Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Res ; 33(5): 355-371, 2023 05.
Article in English | MEDLINE | ID: mdl-36882514

ABSTRACT

Posttranslational modifications add tremendous complexity to proteomes; however, gaps remain in knowledge regarding the function and regulatory mechanism of newly discovered lysine acylation modifications. Here, we compared a panel of non-histone lysine acylation patterns in metastasis models and clinical samples, and focused on 2-hydroxyisobutyrylation (Khib) due to its significant upregulation in cancer metastases. By the integration of systemic Khib proteome profiling in 20 paired primary esophageal tumor and metastatic tumor tissues with CRISPR/Cas9 functional screening, we identified N-acetyltransferase 10 (NAT10) as a substrate for Khib modification. We further showed that Khib modification at lysine 823 in NAT10 functionally contribute to metastasis. Mechanistically, NAT10 Khib modification enhances its interaction with deubiquitinase USP39, resulting in increased NAT10 protein stability. NAT10 in turn promotes metastasis by increasing NOTCH3 mRNA stability in an N4-acetylcytidine-dependent manner. Furthermore, we discovered a lead compound #7586-3507 that inhibited NAT10 Khib modification and showed efficacy in tumor models in vivo at a low concentration. Together, our findings bridge newly identified lysine acylation modifications with RNA modifications, thus providing novel insights into epigenetic regulation in human cancer. We propose that pharmacological inhibition of NAT10 K823 Khib modification constitutes a potential anti-metastasis strategy.


Subject(s)
Lysine , Neoplasms , Humans , Lysine/metabolism , Epigenesis, Genetic , Acylation , Protein Processing, Post-Translational , Acetyltransferases/metabolism , Neoplasms/genetics , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , Ubiquitin-Specific Proteases/genetics
2.
Cancer Res ; 83(11): 1851-1865, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36912612

ABSTRACT

SIGNIFICANCE: Metastatic cancer cells upregulate ANO1 to activate cell-intrinsic and -extrinsic mechanisms that alter cholesterol metabolism and stimulate fibroblasts, which can be targeted with ANO1 inhibitors to inhibit metastatic growth. See related commentary by Singh and Mehla, p. 1759.


Subject(s)
Neoplasm Proteins , Neoplasms , Humans , Neoplasm Proteins/metabolism , Tumor Microenvironment , Cell Line, Tumor , Fibroblasts/metabolism , Cholesterol/metabolism , Anoctamin-1/metabolism , Neoplasms/metabolism
3.
Cell Death Discov ; 8(1): 425, 2022 Oct 22.
Article in English | MEDLINE | ID: mdl-36273228

ABSTRACT

DEAD box RNA helicase 17 (DDX17) has been shown to be an RNA binding protein involved in RNA metabolism and associated with cancer progression. However, the biological role of DDX17 in the pathogenesis of lung adenocarcinoma (LUAD) has not been well characterized. Here, we demonstrated that DDX17 promoted the proliferation, migration and invasion of H1299 and A549 lung adenocarcinoma cells. Analyses of public datasets showed that DDX17 is upregulated in LUAD specimens. Our tumor xenograft models confirmed the in vivo promoting role of DDX17 in the growth and metastasis of LUAD. Mechanistic analyses further revealed that DDX17 protein interacts with the mRNA of MYL9 and MAGEA6 and upregulates their levels. MYL9 could mediate the function of DDX17 to regulate the actin cytoskeleton rearrangement and cell adhesion, particularly by modulating the stress fiber and focal adhesion formation, whereas DDX17 might inhibit the autophagy process through MAGEA6/AMPKα1 axis in LUAD cells. Collectively, our study revealed the oncogenic role and pathways of DDX17 in LUAD.

4.
Front Oncol ; 12: 894786, 2022.
Article in English | MEDLINE | ID: mdl-35646694

ABSTRACT

Hashimoto's thyroiditis (TH) is a risk factor for the occurrence of papillary thyroid carcinoma (PTC), which is considered to be the most common type of thyroid cancer. In recent years, the prevalence of PTC with TH has been increasing, but little is known about the genetic alteration in PTC with TH. This study analyzed the mutation spectrum and mutation signature of somatic single nucleotide variants (SNV) for 10 non-tumor and tumor pair tissues of PTC with TH using whole-exome sequencing. The ANK3 protein expression was evaluated by immunohistochemistry in PTC with TH and PTC samples. Moreover, the functional role of ANK3 in PTC cells was determined by CCK-8 proliferation assay, colony formation assays, cell cycle analysis, cell invasion and migration and in vivo study through overexpression assay. Our results showed three distinct mutational signatures and the C>T/G>A substitution was the most common type of SNV. Gene-set enrichment analysis showed that most of the significantly mutated genes were enriched in the regulation of actin cytoskeleton signaling. Moreover, NCOR2, BPTF, ANK3, and PCSK5 were identified as the significantly mutated genes in PTC with TH, most of which have not been previously characterized. Unexpectedly, it was found that ANK3 was overexpressed in cytoplasm close to the membrane of PTC cells with TH and in almost all PTC cases, suggesting its role as a diagnostic marker of PTC. Ectopic expression of ANK3 suppressed invasion and migration, increased apoptosis of B-CPAP and TPC-1 cells. Moreover, our findings revealed that enhanced ANK3 expression inhibits growth of PTC cells both in vitro and in vivo. Ectopic expression of ANK3 significantly enhanced E-cadherin protein expression and inhibited PTC progression, at least in part, by suppression of epithelial-mesenchymal transition (EMT). Our study shows that ANK3 exerts an anti-oncogenic role in the development of PTC and might be an indolent maintainer of PTC.

5.
Acta Biochim Biophys Sin (Shanghai) ; 53(7): 912-924, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34002210

ABSTRACT

Although initially discovered and extensively studied for its role in inflammation, Annexin A1 (ANXA1) has been reported to be closely related to cancer in recent years, and its role in cancer is specific to tumor types and tissues. In the present study, we identified ANXA1 as an interaction partner of glycogen synthase kinase 3 beta (GSK3ß), a multi-functional serine/threonine kinase tightly associated with cell fate determination and cancer, and assessed the functional significance of GSK3ß-ANXA1 interaction in the metastasis of non-small cell lung cancer (NSCLC). We confirmed the interaction between GSK3ß and ANXA1 in vitro and in H1299 and A549 cells by Glutathione-S-transferase (GST) pull-down assay and co-immunoprecipitation. We found that ANXA1 negatively regulated the phosphorylation of GSK3ß and inhibited the epithelial-mesenchymal transformation (EMT) process and migration and invasion of NSCLC cells. By functional rescue assay, we confirmed that ANXA1 inhibited EMT through the regulation of GSK3ß activity and thereby inhibited the migration and invasion of NSCLC cells. Our study sheds light on the function of ANXA1 and GSK3ß and provides new elements for the understanding of NSCLC pathogenesis.


Subject(s)
Annexin A1/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cytoskeletal Proteins/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , A549 Cells , Annexin A1/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis , Neoplasm Proteins/genetics , Nuclear Proteins/genetics
6.
Proteomics ; 19(15): e1900092, 2019 08.
Article in English | MEDLINE | ID: mdl-31294914

ABSTRACT

Odoroside A (OA) is an active ingredient extracted from the leaves of Nerium oleander Linn. (Apocynaceae). This study aims to examine the anticancer bioactivity of OA against CRC cells and to investigate the action mechanisms involved. As a result, OA can significantly inhibit cellular ability and induce apoptosis of CRC cells in a concentration-dependent manner without any obvious cytotoxicity in normal colorectal epithelial cells. Then, quantitative proteomics combined with bioinformatics is adopted to investigate the alterations of proteins and signaling pathways in response to OA treatment. As suggested by the proteomic analysis, flow cytometry and Western blotting analyses validate that exposure of CRC cells to OA causes cell cycle arrest and apoptosis, accompanied with the activation of the ROS/p53 signaling pathway. This observation demonstrates that OA, as a natural product, can induce oxidative stress to suppress tumor cell growth, implicating a novel therapeutic agent against CRC without obvious side effects.


Subject(s)
Cardenolides/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Proteomics/methods , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology , HT29 Cells , Humans , Signal Transduction/drug effects
7.
Phytomedicine ; 57: 352-363, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30831484

ABSTRACT

BACKGROUND: Nerigoside (NG), a cardenolide isolated from a commonfolk medicine, Nerium oleander Linn. (Apocynaceae), has not been explored for its biological effects. To date, cardenolides have received considerable attention in pharmacology studies due to their direct effects of apoptosis-induction or growth-inhibitory against tumor in vitro and in vivo. Whether and how NG exerts anticancer effects against colorectal cancer remains to be elucidated. PURPOSE: The aim of this study was to investigate the anticancer effect of NG in human colorectal cancer cells. METHODS: To test anticancer effect, we compared potency of NG in two colorectal cancer cell lines, HT29 and SW620 by WST-1 and colony proliferation assays. And we investigated mechanism of anticancer activities by analyzing players in apoptotic and ERK/GSK3ß/ß-catenin signaling pathways in HT29 and SW620 cells treated with NG. RESULTS: In this study, we showed that NG markedly suppressed the cell viability and colony formation of colorectal cancer cells HT29 and SW620, with no significant toxic effect on non-cancer cells NCM460. Annexin V-FITC/PI and CFSE labeling results revealed that NG suppressed cell proliferation in low concentration, along with reducing expression of PCNA, while NG induced apoptosis in high concentration,. Meanwhile, NG significantly arrested cell migration by reversal of EMT and cell cycle on G2/M. Then, we found that the ERK and GSK3ß/ß-catenin signaling pathway were noticeably blocked in CRC cells after treatment with NG. According to western blot, NG upregulated the expression of p-GSK3ß/GSK3ß and decreased especially the expression of ß-catenin in nuclear. In addition, Wnt signaling and its target genes were suppressed in response to NG. Then, the Ser9 phosphorylation of GSK3ß can be reduced / raised by GÖ 6983 / LiCl, respectively. Thus, we further confirmed that the GSK3ß/ß-catenin axis is involved in NG-prevented cell proliferation. CONCLUSION: NG inhibited the growth of colorectal cancer cells by suppressing ERK/GSK3ß/ß-catenin signaling pathway. And the GSK3ß/ß-catenin axis is involved in preventing cell proliferation and migration by NG-treatment. These results suggest that NG may be used to treat colorectal cancer, with better outcome by combining with GSK3ß inhibitor to block Wnt pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , HT29 Cells , Humans , MAP Kinase Signaling System/drug effects , Molecular Targeted Therapy/methods , Nerium/chemistry , Phosphorylation/drug effects , Signal Transduction/drug effects , beta Catenin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...