1
|
Luo L, Zhao Q, Cui X, Dong S, Wang Y, Jiang N, Cai C, Jin J, Liang B. Male-specific lethal 1 (MSL1) promotes Erastin-induced ferroptosis in colon cancer cells by regulating the KCTD12-SLC7A11 axis. Cell Death Dis 2025; 16:281. [PMID: 40221412 PMCID: PMC11993775 DOI: 10.1038/s41419-025-07555-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/22/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
MSL1, a scaffold protein of the MSL histone acetyltransferase complex, is crucial for its structural integrity and enzymatic activity. While MSL1 is highly expressed in various tumors, its role in tumor progression and cell death remains unclear. Here, we provide evidence of a negative regulatory relationship between MSL1 and KCTD12 through biochemical assays and knockdown/overexpression studies. Notably, in colon cancer cells, the ferroptosis inducer Erastin significantly suppressed MSL1 expression, leading to KCTD12 upregulation. Moreover, MSL1 promotes Erastin-induced ferroptosis in HCT116 and SW480 cells via the KCTD12-SLC7A11 axis. Consistently, Erastin-induced changes in ROS, GSH, and MDA levels were regulated by this axis, highlighting its role in ferroptosis. These findings offer potential therapeutic targets and a theoretical foundation for colon cancer treatment.
Collapse
Affiliation(s)
- Lifu Luo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qingzhi Zhao
- School of Nursing, Jilin University, Changchun, Jilin Province, China
- School of Life Sciences, Jilin University, Changchun, Jilin Province, China
| | - Xueli Cui
- School of Life Sciences, Jilin University, Changchun, Jilin Province, China
| | - Shijiao Dong
- School of Life Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yong Wang
- Urology Department, Jilin Province People's Hospital, Changchun, Jilin Province, China
| | - Nan Jiang
- Department of Cardiovascular center, Jilin University First Hospital, Changchun, Jilin Province, China
| | - Chengyu Cai
- School of Life Sciences, Jilin University, Changchun, Jilin Province, China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun, Jilin Province, China.
| | - Bing Liang
- School of Nursing, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
2
|
Nikhil K, Shah K. The significant others of aurora kinase a in cancer: combination is the key. Biomark Res 2024; 12:109. [PMID: 39334449 PMCID: PMC11438406 DOI: 10.1186/s40364-024-00651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
AURKA is predominantly famous as an essential mitotic kinase. Recent findings have also established its critical role in a plethora of other biological processes including ciliogenesis, mitochondrial dynamics, neuronal outgrowth, DNA replication and cell cycle progression. AURKA overexpression in numerous cancers is strongly associated with poor prognosis and survival. Still no AURKA-targeted drug has been approved yet, partially because of the associated collateral toxicity and partly due to its limited efficacy as a single agent in a wide range of tumors. Mechanistically, AURKA overexpression allows it to phosphorylate numerous pathological substrates promoting highly aggressive oncogenic phenotypes. Our review examines the most recent advances in AURKA regulation and focuses on 33 such direct cancer-specific targets of AURKA and their associated oncogenic signaling cascades. One of the common themes that emerge is that AURKA is often involved in a feedback loop with its substrates, which could be the decisive factor causing its sustained upregulation and hyperactivation in cancer cells, an Achilles heel not exploited before. This dynamic interplay between AURKA and its substrates offers potential opportunities for targeted therapeutic interventions. By targeting these substrates, it may be possible to disrupt this feedback loop to effectively reverse AURKA levels, thereby providing a promising avenue for developing safer AURKA-targeted therapeutics. Additionally, exploring the synergistic effects of AURKA inhibition with its other oncogenic and/or tumor-suppressor targets could provide further opportunities for developing effective combination therapies against AURKA-driven cancers, thereby maximizing its potential as a critical drug target.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India.
| | - Kavita Shah
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
3
|
Lam MS, Aw JJ, Tan D, Vijayakumar R, Lim HYG, Yada S, Pang QY, Barker N, Tang C, Ang BT, Sobota RM, Pavesi A. Unveiling the Influence of Tumor Microenvironment and Spatial Heterogeneity on Temozolomide Resistance in Glioblastoma Using an Advanced Human In Vitro Model of the Blood-Brain Barrier and Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302280. [PMID: 37649234 DOI: 10.1002/smll.202302280] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/26/2023] [Indexed: 09/01/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain cancer in adults with a dismal prognosis. Temozolomide (TMZ) is the first-in-line chemotherapeutic; however, resistance is frequent and multifactorial. While many molecular and genetic factors have been linked to TMZ resistance, the role of the solid tumor morphology and the tumor microenvironment, particularly the blood-brain barrier (BBB), is unknown. Here, the authors investigate these using a complex in vitro model for GBM and its surrounding BBB. The model recapitulates important clinical features such as a dense tumor core with tumor cells that invade along the perivascular space; and a perfusable BBB with a physiological permeability and morphology that is altered in the presence of a tumor spheroid. It is demonstrated that TMZ sensitivity decreases with increasing cancer cell spatial organization, and that the BBB can contribute to TMZ resistance. Proteomic analysis with next-generation low volume sample workflows of these cultured microtissues revealed potential clinically relevant proteins involved in tumor aggressiveness and TMZ resistance, demonstrating the utility of complex in vitro models for interrogating the tumor microenvironment and therapy validation.
Collapse
Affiliation(s)
- Maxine Sy Lam
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Joey Jy Aw
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Damien Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Ragavi Vijayakumar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Hui Yi Grace Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Swathi Yada
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Qing You Pang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Nick Barker
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Carol Tang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
- Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Beng Ti Ang
- Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| |
Collapse
|
4
|
Liu P, Liu Z, Luo Q, Fu Q, Zhang X, Yu P, Zhou S, Wang Y, Zhang J, Chen S, Zhang H, Zhu Q, Qin T. A pan-cancer analysis of potassium channel tetramerization domain containing 12 in human cancer. Sci Rep 2023; 13:13898. [PMID: 37626178 PMCID: PMC10457314 DOI: 10.1038/s41598-023-41091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023] Open
Abstract
Abnormal expression of the potassium channel tetramerization domain containing 12 (KCTD12) is closely related to the occurrence and development of various tumors, but a pan-cancer analysis of KCTD12 has not yet been conducted. We explored the association between KCTD12 and more than 30 human malignancies using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. First, the mRNA and protein levels of KCTD12 were examined and their correlations with tumor stage and survival were explored. Second, we analyzed the infiltration of CD8+ and CD4+ T cells and cancer-associated fibroblasts in tumors and explored the correlation between KCTD12 expression and tumor cell stemness, genomic heterogeneity, and diagnostic specificity. Finally, we explored the molecular mechanisms associated with KCTD12 using KEGG/GO analysis. The results showed that KCTD12 mRNA and protein expression levels decreased in most tumors was significantly associated with the prognosis of tumor patients, and the phosphorylation level of KCTD12 decreased in several tumors, such as S200 and T196, pancreatic adenocarcinoma (PAAD), lung adenocarcinoma (LUAD), and breast invasive cancer (BRCA). The expression of KCTD12 was positively correlated with the degree of cancer-associated fibroblasts infiltration in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), head and neck squamous cell carcinoma (HNSC), PAAD, and stomach adenocarcinoma (STAD). The relationship between KCTD12 expression and CD8+ and CD4+ T cell infiltration was also clarified. KCTD12 showed high diagnostic sensitivity for various types of tumors and may be involved in tumor cell biology by affecting tumor cell stemness, tumor burden, and other characteristics. Finally, we analyzed the molecular functions of KCTD12 and possible KEGG/GO signaling pathways. In this study, we developed a biological marker for diagnosis, prognosis, and immune infiltration of the pan-cancers.
Collapse
Affiliation(s)
- Pan Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China
| | - Zhilan Liu
- Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Qiankun Luo
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China
| | - Qiang Fu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China
| | - Xu Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China
| | - Pengfei Yu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China
| | - Shuai Zhou
- Translational Research Institute, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Yingying Wang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, China
| | - Jiali Zhang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, China
| | - Song Chen
- Translational Research Institute, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, China
| | - Hongwei Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China.
| | - Qinghai Zhu
- Zhumadian Central Hospital, No. 747, Zhonghua Road, Zhumadian, 463000, China.
| | - Tao Qin
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450000, China.
| |
Collapse
|
5
|
Smaldone G, Pecoraro G, Pane K, Franzese M, Ruggiero A, Vitagliano L, Salvatore M. The Oncosuppressive Properties of KCTD1: Its Role in Cell Growth and Mobility. BIOLOGY 2023; 12:biology12030481. [PMID: 36979172 PMCID: PMC10045846 DOI: 10.3390/biology12030481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
The KCTD protein family is traditionally regarded as proteins that play key roles in neurological physiopathology. However, new studies are increasingly demonstrating their involvement in many other biological processes, including cancers. This is particularly evident for KCTD proteins not involved in protein ubiquitination and degradation, such as KCTD1. We explored the role of KCTD1 in colorectal cancer by knocking down this protein in the human colon adenocarcinoma cell line, SW480. We re-assessed its ability to downregulate β-catenin, a central actor in the WNT/β-catenin signalling pathway. Interestingly, opposite effects are observed when the protein is upregulated in CACO2 colorectal cancer cells. Moreover, interrogation of the TCGA database indicates that KCTD1 downregulation is associated with β-catenin overexpression in colorectal cancer patients. Indeed, knocking down KCTD1 in SW480 cells led to a significant increase in their motility and stemness, two important tumorigenesis traits, suggesting an oncosuppressor role for KCTD1. It is worth noting that similar effects are induced on colorectal cancer cells by the misregulation of KCTD12, a protein that is distantly related to KCTD1. The presented results further expand the spectrum of KCTD1 involvement in apparently unrelated physiopathological processes. The similar effects produced on colorectal cancer cell lines by KCTD1 and KCTD12 suggest novel, previously unreported analogous activities among members of the KCTD protein family.
Collapse
Affiliation(s)
| | | | - Katia Pane
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143 Naples, Italy
| | | | - Alessia Ruggiero
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
| | | |
Collapse
|
6
|
A nomogram for predicting prognosis of multiple myeloma patients based on a ubiquitin-proteasome gene signature. Aging (Albany NY) 2022; 14:9951-9968. [PMID: 36534449 PMCID: PMC9831738 DOI: 10.18632/aging.204432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is a malignant hematopoietic disease that is usually incurable. However, the ubiquitin-proteasome system (UPS) genes have not yet been established as a prognostic predictor for MM, despite their potential applications in other cancers. METHODS RNA sequencing data and corresponding clinical information were acquired from Multiple Myeloma Research Foundation (MMRF)-COMMPASS and served as a training set (n=787). Validation of the prediction signature were conducted by the Gene Expression Omnibus (GEO) databases (n=1040). To develop a prognostic signature for overall survival (OS), least absolute shrinkage and selection operator regressions, along with Cox regressions, were used. RESULTS A six-gene signature, including KCTD12, SIAH1, TRIM58, TRIM47, UBE2S, and UBE2T, was established. Kaplan-Meier survival analysis of the training and validation cohorts revealed that patients with high-risk conditions had a significantly worse prognosis than those with low-risk conditions. Furthermore, UPS-related signature is associated with a positive immune response. For predicting survival, a simple to use nomogram and the corresponding web-based calculator (https://jiangyanxiamm.shinyapps.io/MMprognosis/) were built based on the UPS signature and its clinical features. Analyses of calibration plots and decision curves showed clinical utility for both training and validation datasets. CONCLUSIONS As a result of these results, we established a genetic signature for MM based on UPS. This genetic signature could contribute to improving individualized survival prediction, thereby facilitating clinical decisions in patients with MM.
Collapse
|
7
|
Zhang CW, Zhou B, Liu YC, Su LW, Meng J, Li SL, Wang XL. LINC00365 inhibited lung adenocarcinoma progression and glycolysis via sponging miR-429/KCTD12 axis. ENVIRONMENTAL TOXICOLOGY 2022; 37:1853-1866. [PMID: 35426242 DOI: 10.1002/tox.23532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
This study researched the function of long non-coding RNA LINC00365 in lung adenocarcinoma (LAD) progression. LINC00365, miR-429, and KCTD12 expression in the LAD clinical tissues and cells were detcetd by qRT-PCR and Western blot. LINC00365, miR-429, and KCTD12 effects on H1975 cells malignant phenotype were detected by cell counting kit-8 assay, clone formation experiment, Transwell experiment, and glycolysis. Dual luciferase reporter gene assay and RNA pull-down assay were implemented. LINC00365 effect on H1975 cells in vivo growth was detected. LINC00365 was low expressed in the LAD patients and cells, associating with poor outcome. LINC00365 up-regulation attenuated H1975 cells proliferation, migration, invasion, glycolysis and in vivo growth. LINC00365 inhibited KCTD12 expression by sponging miR-429. miR-429 up-regulation and KCTD12 down-regulation partial reversed LINC00365 inhibition on H1975 cells malignant phenotype. Thus, LINC00365 inhibited LAD progression and glycolysis via targeting miR-429/KCTD12 axis. LINC00365 might be a potential candidate for LAD target treatment clinically.
Collapse
Affiliation(s)
- Cheng-Wei Zhang
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| | - Bin Zhou
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| | - Yan-Chao Liu
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| | - Li-Wei Su
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| | - Jie Meng
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| | - Shao-Lei Li
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xue-Long Wang
- Department of Thoracic Surgery, Capital Medical University Electric Power Teaching Hospital, Beijing, China
| |
Collapse
|
8
|
Zhou Y, Wang Y. Prognostic implication of an energy metabolism‐related 11‐gene signature in lung cancer. J Biochem Mol Toxicol 2022; 36:e23171. [PMID: 35851973 DOI: 10.1002/jbt.23171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Yang Zhou
- Medical Oncology Department of Gastrointestinal Cancer Cancer Hospital of China Medical University Shenyang China
| | - Yuanhe Wang
- Medical Oncology Department of Gastrointestinal Cancer Cancer Hospital of China Medical University Shenyang China
| |
Collapse
|
9
|
Proteomic Signatures of Diffuse and Intestinal Subtypes of Gastric Cancer. Cancers (Basel) 2021; 13:cancers13235930. [PMID: 34885041 PMCID: PMC8656738 DOI: 10.3390/cancers13235930] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a leading cause of death from cancer globally. Gastric cancer is classified into intestinal, diffuse and indeterminate subtypes based on histology according to the Laurén classification. The intestinal and diffuse subtypes, although different in histology, demographics and outcomes, are still treated in the same fashion. This study was designed to discover proteomic signatures of diffuse and intestinal subtypes. Mass spectrometry-based proteomics using tandem mass tags (TMT)-based multiplexed analysis was used to identify proteins in tumor tissues from patients with diffuse or intestinal gastric cancer with adjacent normal tissue control. A total of 7448 or 4846 proteins were identified from intestinal or diffuse subtype, respectively. This quantitative mass spectrometric analysis defined a proteomic signature of differential expression across the two subtypes, which included gremlin1 (GREM1), bcl-2-associated athanogene 2 (BAG2), olfactomedin 4 (OLFM4), thyroid hormone receptor interacting protein 6 (TRIP6) and melanoma-associated antigen 9 (MAGE-A9) proteins. Although GREM1, BAG2, OLFM4, TRIP6 and MAGE-A9 have all been previously implicated in tumor progression and metastasis, they have not been linked to intestinal or diffuse subtypes of gastric cancer. Using immunohistochemical labelling of a tissue microarray comprising of 124 cases of gastric cancer, we validated the proteomic signature obtained by mass spectrometry in the discovery cohort. Our findings should help investigate the pathogenesis of these gastric cancer subtypes and potentially lead to strategies for early diagnosis and treatment.
Collapse
|
10
|
Angrisani A, Di Fiore A, De Smaele E, Moretti M. The emerging role of the KCTD proteins in cancer. Cell Commun Signal 2021; 19:56. [PMID: 34001146 PMCID: PMC8127222 DOI: 10.1186/s12964-021-00737-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/05/2021] [Indexed: 12/24/2022] Open
Abstract
The human family of Potassium (K+) Channel Tetramerization Domain (KCTD) proteins counts 25 members, and a significant number of them are still only partially characterized. While some of the KCTDs have been linked to neurological disorders or obesity, a growing tally of KCTDs are being associated with cancer hallmarks or involved in the modulation of specific oncogenic pathways. Indeed, the potential relevance of the variegate KCTD family in cancer warrants an updated picture of the current knowledge and highlights the need for further research on KCTD members as either putative therapeutic targets, or diagnostic/prognostic markers. Homology between family members, capability to participate in ubiquitination and degradation of different protein targets, ability to heterodimerize between members, role played in the main signalling pathways involved in development and cancer, are all factors that need to be considered in the search for new key players in tumorigenesis. In this review we summarize the recent published evidence on KCTD members' involvement in cancer. Furthermore, by integrating this information with data extrapolated from public databases that suggest new potential associations with cancers, we hypothesize that the number of KCTD family members involved in tumorigenesis (either as positive or negative modulator) may be bigger than so far demonstrated. Video abstract.
Collapse
Affiliation(s)
| | - Annamaria Di Fiore
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Marta Moretti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Lei X, He Q, Li Z, Zou Q, Xu P, Yu H, Ding Y, Zhu W. Cancer stem cells in colorectal cancer and the association with chemotherapy resistance. Med Oncol 2021; 38:43. [PMID: 33738588 DOI: 10.1007/s12032-021-01488-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The incidence and mortality of colorectal cancer (CRC) have always been among the highest in the world, although the diagnosis and treatment are becoming more and more advanced. At present, the main reason is that patients have acquired drug resistance after long-term conventional drug treatment. An increasing number of evidences confirm the existence of cancer stem cells (CSCs), which are a group of special cells in cancer, only a small part of cancer cells. These special cell populations are not eliminated by chemotherapeutic drugs and result in tumor recurrence and metastasis after drug treatment. CSCs have the ability of self-renewal and multidirectional differentiation, which is associated with the occurrence and development of cancer. CSCs can be screened and identified by related surface markers. In this paper, the characteristic surface markers of CSCs in CRC and the related mechanism of drug resistance will be discussed in detail. A better understanding of the mechanism of CSCs resistance to chemotherapy may lead to better targeted therapy.
Collapse
Affiliation(s)
- Xue Lei
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Qinglian He
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Qian Zou
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Pingrong Xu
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Haibing Yu
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China.
| |
Collapse
|
12
|
K + Channel Tetramerization Domain 5 (KCTD5) Protein Regulates Cell Migration, Focal Adhesion Dynamics and Spreading through Modulation of Ca 2+ Signaling and Rac1 Activity. Cells 2020; 9:cells9102273. [PMID: 33053687 PMCID: PMC7600296 DOI: 10.3390/cells9102273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023] Open
Abstract
Cell migration is critical for several physiological and pathophysiological processes. It depends on the coordinated action of kinases, phosphatases, Rho-GTPases proteins, and Ca2+ signaling. Interestingly, ubiquitination events have emerged as regulatory elements of migration. Thus, the role of proteins involved in ubiquitination processes could be relevant to a complete understanding of pro-migratory mechanisms. KCTD5 is a member of Potassium Channel Tetramerization Domain (KCTD) proteins that have been proposed as a putative adaptor for Cullin3-E3 ubiquitin ligase and a novel regulatory protein of TRPM4 channels. Here, we study whether KCTD5 participates in cell migration-associated mechanisms, such as focal adhesion dynamics and cellular spreading. Our results show that KCTD5 CRISPR/Cas9- and shRNA-based depletion in B16-F10 cells promoted an increase in cell migration and cell spreading, and a decrease in the focal adhesion area, consistent with an increased focal adhesion disassembly rate. The expression of a dominant-negative mutant of Rho-GTPases Rac1 precluded the KCTD5 depletion-induced increase in cell spreading. Additionally, KCTD5 silencing decreased the serum-induced Ca2+ response, and the reversion of this with ionomycin abolished the KCTD5 knockdown-induced decrease in focal adhesion size. Together, these data suggest that KCTD5 acts as a regulator of cell migration by modulating cell spreading and focal adhesion dynamics through Rac1 activity and Ca2+ signaling, respectively.
Collapse
|
13
|
Paulino DSM, Mendes MCS, Camargo JA, Brambilla SR, Wood Dos Santos T, Ribeiro ML, Carvalheira JBC. Diacerein treatment prevents colitis-associated cancer in mice. World J Clin Oncol 2020; 11:732-746. [PMID: 33033695 PMCID: PMC7522546 DOI: 10.5306/wjco.v11.i9.732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/06/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammation is a well-established enabling factor for cancer development and provides a framework for the high prevalence of colon cancer in inflammatory bowel disease. In accordance, chronic inflammation has recently been implicated in the development of cancer stem cells (CSCs). However, the mechanism whereby anti-inflammatory drugs act in the prevention of colitis-associated cancer (CAC) is only partially understood. AIM To evaluate the role of diacerein (DAR), an anti-inflammatory drug that mainly acts through the inhibition of interleukin (IL)-1β expression in the development of CSCs and CAC. METHODS The effects of DAR on colon inflammation in mice with CAC were evaluated by inflammatory index, reverse real-time transcription polymerase chain reaction and western blot. Cytokine levels were measured by enzyme-linked immunosorbent assay. Cells assays evaluated the effects of DAR on CSCs. Immunohistochemistry and apoptosis assays were also used to evaluate the effects of DAR on tumorigenesis associated with inflammation. RESULTS DAR treatment reduced colon inflammation as well as the number and size of tumors in azoxymethane plus dextran sulphate sodium-treated animals. Accordingly, DAR treatment was associated with reduced intracellular signals of inflammation (inhibitor of nuclear factor kappa B kinase and c-Jun N-terminal kinase phosphorylation) in the colon. In addition, DAR treatment was associated with a decrease in colon CSC formation, suggesting that besides reducing colonic inflammation, DAR has a direct effect on the inhibition of colon carcinogenesis. CONCLUSION Together, these data indicate that DAR-mediated IL-1β suppression attenuates inflammation-induced colon cancer and CSC formation, highlighting DAR as a potential candidate for the chemoprevention of CAC.
Collapse
Affiliation(s)
- Daiane S M Paulino
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-970, Brazil
| | - Maria Carolina S Mendes
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-970, Brazil
| | - Juliana A Camargo
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-970, Brazil
| | - Sandra R Brambilla
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-970, Brazil
| | - Tanila Wood Dos Santos
- Department of Clinical Pharmacology and Gastroenterology, Sao Francisco University, Sao Paulo 12916-900, Brazil
| | - Marcelo L Ribeiro
- Department of Clinical Pharmacology and Gastroenterology, Sao Francisco University, Sao Paulo 12916-900, Brazil
| | | |
Collapse
|
14
|
Parachini-Winter C, Bracha S, Ramsey SA, Yang L, Ho E, Leeper HJ, Curran KM. Prospective evaluation of the lymph node proteome in dogs with multicentric lymphoma supplemented with sulforaphane. J Vet Intern Med 2020; 34:2036-2047. [PMID: 32926463 PMCID: PMC7517837 DOI: 10.1111/jvim.15898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Background Lymphoma (LSA) is a common malignancy in dogs. Epigenetic changes are linked to LSA pathogenesis and poor prognosis in humans, and LSA pathogenesis in dogs. Sulforaphane (SFN), an epigenetic‐targeting compound, has recently gained interest in relation to cancer prevention and therapy. Objective Examine the impact of oral supplementation with SFN on the lymph node proteome of dogs with multicentric LSA. Animals Seven client‐owned dogs with multicentric LSA. Methods Prospective, nonrandomized, noncontrolled study in treatment‐naïve dogs with intermediate or large cell multicentric LSA. Lymph node cell aspirates were obtained before and after 7 days of oral supplementation with SFN, and analyzed via label‐free mass spectrometry, immunoblots, and Gene Set Enrichment Analysis. Results There was no clinical response and no adverse events attributed to SFN. For individual dogs, the expression of up to 650 proteins changed by at least 2‐fold (range, 2‐100) after supplementation with SFN. When all dogs where analyzed together, 14 proteins were significantly downregulated, and 10 proteins were significantly upregulated after supplementation with SFN (P < .05). Proteins and gene sets impacted by SFN were commonly involved in immunity, response to oxidative stress, gene transcription, apoptosis, protein transport, maturation and ubiquitination. Conclusions and Clinical Importance Sulforaphane is associated with major changes in the proteome of neoplastic lymphocytes in dogs.
Collapse
Affiliation(s)
- Cyril Parachini-Winter
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Shay Bracha
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Stephen A Ramsey
- Department of Biomedical Sciences, School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, USA
| | - Liping Yang
- Department of Chemistry, College of Science, Oregon State University, Corvallis, Oregon, USA
| | - Emily Ho
- Linus Pauling Institute and College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Haley J Leeper
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Kaitlin M Curran
- Department of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
15
|
Rivas J, Díaz N, Silva I, Morales D, Lavanderos B, Álvarez A, Saldías MP, Pulgar E, Cruz P, Maureira D, Flores G, Colombo A, Blanco C, Contreras HR, Jaña F, Gallegos I, Concha ML, Vergara-Jaque A, Poblete H, González W, Varela D, Trimmer JS, Cáceres M, Cerda O. KCTD5, a novel TRPM4-regulatory protein required for cell migration as a new predictor for breast cancer prognosis. FASEB J 2020; 34:7847-7865. [PMID: 32301552 DOI: 10.1096/fj.201901195rrr] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/23/2022]
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a Ca2+ -activated nonselective cationic channel that regulates cell migration and contractility. Increased TRPM4 expression has been related to pathologies, in which cytoskeletal rearrangement and cell migration are altered, such as metastatic cancer. Here, we identify the K+ channel tetramerization domain 5 (KCTD5) protein, a putative adaptor of cullin3 E3 ubiquitin ligase, as a novel TRPM4-interacting protein. We demonstrate that KCTD5 is a positive regulator of TRPM4 activity by enhancing its Ca2+ sensitivity. We show that through its effects on TRPM4 that KCTD5 promotes cell migration and contractility. Finally, we observed that both TRPM4 and KCTD5 expression are increased in distinct patterns in different classes of breast cancer tumor samples. Together, these data support that TRPM4 activity can be regulated through expression levels of either TRPM4 or KCTD5, not only contributing to increased understanding of the molecular mechanisms involved on the regulation of these important ion channels, but also providing information that could inform treatments based on targeting these distinct molecules that define TRPM4 activity.
Collapse
Affiliation(s)
- José Rivas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile
| | - Nicolás Díaz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Danna Morales
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Alhejandra Álvarez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Eduardo Pulgar
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Guillermo Flores
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Constanza Blanco
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Héctor R Contreras
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fabián Jaña
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Ivan Gallegos
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Miguel L Concha
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Millennium Nucleus on Physics of Active Matter, Santiago, Chile
| | - Ariela Vergara-Jaque
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Horacio Poblete
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Wendy González
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Diego Varela
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
16
|
Ye RY, Kuang XY, Zeng HJ, Shao N, Lin Y, Wang SM. KCTD12 promotes G1/S transition of breast cancer cell through activating the AKT/FOXO1 signaling. J Clin Lab Anal 2020; 34:e23315. [PMID: 32207860 PMCID: PMC7439418 DOI: 10.1002/jcla.23315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/01/2020] [Accepted: 03/05/2020] [Indexed: 01/22/2023] Open
Abstract
Background Sustaining proliferation is the most fundamental step for breast cancer tumor genesis. Accelerated proliferation is usually linked to the uncontrolled cell cycle. However, the internal and external factors linked to the activation of breast cancer cell cycle are still to be investigated. Methods quantitative PCR (qPCR) and Western blotting assay were used to detect the expression of potassium channel tetramerization domain containing 12 (KCTD12) in breast cancer. MTT and colony formation assays were performed to evaluate the effect of KCTD12 on cell proliferation of breast cancer. Anchorage‐independent growth assay was used to examine the in vitro tumorigenesis of breast cancer cells. Flow cytometry assay, qPCR, and Western blotting were used to investigate the detailed mechanisms of KCTD12 on breast cancer progression. Results Herein, the result showed that the level of KCTD12 is significantly decreased in breast cancer tissues and cells, and lower level of KCTD12 predicts poorer survival for patients with breast cancer. Further cell function tests illustrated that downregulation of KCTD12 significantly promotes cell proliferation and in vitro tumor genesis. Besides, molecular biologic experiments showed that downregulation of KCTD12 can enhance the G1/S transition through activating the AKT/FOXO1 signaling. Conclusion Our study inferred that downregulation of KCTD12 can be a novel factor for poor prognosis in breast cancer.
Collapse
Affiliation(s)
- Run-Yi Ye
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xia-Ying Kuang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui-Juan Zeng
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shen-Ming Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Yoon HJ, Kim DH, Kim SJ, Jang JH, Surh YJ. Src-mediated phosphorylation, ubiquitination and degradation of Caveolin-1 promotes breast cancer cell stemness. Cancer Lett 2019; 449:8-19. [DOI: 10.1016/j.canlet.2019.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 12/19/2022]
|
18
|
Tang Y, Chen K, Wu X, Wei Z, Zhang SY, Song B, Zhang SW, Huang Y, Meng J. DRUM: Inference of Disease-Associated m 6A RNA Methylation Sites From a Multi-Layer Heterogeneous Network. Front Genet 2019; 10:266. [PMID: 31001320 PMCID: PMC6456716 DOI: 10.3389/fgene.2019.00266] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/11/2019] [Indexed: 01/27/2023] Open
Abstract
Recent studies have revealed that the RNA N 6-methyladenosine (m6A) modification plays a critical role in a variety of biological processes and associated with multiple diseases including cancers. Till this day, transcriptome-wide m6A RNA methylation sites have been identified by high-throughput sequencing technique combined with computational methods, and the information is publicly available in a few bioinformatics databases; however, the association between individual m6A sites and various diseases are still largely unknown. There are yet computational approaches developed for investigating potential association between individual m6A sites and diseases, which represents a major challenge in the epitranscriptome analysis. Thus, to infer the disease-related m6A sites, we implemented a novel multi-layer heterogeneous network-based approach, which incorporates the associations among diseases, genes and m6A RNA methylation sites from gene expression, RNA methylation and disease similarities data with the Random Walk with Restart (RWR) algorithm. To evaluate the performance of the proposed approach, a ten-fold cross validation is performed, in which our approach achieved a reasonable good performance (overall AUC: 0.827, average AUC 0.867), higher than a hypergeometric test-based approach (overall AUC: 0.7333 and average AUC: 0.723) and a random predictor (overall AUC: 0.550 and average AUC: 0.486). Additionally, we show that a number of predicted cancer-associated m6A sites are supported by existing literatures, suggesting that the proposed approach can effectively uncover the underlying epitranscriptome circuits of disease mechanisms. An online database DRUM, which stands for disease-associated ribonucleic acid methylation, was built to support the query of disease-associated RNA m6A methylation sites, and is freely available at: www.xjtlu.edu.cn/biologicalsciences/drum.
Collapse
Affiliation(s)
- Yujiao Tang
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Kunqi Chen
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of & Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Xiangyu Wu
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of & Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Zhen Wei
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of & Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Song-Yao Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Bowen Song
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of & Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Yufei Huang
- Department of Epidemiology and Biostatistics, University of Texas Health San Antonio, San Antonio, TX, United States
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Jia Meng
- Department of Biological Sciences, Research Center for Precision Medicine, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of & Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Blanc V, Xie Y, Kennedy S, Riordan JD, Rubin DC, Madison BB, Mills JC, Nadeau JH, Davidson NO. Apobec1 complementation factor (A1CF) and RBM47 interact in tissue-specific regulation of C to U RNA editing in mouse intestine and liver. RNA (NEW YORK, N.Y.) 2019; 25:70-81. [PMID: 30309881 PMCID: PMC6298562 DOI: 10.1261/rna.068395.118] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/07/2018] [Indexed: 05/05/2023]
Abstract
Mammalian C to U RNA is mediated by APOBEC1, the catalytic deaminase, together with RNA binding cofactors (including A1CF and RBM47) whose relative physiological requirements are unresolved. Although A1CF complements APOBEC1 for in vitro RNA editing, A1cf-/- mice exhibited no change in apolipoproteinB (apoB) RNA editing, while Rbm47 mutant mice exhibited impaired intestinal RNA editing of apoB as well as other targets. Here we examined the role of A1CF and RBM47 in adult mouse liver and intestine, following deletion of either one or both gene products and also following forced (liver or intestinal) transgenic A1CF expression. There were minimal changes in hepatic and intestinal apoB RNA editing in A1cf-/- mice and no changes in either liver- or intestine-specific A1CF transgenic mice. Rbm47 liver-specific knockout (Rbm47LKO ) mice demonstrated reduced editing in a subset (11 of 20) of RNA targets, including apoB. By contrast, apoB RNA editing was virtually eliminated (<6% activity) in intestine-specific (Rbm47IKO ) mice with only five of 53 targets exhibiting C-to-U RNA editing. Double knockout of A1cf and Rbm47 in liver (ARLKO ) eliminated apoB RNA editing and reduced editing in the majority of other targets, with no changes following adenoviral APOBEC1 administration. Intestinal double knockout mice (ARIKO ) demonstrated further reduced editing (<10% activity) in four of five of the residual APOBEC1 targets identified in ARIKO mice. These data suggest that A1CF and RBM47 each function independently, yet interact in a tissue-specific manner, to regulate the activity and site selection of APOBEC1 dependent C-to-U RNA editing.
Collapse
Affiliation(s)
- Valerie Blanc
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Yan Xie
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Susan Kennedy
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Jesse D Riordan
- Pacific Northwest Research Institute, Seattle, Washington 98122, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Blair B Madison
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Jason C Mills
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| | - Joseph H Nadeau
- Pacific Northwest Research Institute, Seattle, Washington 98122, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63105, USA
| |
Collapse
|
20
|
Wu N, Zhang C, Wang C, Song L, Yao W, Gedanken A, Lin X, Shi D. Zinc-doped copper oxide nanocomposites reverse temozolomide resistance in glioblastoma by inhibiting AKT and ERK1/2. Nanomedicine (Lond) 2018; 13:1303-1318. [PMID: 29949469 DOI: 10.2217/nnm-2017-0359] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM To assess the effect of zinc-doped copper oxide nanocomposites (nZn-CuO NPs) on glioblastoma therapy. MATERIALS & METHODS nZn-CuO NPs were synthesized by sonochemical method and its antitumor effects and underlying molecular mechanisms were investigated both in vitro and in vivo. RESULTS After nZn-CuO NPs treatment, cell proliferation was significantly inhibited in dividing cancer cells but less toxicity was observed in normal cells. In vivo studies show that nZn-CuO NPs inhibited tumor growth in a dose-dependent manner. Further study found that nZn-CuO NPs trigger cell reactive oxygen species (ROS) generation and intrinsic apoptotic pathway. In temozolomide resistance glioblastoma, nZn-CuO NPs disturb cell growth and sphere formation by inhibiting AKT and ERK1/2 activation. CONCLUSION nZn-CuO NPs possess the potential to be developed as a novel anti-tumor agent, especially to treat temozolomide resistance glioblastoma.
Collapse
Affiliation(s)
- Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Drugs & Bioproducts, Qingdao National Laboratory for Marine Science & Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Chunyun Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, China
| | - Changhui Wang
- Shanghai Neuromedical Center, Qingdao University, Shanghai, China
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weicheng Yao
- Department of Neurosurgery, Qingdao University, Qingdao, China
| | - Aharon Gedanken
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Drugs & Bioproducts, Qingdao National Laboratory for Marine Science & Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
21
|
Li H, Li SJ, Shang J, Liu JX, Zheng CH. A Dynamic Scale-Free Network Particle Swarm Optimization for Extracting Features on Multi-Omics Data. J Comput Biol 2018; 26:769-781. [PMID: 30495971 DOI: 10.1089/cmb.2018.0185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mining meaningful and comprehensive molecular characterization of cancers from The Cancer Genome Atlas (TCGA) data has become a bioinformatics bottleneck. Meanwhile, recent progress in cancer analysis shows that multi-omics data can effectively and systematically detect the cancer-related genes at all levels. In this study, we propose an improved particle swarm optimization with dynamic scale-free network, named DSFPSO, to extract features on multi-omics data. The highlights of DSFPSO are taking the dynamic scale-free network as its population structure and diverse velocity updating strategies for fully considering the heterogeneity of particles and their neighbors. Experiments of DSFPSO and its comparison with several state-of-the-art feature extraction approaches are performed on two public data sets from TCGA. Results show that DSFPSO can extract genes associated with cancers effectively.
Collapse
Affiliation(s)
- Huiyu Li
- 1School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Sheng-Jun Li
- 1School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Junliang Shang
- 1School of Information Science and Engineering, Qufu Normal University, Rizhao, China.,2School of Statistics, Qufu Normal University, Qufu, China
| | - Jin-Xing Liu
- 1School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Chun-Hou Zheng
- 3School of Computer Science and Technology, Anhui University, Hefei, China
| |
Collapse
|
22
|
Aronchik I, Dai Y, Labenski M, Barnes C, Jones T, Qiao L, Beebe L, Malek M, Elis W, Shi T, Mavrommatis K, Bray GL, Filvaroff EH. Efficacy of a Covalent ERK1/2 Inhibitor, CC-90003, in KRAS-Mutant Cancer Models Reveals Novel Mechanisms of Response and Resistance. Mol Cancer Res 2018; 17:642-654. [DOI: 10.1158/1541-7786.mcr-17-0554] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 05/31/2018] [Accepted: 09/21/2018] [Indexed: 11/16/2022]
|
23
|
Abbaszadegan MR, Taghehchian N, Li L, Aarabi A, Moghbeli M. Contribution of KCTD12 to esophageal squamous cell carcinoma. BMC Cancer 2018; 18:853. [PMID: 30157793 PMCID: PMC6114029 DOI: 10.1186/s12885-018-4765-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 08/21/2018] [Indexed: 12/30/2022] Open
Abstract
Background It has been shown that the expression of potassium channel tetramerization domain containing 12 (KCTD12) as a regulator of GABAB receptor signaling is reversely associated with gastrointestinal stromal tumors. In present study we examined the probable role of KCTD12 in regulation of several signaling pathways and chromatin remodelers in esophageal squamous cell carcinoma (ESCC). Methods KCTD12 ectopic expression was done in KYSE30 cell line. Comparative quantitative real time PCR was used to assess the expression of stem cell factors and several factors belonging to the WNT/NOTCH and chromatin remodeling in transfected cells in comparison with non-transfected cells. Results We observed that the KCTD12 significantly down regulated expression of NANOG, SOX2, SALL4, KLF4, MAML1, PYGO2, BMI1, BRG1, MSI1, MEIS1, EGFR, DIDO1, ABCC4, ABCG2, and CRIPTO1 in transfected cells in comparison with non-transfected cells. Migration assay showed a significant decrease in cell movement in ectopic expressed cells in comparison with non-transfected cells (p = 0.02). Moreover, KCTD12 significantly decreased the 5FU resistance in transfected cells (p = 0.01). Conclusions KCTD12 may exert its inhibitory role in ESCC through the suppression of WNT /NOTCH, stem cell factors, and chromatin remodelers and can be introduced as an efficient therapeutic marker.
Collapse
Affiliation(s)
- Mohammad Reza Abbaszadegan
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Faculty of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Faculty of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Liping Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330008, People's Republic of China
| | - Azadeh Aarabi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
Cheng Q, Chen A, Du Q, Liao Q, Shuai Z, Chen C, Yang X, Hu Y, Zhao J, Liu S, Wen GR, An J, Jing H, Tuo B, Xie R, Xu J. Novel insights into ion channels in cancer stem cells (Review). Int J Oncol 2018; 53:1435-1441. [PMID: 30066845 DOI: 10.3892/ijo.2018.4500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/28/2018] [Indexed: 11/06/2022] Open
Abstract
Cancer stem cells (CSCs) are immortal cells in tumor tissues that have been proposed as the driving force of tumorigenesis and tumor invasion. Previously, ion channels were revealed to contribute to cancer cell proliferation, migration and apoptosis. Recent studies have demonstrated that ion channels are present in various CSCs; however, the functions of ion channels and their mechanisms in CSCs remain unknown. The present review aimed to focus on the roles of ion channels in the regulation of CSC behavior and the CSC-like properties of cancer cells. Evaluation of the relationship between ion channels and CSCs is critically important for understanding malignancy.
Collapse
Affiliation(s)
- Qijiao Cheng
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Anhai Chen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Zhangli Shuai
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Changmei Chen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Xinrong Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Yaxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Ju Zhao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Songpo Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Guo Rong Wen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxin An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Hai Jing
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
25
|
SUV39H2 promotes colorectal cancer proliferation and metastasis via tri-methylation of the SLIT1 promoter. Cancer Lett 2018; 422:56-69. [PMID: 29458143 DOI: 10.1016/j.canlet.2018.02.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/30/2018] [Accepted: 02/12/2018] [Indexed: 01/01/2023]
Abstract
Suppressor of variegation 3-9 homolog 2 (SUV39H2) is a member of the SUV39H subfamily of lysine methyltransferases. Its role in colorectal cancer (CRC) proliferation and metastasis has remained unexplored. Here, we determined that SUV39H2 was upregulated in CRC tissues compared with that in adjacent non-neoplastic tissues. Further statistical analysis revealed that high SUV39H2 expression was strongly associated with distant metastasis (P = 0.016) and TNM stage (P = 0.038) and predicted a shorter overall survival (OS; P = 0.018) and progression-free survival (PFS; P = 0.018) time for CRC patients. Both in vitro and in vivo assays demonstrated that ectopically expressed SUV39H2 enhanced CRC proliferation and metastasis, while SUV39H2 knockdown inhibited CRC proliferation and metastasis. A molecular screen of SUV39H2 targets found that SUV39H2 negatively regulated the expression of SLIT guidance ligand 1 (SLIT1). Moreover, rescue assays suggested that SLIT1 could antagonize the function of SUV39H2 in CRC. Mechanistic studies indicated that SUV39H2 can directly bind to the SLIT1 promoter, suppressing SLIT1 transcription by catalyzing histone H3 lysine 9 (H3K9) tri-methylation. In summary, we propose that SUV39H2 can predict CRC patient prognosis and stimulate CRC malignant phenotypes via SLIT1 promoter tri-methylation.
Collapse
|
26
|
Sooreshjani MA, Gursoy UK, Aryal UK, Sintim HO. Proteomic analysis of RAW macrophages treated with cGAMP or c-di-GMP reveals differentially activated cellular pathways. RSC Adv 2018; 8:36840-36851. [PMID: 35558957 PMCID: PMC9089301 DOI: 10.1039/c8ra04603d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/23/2018] [Indexed: 12/28/2022] Open
Abstract
Global and quantitative analysis of the proteome help to reveal how host cells sense invading bacteria and respond to bacterial signaling molecules. Here, we performed label free quantitative proteomic analysis of RAW macrophages treated with host-derived cGAMP and bacterial-derived c-di-GMP, in an attempt to identify cellular pathways impacted by these dinucleotides and determine if the host responds differentially to these two cyclic dinucleotides. We identified a total of 3811 proteins of which abundances of 404 proteins in cGAMP and 236 proteins in c-di-GMP treated cells were significantly different compared to the control. Many of the proteins that were strongly and commonly upregulated, such as interferon-induced proteins 47, 202 and 204 (Ifi47, Ifi202, Ifi204), ubiquitin-activating enzyme E7 (Uba7), interferon-induced protein with tetratricopeptide repeats 1, 2 or 3 (Ifit1, Ifit2, Ifit3), ubiquitin-like protein ISG15 (ISG15), might be due to the fact that both dinucleotides promote the production of interferons, which induce the expression of many proteins. However, there were also other proteins that were differentially affected by cGAMP or c-di-GMP treatment, including probable ATP-dependent RNA helicase DHX58 (Dhx58), nuclear autoantigen Sp-100 (Sp100), MARCKS-related protein (Marcksl1) and antigen peptide transporter 2 (Tap2). This is probably due to the differential levels of IFNs produced by the dinucleotides or may indicate that non-STING activation might also contribute to the host's response to c-di-GMP and cGAMP. Interestingly Trex1, a nuclease that degrades DNA (an activator of cGAS to produce cGAMP), was upregulated (3.22 fold) upon cGAMP treatment, hinting at a possible feedback loop to regulate cGAMP synthesis. These results lay a foundation for future studies to better characterize and understand the complex c-di-GMP and cGAMP signaling network. cGAMP modulates proteins involved in antigen presentation and inflammation.![]()
Collapse
Affiliation(s)
| | - Ulvi K. Gursoy
- Department of Periodontology
- Institute of Dentistry
- University of Turku
- Turku
- Finland
| | - Uma K. Aryal
- Purdue Proteomics Facility
- Bindley Bioscience Center
- Purdue University
- West Lafayette
- USA
| | - Herman O. Sintim
- Department of Chemistry
- Purdue University
- West Lafayette
- USA
- Department of Periodontology
| |
Collapse
|
27
|
Duan Q, Zhao H, Zhang Z, Li H, Wu H, Shen Q, Wang C, Yin T. Mechanistic Evaluation and Translational Signature of Gemcitabine-induced Chemoresistance by Quantitative Phosphoproteomics Analysis with iTRAQ Labeling Mass Spectrometry. Sci Rep 2017; 7:12891. [PMID: 29018223 PMCID: PMC5634998 DOI: 10.1038/s41598-017-13330-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/21/2017] [Indexed: 12/19/2022] Open
Abstract
One of the main causations of the poor prognosis of pancreatic cancer is the lack of effective chemotherapies. Gemcitabine is a widely used chemotherapeutic drug, but limited therapeutic efficacy is achieved due to chemoresistance. Recent studies demonstrated that the presence of cancer stem cells may lead to the failure of chemotherapy. Moreover, gemcitabine can promote the stemness of pancreatic cancer cells. We detected the alterations in protein phosphorylation and signaling pathways in pancreatic cancer cells after gemcitabine treatment using iTRAQ labeling LC-MS/MS, because it was featured with the advantages of strong separation ability and analysis range. A total of 232 differentially expressed phosphorylated proteins were identified in this study. Gene Ontology analysis revealed that nuclear lumen, nuclear part and organelle lumen were enriched for cell components and protein binding, poly (A) RNA binding and RNA binding were enriched for molecular function. A variety of signaling pathways were enriched based on KEGG analysis. AMPK, mTOR and PI3K/Akt pathways were verified after gemcitabine exposure. Moreover, we found there were complex interactions of phosphorylated proteins in modulating cancer stemness induced by gemcitabine exposure based on PPIs map. Our experiments may identify potential targets and strategies for sensitizing pancreatic cancer cells to gemcitabine.
Collapse
Affiliation(s)
- Qingke Duan
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengqiang Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhengle Zhang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hehe Li
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiang Shen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunyou Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Yin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
28
|
Zhong Y, Yang J, Xu WW, Wang Y, Zheng CC, Li B, He QY. KCTD12 promotes tumorigenesis by facilitating CDC25B/CDK1/Aurora A-dependent G2/M transition. Oncogene 2017; 36:6177-6189. [PMID: 28869606 PMCID: PMC5671937 DOI: 10.1038/onc.2017.287] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 06/19/2017] [Accepted: 07/14/2017] [Indexed: 02/06/2023]
Abstract
Cell cycle dysregulation leads to uncontrolled cell proliferation and tumorigenesis. Understanding the molecular mechanisms underlying cell cycle progression can provide clues leading to the identification of key proteins involved in cancer development. In this study, we performed proteomics analysis to identify novel regulators of the cell cycle. We found that potassium channel tetramerization domain containing 12 (KCTD12) was significantly upregulated in M phase compared with S phase. We also found that KCTD12 overexpression not only facilitated the G2/M transition and induced cancer cell proliferation, but also promoted the growth of subcutaneous tumors and Ki-67 proliferation index in mice. Regarding the mechanism underlying these phenomena, cyclin-dependent kinase 1 (CDK1) was identified as an interacting partner of KCTD12 by immunoprecipitation and mass spectrometry analysis, which showed that KCTD12 activated CDK1 and Aurora kinase A (Aurora A) and that the effects of KCTD12 on CDK1 phosphorylation and cell proliferation were abrogated by cell division cycle 25B (CDC25B) silencing. In addition, Aurora A phosphorylated KCTD12 at serine 243, thereby initiating a positive feedback loop necessary for KCTD12 to exert its cancer-promoting effects. Furthermore, we analyzed the expression levels of various genes and the correlations between the expression of these genes and survival using tumor tissue microarray and Gene Expression Omnibus (GEO) data sets. The data showed that KCTD12 expression was significantly upregulated in cervical and lung cancers. More importantly, high KCTD12 expression was associated with larger tumor sizes, higher pathological stages and poor patient survival. Collectively, our study demonstrate that KCTD12 binds to CDC25B and activates CDK1 and Aurora A to facilitate the G2/M transition and promote tumorigenesis and that Aurora A phosphorylates KCTD12 at serine 243 to trigger a positive feedback loop, thereby potentiating the effects of KCTD12. Thus, the KCTD12-CDC25B-CDK1-Aurora A axis has important implications for cancer diagnoses and prognoses.
Collapse
Affiliation(s)
- Y Zhong
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China.,Department of Pathology, Medical College, Jinan University, Guangzhou, China
| | - J Yang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - W W Xu
- Institute of Biomedicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Y Wang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - C-C Zheng
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - B Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Q-Y He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
29
|
Luo L, Cui J, Feng Z, Li Y, Wang M, Cai Y, Wu Y, Jin J. Lentiviral-mediated overexpression of KCTD12 inhibits the proliferation of human uveal melanoma OCM-1 cells. Oncol Rep 2016; 37:871-878. [PMID: 28000887 DOI: 10.3892/or.2016.5325] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/02/2016] [Indexed: 11/05/2022] Open
Abstract
Human potassium channel tetramerization domain containing 12 (KCTD12, also known as Pfetin) is a member of the KCTD family which consists of 26 members. It has been reported that KCTD12 regulates agonist potency and kinetics of GABAB receptor signaling. Proteomic analysis indicates that KCTD12 may be a potential biomarker for the diagnosis and prognosis of gastrointestinal stromal tumors. However, little has been reported concerning the role of KCTD12 in the other tumor types. In the present study, we designed and subcloned N-terminally Flag-tagged human KCTD12 into the pLVX‑Puro vector. We then generated a human uveal melanoma cell line (OCM-1) stably expressing KCTD12. Using this stable cell line, we performed a series of experiments including colony formation, invasion, migration and wound-healing assays, flow cytometry and western blotting. Based on the experimental results, we first demonstrated that KCTD12 effectively suppressed the proliferation of OCM-1 cells and limited the spread of OCM-1 cells. In the flow cytometric analysis, prolongation of the progression of G2/M to G1 phase in the KCTD12-overexpressing OCM-1 cells was observed. In addition, inhibition of KCTD12-overexpressing OCM-1 cell xenograft growth in nude mice was observed. Taken together, KCTD12 may serve as a novel therapeutic target for patients with uveal melanoma.
Collapse
Affiliation(s)
- Lifu Luo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jizhe Cui
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhitong Feng
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yana Li
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Mengdi Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yazhen Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
30
|
Chen T, Gong W, Tian H, Wang H, Chu S, Ma J, Yang H, Cheng J, Liu M, Li X, Jiang C. Fibroblast growth factor 18 promotes proliferation and migration of H460 cells via the ERK and p38 signaling pathways. Oncol Rep 2016; 37:1235-1242. [DOI: 10.3892/or.2016.5301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/23/2016] [Indexed: 11/06/2022] Open
|
31
|
Huang GL, Zhang W, Ren HY, Zhou P, Chen Y, Chen QX, Shen DY. Oncogenic retinoic acid receptor α promotes human colorectal cancer growth through simultaneously regulating p21 transcription and GSK3β/β-catenin signaling. Cancer Lett 2016; 388:118-129. [PMID: 27932034 DOI: 10.1016/j.canlet.2016.11.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 12/24/2022]
Abstract
Retinoic acid receptor α (RARα) plays important roles in the progression of several cancers such as leukemia, breast cancer, and lung cancer. In this study, we demonstrated that RARα protein was frequently overexpressed in human CRC specimens and CRC cell lines. RARα knockdown decreased cell survival, proliferation, and colony formation in vitro and tumorigenic potential in nude mice. Specifically, RARα knockdown inhibited cell cycle progression at G1 phase through upregulation of cell cycle inhibitor p21, and downregulation of cyclinD1. Furthermore, RARα was directly recruited to the p21 promoter to inhibit the expression of p21. Simultaneously, RARα contributed to the progression of CRC cells in part due to upregulation of cyclinD1 via activation of GSK3β/β-catenin pathway. Molecular mechanism studies revealed RARα interacted with GSK3β and led to decreased expression of GSK3β at ser9, followed by increased β-catenin expression. Taken together, our results signified the importance of RARα in CRC and demonstrated that RARα promotes CRC progression through suppressing p21 transcription and enhancing GSK3β/β-catenin signaling. RARα might become a potential molecular target for the treatment of CRC.
Collapse
Affiliation(s)
- Gui-Li Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Wei Zhang
- Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Hong-Yue Ren
- Biobank, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Pan Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yun Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qing-Xi Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China.
| | - Dong-Yan Shen
- Biobank, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| |
Collapse
|