personalized identification of wild-type SUMO1-modification sites and revealed the physiological significance of TFII-I SUMOylation in this study.


Assuntos
Neoplasias , Proteína SUMO-1 , Sumoilação , Fatores de Transcrição TFII , Humanos , Anticorpos Monoclonais , Espectrometria de Massas , Neoplasias/genética , Neoplasias/patologia , Peptídeos/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Sumoilação/genética , Fatores de Transcrição TFII/metabolismo
3.
Signal Transduct Target Ther ; 8(1): 426, 2023 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-37925421

RESUMO

Lymph node (LN) metastasis is one of the predominant metastatic routes of non-small cell lung cancer (NSCLC) and is considered as a leading cause for the unsatisfactory prognosis of patients. Although lymphangiogenesis is well-recognized as a crucial process in mediating LN metastasis, the regulatory mechanism involving lymphangiogenesis and LN metastasis in NSCLC remains unclear. In this study, we employed high-throughput sequencing to identify a novel circular RNA (circRNA), circTLCD4-RWDD3, which was significantly upregulated in extracellular vesicles (EVs) from LN metastatic NSCLC and was positively associated with deteriorated OS and DFS of patients with NSCLC from multicenter clinical cohort. Downregulating the expression of EV-packaged circTLCD4-RWDD3 inhibited lymphangiogenesis and LN metastasis of NSCLC both in vitro and in vivo. Mechanically, circTLCD4-RWDD3 physically interacted with hnRNPA2B1 and mediated the SUMO2 modification at K108 residue of hnRNPA2B1 by upregulating UBC9. Subsequently, circTLCD4-RWDD3-induced SUMOylated hnRNPA2B1 was recognized by the SUMO interaction motif (SIM) of ALIX and activated ALIX to recruit ESCRT-III, thereby facilitating the sorting of circTLCD4-RWDD3 into NSCLC cell-derived EVs. Moreover, EV-packaged circTLCD4-RWDD3 was internalized by lymphatic endothelial cells to activate the transcription of PROX1, resulting in the lymphangiogenesis and LN metastasis of NSCLC. Importantly, blocking EV-mediated transmission of circTLCD4-RWDD3 via mutating SIM in ALIX or K108 residue of hnRNPA2B1 inhibited the lymphangiogenesis and LN metastasis of NSCLC in vivo. Our findings reveal a precise mechanism underlying SUMOylated hnRNPA2B1-induced EV packaging of circTLCD4-RWDD3 in facilitating LN metastasis of NSCLC, suggesting that EV-packaged circTLCD4-RWDD3 could be a potential therapeutic target against LN metastatic NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Vesículas Extracelulares , Neoplasias Pulmonares , RNA Circular , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Células Endoteliais/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Linfática , Sumoilação/genética , Fatores de Transcrição , RNA Circular/genética
4.
Hum Mol Genet ; 33(1): 1-11, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37694858

RESUMO

MeCP2 (Methyl CpG binding protein 2) is an intrinsically disordered protein that binds to methylated genome regions. The protein is a critical transcriptional regulator of the brain, and its mutations account for 95% of Rett syndrome (RTT) cases. Early studies of this neurodevelopmental disorder revealed a close connection with dysregulations of the ubiquitin system (UbS), notably as related to UBE3A, a ubiquitin ligase involved in the proteasome-mediated degradation of proteins. MeCP2 undergoes numerous post-translational modifications (PTMs), including ubiquitination and sumoylation, which, in addition to the potential functional outcomes of their monomeric forms in gene regulation and synaptic plasticity, in their polymeric organization, these modifications play a critical role in proteasomal degradation. UbS-mediated proteasomal degradation is crucial in maintaining MeCP2 homeostasis for proper function and is involved in decreasing MeCP2 in some RTT-causing mutations. However, regardless of all these connections to UbS, the molecular details involved in the signaling of MeCP2 for its targeting by the ubiquitin-proteasome system (UPS) and the functional roles of monomeric MeCP2 ubiquitination and sumoylation remain largely unexplored and are the focus of this review.


Assuntos
Proteína 2 de Ligação a Metil-CpG , Síndrome de Rett , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Sumoilação/genética , Complexo de Endopeptidases do Proteassoma/genética , Síndrome de Rett/metabolismo , Ubiquitinação/genética , Ubiquitina/metabolismo
5.
J Struct Biol ; 215(2): 107961, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37059313

RESUMO

AKT/PKB is a kinase involved in the regulation of a plethora of cell processes. Particularly, in embryonic stem cells (ESCs), AKT is crucial for the maintenance of pluripotency. Although the activation of this kinase relies on its recruitment to the cellular membrane and subsequent phosphorylation, multiple other post-translational modifications (PTMs), including SUMOylation, fine-tune its activity and target specificity. Since this PTM can also modify the localization and availability of different proteins, in this work we explored if SUMOylation impacts on the subcellular compartmentalization and distribution of AKT1 in ESCs. We found that this PTM does not affect AKT1 membrane recruitment, but it modifies the AKT1 nucleus/cytoplasm distribution, increasing its nuclear presence. Additionally, within this compartment, we found that AKT1 SUMOylation also impacts on the chromatin-binding dynamics of NANOG, a central pluripotency transcription factor. Remarkably, the oncogenic E17K AKT1 mutant produces major changes in all these parameters increasing the binding of NANOG to its targets, also in a SUMOylation dependent manner. These findings demonstrate that SUMOylation modulates AKT1 subcellular distribution, thus adding an extra layer of regulation of its function, possibly by affecting the specificity and interaction with its downstream targets.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Sumoilação , Mutação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sumoilação/genética , Cromatina/genética , Células-Tronco Embrionárias/metabolismo
6.
Comb Chem High Throughput Screen ; 26(3): 595-609, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35598238

RESUMO

BACKGROUND: Endometrial cancer is ranked fourth in women's cancers worldwide. SUMOylation is a process of post-translational modification and some evidence indicate that SUMOylation may influence the occurrence and development of cancer. Until now, the prognostic value of SUMOylation-related genes in endometrial cancer remains unclear. Therefore, we aimed at exploring the prognostic value of SUMOylation-related genes in endometrial cancer in this study. METHODS: The transcriptome of endometrial cancer from TCGA database was downloaded and then differentially expressed SUMOylation-related genes were extracted. The risk model was constructed with the use of the least absolute shrinkage and selection operator Cox regression. Samples were divided into low-risk and high-risk group based on the risk score. Survival analysis and Cox analysis were performed between groups. A validation cohort from Fudan University Shanghai Cancer Center were obtained to verify the model. Gene ontology and Kyoto Encylopedia of Genes and Genomes analyses were conducted based on differentially expressed genes between groups. RESULTS: Samples in low-risk group possess better outcome than in high-risk group. (P<0.001) The results of univariate (P<0.001) and multivariate (P=0.018) analysis showed that the risk score was independently correlated to worse outcome for patients with endometrial cancer. In Fudan University Shanghai Cancer Center validation cohort, the low-risk group possessed better survival outcome than the high-risk group (P=0.0393). Functional analysis demonstrated that most of the immune cell infiltration levels and immune pathways activity in low-risk group were higher than in high-risk group. CONCLUSIONS: In short, the SUMOylation-related signature had good predictability in endometrial cancer and SUMOylation-related genes play important roles in tumour immunity. Also, our study might have some merits in elucidating potential mechanism of SUMOylation in endometrial cancer.


Assuntos
Neoplasias do Endométrio , Sumoilação , Humanos , Feminino , Sumoilação/genética , China , Neoplasias do Endométrio/genética , Fatores de Risco , Bases de Dados Factuais
7.
Cancer Commun (Lond) ; 42(11): 1141-1161, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36209503

RESUMO

BACKGROUND: Gastric cancer (GC) is among the most malignant tumors, yet the pathogenesis is not fully understood, especially the lack of detailed information about the mechanisms underlying long non-coding RNA (lncRNA)-mediated post-translational modifications. Here, the molecular mechanisms and clinical significance of the novel lncRNA syndecan-binding protein 2-antisense RNA 1 (SDCBP2-AS1) in the tumorigenesis and progression of GC were investigated. METHODS: The expression levels of SDCBP2-AS1 in 132 pairs of GC and adjacent normal tissues were compared, and the biological functions were assessed in vitro and in vivo. RNA pull-down and immunoprecipitation assays were conducted to clarify the interactions of SDCBP2-AS1 and heterogeneous nuclear ribonucleoprotein (hnRNP) K. RNA-sequencing, immunoprecipitation, immunofluorescence, and luciferase analyses were performed to investigate the functions of SDCBP2-AS1. RESULTS: SDCBP2-AS1 was significantly downregulated in GC tissues and predictive of poor patient prognosis. Silencing of SDCBP2-AS1 promoted the proliferation and migration of GC cells both in vitro and in vivo. Mechanically, SDCBP2-AS1 physically bound to hnRNP K to repress SUMOylation of hnRNP K and facilitated ubiquitination of hnRNP K and ß-catenin, thereby promoting the degradation of ß-catenin in the cytoplasm. Silencing of SDCBP2-AS1 caused SUMOylation of hnRNP K and stabilized ß-catenin activity, which altered transcription of downstream genes, resulting in tumorigenesis and metastasis of GC. Moreover, the knockdown of hnRNP K partially abrogated the effects of SDCBP2-AS1. CONCLUSIONS: SDCBP2-AS1 interacts with hnRNP K to suppress tumorigenesis and metastasis of GC and regulates post-transcriptional modifications of hnRNP K to destabilize ß-catenin. These findings suggest SDCBP2-AS1 as a potential target for the treatment of GC.


Assuntos
RNA Longo não Codificante , Neoplasias Gástricas , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Sumoilação/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Neoplasias Gástricas/patologia , Carcinogênese/genética , Carcinogênese/metabolismo , Sinteninas/genética , Sinteninas/metabolismo
8.
Nat Commun ; 13(1): 5133, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36050397

RESUMO

DNA end resection is delicately regulated through various types of post-translational modifications to initiate homologous recombination, but the involvement of SUMOylation in this process remains incompletely understood. Here, we show that MRE11 requires SUMOylation to shield it from ubiquitin-mediated degradation when resecting damaged chromatin. Upon DSB induction, PIAS1 promotes MRE11 SUMOylation on chromatin to initiate DNA end resection. Then, MRE11 is deSUMOylated by SENP3 mainly after it has moved away from DSB sites. SENP3 deficiency results in MRE11 degradation failure and accumulation on chromatin, causing genome instability. We further show that cancer-related MRE11 mutants with impaired SUMOylation exhibit compromised DNA repair ability. Thus, we demonstrate that MRE11 SUMOylation in coordination with ubiquitylation is dynamically controlled by PIAS1 and SENP3 to facilitate DNA end resection and maintain genome stability.


Assuntos
Cromatina , Proteína Homóloga a MRE11 , Sumoilação , Ubiquitinação , Cisteína Endopeptidases/metabolismo , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Instabilidade Genômica , Homeostase , Humanos , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Sumoilação/genética , Sumoilação/fisiologia , Ubiquitinação/genética , Ubiquitinação/fisiologia
9.
PLoS Genet ; 18(6): e1010275, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35696436

RESUMO

Timely completion of eukaryotic genome duplication requires coordinated DNA replication initiation at multiple origins. Replication begins with the loading of the Mini-Chromosome Maintenance (MCM) complex, proceeds by the activation of the Cdc45-MCM-GINS (CMG) helicase, and ends with CMG removal after chromosomes are fully replicated. Post-translational modifications on the MCM and associated factors ensure an orderly transit of these steps. Although the mechanisms of CMG activation and removal are partially understood, regulated MCM loading is not, leaving an incomplete understanding of how DNA replication begins. Here we describe a site-specific modification of Mcm3 by the Small Ubiquitin-like MOdifier (SUMO). Mutations that prevent this modification reduce the MCM loaded at replication origins and lower CMG levels, resulting in impaired cell growth, delayed chromosomal replication, and the accumulation of gross chromosomal rearrangements (GCRs). These findings demonstrate the existence of a SUMO-dependent regulation of origin-bound MCM and show that this pathway is needed to prevent genome rearrangements.


Assuntos
Replicação do DNA , Sumoilação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA Helicases/genética , Replicação do DNA/genética , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Origem de Replicação/genética , Sumoilação/genética
10.
PLoS Genet ; 18(6): e1009978, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35666766

RESUMO

The sumoylation (SUMO) pathway is involved in a variety of processes during C. elegans development, such as gonadal and vulval fate specification, cell cycle progression and maintenance of chromosome structure. The ubiquitous expression and pleiotropic effects have made it difficult to dissect the tissue-specific functions of the SUMO pathway and identify its target proteins. To overcome these challenges, we have established tools to block protein sumoylation and degrade sumoylated target proteins in a tissue-specific and temporally controlled manner. We employed the auxin-inducible protein degradation system (AID) to down-regulate the SUMO E3 ligase GEI-17 or the SUMO ortholog SMO-1, either in the vulval precursor cells (VPCs) or in the gonadal anchor cell (AC). Our results indicate that the SUMO pathway acts in multiple tissues to control different aspects of vulval development, such as AC positioning, basement membrane (BM) breaching, VPC fate specification and morphogenesis. Inhibition of protein sumoylation in the VPCs resulted in abnormal toroid formation and ectopic cell fusions during vulval morphogenesis. In particular, sumoylation of the ETS transcription factor LIN-1 at K169 is necessary for the proper contraction of the ventral vulA toroids. Thus, the SUMO pathway plays several distinct roles throughout vulval development.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Feminino , Organogênese , Sumoilação/genética , Fatores de Transcrição/genética , Vulva
11.
Proc Natl Acad Sci U S A ; 119(26): e2202631119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35733256

RESUMO

Angiogenesis contributes fundamentally to embryonic development, tissue homeostasis, and wound healing. Basic fibroblast growth factor (FGF2) is recognized as the first proangiogenic molecule discovered, and it facilitates angiogenesis by activating FGF receptor 1 (FGFR1) signaling in endothelial cells. However, the precise roles of FGFR and the FGF/FGFR signaling axis in angiogenesis remain unclear, especially because of the contradictory phenotypes of in vivo FGF and FGFR gene deficiency models. Our previous study results suggested a potential role of posttranslational small ubiquitin-like modifier modification (SUMOylation), with highly dynamic regulatory features, in vascular development and disorder. Here, we identified SENP1-regulated endothelial FGFR1 SUMOylation at conserved lysines responding to proangiogenic stimuli, while SENP1 functioned as the deSUMOylase. Hypoxia-enhanced FGFR1 SUMOylation restricted the tyrosine kinase activation of FGFR1 by modulating the dimerization of FGFR1 and FGFR1 binding with its phosphatase PTPRG. Consequently, it facilitated the recruitment of FRS2α to VEGFR2 but limited additional recruitment of FRS2α to FGFR1, supporting the activation of VEGFA/VEGFR2 signaling in endothelial cells. Furthermore, SUMOylation-defective mutation of FGFR1 resulted in exaggerated FGF2/FGFR1 signaling but suppressed VEGFA/VEGFR2 signaling and the angiogenic capabilities of endothelial cells, which were rescued by FRS2α overexpression. Reduced angiogenesis and endothelial sprouting in mice bearing an endothelial-specific, FGFR1 SUMOylation-defective mutant confirmed the functional significance of endothelial FGFR1 SUMOylation in vivo. Our findings identify the reversible SUMOylation of FGFR1 as an intrinsic fine-tuned mechanism in coordinating endothelial angiogenic signaling during neovascularization; SENP1-regulated FGFR1 SUMOylation and deSUMOylation controls the competitive recruitment of FRS2α by FGFR1 and VEGFR2 to switch receptor-complex formation responding to hypoxia and normoxia angiogenic environments.


Assuntos
Células Endoteliais , Neovascularização Fisiológica , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Sumoilação , Animais , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hipóxia/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Sumoilação/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
BMC Cancer ; 22(1): 572, 2022 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-35606717

RESUMO

BACKGROUND: Post-translational modification plays an important role in the occurrence and development of various tumors. However, few researches were focusing on the SUMOylation regulatory genes as tumor biomarkers to predict the survival for specific patients. Here, we constructed and validated a two-gene signature to predict the overall survival (OS) of non-small cell lung cancer (NSCLC) patients. METHODS: The datasets analyzed in this study were downloaded from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) Cox regression was used to construct the two-gene signature. Gene set enrichment analysis (GSEA) and Gene Ontology (GO) was used to identify hub pathways associated with risk genes. The CCK-8 assay, cell cycle analysis, and transwell assay was used to validate the function of risk genes in NSCLC cell lines. RESULTS: Firstly, most of the SUMOylation regulatory genes were highly expressed in various tumors through the R package 'limma' in the TCGA database. Secondly, our study found that the two gene signature constructed by LASSO regression analysis, as an independent prognostic factor, could predict the OS in both the TCGA training cohort and GEO validation cohorts (GSE68465, GSE37745, and GSE30219). Furthermore, functional enrichment analysis suggests that high-risk patients defined by the risk score system were associated with the malignant phenomenon, such as DNA replication, cell cycle regulation, p53 signaling pathway. Finally, the results of the CCK-8 assay, cell cycle analysis, and transwell assay demonstrated that the two risk genes, SAE1 and UBA2, could promote proliferation and migration in non-small cell lung cancer cells. CONCLUSIONS: The two-gene signature constructed in our study could predict the OS and may provide valuable clinical guidance for the treatment of NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Genes Reguladores , Humanos , Neoplasias Pulmonares/genética , Prognóstico , Sumoilação/genética , Enzimas Ativadoras de Ubiquitina/genética
13.
Nat Struct Mol Biol ; 29(2): 130-142, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35173350

RESUMO

Nuclear Argonaute proteins, guided by small RNAs, mediate sequence-specific heterochromatin formation. The molecular principles that link Argonaute-small RNA complexes to cellular heterochromatin effectors on binding to nascent target RNAs are poorly understood. Here, we explain the mechanism by which the PIWI-interacting RNA (piRNA) pathway connects to the heterochromatin machinery in Drosophila. We find that Panoramix, a corepressor required for piRNA-guided heterochromatin formation, is SUMOylated on chromatin in a Piwi-dependent manner. SUMOylation, together with an amphipathic LxxLL motif in Panoramix's intrinsically disordered repressor domain, are necessary and sufficient to recruit Small ovary (Sov), a multi-zinc-finger protein essential for general heterochromatin formation and viability. Structure-guided mutations that eliminate the Panoramix-Sov interaction or that prevent SUMOylation of Panoramix uncouple Sov from the piRNA pathway, resulting in viable but sterile flies in which Piwi-targeted transposons are derepressed. Thus, Piwi engages the heterochromatin machinery specifically at transposon loci by coupling recruitment of a corepressor to nascent transcripts with its SUMOylation.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Motivos de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sítios de Ligação/genética , Cromatina/genética , Cromatina/metabolismo , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/química , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Inativação Gênica , Genes de Insetos , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/metabolismo , Modelos Moleculares , Mutação , Proteínas Nucleares/química , Células-Tronco de Oogônios/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas de Ligação a RNA/química , Sumoilação/genética , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
14.
J Gastroenterol Hepatol ; 37(4): 700-713, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35030640

RESUMO

BACKGROUND AND AIM: Colorectal cancer (CRC) is one of the major health issues in the world. Circ_0000677 has been shown to be upregulated in CRC with unclarified function and mechanism. Methyltransferase-like 3 (METTL3) acts as a regulator for gene expression via the mechanism of RNA N6 -methyladenosine (m6 A) in different types of cancer, which is under the control of SUMO1-based SUMOylation. We aim to investigate their roles in CRC progression. METHODS: Quantitative real-time polymerase chain reaction and Western blot were used to detect the expressions of METTL3, circ_0000677, and ATP binding cassette subfamily c member 1(ABCC1) in CRC patients' tissues and cell lines. The functions of ABCC1 and circ_0000677 in CRC were studied by manipulating their level via knocking down or overexpression. RNA pull-down and RNA immunoprecipitation assays were performed to identify the specific binding of target genes. The biological function of SUMOylation of METTL3 was investigated in vivo by xenograft mice tumor model. RESULTS: METTL3, circ_0000677, and ABCC1 were upregulated in CRC patients' samples and cell lines. Circ_0000677 positively regulates CRC cell proliferation and drug resistance via affecting ABCC1 expression. METTL3 facilitated circ_0000677 level via m6 A modification. METTL3 was regulated by SUMO1-mediated SUMOylation in CRC. Mutation of METTL3-K459 could suppress tumor growth in vivo via regulating circ_0000677/ABCC1 axis. CONCLUSIONS: Overall, our study revealed that circ_0000677 and its downstream target ABCC1 were upregulated in CRC cells, induced by the METTL3-mediated m6 A modification of circ_0000677 and SUMO1-mediated SUMOylation of METTL3. This work provided a new strategy for the therapeutic treatment of CRC.


Assuntos
Neoplasias Colorretais , Metiltransferases , Animais , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Humanos , Metiltransferases/genética , Camundongos , Sumoilação/genética
15.
Life Sci Alliance ; 5(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35022246

RESUMO

CRISPR/Cas9 is a popular genome editing technology. Although widely used, little is known about how this prokaryotic system behaves in humans. An unwanted consequence of eukaryotic Cas9 expression is off-target DNA binding leading to mutagenesis. Safer clinical implementation of CRISPR/Cas9 necessitates a finer understanding of the regulatory mechanisms governing Cas9 behavior in humans. Here, we report our discovery of Cas9 sumoylation and ubiquitylation, the first post-translational modifications to be described on this enzyme. We found that the major SUMO2/3 conjugation site on Cas9 is K848, a key positively charged residue in the HNH nuclease domain that is known to interact with target DNA and contribute to off-target DNA binding. Our results suggest that Cas9 ubiquitylation leads to decreased stability via proteasomal degradation. Preventing Cas9 sumoylation through conversion of K848 into arginine or pharmacologic inhibition of cellular sumoylation enhances the enzyme's turnover and diminishes guide RNA-directed DNA binding efficacy, suggesting that sumoylation at this site regulates Cas9 stability and DNA binding. More research is needed to fully understand the implications of these modifications for Cas9 specificity.


Assuntos
Proteína 9 Associada à CRISPR , DNA/metabolismo , Lisina , Sumoilação/genética , Proteína 9 Associada à CRISPR/química , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Células HEK293 , Humanos , Lisina/química , Lisina/genética , Estabilidade Proteica , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo
16.
Nat Commun ; 13(1): 281, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-35022408

RESUMO

SUMOylation is a post-translational modification of proteins that regulates these proteins' localization, turnover or function. Aberrant SUMOylation is frequently found in cancers but its origin remains elusive. Using a genome-wide transposon mutagenesis screen in a MYC-driven B-cell lymphoma model, we here identify the SUMO isopeptidase (or deconjugase) SENP6 as a tumor suppressor that links unrestricted SUMOylation to tumor development and progression. Notably, SENP6 is recurrently deleted in human lymphomas and SENP6 deficiency results in unrestricted SUMOylation. Mechanistically, SENP6 loss triggers release of DNA repair- and genome maintenance-associated protein complexes from chromatin thereby impairing DNA repair in response to DNA damages and ultimately promoting genomic instability. In line with this hypothesis, SENP6 deficiency drives synthetic lethality to Poly-ADP-Ribose-Polymerase (PARP) inhibition. Together, our results link SENP6 loss to defective genome maintenance and reveal the potential therapeutic application of PARP inhibitors in B-cell lymphoma.


Assuntos
Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Mutação , Sumoilação/fisiologia , Animais , Biomarcadores Tumorais , Carbono-Nitrogênio Liases/genética , Carbono-Nitrogênio Liases/metabolismo , Cromatina , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Instabilidade Genômica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Processamento de Proteína Pós-Traducional , Sumoilação/efeitos dos fármacos , Sumoilação/genética , Mutações Sintéticas Letais , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Plant Sci ; 314: 111099, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34895537

RESUMO

Abiotic and biotic stresses are the major factors limiting plant growth. Arabidopsis E3 SUMO ligase SIZ1 plays an essential role in plant stress tolerance. Herein, we identified a SIZ/PAIS-type protein in pepper (Capsicum annuum), namely CaSIZ1, which shares 60 % sequence identity with AtSIZ1. The stems and flowers of pepper had a relatively higher expression of CaSIZ1 than the fruits, leaves, and roots. ABA and NaCl treatments induced CaSIZ1. CaSIZ1 protein was localized in the nucleus and partially rescued the dwarf and ABA-sensitive phenotypes of Atsiz1-2, suggesting the functional replacement of CaSIZ1 with AtSIZ1. We found that CaSIZ1 interacted with CaABI5, and ABA promoted the accumulation of SUMO conjugates in pepper. CaSIZ1 knockdown did not only reduce ABA-induced SUMOylation, but also attenuated the salt tolerance of pepper. Overall, the results of this study suggest that CaSIZ1 has a significant role in ABA-induced SUMOylation and stress response.


Assuntos
Ácido Abscísico/metabolismo , Capsicum/genética , Capsicum/metabolismo , Estresse Salino/efeitos dos fármacos , Tolerância ao Sal/genética , Sumoilação/efeitos dos fármacos , Arabidopsis/genética , Arabidopsis/metabolismo , Produtos Agrícolas/genética , Produtos Agrícolas/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Genes de Plantas , Folhas de Planta/metabolismo , Raízes de Plantas/metabolismo , Caules de Planta/metabolismo , Plantas Geneticamente Modificadas/genética , Sumoilação/genética , Nicotiana/genética , Nicotiana/metabolismo , Verduras/genética , Verduras/metabolismo
18.
Cell Rep ; 37(8): 110034, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818558

RESUMO

Endogenous metabolites, environmental agents, and therapeutic drugs promote formation of covalent DNA-protein crosslinks (DPCs). Persistent DPCs compromise genome integrity and are eliminated by multiple repair pathways. Aberrant Top1-DNA crosslinks, or Top1ccs, are processed by Tdp1 and Wss1 functioning in parallel pathways in Saccharomyces cerevisiae. It remains obscure how cells choose between diverse mechanisms of DPC repair. Here, we show that several SUMO biogenesis factors (Ulp1, Siz2, Slx5, and Slx8) control repair of Top1cc or an analogous DPC lesion. Genetic analysis reveals that SUMO promotes Top1cc processing in the absence of Tdp1 but has an inhibitory role if cells additionally lack Wss1. In the tdp1Δ wss1Δ mutant, the E3 SUMO ligase Siz2 stimulates sumoylation in the vicinity of the DPC, but not SUMO conjugation to Top1. This Siz2-dependent sumoylation inhibits alternative DPC repair mechanisms, including Ddi1. Our findings suggest that SUMO tunes available repair pathways to facilitate faithful DPC repair.


Assuntos
Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Cisteína Endopeptidases/metabolismo , DNA/metabolismo , Reparo do DNA/genética , DNA Topoisomerases Tipo I/metabolismo , Proteínas de Ligação a DNA/genética , Diester Fosfórico Hidrolases/metabolismo , Proteína SUMO-1/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/genética , Sumoilação/fisiologia , Ubiquitina-Proteína Ligases/metabolismo
19.
Cell Death Dis ; 12(11): 986, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34686655

RESUMO

MAPK/p38 is an important mammalian signaling cascade that responds to a variety of intracellular or extracellular stimuli, such as reactive oxygen species (ROS), and participates in numerous physiological and pathological processes. However, the biological function of p38 in different tumors, and even at different stages of the same tumor, remains elusive. To further understand the regulatory mechanism of p38 and oxidative stress in the occurrence and development of gastric cancer, we report SUMOylation as a novel post-translational modification occurring on lysine 152 of MAPK14/p38α through immunoprecipitation and series of pull-down assays in vitro and in vivo. Importantly, we determine that p38α-SUMOylation functions as an authentic sensor and accelerator of reactive oxygen species generation via interaction with and activation of MK2 in the nucleus, and the ROS accumulation, in turn, promotes the SUMOylation of p38α by stabilizing the PIASxα protein. This precise regulatory mechanism is exploited by gastric cancer cells to create an internal environment for survival and, ultimately, metastasis. This study reveals novel insights into p38α-SUMOylation and its association with the intracellular oxidative stress response, which is closely related to the processes of gastric cancer. Furthermore, the PIASxα/p38α-SUMOylation/MK2 cis-axis may serve as a desirable therapeutic target in gastric cancer as targeting PIASxα, MK2, or a specific peptide region of p38α may reconcile the aberrant oxidative stress response in gastric cancer cells.


Assuntos
Neoplasias Gástricas/genética , Sumoilação/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Feminino , Humanos , Camundongos , Metástase Neoplásica , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/patologia
20.
PLoS Genet ; 17(10): e1009830, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34695110

RESUMO

The post-translational addition of SUMO plays essential roles in numerous eukaryotic processes including cell division, transcription, chromatin organization, DNA repair, and stress defense through its selective conjugation to numerous targets. One prominent plant SUMO ligase is METHYL METHANESULFONATE-SENSITIVE (MMS)-21/HIGH-PLOIDY (HPY)-2/NON-SMC-ELEMENT (NSE)-2, which has been connected genetically to development and endoreduplication. Here, we describe the potential functions of MMS21 through a collection of UniformMu and CRISPR/Cas9 mutants in maize (Zea mays) that display either seed lethality or substantially compromised pollen germination and seed/vegetative development. RNA-seq analyses of leaves, embryos, and endosperm from mms21 plants revealed a substantial dysregulation of the maize transcriptome, including the ectopic expression of seed storage protein mRNAs in leaves and altered accumulation of mRNAs associated with DNA repair and chromatin dynamics. Interaction studies demonstrated that MMS21 associates in the nucleus with the NSE4 and STRUCTURAL MAINTENANCE OF CHROMOSOMES (SMC)-5 components of the chromatin organizer SMC5/6 complex, with in vitro assays confirming that MMS21 will SUMOylate SMC5. Comet assays measuring genome integrity, sensitivity to DNA-damaging agents, and protein versus mRNA abundance comparisons implicated MMS21 in chromatin stability and transcriptional controls on proteome balance. Taken together, we propose that MMS21-directed SUMOylation of the SMC5/6 complex and other targets enables proper gene expression by influencing chromatin structure.


Assuntos
Proteínas de Arabidopsis/genética , Genoma de Planta/genética , Instabilidade Genômica/genética , Ligases/genética , Proteína SUMO-1/genética , Sumoilação/genética , Zea mays/genética , Cromatina/genética , Cromossomos de Plantas/genética , Proteoma/genética , Transcrição Gênica/genética , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...