1
|
Venati SR, Uversky VN. Exploring Intrinsic Disorder in Human Synucleins and Associated Proteins. Int J Mol Sci 2024; 25:8399. [PMID: 39125972 PMCID: PMC11313516 DOI: 10.3390/ijms25158399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, β-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human β- and γ- synucleins and revealed that, relative to the β- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.
Collapse
Affiliation(s)
- Sriya Reddy Venati
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Zhang X, Zhang X, Li X, Bao H, Li G, Li N, Li H, Dou J. NUCKS1 Acts as a Promising Novel Biomarker for the Prognosis of Patients with Hepatocellular Carcinoma. Cancer Biother Radiopharm 2023; 38:720-725. [PMID: 33601927 DOI: 10.1089/cbr.2020.4226] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objective: Nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) is highly expressed in some tumors, including hepatocellular carcinoma (HCC). However, its clinical significance in HCC prognosis is still unclear. The aim of this study was to explore the expression and prognostic value of NUCKS1 in HCC. Materials and Methods: Quantitative real-time polymerase chain reaction was used to detect relative expression of NUCKS1 mRNA in HCC tissues and corresponding adjacent normal tissues. The relationship between NUCKS1 expression and clinical characteristics of patients was analyzed by χ2 test. Kaplan-Meier method and Cox regression analysis were applied to estimate prognostic value of NUCKS1 in HCC. Results: Compared with normal ones, the expression of NUCKS1 mRNA was significantly upregulated in HCC tissues (p < 0.001). Besides, NUCKS1 expression was closely associated with tumor differentiation, tumor node metastasis stage, vascular invasion, and metastasis (p < 0.05). Kaplan-Meier analysis revealed that overall survival was obviously longer in HCC patients with low expression of NUCKS1 than those with high NUCKS1 expression (log rank test, p = 0.001). NUCKS1 might be an independent prognostic factor for HCC patients (HR = 1.905, 95% CI = 1.106-3.283, p = 0.020). Conclusions: NUCKS1 may be correlated with the progression of HCC and serve as a potential predictive factor for the prognosis of this disease.
Collapse
Affiliation(s)
- Xianfeng Zhang
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Xianjun Zhang
- Department of Gynaecology, Harrison International Peace Hospital, Hengshui, China
| | - Xinguo Li
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Hongbing Bao
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Guang Li
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Ning Li
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Hengli Li
- Department of Hepatopancreatobiliary Surgery and Harrison International Peace Hospital, Hengshui, China
| | - Jian Dou
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
3
|
Zheng S, Ji R, He H, Li N, Han C, Han J, Li X, Zhang L, Wang Y, Zhao W. NUCKS1, a LINC00629-upregulated gene, facilitated osteosarcoma progression and metastasis by elevating asparagine synthesis. Cell Death Dis 2023; 14:489. [PMID: 37528150 PMCID: PMC10393983 DOI: 10.1038/s41419-023-06010-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 08/03/2023]
Abstract
Nuclear ubiquitous casein and cyclin-dependent kinase substrate 1 (NUCKS1) has been reported to play an oncogenic role in several cancers. However, the biological functions and regulatory mechanism of NUCKS1 in osteosarcoma have not been fully understood. In this study, we reported that NUCKS1 was significantly increased in osteosarcoma. Depletion of NUCKS1 decreased osteosarcoma cell proliferation and metastasis in vivo and in vitro. Overexpression of NUCKS1 accelerated osteosarcoma cell aggressiveness. Mechanistically, NUCKS1 facilitated asparagine (Asn) synthesis by transcriptionally upregulating asparagine synthetase (ASNS) expression and elevating the levels of Asn in osteosarcoma cells, leading to increased cell growth and metastasis. Inhibition of ASNS or reduction of Asn decreased osteosarcoma cell aggressiveness and impaired the promoting effects of NUCKS1 on tumorigenesis and metastasis. Furthermore, we also found that by acting as a sponge for miR-4768-3p, LINC00629 promoted NUCKS1 expression. Collectively, our findings highlight the role of NUCKS1 in regulating asparagine metabolism and reveal that LINC00629 is an important regulator of NUCKS1 that contributes to NUCKS1 upregulation in osteosarcoma.
Collapse
Affiliation(s)
- Shuo Zheng
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China
| | - Renchen Ji
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China
| | - Hongtao He
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China
| | - Na Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Chuanchun Han
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China
| | - Jian Han
- Department of Orthopedics, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, Liaoning, 116033, P.R. China
| | - Xiaodong Li
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China.
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China.
| | - Lu Zhang
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China.
| | - Yuan Wang
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China.
| | - Wenzhi Zhao
- The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116044, P.R. China.
| |
Collapse
|
4
|
Guo J, Zhou X, Cheng L, Gao X. Construction of a miRNA-mRNA network related to exosomes in metastatic hepatocellular carcinoma. Heliyon 2023; 9:e15428. [PMID: 37101627 PMCID: PMC10123261 DOI: 10.1016/j.heliyon.2023.e15428] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023] Open
Abstract
Aims This study aimed to construct a miRNA-mRNA network to elucidate the molecular mechanism of exosome function in metastatic HCC. Methods We explored the Gene Expression Omnibus (GEO) database and then analyzed the RNAs of 50 samples to obtain differentially expressed miRNAs (DEMs) and mRNAs (DEGs) involved in the progression of metastatic HCC. Next, a miRNA-mRNA network related to exosomes in metastatic HCC was constructed on the basis of the identified DEMs and DEGs. Finally, the function of the miRNA-mRNA network was explored by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Immunohistochemistry was performed to validate expression of NUCKS1 in HCC specimens. Based on the immunohistochemistry, the score of the NUCKS1 expression was calculated, and the patients were divided into high- and low-expression patients, and the differences in survival between the two groups were compared. Results Through our analysis, 149 DEMs and 60 DEGs were identified. In addition, a miRNA-mRNA network, including 23 miRNAs and 14 mRNAs, was constructed. Low expression of NUCKS1 was validated in the majority of HCCs compared with their matched adjacent cirrhosis specimens (P < 0.001), which was consistent with our result of differential expression analyses. HCC patients with low expression of NUCKS1 had shorter overall survival than those with high NUCKS1 expression (P = 0.0441). Conclusions The novel miRNA-mRNA network will provide new insights into the underlying molecular mechanisms of exosomes in metastatic HCC. NUCKS1 might serve a potential therapeutic target to restrain the development of HCC.
Collapse
Affiliation(s)
- Jiang Guo
- Department of Interventional Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Long Cheng
- Department of Interventional Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xuesong Gao
- Department of General Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Corresponding author. No. 8 Jingshun East Street, Chaoyang District, Beijing, China.
| |
Collapse
|
5
|
Østvold AC, Grundt K, Wiese C. NUCKS1 is a highly modified, chromatin-associated protein involved in a diverse set of biological and pathophysiological processes. Biochem J 2022; 479:1205-1220. [PMID: 35695515 PMCID: PMC10016235 DOI: 10.1042/bcj20220075] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 11/17/2022]
Abstract
The Nuclear Casein and Cyclin-dependent Kinase Substrate 1 (NUCKS1) protein is highly conserved in vertebrates, predominantly localized to the nucleus and one of the most heavily modified proteins in the human proteome. NUCKS1 expression is high in stem cells and the brain, developmentally regulated in mice and associated with several diverse malignancies in humans, including cancer, metabolic syndrome and Parkinson's disease. NUCKS1 function has been linked to modulating chromatin architecture and transcription, DNA repair and cell cycle regulation. In this review, we summarize and discuss the published information on NUCKS1 and highlight the questions that remain to be addressed to better understand the complex biology of this multifaceted protein.
Collapse
Affiliation(s)
- Anne Carine Østvold
- Institute of Basic Medical Science, Dept. of Biochemistry, University of Oslo, P.O box 1110 Blindern, 0317 Oslo, Norway
| | - Kirsten Grundt
- Institute of Basic Medical Science, Dept. of Biochemistry, University of Oslo, P.O box 1110 Blindern, 0317 Oslo, Norway
| | - Claudia Wiese
- Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| |
Collapse
|
6
|
Yu J, Zhang H, Zhao C, Li G, Zhang Y, Sun Y. CircRNA circ_0008037 facilitates tumor growth and the Warburg effect via upregulating NUCKS1 by binding to miR-433-3p in non-small cell lung cancer. Thorac Cancer 2021; 13:162-172. [PMID: 34850570 PMCID: PMC8758425 DOI: 10.1111/1759-7714.14235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) participate in the genesis and progression of tumors, including non-small cell lung cancer (NSCLC). At present, the role and regulatory mechanisms of circRNAs in NSCLC have not been fully elucidated. The aim of this study was to explore the role and regulatory mechanism of circRNA hsa_circ_0008037 (circ_0008037) in NSCLC. METHODS Expression of circ_0008037 in NSCLC tissues and cells was detected by quantitative real-time polymerase chain reaction (RT-qPCR). Loss-of-function experiments were performed to analyze the influence of circ_0008037 knockdown on proliferation, migration, invasion, and the Warburg effect of NSCLC cells. Western blotting was utilized for protein analysis. The regulatory mechanism of circ_0008037 was surveyed by bioinformatics analysis, RNA pulldown assay, and dual-luciferase reporter assay. Xenograft assay was used to validate the oncogenicity of circ_0008037 in NSCLC in vivo. RESULTS Circ_0008037 was upregulated in NSCLC tissues and cells. Circ_0008037 downregulation reduced tumor growth in vivo and repressed proliferation, migration, invasion, and decreased the Warburg effect of NSCLC cells in vitro. Mechanically, circ_0008037 regulated nuclear ubiquitous casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) expression via sponging miR-433-3p. Furthermore, MiR-433-3p inhibitor reversed the inhibiting influence of circ_0008037 silencing on proliferation, migration, invasion, and the Warburg effect of NSCLC cells. Also, NUCKS1 elevation overturned the repressive influence of miR-433-3p mimic on proliferation, migration, invasion, and the Warburg effect of NSCLC cells. CONCLUSION Circ_0008037 accelerated tumor growth and elevated the Warburg effect via regulating NUCKS1 expression by adsorbing miR-433-3p, providing an underlying target for NSCLC treatment.
Collapse
Affiliation(s)
- Jia Yu
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Haining Zhang
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Chunsheng Zhao
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Guanghui Li
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Yingying Zhang
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Yang Sun
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| |
Collapse
|
7
|
Hume S, Grou CP, Lascaux P, D'Angiolella V, Legrand AJ, Ramadan K, Dianov GL. The NUCKS1-SKP2-p21/p27 axis controls S phase entry. Nat Commun 2021; 12:6959. [PMID: 34845229 PMCID: PMC8630071 DOI: 10.1038/s41467-021-27124-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Efficient entry into S phase of the cell cycle is necessary for embryonic development and tissue homoeostasis. However, unscheduled S phase entry triggers DNA damage and promotes oncogenesis, underlining the requirement for strict control. Here, we identify the NUCKS1-SKP2-p21/p27 axis as a checkpoint pathway for the G1/S transition. In response to mitogenic stimulation, NUCKS1, a transcription factor, is recruited to chromatin to activate expression of SKP2, the F-box component of the SCFSKP2 ubiquitin ligase, leading to degradation of p21 and p27 and promoting progression into S phase. In contrast, DNA damage induces p53-dependent transcriptional repression of NUCKS1, leading to SKP2 downregulation, p21/p27 upregulation, and cell cycle arrest. We propose that the NUCKS1-SKP2-p21/p27 axis integrates mitogenic and DNA damage signalling to control S phase entry. The Cancer Genome Atlas (TCGA) data reveal that this mechanism is hijacked in many cancers, potentially allowing cancer cells to sustain uncontrolled proliferation.
Collapse
Affiliation(s)
- Samuel Hume
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK
| | - Claudia P Grou
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK
| | - Pauline Lascaux
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK
| | - Vincenzo D'Angiolella
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK
| | - Arnaud J Legrand
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK.
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| | - Kristijan Ramadan
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK.
| | - Grigory L Dianov
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK.
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Lavrentieva 10, 630090, Novosibirsk, Russia.
- Novosibirsk State University, Novosibirsk, Russian Federation, 630090, Russia.
| |
Collapse
|
8
|
Ma H, Xu J, Zhao R, Qi Y, Ji Y, Ma K. Upregulation of NUCKS1 in Lung Adenocarcinoma is Associated with a Poor Prognosis. Cancer Invest 2021; 39:435-444. [PMID: 33683970 DOI: 10.1080/07357907.2021.1899199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
To evaluate the clinicopathologic features and survival analysis of NUCKS1 expression in human lung adenocarcinoma (LA), we used bioinformatic methods to obtain NUCKS1 gene status and correlated it with prognosis in LA. We compared NUCKS1 expression in 70 samples of LA with intrinsically normal lung alveoli (NLA) by immunohistochemistry, and analyzed their clinicopathologic features. NUCKS1 was overexpressed in LA components(LACs) relative to NLA, and was significantly correlated to patient with 5-year disease-free survival (DFS) and overall survival(OS). Elevated NUCKS1 expression in LACs was shown to be an independent prognostic indicator for OS and a biomarker in LA.
Collapse
Affiliation(s)
- Hongfei Ma
- Department of Thoracic Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China.,Department of Thoracic Surgery, The Affiliated Hospital, Qingdao University, Qingdao, China
| | - Jing Xu
- Department of Pathology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Ruixia Zhao
- Department of Pathology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Yongyun Qi
- Department of Pathology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Yong Ji
- Medical Department, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Shenzhen, China
| | - Kai Ma
- Department of Thoracic Surgery, The Affiliated Hospital, Qingdao University, Qingdao, China.,Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
9
|
Zhao S, Wang B, Ma Y, Kuang J, Liang J, Yuan Y. NUCKS1 Promotes Proliferation, Invasion and Migration of Non-Small Cell Lung Cancer by Upregulating CDK1 Expression. Cancer Manag Res 2021; 12:13311-13323. [PMID: 33380837 PMCID: PMC7769091 DOI: 10.2147/cmar.s282181] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is a predominant type of lung cancer with a high mortality rate. Objective The aim of this study is to investigate the roles of nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) in NSCLC and to identify the potential mechanisms. Materials and Methods The expression of NUCKS1 in several NSCLC cells was detected firstly. Then, NUCKS1 was overexpressed or silenced in both A549 and NCI-H460 cells, where cell proliferation, invasion and migration were, respectively, determined, using CCK-8, colony formation assay, transwell and wound healing assays. Cell cycle analysis was performed, and the expression-associated proteins were detected by Western blotting. Subsequently, NCI-H460 cells with NUCKS1 overexpression for the subsequent tumor-bearing experiment. And the NUCKS1 expression in tumor tissues was measured by means of immunohistochemistry and Western blotting. Additionally, the STRING database predicted that Cyclin-Dependent Kinase 1 (CDK1) would bind to NUSK1, which was verified by the co-immunoprecipitation assay. Then, CDK1 was silenced by transfection with short hairpin RNA (shRNA)-CDK-1 or by exposure to CDK1 inhibitor p2767-00. And the biological characteristics of proliferation, invasion and migration were examined. Results Results indicated that NUCKS1 was overly expressed in NSCLC cells, and its overexpression promoted proliferation, invasion and migration of both A549 and NCI-H460 cells while NUCKS1 knockdown displayed the opposite effects. Moreover, the results of the xenograft experiments revealed that NUCKS1-upregulation promoted the tumor growth. Furthermore, the immunoprecipitation assay verified CDK1’s interaction with NUCKS1, and CDK1 knockdown alleviates the impact of NUCKS1 overexpression on NSCLC cell proliferation, invasion and migration. Conclusion Taken together, these findings demonstrated that NUCKS1 promotes proliferation, invasion and migration of NSCLC by upregulating CDK1, providing a novel putative target for the clinical treatment of NSCLC.
Collapse
Affiliation(s)
- Shufen Zhao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Baiyao Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou City, Guangdong Province 510095, People's Republic of China
| | - Yanning Ma
- Shunde Hospital, Southern Medical University, Foshan City, Guangdong Province 528308, People's Republic of China
| | - Junjie Kuang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Jiyun Liang
- Shunde Hospital, Southern Medical University, Foshan City, Guangdong Province 528308, People's Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510515, People's Republic of China.,Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou City, Guangdong Province 510095, People's Republic of China
| |
Collapse
|
10
|
Zhao E, Feng L, Bai L, Cui H. NUCKS promotes cell proliferation and suppresses autophagy through the mTOR-Beclin1 pathway in gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:194. [PMID: 32958058 PMCID: PMC7504682 DOI: 10.1186/s13046-020-01696-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
Background Nuclear casein kinase and cyclin-dependent kinase substrate (NUCKS), a novel gene first reported in 2001, is a member of the high mobility group (HMG) family. Although very little is known regarding the biological roles of NUCKS, emerging clinical evidence suggests that the NUCKS protein can be used as a biomarker and therapeutic target in various human ailments, including several types of cancer. Methods We first assessed the potential correlation between NUCKS expression and gastric cancer prognosis. Then functional experiments were conducted to evaluate the effects of NUCKS in cell proliferation, cell cycle, apoptosis and autophagy. Finally, the roles of NUCKS on gastric cancer were examined in vivo. Results We found that NUCKS was overexpressed in gastric cancer patients with poor prognosis. Through manipulating NUCKS expression, it was observed to be positively associated with cell proliferation in vitro and in vivo. NUCKS knockdown could induce cell cycle arrest and apoptosis. Then further investigation indicated that NUCKS knockdown could also significantly induce a marked increase in autophagy though the mTOR-Beclin1 pathway, which could be was rescued by NUCKS restoration. Moreover, silencing Beclin1 in NUCKS knockdown cells or adding rapamycin in NUCKS-overexpressed cells also confirmed these results. Conclusions Our findings revealed that NUCKS functions as an oncogene and an inhibitor of autophagy in gastric cancer. Thus, the downregulation or inhibition of NUCKS may be a potential therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Erhu Zhao
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400716, China.,Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, 400716, China.,NHC Key Laboratory of Birth Defects and Reproductive Health (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute), Chongqing, 400020, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400716, China
| | - Liying Feng
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400716, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400716, China.,Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400715, China
| | - Longchang Bai
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400716, China.,Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, 400716, China.,Westa College, Southwest University, Chongqing, 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400716, China. .,Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, 400716, China. .,NHC Key Laboratory of Birth Defects and Reproductive Health (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute), Chongqing, 400020, China. .,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400716, China. .,Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400715, China.
| |
Collapse
|
11
|
miR-137: A Novel Therapeutic Target for Human Glioma. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:614-622. [PMID: 32736290 PMCID: PMC7393316 DOI: 10.1016/j.omtn.2020.06.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/18/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
MicroRNA (miR)-137 is highly expressed in the brain and plays a crucial role in the development and prognosis of glioma. In this review, we aim to summarize the latest findings regarding miR-137 in glioma cell apoptosis, proliferation, migration, invasion, angiogenesis, drug resistance, and cancer treatment. In addition, we focus on the identified miR-137 targets and pathways in the occurrence and development of glioma. Finally, future implications for the diagnostic and therapeutic potential of miR-137 in glioma were discussed.
Collapse
|
12
|
Wu L, Wang J, Cai Q, Cavazos TB, Emami NC, Long J, Shu XO, Lu Y, Guo X, Bauer JA, Pasaniuc B, Penney KL, Freedman ML, Kote-Jarai Z, Witte JS, Haiman CA, Eeles RA, Zheng W. Identification of Novel Susceptibility Loci and Genes for Prostate Cancer Risk: A Transcriptome-Wide Association Study in Over 140,000 European Descendants. Cancer Res 2019; 79:3192-3204. [PMID: 31101764 PMCID: PMC6606384 DOI: 10.1158/0008-5472.can-18-3536] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/04/2019] [Accepted: 05/09/2019] [Indexed: 11/16/2022]
Abstract
Genome-wide association study-identified prostate cancer risk variants explain only a relatively small fraction of its familial relative risk, and the genes responsible for many of these identified associations remain unknown. To discover novel prostate cancer genetic loci and possible causal genes at previously identified risk loci, we performed a transcriptome-wide association study in 79,194 cases and 61,112 controls of European ancestry. Using data from the Genotype-Tissue Expression Project, we established genetic models to predict gene expression across the transcriptome for both prostate models and cross-tissue models and evaluated model performance using two independent datasets. We identified significant associations for 137 genes at P < 2.61 × 10-6, a Bonferroni-corrected threshold, including nine genes that remained significant at P < 2.61 × 10-6 after adjusting for all known prostate cancer risk variants in nearby regions. Of the 128 remaining associated genes, 94 have not yet been reported as potential target genes at known loci. We silenced 14 genes and many showed a consistent effect on viability and colony-forming efficiency in three cell lines. Our study provides substantial new information to advance our understanding of prostate cancer genetics and biology. SIGNIFICANCE: This study identifies novel prostate cancer genetic loci and possible causal genes, advancing our understanding of the molecular mechanisms that drive prostate cancer.
Collapse
Affiliation(s)
- Lang Wu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Jifeng Wang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Urology, The Fifth People's Hospital of Shanghai, Shanghai, China
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Taylor B Cavazos
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
| | - Nima C Emami
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yingchang Lu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xingyi Guo
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua A Bauer
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Institute of Chemical Biology, High-Throughput Screening Facility, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - John S Witte
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Christopher A Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, California
| | - Rosalind A Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
13
|
Giunti L, Da Ros M, De Gregorio V, Magi A, Landini S, Mazzinghi B, Buccoliero AM, Genitori L, Giglio S, Sardi I. A microRNA profile of pediatric glioblastoma: The role of NUCKS1 upregulation. Mol Clin Oncol 2019; 10:331-338. [PMID: 30847170 PMCID: PMC6388501 DOI: 10.3892/mco.2019.1795] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are a novel class of gene regulators that may be involved in tumor chemoresistance. Recently, specific miRNA expression profiles have been identified in adult glioblastoma (aGBM), but there are only limited data available on the role of miRNAs in pediatric GBM (pGBM). In the present study, the expression profile of miRNAs was examined in seven pGBMs and three human GBM cell lines (U87MG, A172 and T98G), compared with a non-tumoral pool of pediatric cerebral cortex samples by microarray analysis. A set of differentially expressed miRNAs was identified, including miR-490, miR-876-3p, miR-876-5p, miR-448 and miR-137 (downregulated), as well as miR-501-3p (upregulated). Through bioinformatics analysis, a series of target genes was predicted. In addition, similar gene expression patterns in pGBMs and cell lines was confirmed. Of note, drug resistant T98G cells had upregulated nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) expression, a protein overexpressed in many tumors that serves an important role in cell proliferation and progression. On the basis of the present preliminary report, it could be intriguing to further investigate the relationship between each of the identified differentially expressed miRNAs and NUCKS1, in order to clarify their involvement in the multi-drug resistance mechanism of pGBMs.
Collapse
Affiliation(s)
- Laura Giunti
- Medical Genetics Unit, Meyer Children's University Hospital, I-50139 Florence, Italy
| | - Martina Da Ros
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's University Hospital, I-50139 Florence, Italy
| | - Veronica De Gregorio
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's University Hospital, I-50139 Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, I-50139 Florence, Italy
| | - Samuela Landini
- Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences 'Mario Serio', University of Florence, I-50139 Florence, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, I-50139 Florence, Italy
| | | | - Lorenzo Genitori
- Neurosurgery Unit, Meyer Children's University Hospital, I-50139 Florence, Italy
| | - Sabrina Giglio
- Medical Genetics Unit, Meyer Children's University Hospital, I-50139 Florence, Italy.,Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences 'Mario Serio', University of Florence, I-50139 Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's University Hospital, I-50139 Florence, Italy
| |
Collapse
|
14
|
Huang YK, Kang WM, Ma ZQ, Liu YQ, Zhou L, Yu JC. NUCKS1 promotes gastric cancer cell aggressiveness by upregulating IGF-1R and subsequently activating the PI3K/Akt/mTOR signaling pathway. Carcinogenesis 2018; 40:370-379. [PMID: 30371738 DOI: 10.1093/carcin/bgy142] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/17/2018] [Indexed: 01/13/2023] Open
Affiliation(s)
- Ya-Kai Huang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Wei-Ming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Zhi-Qiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Yu-Qin Liu
- Cell Culture Centre, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Jian-Chun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| |
Collapse
|
15
|
Nucks1 synergizes with Trp53 to promote radiation lymphomagenesis in mice. Oncotarget 2018; 7:61874-61889. [PMID: 27542204 PMCID: PMC5308697 DOI: 10.18632/oncotarget.11297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 12/22/2022] Open
Abstract
NUCKS1 is a 27 kD vertebrate-specific protein, with a role in the DNA damage response. Here, we show that after 4 Gy total-body X-irradiation, Trp53+/− Nucks1+/− mice more rapidly developed tumors, particularly thymic lymphoma (TL), than Trp53+/− mice. TLs in both cohorts showed loss of heterozygosity (LOH) of the Trp53+ allele in essentially all cases. In contrast, LOH of the Nucks1+ allele was rare. Nucks1 expression correlated well with Nucks1 gene dosage in normal thymi, but was increased in the majority of TLs from Trp53+/− Nucks1+/− mice, suggesting that elevated Nucks1 message may be associated with progression towards malignancy in vivo. Trp53+/− Nucks1+/− mice frequently succumbed to CD4- CD8- TLs harboring translocations involving Igh but not Tcra/d, indicating TLs in Trp53+/− Nucks1+/− mice mostly originated prior to the double positive stage and at earlier lineage than TLs in Trp53+/- mice. Monoclonal rearrangements at Tcrb were more prevalent in TLs from Trp53+/− Nucks1+/− mice, as was infiltration of primary TL cells to distant organs (liver, kidney and spleen). We propose that, in the context of Trp53 deficiency, wild type levels of Nucks1 are required to suppress radiation-induced TL, likely through the role of the NUCKS1 protein in the DNA damage response.
Collapse
|
16
|
Roles of NUCKS1 in Diseases: Susceptibility, Potential Biomarker, and Regulatory Mechanisms. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7969068. [PMID: 29619377 PMCID: PMC5830027 DOI: 10.1155/2018/7969068] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/31/2017] [Indexed: 12/16/2022]
Abstract
Nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) is a 27 kD chromosomal, highly conserved, and vertebrate-specific protein. NUCKS1 gene encodes a nuclear protein and the conserved regions of NUCKS1 contain several consensus phosphorylation sites for casein kinase II (CK2) and cyclin-dependent kinases (Cdk) and a basic DNA-binding domain. NUCKS1 is similar to the high mobility group (HMG) family which dominates chromatin remodeling and regulates gene transcription. Meanwhile, NUCKS1 is a RAD51 associated protein 1 (RAD51AP1) paralog that is significant for homologous recombination (HR) and genome stability and also a transcriptional regulator of the insulin signaling components. NUCKS1 plays an important role in DNA damage response and metabolism, participates in inflammatory immune response, and correlates with microRNA. Although there is still not enough functional information on NUCKS1, evidences suggest that NUCKS1 can be used as the biomarker of several cancers. This review summarizes the latest research on NUCKS1 about its susceptibility in diseases, expression levels, and regulatory mechanisms as well as the possible functions in reference to diseases.
Collapse
|
17
|
Clawson GA, Matters GL, Xin P, McGovern C, Wafula E, dePamphilis C, Meckley M, Wong J, Stewart L, D’Jamoos C, Altman N, Imamura Kawasawa Y, Du Z, Honaas L, Abraham T. "Stealth dissemination" of macrophage-tumor cell fusions cultured from blood of patients with pancreatic ductal adenocarcinoma. PLoS One 2017; 12:e0184451. [PMID: 28957348 PMCID: PMC5619717 DOI: 10.1371/journal.pone.0184451] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022] Open
Abstract
Here we describe isolation and characterization of macrophage-tumor cell fusions (MTFs) from the blood of pancreatic ductal adenocarcinoma (PDAC) patients. The MTFs were generally aneuploidy, and immunophenotypic characterizations showed that the MTFs express markers characteristic of PDAC and stem cells, as well as M2-polarized macrophages. Single cell RNASeq analyses showed that the MTFs express many transcripts implicated in cancer progression, LINE1 retrotransposons, and very high levels of several long non-coding transcripts involved in metastasis (such as MALAT1). When cultured MTFs were transplanted orthotopically into mouse pancreas, they grew as obvious well-differentiated islands of cells, but they also disseminated widely throughout multiple tissues in "stealth" fashion. They were found distributed throughout multiple organs at 4, 8, or 12 weeks after transplantation (including liver, spleen, lung), occurring as single cells or small groups of cells, without formation of obvious tumors or any apparent progression over the 4 to 12 week period. We suggest that MTFs form continually during PDAC development, and that they disseminate early in cancer progression, forming "niches" at distant sites for subsequent colonization by metastasis-initiating cells.
Collapse
Affiliation(s)
- Gary A. Clawson
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Gail L. Matters
- Department of Biochemistry & Molecular Biology, HMC, PSU, Hershey, PA, United States of America
| | - Ping Xin
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Christopher McGovern
- Department of Biochemistry & Molecular Biology, HMC, PSU, Hershey, PA, United States of America
| | - Eric Wafula
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Claude dePamphilis
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Morgan Meckley
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Joyce Wong
- Department of Surgery, HMC, PSU, Hershey, PA, United States of America
| | - Luke Stewart
- Applications Support, Fluidigm Corporation, South San Francisco, CA, United States of America
| | - Christopher D’Jamoos
- Applications Support, Fluidigm Corporation, South San Francisco, CA, United States of America
| | - Naomi Altman
- Department of Statistics, Eberly College, UP, PSU, University Park, PA, United States of America
| | - Yuka Imamura Kawasawa
- Department of Pharmacology and Biochemistry & Molecular Biology, Institute for Personalized Medicine, HMC, PSU, Hershey, PA, United States of America
| | - Zhen Du
- Gittlen Cancer Research Laboratories and the Department of Pathology, Hershey Medical Center (HMC), Pennsylvania State University (PSU), Hershey, PA, United States of America
| | - Loren Honaas
- Department of Biology, Eberly College, University Park (UP), Pennsylvania State University, University Park, PA, United States of America
| | - Thomas Abraham
- Department of Neural & Behavioral Sciences and Microscopy Imaging Facility, HMC, PSU, Hershey, PA, United States of America
| |
Collapse
|
18
|
Shi C, Qin L, Gao H, Gu L, Yang C, Liu H, Liu T. NUCKS nuclear elevated expression indicates progression and prognosis of ovarian cancer. Tumour Biol 2017; 39:1010428317714631. [PMID: 28877654 DOI: 10.1177/1010428317714631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
NUCKS (nuclear, casein kinase, and cyclin-dependent kinase substrate) is implicated in the tumorigenesis of several human malignancies, but its role in ovarian cancer remains unknown. We aim to investigate NUCKS expression and its clinical significance in ovarian cancer. The messenger RNA expression of NUCKS was determined in normal and malignant ovarian tissues using quantitative polymerase chain reaction assay. Immunohistochemistry was applied to detect the status of NUCKS protein expression in 121 ovarian cancer tissues. NUCKS protein high expression was detected in 52 (43.0%) of 121 patients. NUCKS messenger RNA expression was gradually upregulated in non-metastatic ovarian cancers ( n = 20), metastatic ovarian cancers ( n = 20), and its matched metastatic lesions ( n = 20) in comparison with that in normal ovarian tissues ( n = 10; p < 0.05). Elevated expression of NUCKS in ovarian cancer was associated significantly with the Federation of Gynecology and Obstetrics stage ( p = 0.037), histological grade ( p = 0.003), residual disease ( p = 0.013), lymph node metastasis ( p = 0.002), response to chemotherapy ( p < 0.001), and recurrence ( p = 0.013). In the multivariate Cox analysis, NUCKS expression was an independent prognostic marker for overall survival and disease-free survival in ovarian cancer with p values of <0.001 for both. Especially, NUCKS overexpression had prognostic potential for overall survival and disease-free survival ( p < 0.001 for both) in advanced ovarian cancers and only for disease-free survival in early ovarian cancers ( p = 0.017). Our data suggest that NUCKS overexpression may contribute to progression and poor prognosis in ovarian cancer especially in advanced ovarian cancer.
Collapse
Affiliation(s)
- Ce Shi
- 1 Department of Leukemia, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ling Qin
- 2 Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongyu Gao
- 3 Department of Gastroenterologic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Gu
- 4 Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chang Yang
- 4 Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hebing Liu
- 5 Department of Biostatistics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tianbo Liu
- 4 Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
19
|
Grundt K, Thiede B, Østvold AC. Identification of kinases phosphorylating 13 sites in the nuclear, DNA-binding protein NUCKS. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:359-369. [PMID: 28011258 DOI: 10.1016/j.bbapap.2016.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/05/2016] [Accepted: 12/19/2016] [Indexed: 12/27/2022]
Abstract
NUCKS is a vertebrate specific, nuclear and DNA-binding phospho protein. The protein is highly expressed in rapidly dividing cells, and is overexpressed in a number of cancer tissues. The phosphorylation of NUCKS is cell cycle and DNA-damage regulated, but little is known about the responsible kinases. By utilizing in vitro and in vivo phosphorylation assays using isolated NUCKS as well as synthetic NUCKS-derived peptides in combination with mass spectrometry, phosphopeptide mapping, phosphphoamino acid analyses, phosphospecific antibodies and the use of specific kinase inhibitors, we found that NUCKS is phosphorylated on 11 sites by CK2. At least 7 of the CK2 sites are phosphorylated in vivo. We also found that NUCKS is phosphorylated on two sites by ATM kinase and DNA-PK in vitro, and is phosphorylated in vivo by ATM kinase in γ-irradiated cells. All together, we identified three kinases phosphorylating 13 out of 39 in vivo phosphorylated sites in mammalian NUCKS. The identification of CK2 and PIKK kinases as kinases phosphorylating NUCKS in vivo provide further evidence for the involvement of NUCKS in cell cycle control and DNA repair.
Collapse
Affiliation(s)
- Kirsten Grundt
- University of Oslo, Institute of Basic Medical Sciences, Department of Biochemistry, P.O. Box 1112, Blindern N-0317, Oslo, Norway
| | - Bernd Thiede
- University of Oslo, Department of Biosciences, P.O. Box 1066, Blindern N-0316, Oslo, Norway
| | - Anne Carine Østvold
- University of Oslo, Institute of Basic Medical Sciences, Department of Biochemistry, P.O. Box 1112, Blindern N-0317, Oslo, Norway.
| |
Collapse
|