1
|
Wang T, Li W, Wu Y, You L, Zheng C, Zhang J, Qu L, Sun X. Construction of a prognostic model based on disulfidptosis-related genes and identification of CCNA2 as a novel biomarker for hepatocellular carcinoma. Biol Direct 2024; 19:128. [PMID: 39695705 DOI: 10.1186/s13062-024-00569-9] [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: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Disulfidptosis, identified as an innovative form of cellular death subsequent to cuproptosis, is currently under investigation for its mechanisms in oncological contexts. In-depth analyses exploring the relationship between disulfidptosis-related genes (DRGs) and hepatocellular carcinoma (HCC) are currently limited. METHODS Transcriptomic data and clinical information were retrieved from the TCGA and GEO databases (GSE76427 and GSE54236), concentrating on the expression levels of 24 DRGs. Subsequently, multifactor and LASSO regression analyses were utilized to construct the 5-DRG prognostic signature. Immunohistochemistry (IHC) was employed to assess Cyclin A2 (CCNA2) protein expression levels. Quantitative real-time PCR (qRT-PCR) and western blot analyses were conducted to detect transcriptomic and protein expression of CCNA2-targeting short interfering RNA (siRNA). The Cell Counting Kit-8 (CCK-8) assay, EdU staining, and scratch experiments were employed to observe the proliferation and migration of hepatoma cell lines subsequent to CCNA2 inhibition. RESULTS Three HCC patterns were identified, among which pattern B exhibited the the most unfavorable survival outcomes. Five DRGs (STC2, PBK, CCNA2, SERPINE1, and SLC6A1) were involved to establish the 5-DRG prognostic signature. High-risk groups (HRGs) exhibited prolonged survival durations in comparison to low-risk groups (LRGs). Both bioinformatics analyses and experimental methodologies corroborated the association of CCNA2 with poor prognosis in HCC patients. Functional studies elucidated that interference with CCNA2 significantly inhibited proliferation and migration, while simultaneously promoting apoptosis in hepatoma cells and resulting in the downregulation of epithelial-mesenchymal transition (EMT)-related protein markers. CONCLUSIONS The 5-DRG prognostic signature is proficient in predicting clinical outcomes, informing therapeutic strategies, and elucidating the characteristics of the immune microenvironment in HCC patients. Furthermore, this study elucidates the potential of CCNA2 as an innovative biomarker for HCC.
Collapse
Affiliation(s)
- Tao Wang
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenxuan Li
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuelan Wu
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Liping You
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao Zheng
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinghao Zhang
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lihong Qu
- Department of Infectious Diseases, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xuehua Sun
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
2
|
Ali F, Iqbal A, Azhar I, Qayyum A, Hassan SA, Hasan MSA, Jawi M, Hassan HM, Al-Emam A, Sajid M. Exploring a novel four-gene system as a diagnostic and prognostic biomarker for triple-negative breast cancer, using clinical variables. Comput Biol Chem 2024; 113:108247. [PMID: 39427606 DOI: 10.1016/j.compbiolchem.2024.108247] [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: 08/03/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. This research aims to find real hub genes for prognostic biomarkers of TNBC therapy. The GEO datasets GSE27447 and GSE233242 were analyzed using R package limma to explore DEGs. The PPI was generated using the STRING database. Cytoscape software plug-ins were used to screen the hub genes. Using the DAVID database, GO functional enrichment and KEGG pathway enrichment analysis were performed. Different online expression databases were employed to investigate the functions of real hub genes in tumor driving, diagnosis, and prognosis in TNBC patients with various clinicopathologic characteristics. A total of one hundred DEGs were identified between both datasets. The seven hub genes were identified after the topological parameter analysis of the PPI network. The KEGG pathway and GO analysis suggest that four genes (PSMB1, PSMC1, PSMF1, and PSMD8) are highly enriched in proteasome and were finally considered as real hub genes. Additionally, the expression analysis demonstrated that hub genes were notably up-regulated in TNBC patients compared to controls. Furthermore, correlational analyses revealed the positive and negative correlations among the expression of the real hub genes and various ancillary data, including tumor purity, promoter methylation status, overall survival (OS), genetic alterations, infiltration of CD8+ T and CD4+ immune cells, and a few more, across TNBC samples. Finally, our analysis identified a couple of significant chemotherapeutic drugs, miRNAs and transcription factors (TFS) with intriguing curative potential. In conclusion, we identified four real hub genes as novel biomarkers to overcome heterogenetic-particular challenges in diagnosis, prognosis, and therapy for TNBC patients.
Collapse
Affiliation(s)
- Faisal Ali
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Azhar Iqbal
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Iqra Azhar
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Adiba Qayyum
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Syed Ali Hassan
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science And Technology University, Gopalgonj, Dhaka 8100, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh.
| | - Motasim Jawi
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia.
| | - Muhammad Sajid
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, Punjab 56300, Pakistan.
| |
Collapse
|
3
|
Zhang J, Che T, Wang L, Sun W, Zhao J, Chen J, Liu Y, Pu Q, Zhang Y, Li J, Li Z, Zhu Z, Fu Q, Wang X, Yuan J. Proteomics coupled transcriptomics reveals Slc34a1 and Slc34a3 downregulation as potential features of nephrotoxin-induced acute kidney injury. J Proteomics 2024; 302:105203. [PMID: 38782357 DOI: 10.1016/j.jprot.2024.105203] [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: 08/07/2023] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Acute kidney injury (AKI) stands as a prevalent and economically burdensome condition worldwide, yet its complex molecular mechanisms remain incompletely understood. To address this gap, our study employs a multifaceted approach, combining mass spectrometry and RNA sequencing technologies, to elucidate the intricate molecular landscape underlying nephrotoxin-induced AKI in mice by cisplatin- and LPS-induced. By examining the protein and RNA expression profiles, we aimed to uncover novel insights into the pathogenesis of AKI and identify potential diagnostic and therapeutic targets. Our results demonstrate significant down-regulation of Slc34a1 and Slc34a3, shedding light on their crucial roles in AKI pathology and highlighting their promise as actionable targets for diagnosis and treatment. This comprehensive analysis not only enhances our understanding of AKI pathophysiology but also offers valuable avenues for the development of targeted interventions to mitigate its clinical impact. SIGNIFICANCE: Nephrotoxicity acute kidney injury (AKI) is a common clinical condition whose pathogenesis is the process by which some drugs, chemicals or other factors cause damage to the kidneys, resulting in impaired kidney function. Although it has been proved that different nephrotoxic substances can affect the kidney through different pathways, whether they have a commonality has not been registered. Here, we combined transcriptomics and proteomics to study the molecular mechanism of LPS and cisplatin-induced nephrotoxic acute kidney injury finding that the down-regulation of Slc34a1 and Slc34a3 may be a critical link in nephrotoxic acute kidney injury, which can be used as a marker for its early diagnosis.
Collapse
Affiliation(s)
- Junying Zhang
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Che
- Chongqing Nanan District Center for Diseases Control and Prevention, Chongqing 401336, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Qi Pu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yu Zhang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiani Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhangfu Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China
| | - Zhaojing Zhu
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Qihuan Fu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China..
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China.; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
4
|
Zhang XY, Li Y. PHD-BAH Domain in ASH1L Could Recognize H3K4 Methylation and Regulate the Malignant Behavior of Cholangiocarcinoma. Anticancer Agents Med Chem 2024; 24:1264-1274. [PMID: 39034728 DOI: 10.2174/0118715206312004240712072532] [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: 04/03/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Histone methyltransferase absent, small, or homeotic discs1-like (ASH1L) is composed of su(var)3-9, enhancer of zeste, trithorax (SET) domain, pleckstrin homology domain (PHD) domain, middle (MID) domain, and bromo adjacent homology (BAH) domain. The SET domain of ASH1L is known to mediate mediate H3K36 dimethylation (H3K36me2) modification. However, the specific functions of the PHD-BAH domain remain largely unexplored. This study aimed to explore the biological function of the PHD-BAH domain in ASH1L. METHODS We employed a range of techniques, including a prokaryotic fusion protein expression purification system, pull-down assay, Isothermal Titration Calorimetry (ITC), polymerase chain reaction (PCR), and sitedirected mutagenesis, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR-Cas9) gene editing, cell culture experiment, western blot, cell proliferation assay, and cell apoptosis test. RESULTS The PHD-BAH domain in ASH1L preferentially binds to the H3K4me2 peptide over H3K4 monomethylation (H3K4me1) and H3K4 trimethylation (H3K4me3) peptide. Notably, the W2603A mutation within the PHD-BAH domain could disrupt the interaction with H3K4me2 in vitro. Compared with wild-type Cholangiocarcinoma (CHOL) cells, deletion of the PHD-BAH domain in ASH1L led to increased CHOL cell apoptosis and reduced cell proliferation (P < 0.001). Additionally, the W2603A mutation affected the regulation of the proteasome 20S subunit beta (PSMB) family gene set. CONCLUSION W2603A mutation was crucial for the interaction between the PHD-BAH domain and the H3K4me2 peptide. ASH1L regulated CHOL cell survival and proliferation through its PHD-BAH domain by modulating the expression of the PSMB family gene set.
Collapse
Affiliation(s)
- Xiang-Yu Zhang
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yue Li
- External Cooperation Liaison Office, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, China
| |
Collapse
|
5
|
Zhong Y, Yu F, Yang L, Wang Y, Liu L, Jia C, Cai H, Yang J, Sheng S, Lv Z, Weng L, Wu B, Zhang X. HOXD9/miR-451a/PSMB8 axis is implicated in the regulation of cell proliferation and metastasis via PI3K/AKT signaling pathway in human anaplastic thyroid carcinoma. J Transl Med 2023; 21:817. [PMID: 37974228 PMCID: PMC10652604 DOI: 10.1186/s12967-023-04538-0] [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: 06/25/2023] [Accepted: 09/17/2023] [Indexed: 11/19/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a deadly disease with a poor prognosis. Thus, there is a pressing need to determine the mechanism of ATC progression. The homeobox D9 (HOXD9) transcription factor has been associated with numerous malignancies but its role in ATC is unclear. In the present study, the carcinogenic potential of HOXD9 in ATC was investigated. We assessed the differential expression of HOXD9 on cell proliferation, migration, invasion, apoptosis, and epithelial-mesenchymal transition (EMT) in ATC and explored the interactions between HOXD9, microRNA-451a (miR-451a), and proteasome 20S subunit beta 8 (PSMB8). In addition, subcutaneous tumorigenesis and lung metastasis in mouse models were established to investigate the role of HOXD9 in ATC progression and metastasis in vivo. HOXD9 expression was enhanced in ATC tissues and cells. Knockdown of HOXD9 inhibited cell proliferation, migration, invasion, and EMT but increased apoptosis in ATC cells. The UCSC Genome Browser and JASPAR database identified HOXD9 as an upstream regulator of miR-451a. The direct binding of miR-451a to the untranslated region (3'-UTR) of PSMB8 was established using a luciferase experiment. Blocking or activation of PI3K by LY294002 or 740Y-P could attenuate the effect of HOXD9 interference or overexpression on ATC progression. The PI3K/AKT signaling pathway was involved in HOXD9-stimulated ATC cell proliferation and EMT. Consistent with in vitro findings, the downregulation of HOXD9 in ATC cells impeded tumor growth and lung metastasis in vivo. Our research suggests that through PI3K/AKT signaling, the HOXD9/miR-451a/PSMB8 axis may have significance in the control of cell proliferation and metastasis in ATC. Thus, HOXD9 could serve as a potential target for the diagnosis of ATC.
Collapse
Affiliation(s)
- Yong Zhong
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Fan Yu
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Ling Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin Liu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Chengyou Jia
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Haidong Cai
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Jianshe Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Shiyang Sheng
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University and Shanghai Center of Thyroid Diseases, No. 301 Middle Yanchang Road, Shanghai, 200072, China.
| | - Li Weng
- Department of Intervention, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Bo Wu
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
- Center of Thyroid, Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Xiaoping Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
6
|
Monittola F, Bianchi M, Nasoni MG, Luchetti F, Magnani M, Crinelli R. Gastric cancer cell types display distinct proteasome/immunoproteasome patterns associated with migration and resistance to proteasome inhibitors. J Cancer Res Clin Oncol 2023; 149:10085-10097. [PMID: 37261527 PMCID: PMC10423134 DOI: 10.1007/s00432-023-04948-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE Gastric cancers (GC) display histological and molecular differences. This heterogeneity has limited the development of new therapeutic strategies which requires the identification of the molecular players involved in GC pathogenesis and the investigation of their responsiveness to drugs. Several proteasome subunits have been identified as prognostic markers in GC and their role studied by gene knockdown. However, proteasomes are multi-subunit protein complexes co-existing in multiple forms with distinct activity/specificity and ability to change in response to inhibitors. Information on the role of different proteasome particles in cancer and their relevance as therapeutic targets is limited. METHODS Based on this evidence, subunit assembly into proteasome complexes and activity were investigated by native PAGE followed by immunoblotting, and by using fluorogenic substrates, respectively. RESULTS Here we show that GC cell lines with epithelial and/or diffuse Lauren's histotype express different levels of immunoproteasome subunits and equal amounts of constitutive counterparts. Immunoproteasome subunits were highly expressed and preferentially assembled into 19S capped complexes in diffuse-type cells, where most of the activity was catalyzed by the 26S and 30S particles. In epithelial cells, activity appeared equally distributed between 19S- and 11S-capped proteolytic particles. This proteasome pattern was associated with higher resistance of diffuse-type cells to proteasome inhibition. Immunoproteasome inhibition by ONX 0914 did not influence cell viability but affected metastatic cell migration. CONCLUSIONS These results suggest that pharmacological inhibition of the immunoproteasome may be useful in treating metastatic gastric cancers.
Collapse
Affiliation(s)
- Francesca Monittola
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Marzia Bianchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy.
| |
Collapse
|
7
|
Fan S, Liu Y, Lin Z, Zhang Y, Zhang N, Zhao Y, Zhou J, Mao A, Wang L, Feng Y, He X, Wang L, Pan Q. ZNF655 promotes the progression of hepatocellular carcinoma through PSMB8. Cell Biol Int 2023; 47:1535-1546. [PMID: 37272200 DOI: 10.1002/cbin.12050] [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: 06/20/2022] [Revised: 04/18/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a type of liver cancer that is associated with high mortality rates. This study aims to investigate the role of ZNF655, a member of the zinc finger protein family, in the development of HCC. Immunohistochemical staining analysis was conducted to evaluate the expression of ZNF655 in HCC patient samples. Lentivirus-mediated ZNF655 knockdown was established in HCC cell lines (BEL-7402 and HCCLM3). The effects of ZNF655 on different aspects of HCC cell behavior such as proliferation, apoptosis, cycle, migration and tumor formation were examined. Downstream targets of ZNF655 in HCC were identified and verified through loss/gain-of-function experiments. Clinically, ZNF655 expression was elevated in HCC and increased with the severity of the disease. Functionally, inhibition of ZNF655 expression reduced the progression of HCC cells by decreasing proliferation, causing apoptosis, arresting cell cycle retention in G2, suppressing migration, and attenuating tumor formation in mice. Mechanistically, the proteasome subunit beta type-8 (PSMB8) was found to be co-expressed with ZNF655 in HCC, and PSMB8 knockdown weakened the promotion of ZNF655 overexpression on HCC. In summary, these findings suggest that ZNF655 promotes the progression of HCC through PSMB8, and inhibition of its expression may be a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Shasha Fan
- Department of Oncology, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, Hunan, China
| | - Yu Liu
- Department of Pathology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Zhenhai Lin
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongfa Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiming Zhao
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaming Zhou
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anrong Mao
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Longrong Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Feng
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xigan He
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Pan
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Mottaghi-Dastjerdi N, Ghorbani A, Montazeri H, Guzzi PH. A systems biology approach to pathogenesis of gastric cancer: gene network modeling and pathway analysis. BMC Gastroenterol 2023; 23:248. [PMID: 37482618 PMCID: PMC10364406 DOI: 10.1186/s12876-023-02891-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks among the most common malignancies worldwide. This study aimed to find critical genes/pathways in GC pathogenesis. METHODS Gene interactions were analyzed, and the protein-protein interaction network was drawn. Then enrichment analysis of the hub genes was performed and network cluster analysis and promoter analysis of the hub genes were done. Age/sex analysis was done on the identified genes. RESULTS Eleven hub genes in GC were identified in the current study (ATP5A1, ATP5B, ATP5D, MT-ATP8, COX7A2, COX6C, ND4, ND6, NDUFS3, RPL8, and RPS16), mostly involved in mitochondrial functions. There was no report on the ATP5D, ND6, NDUFS3, RPL8, and RPS16 in GC. Our results showed that the most affected processes in GC are the metabolic processes, and the oxidative phosphorylation pathway was considerably enriched which showed the significance of mitochondria in GC pathogenesis. Most of the affected pathways in GC were also involved in neurodegenerative diseases. Promoter analysis showed that negative regulation of signal transduction might play an important role in GC pathogenesis. In the analysis of the basal expression pattern of the selected genes whose basal expression presented a change during the age, we found that a change in age may be an indicator of changes in disease insurgence and/or progression at different ages. CONCLUSIONS These results might open up new insights into GC pathogenesis. The identified genes might be novel diagnostic/prognostic biomarkers or potential therapeutic targets for GC. This work, being based on bioinformatics analysis act as a hypothesis generator that requires further clinical validation.
Collapse
Affiliation(s)
- Negar Mottaghi-Dastjerdi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran.
| | - Abozar Ghorbani
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute (NSTRI), Karaj, Iran.
| | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Pietro Hiram Guzzi
- Department of Surgical and Medical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
9
|
Ajay AK, Chu P, Patel P, Deban C, Roychowdhury C, Heda R, Halawi A, Saad A, Younis N, Zhang H, Jiang X, Nasr M, Hsiao LL, Lin G, Azzi JR. High-Throughput/High Content Imaging Screen Identifies Novel Small Molecule Inhibitors and Immunoproteasomes as Therapeutic Targets for Chordoma. Pharmaceutics 2023; 15:1274. [PMID: 37111759 PMCID: PMC10145398 DOI: 10.3390/pharmaceutics15041274] [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: 02/06/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Chordomas account for approximately 1-4% of all malignant bone tumors and 20% of primary tumors of the spinal column. It is a rare disease, with an incidence estimated to be approximately 1 per 1,000,000 people. The underlying causative mechanism of chordoma is unknown, which makes it challenging to treat. Chordomas have been linked to the T-box transcription factor T (TBXT) gene located on chromosome 6. The TBXT gene encodes a protein transcription factor TBXT, or brachyury homolog. Currently, there is no approved targeted therapy for chordoma. Here, we performed a small molecule screening to identify small chemical molecules and therapeutic targets for treating chordoma. We screened 3730 unique compounds and selected 50 potential hits. The top three hits were Ribociclib, Ingenol-3-angelate, and Duvelisib. Among the top 10 hits, we found a novel class of small molecules, including proteasomal inhibitors, as promising molecules that reduce the proliferation of human chordoma cells. Furthermore, we discovered that proteasomal subunits PSMB5 and PSMB8 are increased in human chordoma cell lines U-CH1 and U-CH2, confirming that the proteasome may serve as a molecular target whose specific inhibition may lead to better therapeutic strategies for chordoma.
Collapse
Affiliation(s)
- Amrendra K. Ajay
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Philip Chu
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
| | - Poojan Patel
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Christa Deban
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
| | - Chitran Roychowdhury
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
| | - Radhika Heda
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
| | - Ahmad Halawi
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Anis Saad
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nour Younis
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xiuju Jiang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mahmoud Nasr
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Li-Li Hsiao
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Lin
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jamil R. Azzi
- Transplant Research Centre, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA (R.H.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Li D, Li X, Li S, Qi M, Sun X, Hu G. Relationship between the deep features of the full-scan pathological map of mucinous gastric carcinoma and related genes based on deep learning. Heliyon 2023; 9:e14374. [PMID: 36942252 PMCID: PMC10023952 DOI: 10.1016/j.heliyon.2023.e14374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Background Long-term differential expression of disease-associated genes is a crucial driver of pathological changes in mucinous gastric carcinoma. Therefore, there should be a correlation between depth features extracted from pathology-based full-scan images using deep learning and disease-associated gene expression. This study tried to provides preliminary evidence that long-term differentially expressed (disease-associated) genes lead to subtle changes in disease pathology by exploring their correlation, and offer a new ideas for precise analysis of pathomics and combined analysis of pathomics and genomics. Methods Full pathological scans, gene sequencing data, and clinical data of patients with mucinous gastric carcinoma were downloaded from TCGA data. The VGG-16 network architecture was used to construct a binary classification model to explore the potential of VGG-16 applications and extract the deep features of the pathology-based full-scan map. Differential gene expression analysis was performed and a protein-protein interaction network was constructed to screen disease-related core genes. Differential, Lasso regression, and extensive correlation analyses were used to screen for valuable deep features. Finally, a correlation analysis was used to determine whether there was a correlation between valuable deep features and disease-related core genes. Result The accuracy of the binary classification model was 0.775 ± 0.129. A total of 24 disease-related core genes were screened, including ASPM, AURKA, AURKB, BUB1, BUB1B, CCNA2, CCNB1, CCNB2, CDCA8, CDK1, CENPF, DLGAP5, KIF11, KIF20A, KIF2C, KIF4A, MELK, PBK, RRM2, TOP2A, TPX2, TTK, UBE2C, and ZWINT. In addition, differential, Lasso regression, and extensive correlation analyses were used to screen eight valuable deep features, including features 51, 106, 109, 118, 257, 282, 326, and 487. Finally, the results of the correlation analysis suggested that valuable deep features were either positively or negatively correlated with core gene expression. Conclusion The preliminary results of this study support our hypotheses. Deep learning may be an important bridge for the joint analysis of pathomics and genomics and provides preliminary evidence for long-term abnormal expression of genes leading to subtle changes in pathology.
Collapse
Affiliation(s)
- Ding Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyuan Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shifang Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mengmeng Qi
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaowei Sun
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guojie Hu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
11
|
Alsadat Mahmoudian R, Amirhosein M, Mahmoudian P, Fardi Golyan F, Mokhlessi L, Maftooh M, Khazaei M, Nassiri M, Mahdi Hassanian S, Ghayour-Mobarhan M, Ferns GA, Shahidsales S, Avan A. The therapeutic potential value of Cancer-testis antigens in immunotherapy of gastric cancer. Gene 2023; 853:147082. [PMID: 36464170 DOI: 10.1016/j.gene.2022.147082] [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: 09/27/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Gastric cancer (GC) is the fourth most common cause of mortality and the fifth for incidence, globally. Diagnosis, early prognosis, and therapy remains challenging for this condition, and new tumor-associated antigens are required for its detection and immunotherapy. Cancer-testis antigens (CTAs) are a subfamily of tumor-associated antigens (TAAs) that have been identified as potential biomarkers and targets for cancer immunotherapy. The CTAs-restricted expression pattern in tumor cells and their potential immunogenicity identify them as attractive target candidates in CTA-based diagnosis or prognosis or immunotherapy. To date, numerous studies have reported the dysregulation of CTAs in GC. Several clinical trials have been done to assess CTA-based immunotherapeutic potential in the treatment of GC patients. NY-ESO-1, MAGE, and KK-LC-1 have been used in GC clinical trials. We review recent studies that have investigated the potential of the CTAs in GC regarding the expression, function, aggressive phenotype, prognosis, and immunological responses as well as their possible clinical significance as immunotherapeutic targets with a focus on challenges and future interventions.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Maharati Amirhosein
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leila Mokhlessi
- Centre for Biomedical Education and Research, Institute of Pharmacology and Toxicology, Witten/Herdecke University, Witten, Germany.
| | - Mina Maftooh
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Ghayour-Mobarhan
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK.
| | | | - Amir Avan
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Identification of Anoikis-Related Subgroups and Prognosis Model in Liver Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24032862. [PMID: 36769187 PMCID: PMC9918018 DOI: 10.3390/ijms24032862] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 02/05/2023] Open
Abstract
Resistance to anoikis is a key characteristic of many cancer cells, promoting cell survival. However, the mechanism of anoikis in hepatocellular carcinoma (HCC) remains unknown. In this study, we applied differentially expressed overlapping anoikis-related genes to classify The Cancer Genome Atlas (TCGA) samples using an unsupervised cluster algorithm. Then, we employed weighted gene coexpression network analysis (WGCNA) to identify highly correlated genes and constructed a prognostic risk model based on univariate Cox proportional hazards regression. This model was validated using external datasets from the International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO). Finally, we used a CIBERSORT algorithm to investigate the correlation between risk score and immune infiltration. Our results showed that the TCGA cohorts could be divided into two subgroups, with subgroup A having a lower survival probability. Five genes (BAK1, SPP1, BSG, PBK and DAP3) were identified as anoikis-related prognostic genes. Moreover, the prognostic risk model effectively predicted overall survival, which was validated using ICGC and GEO datasets. In addition, there was a strong correlation between infiltrating immune cells and prognostic genes and risk score. In conclusion, we identified anoikis-related subgroups and prognostic genes in HCC, which could be significant for understanding the molecular mechanisms and treatment of HCC.
Collapse
|
13
|
The dichotomous role of immunoproteasome in cancer: Friend or foe? Acta Pharm Sin B 2022; 13:1976-1989. [DOI: 10.1016/j.apsb.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022] Open
|
14
|
Liu R, Liu R, Guo Z, Ren J, Huang J, Luo Q, Tan Q. shRNA‑mediated knockdown of KNTC1 inhibits non-small-cell lung cancer through regulating PSMB8. Cell Death Dis 2022; 13:685. [PMID: 35933405 PMCID: PMC9357013 DOI: 10.1038/s41419-022-05140-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/21/2023]
Abstract
In view of the important roles played by Kinetochore proteins in mitosis, we believed that they may contribute to the development and progression of human cancers, which has been reported recently elsewhere. Kinetochore-associated 1 (KNTC1) participates in the segregation of sister chromatids during mitosis, the effects of which on non-small-cell lung cancer (NSCLC) remain unclear. Here, we sought to identify the biological significance of KNTC1 in NSCLC. KNTC1 protein expression in NSCLC tissues was investigated by immunohistochemistry. Lentivirus delivered short hairpin RNA (shRNA) was utilized to establish KNTC1 silence NSCLC cell lines. The effects of KNTC1 depletion on NSCLC cell proliferation, migration, apoptosis, and tumor formation were analyzed by MTT assay, wound-healing assay, transwell assay, flow cytometry assay, and in nude mouse models in vivo. After KNTC1 reduction, NSCLC cell viability, proliferation, migration, and invasion were restrained. A xenograft tumor model was also provided to demonstrate the inhibited tumorigenesis in NSCLC. In addition, the downstream mechanism analysis indicated that KNTC1 depletion was positively associated with PSMB8. The findings of the present study suggested that KNTC1 may have a pivotal role in mediating NSCLC progression and may act as a novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Ruijun Liu
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Ruili Liu
- Department of Stomatology, Ordos central hospital, Ordos, Inner Mongolia, 017000, P. R. China
| | - Zhiyi Guo
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Jianghao Ren
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Jia Huang
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Qingquan Luo
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| | - Qiang Tan
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| |
Collapse
|
15
|
Sereda EE, Kolegova ES, Kakurina GV, Korshunov DA, Sidenko EA, Doroshenko AV, Slonimskaya EM, Kondakova IV. Five-year survival in luminal breast cancer patients: relation with intratumoral activity of proteasomes. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2022; 3:23. [PMID: 38751528 PMCID: PMC11093047 DOI: 10.21037/tbcr-22-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/14/2022] [Indexed: 05/18/2024]
Abstract
Background The purpose of the study was to analyze the relationship between the caspase-like (CL) and chymotrypsin-like (ChTL) activities of proteasomes and the 5-year overall and metastasis-free survival rates in patients with luminal breast cancer. Methods The study included 117 patients with primary operable invasive breast cancer (T1-2N0-1M0). Tissue samples from breast cancer patients were obtained as a result of the radical mastectomy or breast conserving surgery, which was a first line of therapy. The ChTL and CL proteasomes activities in the tumor tissue and in the surrounding adjacent breast tissues were assessed using the fluorometric method. The coefficients of ChTL (cChTL) and CL (cCL) proteasomes activities were also determined. The coefficients were calculated as the ratio of the corresponding proteasomes activity in the tumor tissue to the surrounding adjacent breast tissues. Within 5 years of follow-up, hematogenous metastases occurred in 14% of patients with luminal A breast cancer, in 31% of patients with luminal B human epidermal growth factor receptor-2 (HER-2) negative and in 23% of patients with luminal B HER-2 positive breast cancers. The study protocol was approved by the Local Ethics Committee of the Cancer Research Institute of Tomsk National Research Medical Center. Written informed consent was obtained from all patients. Results An increase in the ChTL and CL proteasomes activities was shown in all studied molecular subtypes of breast cancer compared to adjacent tissues. It was found that the cChTL of >35.9 U/mg protein and the cCL of >2.21 in breast cancer patients were associated with the development of distant metastases. In patients with luminal A breast cancer, the 5-year metastasis-free survival rates were associated only with the value of cCL of proteasomes (log-rank test: P=0.008). In patients with luminal B HER-2 negative breast cancer, the 5-year metastasis-free survival rates were associated with the levels of ChTL and cCL proteasomes activities (log-rank test: P=0.02 and P=0.04, respectively). Conclusions The data obtained on the correlation of 5-year metastasis-free survival rates with the level of proteasomes activities indicate the possibility of their use as additional prognostic criteria for breast cancer.
Collapse
Affiliation(s)
- Elena E. Sereda
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Biology, Siberian State Medical University, Tomsk, Russia
| | - Elena. S. Kolegova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Gelena V. Kakurina
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Biology, Siberian State Medical University, Tomsk, Russia
| | - Dmitriy A. Korshunov
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Evgenia A. Sidenko
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Artem V. Doroshenko
- General Oncology Department, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Elena M. Slonimskaya
- General Oncology Department, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Irina V. Kondakova
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
16
|
PSMB2 knockdown suppressed proteasome activity and cell proliferation, promoted apoptosis, and blocked NRF1 activation in gastric cancer cells. Cytotechnology 2022; 74:491-502. [PMID: 36110152 PMCID: PMC9374866 DOI: 10.1007/s10616-022-00538-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/18/2022] [Indexed: 11/03/2022] Open
Abstract
Proteasome 20S Subunit Beta 2 (PSMB2) has been suggested to play several roles in cancer. However, the role of PSMB2 and its underlying mechanisms in gastric cancer have not been studied. In this study, qRT-PCR was employed to detect the expression of genes that encode for 26 s proteasome subunit proteins. PSMB2 expression and its prognostic ability were assessed by collecting patient tissue samples and reviewing the TCGA and Kaplan-Meier Plotter databases. Immunofluorescence and western blotting experiments were performed to evaluate the expression of PSMB2 in human gastric cancer cells and normal gastric epithelial cells. Subsequently, PSMB2 was knocked down in HGC-27 and SNU-1 cells and overexpressed in N-87 and AGS cells. Proteasome activity assays, 5-Ethynyl-2'-deoxyuridine staining, and TUNEL assays were used to assess proteasome activity, cell proliferation, and apoptosis. Tumor xenograft assays were conducted to evaluate PSMB2 function in vivo. Our results showed that a total of 8 genes encoding for the 26 s proteasome subunit protein were highly expressed in a variety of gastric cancer cells. Next, PSMB2 was selected as the focus of subsequent studies which showed that PSMB2 was highly expressed in samples of gastric cancer tissue. Furthermore, a review of the TCGA database revealed that a high level of PSMB2 expression was associated with a poor clinical prognosis. Our results indicated that PSMB2 overexpression promoted proteasome activity, cell proliferation, and suppressed the apoptosis of gastric cancer cells, while those effects were reversed by treatment with a proteasome inhibitor (MG132). In contrast, PSMB2 knockdown produced the opposite effects and also blocked NRF1 activation. Moreover, PSMB2 knockdown inhibited tumor growth in vivo, decreased PSMB2 expression and cell proliferation, and promoted apoptosis in tumor tissues. Our findings revealed the role played by PSMB2 in gastric cancer and suggest PSMB2 as a new target molecule for use in diagnosing and treating gastric cancer.
Collapse
|
17
|
Cao J, Zhang R, Zhang Y, Wang Y. Combined screening analysis of aberrantly methylated-differentially expressed genes and pathways in hepatocellular carcinoma. J Gastrointest Oncol 2022; 13:311-325. [PMID: 35284134 PMCID: PMC8899745 DOI: 10.21037/jgo-21-866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Methylation plays an important role in hepatocellular carcinoma (HCC) by altering the expression of key genes. The aim of this study was to screen the aberrantly methylated-differentially expressed genes (DEGs) in HCC and elucidate their underlying molecular mechanism. METHODS Gene expression microarrays (GSE101685) and gene methylation microarrays (GSE44909) were selected. DEGs and differentially methylated genes (DMGs) were screened. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the Database for Annotation, Visualization, and Integrated discovery (DAVID). The Search Tool for the Retrieval of Interacting Genes (STRING) database was used to analyze the functional protein-protein interaction (PPI) network. Molecular Complex Detection (MCODE) analysis was performed using the Cytoscape software. Hub genes were verified in The Cancer Genome Atlas (TCGA) database. RESULTS A total of 80 hypomethylation-high expression genes (Hypo-HGs) were identified. Pathway enrichment analysis showed DNA replication, cell cycle, viral carcinogenesis, and the spliceosome. The top 5 hub genes were minichromosome maintenance complex component 3 (MCM3), checkpoint kinase 1 (CHEK1), kinesin family member 11 (KIF11), PDZ binding kinase (PBK), and Rac GTPase activating protein 1 (RACGAP1). In addition, 189 hypermethylation-low expression genes (Hyper-LGs) were identified. Pathway enrichment analysis indicated enrichment in metabolic pathways, drug metabolism-other enzymes, and chemical carcinogenesis. The top 5 hub genes were leukocyte immunoglobulin like receptor B2 (LILRB2), formyl peptide receptor 1 (FPR1), S100 calcium binding protein A9 (S100A9), S100 calcium binding protein A8 (S100A8), and myeloid cell nuclear differentiation antigen (MNDA). The methylation status and mRNA expression of MCM3, CHEK1, KIF11, PBK, and S100A9 were consistent in the TCGA database and significantly correlated with the prognosis of patients. CONCLUSIONS Combined screening of aberrantly methylated-DEGs based on bioinformatic analysis may provide new clues for elucidating the epigenetic mechanism in HCC. Hub genes, including MCM3, CHEK1, KIF11, PBK, and S100A9, may serve as biomarkers for the precise diagnosis of HCC.
Collapse
Affiliation(s)
- Jisen Cao
- Department of Hepatobiliary Surgery, The Third Central Hospital of Tianjin, Tianjin, China
| | - Ruiqiang Zhang
- Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, China
| | - Ye Zhang
- Department of Hepatobiliary Surgery, The Third Central Hospital of Tianjin, Tianjin, China
| | - Yijun Wang
- Department of Hepatobiliary Surgery, The Third Central Hospital of Tianjin, Tianjin, China
| |
Collapse
|
18
|
Tian W, Chen K, Yan G, Han X, Liu Y, Zhang Q, Liu M. A Novel Prognostic Tool for Glioma Based on Enhancer RNA-Regulated Immune Genes. Front Cell Dev Biol 2022; 9:798445. [PMID: 35127714 PMCID: PMC8811171 DOI: 10.3389/fcell.2021.798445] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Gliomas are the most malignant tumors of the nervous system. Even though their survival outcome is closely affected by immune-related genes (IRGs) in the tumor microenvironment (TME), the corresponding regulatory mechanism remains poorly characterized. Methods: Specific enhancer RNAs (eRNAs) can be found in tumors, where they control downstream genes. The present study aimed to identify eRNA-regulated IRGs, evaluate their influence on the TME, and use them to construct a novel prognostic model for gliomas. Results: Thirteen target genes (ADCYAP1R1, BMP2, BMPR1A, CD4, DDX17, ELN, FGF13, MAPT, PDIA2, PSMB8, PTPN6, SEMA6C, and SSTR5) were identified and integrated into a comprehensive risk signature, which distinguished two risk subclasses. Discrepancies between these subclasses were compared to explore potential mechanisms attributed to eRNA-regulated genes, including immune cell infiltration, clinicopathological features, survival outcomes, and chemotherapeutic drug sensitivity. Furthermore, the risk signature was used to construct a prognostic tool that was evaluated by calibration curve, clinical utility, Harrell’s concordance index (0.87; 95% CI: 0.84–0.90), and time-dependent receiver operator characteristic curves (AUCs: 0.93 and 0.89 at 3 and 5 years, respectively). The strong reliability and robustness of the established prognostic tool were validated in another independent cohort. Finally, potential subtypes were explored in patients with grade III tumors. Conclusion: Overall, eRNAs were associated with immune-related dysfunctions in the TME. Targeting of IRGs regulated by eRNAs could improve immunotherapeutic/therapeutic outcomes.
Collapse
Affiliation(s)
- Wei Tian
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Kegong Chen
- Department of Cardio-Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangcan Yan
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Xinhao Han
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Yanlong Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiuju Zhang
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Meina Liu
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Koshino A, Nagano A, Ota A, Hyodo T, Ueki A, Komura M, Sugimura-Nagata A, Ebi M, Ogasawara N, Kasai K, Hosokawa Y, Kasugai K, Takahashi S, Inaguma S. PBK Enhances Cellular Proliferation With Histone H3 Phosphorylation and Suppresses Migration and Invasion With CDH1 Stabilization in Colorectal Cancer. Front Pharmacol 2022; 12:772926. [PMID: 35115926 PMCID: PMC8804381 DOI: 10.3389/fphar.2021.772926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequent gastrointestinal malignancies with high morbidity and mortality rates. Several biological markers for the prognostication of patient outcome of CRCs are available. Recently, our group identified two favorable factors for the survival of CRC patients: PDZ-binding kinase (PBK) and phospho-histone H3 (PHH3). Both showed a significant inverse association to pT stage. The aim of this study was to uncover the mechanism through which these cellular proliferation–associated protein expressions lead to favorable clinical outcome in CRC patients. We first confirmed co-expression of PBK and PHH3 in CRC cells. Further investigation showed that aberrantly expressed PBK up-regulated the cellular proliferation of CRC cells with accumulation of PHH3. The PBK inhibitor OTS514 suppressed cellular proliferation of CRC cells through down-regulation of PHH3 and induction of apoptosis. In vitro studies revealed that PBK suppressed the migration and invasion of CRC cells with suppression of Wnt/β-catenin signaling and CDH1 stabilization. Exogeneous PBK up-regulated the phosphorylated CDH1 at S840, S846, and S847 residues in cultured cells. Recombinant PBK directly phosphorylated HH3; however, it failed to phosphorylate CDH1 directly in vitro. The present study demonstrated the association of two markers PBK and PHH3 in CRC. We further identified one of the potential mechanisms by which higher expression of these cellular proliferation–associated proteins leads to the better survival of CRC patients, which likely involves PBK-mediated suppression of the migration and invasion of CRC cells. Our findings suggest that PBK-targeting therapeutics may be useful for the treatment of CRC patients with PBK-expressing tumors.
Collapse
Affiliation(s)
- Akira Koshino
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Aya Nagano
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akane Ueki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masayuki Komura
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akane Sugimura-Nagata
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Masahide Ebi
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Naotaka Ogasawara
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kunio Kasugai
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shingo Inaguma
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
- Department of Pathology, Nagoya City University East Medical Center, Nagoya, Japan
- *Correspondence: Shingo Inaguma,
| |
Collapse
|
20
|
Guo JY, Jing ZQ, Li XJ, Liu LY. Bioinformatic Analysis Identifying PSMB 1/2/3/4/6/8/9/10 as Prognostic Indicators in Clear Cell Renal Cell Carcinoma. Int J Med Sci 2022; 19:796-812. [PMID: 35693739 PMCID: PMC9149646 DOI: 10.7150/ijms.71152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022] Open
Abstract
Renal cancer incidence has been increasing across the world, clear cell renal cell carcinoma (ccRCC) represents the major subtype of renal cancer. The proteasome is involved in onset, metabolism and survival of tumor and has been recognized as a therapeutic target for various malignancies, while the role of β subunits of proteasome, PSMB gene family, in ccRCC has not been fully unveiled. Herein we investigated the expression and the prognostic role of PSMBs in ccRCC by analyzing a series of databases, including ONCOMINE, UALCAN, cBioPortal, STRING, GEPIA, GO and KEGG. Over-expressions of PSMB1/2/4/7/8/9/10 mRNA were found in ccRCC tissues compared to normal tissues, transcriptional levels of PSMB2/3/4/6/8/9/10 were significantly positively associated with patients' individual cancer stages and grades. Similar or higher levels of proteins encoded by PSMB1/2/3/7/8/9/10 were observed in tumor tissues compared to normal renal tissues. Further, high mRNA levels of PSMB1/2/3/4/6/10 were correlated with shorter overall survival in univariate analysis. Taken together, the results of our analysis implied that overexpression of PSMB1/2/3/4/6/8/9/10 were indicative of worse prognosis of ccRCC. However, further researches were required to validate our findings.
Collapse
Affiliation(s)
- Jing-Yi Guo
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Zuo-Qian Jing
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xue-Jie Li
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Li-Yuan Liu
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
21
|
Sauerer T, Lischer C, Weich A, Berking C, Vera J, Dörrie J. Single-Molecule RNA Sequencing Reveals IFNγ-Induced Differential Expression of Immune Escape Genes in Merkel Cell Polyomavirus-Positive MCC Cell Lines. Front Microbiol 2021; 12:785662. [PMID: 35003017 PMCID: PMC8727593 DOI: 10.3389/fmicb.2021.785662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Merkel cell carcinoma (MCC) is a rare and highly aggressive cancer, which is mainly caused by genomic integration of the Merkel cell polyomavirus and subsequent expression of a truncated form of its large T antigen. The resulting primary tumor is known to be immunogenic and under constant pressure to escape immune surveillance. Because interferon gamma (IFNγ), a key player of immune response, is secreted by many immune effector cells and has been shown to exert both anti-tumoral and pro-tumoral effects, we studied the transcriptomic response of MCC cells to IFNγ. In particular, immune modulatory effects that may help the tumor evade immune surveillance were of high interest to our investigation. The effect of IFNγ treatment on the transcriptomic program of three MCC cell lines (WaGa, MKL-1, and MKL-2) was analyzed using single-molecule sequencing via the Oxford Nanopore platform. A significant differential expression of several genes was detected across all three cell lines. Subsequent pathway analysis and manual annotation showed a clear upregulation of genes involved in the immune escape of tumor due to IFNγ treatment. The analysis of selected genes on protein level underlined our sequencing results. These findings contribute to a better understanding of immune escape of MCC and may help in clinical treatment of MCC patients. Furthermore, we demonstrate that single-molecule sequencing can be used to assess characteristics of large eukaryotic transcriptomes and thus contribute to a broader access to sequencing data in the community due to its low cost of entry.
Collapse
Affiliation(s)
- Tatjana Sauerer
- RNA-based Immunotherapy, Hautklinik, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christopher Lischer
- Systems Tumor Immunology, Hautklinik, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Adrian Weich
- Systems Tumor Immunology, Hautklinik, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Carola Berking
- Hautklinik, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Julio Vera
- Systems Tumor Immunology, Hautklinik, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Jan Dörrie
- RNA-based Immunotherapy, Hautklinik, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg, Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
22
|
Tripathi SC, Vedpathak D, Ostrin EJ. The Functional and Mechanistic Roles of Immunoproteasome Subunits in Cancer. Cells 2021; 10:cells10123587. [PMID: 34944095 PMCID: PMC8700164 DOI: 10.3390/cells10123587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
Cell-mediated immunity is driven by antigenic peptide presentation on major histocompatibility complex (MHC) molecules. Specialized proteasome complexes called immunoproteasomes process viral, bacterial, and tumor antigens for presentation on MHC class I molecules, which can induce CD8 T cells to mount effective immune responses. Immunoproteasomes are distinguished by three subunits that alter the catalytic activity of the proteasome and are inducible by inflammatory stimuli such as interferon-γ (IFN-γ). This inducible activity places them in central roles in cancer, autoimmunity, and inflammation. While accelerated proteasomal degradation is an important tumorigenic mechanism deployed by several cancers, there is some ambiguity regarding the role of immunoproteasome induction in neoplastic transformation. Understanding the mechanistic and functional relevance of the immunoproteasome provides essential insights into developing targeted therapies, including overcoming resistance to standard proteasome inhibition and immunomodulation of the tumor microenvironment. In this review, we discuss the roles of the immunoproteasome in different cancers.
Collapse
Affiliation(s)
- Satyendra Chandra Tripathi
- Department of Biochemistry, All India Institute of Medical Sciences Nagpur, Nagpur 441108, MH, India;
- Correspondence: (S.C.T.); (E.J.O.)
| | - Disha Vedpathak
- Department of Biochemistry, All India Institute of Medical Sciences Nagpur, Nagpur 441108, MH, India;
| | - Edwin Justin Ostrin
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (S.C.T.); (E.J.O.)
| |
Collapse
|
23
|
Immune Score-based Molecular Subtypes and Signature Associated with Clinical Outcome in Hepatoblastoma. HEPATITIS MONTHLY 2021. [DOI: 10.5812/hepatmon.118268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Background: This study aimed to identify genes related to the immune score of hepatoblastoma, examine the characteristics of the immune microenvironment of hepatoblastoma, and construct a risk scoring system for predicting the prognosis of hepatoblastoma. Methods: Through using the gene chip data of patients with hepatoblastoma with survival data in the ArrayExpress and GEO databases, the immune score of hepatoblastoma was calculated by the ESITIMATE algorithm, and the prognostic value of immune score in patients with hepatoblastoma was studied by the survival analysis. Genes related to the immune score were identified by the WGCNA algorithm. According to these genes, patients with hepatoblastoma were clustered unsupervised. Finally, the risk scoring system was constructed according to the immune score-related genes. Results: The immune score calculated by the ESTIMATE algorithm had a good prognostic value in patients with hepatoblastoma. Patients with high immune scores had better OS than those with low immune scores (P < 0.001). A total of 146 immune score-related genes were identified by WGCNA analysis, and univariate COX regression analysis indicated that 59 of the genes had prognostic value. According to the unsupervised clustering results of the 146 immune score-related genes, patients with hepatoblastoma could be divided into two subtypes with different prognoses, namely molecular subtype 1 and subtype 2, with molecular subtype 1 having a better prognosis. The immunocyte infiltration analysis results showed that the difference between the two subtypes was mainly in activated CD4 T cells, activated dendritic cells, CD56 bright natural killer cells, the macrophage, and regulatory T cells. According to the immune score-related genes, a risk scoring system was constructed based on a five-gene signature. After the cut-off value was determined, patients with hepatoblastoma were divided into a high-risk group and a low-risk group. The prognosis of the two groups was different. Conclusions: The immune score has a good prognostic value in patients with hepatoblastoma. Based on the different expression patterns of immune score-related genes, hepatoblastoma can be divided into two different prognostic molecular subtypes, showing different immunocyte infiltration patterns. The established risk scoring system based on a five-gene signature has a good predictive value in patients with hepatoblastoma.
Collapse
|
24
|
Han X, Wang DZ, Yuan M, Bai WJ. Lemur tyrosine kinase 2 silencing inhibits the proliferation of gastric cancer cells by regulating GSK-3β phosphorylation and β-catenin nuclear translocation. Bioengineered 2021; 13:6231-6243. [PMID: 34719320 PMCID: PMC8982461 DOI: 10.1080/21655979.2021.1999375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Previous studies on the mechanism of proliferation and cell cycle progression of gastric cancer cells have shown promising perspectives for the prevention and treatment of gastric cancer. The aim of the present study was to investigate the role of lemur tyrosine kinase 2 (LMTK2) in gastric cancer cell proliferation and cell cycle progression, as well as in tumor-bearing nude mouse models. The expression levels of LMTK2 were determined in gastric cancer cell lines. In addition, the effects of LMTK2 silencing or overexpression on cell proliferation were measured using Cell Counting Kit-8, BrdU and colony formation assays. Cell cycle progression was analyzed using flow cytometry and western blotting. The expression levels of proteins associated with the β-catenin pathway were assessed using western blot analysis. A tumor-bearing nude mouse model was established by injecting gastric cancer cells, and the effect of LMTK2 knockdown or overexpression on tumor growth was examined. The expression levels of LMTK2 were found to be upregulated in all gastric cancer cell lines. Moreover, LMTK2 knockdown inhibited cell proliferation, colony formation and cell cycle progression. LMTK2 knockdown also inhibited the activation of GSK-3β/β-catenin signaling, as evidenced by reduced GSK-3β phosphorylation and nuclear β-catenin levels. LMTK2 knockdown also suppressed tumor growth, whereas overexpression accelerated this process. In conclusion, LMTK2 silencing can inhibit the proliferation of gastric cancer cells in vitro and tumor growth in vivo by regulating GSK-3β phosphorylation and β-catenin nuclear translocation.
Collapse
Affiliation(s)
- Xin Han
- Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Da-Zhong Wang
- Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Meng Yuan
- Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Wei-Jun Bai
- Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
25
|
Adam RS, Blomberg I, Ten Hoorn S, Bijlsma MF, Vermeulen L. The recurring features of molecular subtypes in distinct gastrointestinal malignancies-A systematic review. Crit Rev Oncol Hematol 2021; 164:103428. [PMID: 34284100 DOI: 10.1016/j.critrevonc.2021.103428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/26/2022] Open
Abstract
In colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC) and gastric cancer (GC) multiple studies of inter-tumor heterogeneity have identified molecular subtypes, which correlate with clinical features. Our aim was to investigate the attributes of molecular subtypes across three different gastrointestinal cancer types. We performed a systematic search for publications on molecular subtypes or classifications in PDAC and GC and compared the described subtypes with the established consensus molecular subtypes of CRC. Examining the characteristics of subtypes across CRC, PDAC and GC resulted in four categories of subtypes. We describe uniting and distinguishing features within a mesenchymal, an epithelial, an immunogenic and a metabolic and digestive subtype category. We conclude that molecular subtypes of CRC, PDAC and GC display relevant overlap in molecular features and clinical outcomes. This finding encourages quantitative studies on subtypes across different cancer types and could lead to a paradigm shift in future treatment strategies.
Collapse
Affiliation(s)
- Ronja S Adam
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Ilse Blomberg
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Sanne Ten Hoorn
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
26
|
Deng M, Li S, Mei J, Lin W, Zou J, Wei W, Guo R. High SGO2 Expression Predicts Poor Overall Survival: A Potential Therapeutic Target for Hepatocellular Carcinoma. Genes (Basel) 2021; 12:genes12060876. [PMID: 34200261 PMCID: PMC8226836 DOI: 10.3390/genes12060876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Shugoshin2 (SGO2) may participate in the occurrence and development of tumors by regulating abnormal cell cycle division, but its prognostic value in hepatocellular carcinoma (HCC) remains unclear. In this study, we accessed The Cancer Genome Atlas (TCGA) database to get the clinical data and gene expression profile of HCC. The expression of SGO2 in HCC tissues and nontumor tissues and the relationship between SGO2 expression, survival, and clinicopathological parameters were analyzed. The SGO2 expression level was significantly higher in HCC tissues than in nontumor tissues (p < 0.001). An analysis from the Oncomine and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) databases also demonstrated that SGO2 was upregulated in HCC (all p < 0.001). A logistic regression analysis revealed that the high expression of SGO2 was significantly correlated with gender, tumor grade, pathological stage, T classification, and Eastern Cancer Oncology Group (ECOG) score (all p < 0.05). The overall survival (OS) of HCC patients with higher SGO2 expression was significantly poor (p < 0.001). A multivariate analysis showed that age and high expression of SGO2 were independent predictors of poor overall survival (all p < 0.05). Twelve signaling pathways were significantly enriched in samples with the high-SGO2 expression phenotype. Ten proteins and 34 genes were significantly correlated with SGO2. In conclusion, the expression of SGO2 is closely related to the survival of HCC. It may be used as a potential therapeutic target and prognostic marker of HCC.
Collapse
Affiliation(s)
- Min Deng
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Shaohua Li
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jie Mei
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wenping Lin
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jingwen Zou
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wei Wei
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Rongping Guo
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (M.D.); (S.L.); (J.M.); (W.L.); (J.Z.); (W.W.)
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Correspondence: ; Tel.: +86-188-1980-9988
| |
Collapse
|
27
|
Zhou Y, Xu B, Wu S, Liu Y. Prognostic Immune-Related Genes of Patients With Ewing's Sarcoma. Front Genet 2021; 12:669549. [PMID: 34122521 PMCID: PMC8194304 DOI: 10.3389/fgene.2021.669549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/30/2021] [Indexed: 01/21/2023] Open
Abstract
Ewing's sarcoma (ES) is an extremely aggressive malignant bone tumor with a high incidence among children and adolescents. The immune microenvironment plays an important role in ES development. The aim of the current study was to investigate the immune microenvironment in ES patients to identify immune-related gene signatures. Single-sample gene set enrichment analysis (ssGSEA) was used to cluster the RNA sequences of 117 ES patients, and their immune cell infiltration data were downloaded and evaluated based on the Gene Expression Omnibus (GEO) database. High, medium, and low immune cell infiltration clusters were identified. Based on the comparison of clusters with high and low immune cell infiltration, normal skeletal muscle cells, and ES, we identified 198 common differentially expressed genes. GO and KEGG enrichment analyses indicated the underlying immune mechanism in ES. Cox and LASSO regression analyses were conducted to select immune-related prognostic genes. An external dataset from the International Cancer Genome Consortium (ICGC) was used to validate our results. Ten immune-related, independent prognostic genes (FMO2, GLCE, GPR64, IGFBP4, LOXHD1, PBK, SNAI2, SPP1, TAPT1-AS1, and ZIC2) were selected for analysis. These 10 immune-related genes signature were determined to exhibit independent prognostic significance for ES. The results of this study provide an approach for predicting the prognosis and survival of ES patients, and the elucidated genes may be a promising target for immunotherapy.
Collapse
Affiliation(s)
- Yangfan Zhou
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bin Xu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shusheng Wu
- The First Affiliated Hospital of USTC, Hefei, China
| | - Yulian Liu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
28
|
Cao H, Yang M, Yang Y, Fang J, Cui Y. PBK/TOPK promotes chemoresistance to oxaliplatin in hepatocellular carcinoma cells by regulating PTEN. Acta Biochim Biophys Sin (Shanghai) 2021; 53:584-592. [PMID: 33772548 DOI: 10.1093/abbs/gmab028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Indexed: 11/14/2022] Open
Abstract
Oxaliplatin (OXA) resistance limits the efficiency of treatment for hepatocellular carcinoma (HCC). Studies have shown that the PDZ-binding kinase (PBK) plays important roles in tumors. However, the role of PBK in HCC is still a problem. In this study, we explored whether PBK is involved in the chemoresistance to OXA in HCC. Expressions of PBK in six HCC cell lines and one human hepatocytes line were determined by real-time quantitative PCR and western blot analysis. SNU-182 and HepG2 cells were chosen to induce OXA resistance. PBK was silenced or overexpressed in OXA-resistant and sensitive cell lines. Then, cell proliferation, migration, and invasion were measured by cholecystokinin-8 assay and Transwell assay, respectively. The Cancer Genome Atlas dataset showed that PBK is highly expressed in HCC and signifies poor prognosis to patient with HCC. Results showed that expression of PBK in HCC cells was significantly higher than that in THLE2 cells, and it was further increased in OXA-resistant HCC cells. Silencing of PBK promoted the sensitivity of drug-resistant HCC cells to OXA. Overexpression of PBK relieved the apoptosis induced by OXA and promoted the migration and invasion of OXA-sensitive HCC cells. Thus, this study revealed that high PBK expression is correlated with OXA resistance in HCC cells, which may provide a promising therapeutic target for treating HCC.
Collapse
Affiliation(s)
- Hongmin Cao
- Oncology Department, SSL Central Hospital of Dongguan City, The Third People’s Hospital of Dongguan City, Dongguan 523326, China
| | - Mei Yang
- Oncology Department, SSL Central Hospital of Dongguan City, The Third People’s Hospital of Dongguan City, Dongguan 523326, China
| | - Yufeng Yang
- Department of Pathology, SSL Central Hospital of Dongguan City, The Third People’s Hospital of Dongguan City, Dongguan 523326, China
| | - Jiayan Fang
- Oncology Department, SSL Central Hospital of Dongguan City, The Third People’s Hospital of Dongguan City, Dongguan 523326, China
| | - Yejia Cui
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, The Third People’s Hospital of Dongguan City, Dongguan 523326, China
| |
Collapse
|
29
|
Dutta D, VandeHaar P, Fritsche LG, Zöllner S, Boehnke M, Scott LJ, Lee S. A powerful subset-based method identifies gene set associations and improves interpretation in UK Biobank. Am J Hum Genet 2021; 108:669-681. [PMID: 33730541 DOI: 10.1016/j.ajhg.2021.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
Tests of association between a phenotype and a set of genes in a biological pathway can provide insights into the genetic architecture of complex phenotypes beyond those obtained from single-variant or single-gene association analysis. However, most existing gene set tests have limited power to detect gene set-phenotype association when a small fraction of the genes are associated with the phenotype and cannot identify the potentially "active" genes that might drive a gene set-based association. To address these issues, we have developed Gene set analysis Association Using Sparse Signals (GAUSS), a method for gene set association analysis that requires only GWAS summary statistics. For each significantly associated gene set, GAUSS identifies the subset of genes that have the maximal evidence of association and can best account for the gene set association. Using pre-computed correlation structure among test statistics from a reference panel, our p value calculation is substantially faster than other permutation- or simulation-based approaches. In simulations with varying proportions of causal genes, we find that GAUSS effectively controls type 1 error rate and has greater power than several existing methods, particularly when a small proportion of genes account for the gene set signal. Using GAUSS, we analyzed UK Biobank GWAS summary statistics for 10,679 gene sets and 1,403 binary phenotypes. We found that GAUSS is scalable and identified 13,466 phenotype and gene set association pairs. Within these gene sets, we identify an average of 17.2 (max = 405) genes that underlie these gene set associations.
Collapse
Affiliation(s)
- Diptavo Dutta
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Peter VandeHaar
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lars G Fritsche
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sebastian Zöllner
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael Boehnke
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura J Scott
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seunggeun Lee
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Graduate School of Data Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
30
|
Nagano-Matsuo A, Inoue S, Koshino A, Ota A, Nakao K, Komura M, Kato H, Naiki-Ito A, Watanabe K, Nagayasu Y, Hosokawa Y, Takiguchi S, Kasugai K, Kasai K, Inaguma S, Takahashi S. PBK expression predicts favorable survival in colorectal cancer patients. Virchows Arch 2021; 479:277-284. [PMID: 33638656 DOI: 10.1007/s00428-021-03062-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/05/2021] [Accepted: 02/15/2021] [Indexed: 01/02/2023]
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal cancers worldwide with high morbidity and mortality rates. The discovery of small molecule anticancer reagents has significantly affected cancer therapy. However, the anticancer effects of these therapies are not sufficient to completely cure CRC. PDZ-binding kinase (PBK) was initially identified as a mitotic kinase for mitogen-activated protein kinase and is involved in cytokinesis and spermatogenesis. Aberrant expression of PBK has been reported to be closely associated with malignant phenotypes of many cancers and/or patient survival. However, the expression of PBK and its association to patient survival in CRC have not been fully elucidated. In the present study, 269 primary CRCs were evaluated immunohistochemically for PBK expression to assess its ability as a prognostic factor. CRC tumor cells variably expressed PBK (range, 0-100%; median, 32%) in the nucleus and cytoplasm. Univariate analyses identified a significant inverse correlation between PBK expression and pT stage (P<0.0001). Furthermore, patients carrying CRC with higher PBK expression showed significantly favorable survival (P=0.0094). Multivariate Cox proportional hazards regression analysis revealed high PBK expression (HR, 0.52; P=0.015) as one of the potential favorable factors for CRC patients. PBK expression showed significant correlation to Ki-67 labeling indices (ρ=0.488, P<0.0001). In vitro, the PBK inhibitor OTS514 suppressed cellular proliferation of CRC cells with PBK expression through downregulation of P-ERK and induction of apoptosis. These results suggest that PBK-targeting therapeutics may be useful for the treatment of PBK-expressing CRC patients.
Collapse
Affiliation(s)
- Aya Nagano-Matsuo
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Inoue
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akira Koshino
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenju Nakao
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masayuki Komura
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kawori Watanabe
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuko Nagayasu
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kunio Kasugai
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shingo Inaguma
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan. .,Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan. .,Department of Pathology, Nagoya City East Medical Center, Nagoya, Japan. .,Educational Research Center for Advanced Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
31
|
Wu M, Chen P, Liu F, Lv B, Ge M, Jiang P, Xu W, Liu X, Yang D. ONX0912, a selective oral proteasome inhibitor, triggering mitochondrial apoptosis and mitophagy in liver cancer. Biochem Biophys Res Commun 2021; 547:102-110. [PMID: 33610037 DOI: 10.1016/j.bbrc.2021.02.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 11/29/2022]
Abstract
Proteasome inhibitors represent effective anti-tumor drugs. ONX0912 is a novel oral proteasome inhibitor that selectively targets the chymotrypsin-like activity of 20S proteasome subunits β5 and LMP7 (Low molecular mass polypeptide-7). It has been shown to be effective in hematologic malignancies. However, its anti-tumor effect in solid tumors remains unclear. Here, we discovered that ONX0912 suppressed the expansion of liver cancer cells. ONX0912 treatment led to an increased level of mitochondrial membrane potential collapse and mitochondrial ROS in tumor cells in a concentration- and exposure time-dependent manner, indicating ONX0912 triggers apoptosis through the intrinsic mitochondrial pathway. ONX0912 also induced mitophagy by activating Parkin/Pink pathway. Silencing mitophagy receptor protein, p62, aggravated the ONX0912-mediated apoptosis, which implied a new mechanism for the conversion between autophagy and apoptosis. Furthermore, we found that the ONX0912 target protein, LMP7 was overexpressed in liver cancer tissues compared to their adjacent tissues and increased level of LMP7 predicted worse clinical characteristics and poorer prognosis. In conclusion, we demonstrated that ONX0912 suppressed liver cancer cell expansion by inducing apoptosis and mitophagy. Our data also revealed ONX0912 as a potential clinical therapeutic drug for liver cancer therapy, and inhibition of mitophagy may sensitize the anti-tumor effect of ONX0912.
Collapse
Affiliation(s)
- Mengmeng Wu
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ping Chen
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, 200040, China; Institute of Biomedical Science of Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Bin Lv
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mengxiao Ge
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Peicheng Jiang
- Department of Digestive Diseases of Jinshan Hospital, Fudan University, Shanghai, 200540, China
| | - Wei Xu
- Department of Immunology, Fudan University, Shanghai, 200032, China
| | - Xiuping Liu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
| | - Dongqin Yang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
32
|
Chang HH, Cheng YC, Tsai WC, Chen Y. PSMB8 inhibition decreases tumor angiogenesis in glioblastoma through vascular endothelial growth factor A reduction. Cancer Sci 2020; 111:4142-4153. [PMID: 32816328 PMCID: PMC7648028 DOI: 10.1111/cas.14625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma, also known as glioblastoma multiforme (GBM), is a fast‐growing tumor and the most aggressive brain malignancy. Proteasome subunit beta type‐8 (PSMB8) is one of the 17 essential subunits for the complete assembly of the 20S proteasome complex. The aim of the present study was to evaluate the role of PSMB8 expression in GBM progression and angiogenesis. PSMB8 expression in glioblastoma LN229 and U87MG was knocked down by siRNA or inducible shRNA both in vitro and in vivo. After PSMB8 reduction, cell survival, migration, invasion, angiogenesis, and the related signaling cascades were evaluated. An orthotopic mouse tumor model was also provided to examine the angiogenesis within tumors. A GEO profile analysis indicated that high expression of PSMB8 mRNA in GBM patients was correlated with a low survival rate. In immunohistochemistry analysis, PSMB8 expression was higher in high‐grade than in low‐grade brain tumors. The proliferation, migration, and angiogenesis of human GBM cells were decreased by PSMB8 knockdown in vitro. Furthermore, phosphorylated focal adhesion kinase (p‐FAK), p‐paxillin, MMP2, MMP9, and cathepsin B were significantly reduced in LN229 cells. Integrin β1 and β3 were reduced in HUVEC after incubation with LN229‐conditioned medium. In an orthotopic mouse tumor model, inducible knockdown of PSMB8 reduced the expression of vascular endothelial growth factor (VEGF), VEGF receptor, and CD31 as well as the progression of human glioblastoma. In this article, we demonstrated the role of PSMB8 in glioblastoma progression, especially neovascularization in vitro and in vivo. These results may provide a target for the anti–angiogenic effect of PSMB8 in glioblastoma therapy in the future.
Collapse
Affiliation(s)
- Hsin-Han Chang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chen Cheng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
33
|
Ota A, Hanamura I, Karnan S, Inaguma S, Takei N, Lam VQ, Mizuno S, Kanasugi J, Wahiduzzaman M, Rahman ML, Hyodo T, Konishi H, Tsuzuki S, Ikeda H, Takami A, Hosokawa Y. Novel Interleukin-6 Inducible Gene PDZ-Binding Kinase Promotes Tumor Growth of Multiple Myeloma Cells. J Interferon Cytokine Res 2020; 40:389-405. [PMID: 32721246 PMCID: PMC7462034 DOI: 10.1089/jir.2020.0111] [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] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) remains an intractable hematological malignancy, despite recent advances in anti-MM drugs. Here, we show that role of PDZ binding kinase (PBK) in MM tumor growth. We identified that interleukin-6 (IL-6) readily increases PBK expression. Kaplan–Meier analysis showed that the MM patients with higher expression of PBK have a significant shorter survival time compared with those with moderate/lower expression of PBK. Knockout of PBK dramatically suppressed in vivo tumor growth in MM cells, while genome editing of PBK changing from asparagine to serine substitution (rs3779620) slightly suppresses the tumor formation. Mechanistically, loss of PBK increased the number of apoptotic cells with concomitant decrease in the phosphorylation level of Stat3 as well as caspase activities. A novel PBK inhibitor OTS514 significantly decreased KMS-11-derived tumor growth. These findings highlight the novel oncogenic role of PBK in tumor growth of myeloma, and it might be a novel therapeutic target for the treatment of patients with MM.
Collapse
Affiliation(s)
- Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shingo Inaguma
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Norio Takei
- Institute for Animal Experimentation, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Vu Quang Lam
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shohei Mizuno
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Jo Kanasugi
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Md Lutfur Rahman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroshi Ikeda
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akiyoshi Takami
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
34
|
Liu Y, Yang HZ, Jiang YJ, Xu LQ. miR-451a is downregulated and targets PSMB8 in prostate cancer. Kaohsiung J Med Sci 2020; 36:494-500. [PMID: 32128987 DOI: 10.1002/kjm2.12196] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/31/2019] [Accepted: 02/09/2020] [Indexed: 01/05/2023] Open
Abstract
Abnormal expression of microRNAs (miRNAs) is frequently occurred in prostate cancer (PCa). This study was aimed to investigate the biological roles of miR-451a in PCa. Quantitative real-time PCR (qRT-PCR) and Western blot were employed to investigate the expression levels of miR-451a and proteasome (prosome, macropain) subunit, beta type, 8 (PSMB8) in PCa cell lines. Luciferase activity reporter assay was used to verify the connection between miR-451a and PSMB8. in vitro functional experiments were performed to measure the effects of miR-451a or PSMB8 on PCa cell proliferation, colony formation ability, cell invasion, and cell apoptosis. miR-451a expression was downregulated, whereas PSMB8 expression was upregulated in PCa cell lines. Luciferase activity reporter assay confirmed the direct connection between miR-451a and PSMB8. Overexpression of miR-451a inhibits PCa cell proliferation, colony formation, cell invasion and promotes cell apoptosis, while the overexpression of PSMB8 caused the opposite effects. Moreover, rescue experiments confirmed PSMB8 was a functional target of miR-451a. In conclusion, this study provides novel insights into the role of miR-451a in PCa, and the results demonstrated miR-451a could inhibit PCa progression by targeting PSMB8.
Collapse
Affiliation(s)
- Yun Liu
- Department of Urinary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Huan-Zhi Yang
- Department of Neurology, Central Hospital of Binzhou, Binzhou, Shandong, People's Republic of China
| | - Yong-Jun Jiang
- Department of Urinary Surgery, The Third People's Hospital of LinYi, Linyi, Shandong, People's Republic of China
| | - Li-Qi Xu
- Department of Urologic Surgery, No. 906 Hospital of PLA, Ningbo, Zhejiang, People's Republic of China
| |
Collapse
|
35
|
Bioinformatics Analysis of Potential Key Genes in Trastuzumab-Resistant Gastric Cancer. DISEASE MARKERS 2019; 2019:1372571. [PMID: 31949544 PMCID: PMC6948351 DOI: 10.1155/2019/1372571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/03/2019] [Accepted: 11/09/2019] [Indexed: 12/24/2022]
Abstract
Background This study was performed to identify genes related to acquired trastuzumab resistance in gastric cancer (GC) and to analyze their prognostic value. Methods The gene expression profile GSE77346 was downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were obtained by using GEO2R. Functional and pathway enrichment was analyzed by using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Search Tool for the Retrieval of Interacting Genes (STRING), Cytoscape, and MCODE were then used to construct the protein-protein interaction (PPI) network and identify hub genes. Finally, the relationship between hub genes and overall survival (OS) was analyzed by using the online Kaplan-Meier plotter tool. Results A total of 327 DEGs were screened and were mainly enriched in terms related to pathways in cancer, signaling pathways regulating stem cell pluripotency, HTLV-I infection, and ECM-receptor interactions. A PPI network was constructed, and 18 hub genes (including one upregulated gene and seventeen downregulated genes) were identified based on the degrees and MCODE scores of the PPI network. Finally, the expression of four hub genes (ERBB2, VIM, EGR1, and PSMB8) was found to be related to the prognosis of HER2-positive (HER2+) gastric cancer. However, the prognostic value of the other hub genes was controversial; interestingly, most of these genes were interferon- (IFN-) stimulated genes (ISGs). Conclusions Overall, we propose that the four hub genes may be potential targets in trastuzumab-resistant gastric cancer and that ISGs may play a key role in promoting trastuzumab resistance in GC.
Collapse
|
36
|
Lei M, Jingjing Z, Tao J, Jianping M, Yuanxin Z, Jifeng W, Lianguo X, Lidong Z, Ying W. LncRNA HCP5 promotes LAML progression via PSMB8-mediated PI3K/AKT pathway activation. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:1025-1032. [PMID: 31836918 DOI: 10.1007/s00210-019-01788-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/03/2019] [Indexed: 02/08/2023]
Abstract
Acute myeloid leukemia is an aggressive myeloid malignancy, characterized by rapid cellular proliferation and generally high mortality. Due to the lack of a complete understanding of AML, its clinical outcomes are still not satisfactory. In this study, we examined the function of the long non-coding RNA-HLA complex P5 (HCP5) on AML by analyzing the clinical samples, TCGA data, and by shRNA-mediated HCP5 deficiency in vitro. Our results showed that HCP5 is highly expressed in AML and is positive associated with poor prognosis, and HCP5 knockdown was significantly suppressing AML cell line proliferation and inducing G1/S arrest in vitro. In mechanism, the proteasome subunit beta type 8 (PSMB8) expression was dramatically inhibited in HCP5 knockdown cells while increased in HCP5 overexpression cells. PSMB8 was also highly expressed in AML and with poor prognosis. Furthermore, HCP5 regulates PI3K/AKT pathway activation depending on PSMB8. Our results showed a promoting function of HCP5 on AML and may provide a compelling biomarker and therapy target for AML.
Collapse
Affiliation(s)
- Miao Lei
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Zhao Jingjing
- Department of Oncology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Jia Tao
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Mao Jianping
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Zhu Yuanxin
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Wei Jifeng
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Xue Lianguo
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Zhao Lidong
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Wang Ying
- Department of Hematology, The First People's Hospital of Lianyungang, No.182 Tongguan North Road, Lianyungang, 222000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
37
|
PBK as a Potential Biomarker Associated with Prognosis of Glioblastoma. J Mol Neurosci 2019; 70:56-64. [DOI: 10.1007/s12031-019-01400-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/14/2019] [Indexed: 01/04/2023]
|
38
|
Kar A, Zhang Y, Yacob BW, Saeed J, Tompkins KD, Bagby SM, Pitts TM, Somerset H, Leong S, Wierman ME, Kiseljak-Vassiliades K. Targeting PDZ-binding kinase is anti-tumorigenic in novel preclinical models of ACC. Endocr Relat Cancer 2019; 26:765-778. [PMID: 31325906 PMCID: PMC6938568 DOI: 10.1530/erc-19-0262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
Adrenocortical carcinoma (ACC) is an aggressive orphan malignancy with less than 35% 5-year survival and 75% recurrence. Surgery remains the primary therapy and mitotane, an adrenolytic, is the only FDA-approved drug with wide-range toxicities and poor tolerability. There are no targeted agents available to date. For the last three decades, H295R cell line and its xenograft were the only available preclinical models. We recently developed two new ACC patient-derived xenograft mouse models and corresponding cell lines (CU-ACC1 and CU-ACC2) to advance research in the field. Here, we have utilized these novel models along with H295R cells to establish the mitotic PDZ-binding kinase (PBK) as a promising therapeutic target. PBK is overexpressed in ACC samples and correlates with poor survival. We show that PBK is regulated by FOXM1 and targeting PBK via shRNA decreased cell proliferation, clonogenicity and anchorage-independent growth in ACC cell lines. PBK silencing inhibited pAkt, pp38MAPK and pHistone H3 altering the cell cycle. Therapeutically, targeting PBK with the small-molecule inhibitor HITOPK032 phenocopied PBK-specific modulation of pAkt and pHistone H3, but also induced apoptosis via activation of JNK. Consistent with in vitro findings, treatment of CU-ACC1 PDXs with HITOPK032 significantly reduced tumor growth by 5-fold (P < 0.01). Treated tumor tissues demonstrated increased rates of apoptosis and JNK activation, with decreased pAkt and Histone H3 phosphorylation, consistent with effects observed in ACC cell lines. Together these studies elucidate the mechanism of PBK in ACC tumorigenesis and establish the potential therapeutic potential of HITOPK032 in ACC patients.
Collapse
Affiliation(s)
- Adwitiya Kar
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Yu Zhang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Betelehem W. Yacob
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Jordan Saeed
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Kenneth D. Tompkins
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Stacey M. Bagby
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Todd M. Pitts
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Hilary Somerset
- Department of Pathology, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Stephen Leong
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
| | - Margaret E. Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus Aurora, CO 80045
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045
| |
Collapse
|
39
|
Balakrishnan K, Panneerpandian P, Devanandan HJ, Sekar BT, Rayala SK, Ganesan K. Salt-mediated transcriptional and proteasomal dysregulations mimic the molecular dysregulations of stomach cancer. Toxicol In Vitro 2019; 61:104588. [PMID: 31279909 DOI: 10.1016/j.tiv.2019.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Karthik Balakrishnan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Ponmathi Panneerpandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Helen Jemimah Devanandan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Balaji T Sekar
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamilnadu, India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India.
| |
Collapse
|
40
|
Mascolo MG, Perdichizzi S, Vaccari M, Rotondo F, Zanzi C, Grilli S, Paparella M, Jacobs MN, Colacci A. The transformics assay: first steps for the development of an integrated approach to investigate the malignant cell transformation in vitro. Carcinogenesis 2019; 39:955-967. [PMID: 29554273 PMCID: PMC6031005 DOI: 10.1093/carcin/bgy037] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 03/14/2018] [Indexed: 12/20/2022] Open
Abstract
The development of alternative methods to animal testing is a priority in the context of regulatory toxicology. Carcinogenesis is a field where the demand for alternative methods is particularly high. The standard rodent carcinogenicity bioassay requires a large use of animals, high costs, prolonged duration and shows several limitations, which can affect the comprehension of the human relevance of animal carcinogenesis. The cell transformation assay (CTA) has long been debated as a possible in vitro test to study carcinogenesis. This assay provides an easily detectable endpoint of oncotransformation, which can be used to anchor the exposure to the acquisition of the malignant phenotype. However, the current protocols do not provide information on either molecular key events supporting the carcinogenesis process, nor the mechanism of action of the test chemicals. In order to improve the use of this assay in the integrated testing strategy for carcinogenesis, we developed the transformics method, which combines the CTA and transcriptomics, to highlight the molecular steps leading to in vitro malignant transformation. We studied 3-methylcholanthrene (3-MCA), a genotoxic chemical able to induce in vitro cell transformation, at both transforming and subtransforming concentrations in BALB/c 3T3 cells and evaluated the gene modulation at critical steps of the experimental protocol. The results gave evidence for the potential key role of the immune system and the possible involvement of the aryl hydrocarbon receptor (AhR) pathway as the initial steps of the in vitro transformation process induced by 3-MCA, suggesting that the initiating events are related to non-genotoxic mechanisms.
Collapse
Affiliation(s)
- Maria Grazia Mascolo
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| | - Stefania Perdichizzi
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| | - Monica Vaccari
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| | - Francesca Rotondo
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| | - Cristina Zanzi
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| | - Sandro Grilli
- Department of Experimental, Diagnostic and Specialty Medicine, Section of Cancerology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Martin Paparella
- Chemicals and Biocides, Environment Agency Austria, Vienna, Austria
| | - Miriam N Jacobs
- Department of Toxicology, Centre for Radiation, Chemical and Environmental Hazards Public Health England, Chilton, Oxfordshire, UK
| | - Annamaria Colacci
- Center for Environmental Toxicology, Agency for Prevention, Environment and Energy, Emilia-Romagna, Viale Filopanti, Bologna, Italy
| |
Collapse
|
41
|
Zhang Y, Yang X, Wang R, Zhang X. Prognostic Value of PDZ-Binding Kinase/T-LAK Cell-Originated Protein Kinase (PBK/TOPK) in Patients with Cancer. J Cancer 2019; 10:131-137. [PMID: 30662533 PMCID: PMC6329853 DOI: 10.7150/jca.28216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/24/2018] [Indexed: 01/01/2023] Open
Abstract
Background: PDZ-binding kinase/T-LAK cell-originated protein kinase (PBK/TOPK) plays a critical role in tumorigenesis and cancer progression. However, the prognostic roles in cancer patients are inconsistent or even controversial. Therefore, we performed a meta-analysis to investigate the prognostic value of PBK/TOPK in cancers. Methods: Literature search was performed using several online databases (PubMed, Web of Science, Embase, Cochrane Library, and Google Scholar, National Knowledge Infrastructure and Wanfang) for eligible articles published up to May 1, 2018. The relationship between PBK/TOPK expression and prognosis in cancers was investigated by using pooled hazard ratios (HRs) with 95% confidence intervals (CIs) through STATA 12.0 software. Results: Totally 20 eligible studies were included in this meta-analysis. The pooled results showed that carriers with high protein expression of PBK/TOPK were significantly associated with poor OS (HR: 1.69, 95% CI: 1.33-2.04) in various cancers, and patients with increased PBK/TOPK protein expression were significantly correlated with inferior RFS (HR: 1.63, 95% CI: 1.02-2.24) and short DFS (HR: 1.69, 95% CI: 1.16-2.23). Conclusions: The findings suggest that PBK/TOPK protein expression might serve as a prognostic tumor marker in cancers.
Collapse
Affiliation(s)
- Yi Zhang
- Department of General Surgery, the First People's Hospital of Neijiang, Neijiang 641000, Sichuan Province, P. R. China
| | - Xianjin Yang
- Department of General Surgery, the First People's Hospital of Neijiang, Neijiang 641000, Sichuan Province, P. R. China
| | - Rong Wang
- Department of General Surgery, the First People's Hospital of Neijiang, Neijiang 641000, Sichuan Province, P. R. China
| | - Xu Zhang
- Department of General Surgery, the First People's Hospital of Neijiang, Neijiang 641000, Sichuan Province, P. R. China
| |
Collapse
|
42
|
Prince C, Hammerton G, Taylor AE, Anderson EL, Timpson NJ, Davey Smith G, Munafò MR, Relton CL, Richmond RC. Investigating the impact of cigarette smoking behaviours on DNA methylation patterns in adolescence. Hum Mol Genet 2019; 28:155-165. [PMID: 30215712 PMCID: PMC6298233 DOI: 10.1093/hmg/ddy316] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/02/2018] [Accepted: 09/03/2018] [Indexed: 01/09/2023] Open
Abstract
Smoking usually begins in adolescence, and early onset of smoking has been linked to increased risk of later life disease. There is a need to better understand the biological impact of cigarette smoking behaviours in adolescence. DNA methylation profiles related to smoking behaviours and cessation in adulthood have been previously identified, but alterations arising from smoking initiation have not been thoroughly investigated. We aimed to investigate DNA methylation in the Avon Longitudinal Study of Parents and Children in relation to (1) different smoking measures, (2) time since smoking initiation and frequency of smoke exposure and (3) latent classes of smoking behaviour. Using 2620 CpG sites previously associated with cigarette smoking, we investigated DNA methylation change in relation to own smoking measures, smoke exposure duration and frequency, and using longitudinal latent class analysis of different smoking behaviour patterns in 968 adolescents. Eleven CpG sites located in seven gene regions were differentially methylated in relation to smoking in adolescence. While only AHRR (cg05575921) showed a robust pattern of methylation in relation to weekly smoking, several CpGs showed differences in methylation among individuals who had tried smoking compared with non-smokers. In relation to smoke exposure duration and frequency, cg05575921 showed a strong dose-response relationship, while there was evidence for more immediate methylation change at other sites. Our findings illustrate the impact of cigarette smoking behaviours on DNA methylation at some smoking-responsive CpG sites, even among individuals with a short smoking history.
Collapse
Affiliation(s)
| | - Gemma Hammerton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Amy E Taylor
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Emma L Anderson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - George Davey Smith
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Tobacco and Alcohol Research Group, School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Caroline L Relton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Rebecca C Richmond
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
43
|
Chakraborty S, Ghosh Z. A systemic insight into astrocytoma biology across different grades. J Cell Physiol 2018; 234:4243-4255. [DOI: 10.1002/jcp.27193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/17/2018] [Indexed: 01/05/2023]
Affiliation(s)
| | - Zhumur Ghosh
- Bioinformatics Centre, Bose Institute Kolkata India
| |
Collapse
|
44
|
Yang YF, Pan YH, Cao Y, Fu J, Yang X, Zhang MF, Tian QH. PDZ binding kinase, regulated by FoxM1, enhances malignant phenotype via activation of β-Catenin signaling in hepatocellular carcinoma. Oncotarget 2018; 8:47195-47205. [PMID: 28525379 PMCID: PMC5564556 DOI: 10.18632/oncotarget.17587] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022] Open
Abstract
Deregulation of serine/threonine kinase contributes to the development and progression of human diseases. PDZ-binding kinase (PBK) has been implicated in the malignant process of cancers, but its role and clinical significance in hepatocellular carcinoma (HCC) remains unclear. Here we show that PBK expression is increased and associated with larger tumor size, presence of vascular invasion, lymph node metastasis and poor overall and disease-free survivals in two independent cohorts of 879 patients with HCC. In vitro and in vivo data demonstrate PBK exerts oncogenic functions in HCC via activation of β-Catenin signaling pathway. The inhibition of β-Catenin by siRNAs or XAV-939 significantly attenuates PBK-mediated malignant phenotypes. PBK is further identified as a downstream effector of FoxM1. In clinical samples, PBK expression is positively correlated with the expression of FoxM1 and nuclear β-Catenin. Collectively, these findings suggest PBK functions as an oncogene in HCC and the newly identified FoxM1/PBK/β-Catenin axis serves as a promising prognostic factor as well as therapeutic intervention for HCC.
Collapse
Affiliation(s)
- Yu-Feng Yang
- Department of Pathology, Dongguan Third People's Hospital, Dongguan, China
| | - Ying-Hua Pan
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yun Cao
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jia Fu
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xia Yang
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Mei-Fang Zhang
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qiu-Hong Tian
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
45
|
Liew PL, Huang RL, Weng YC, Fang CL, Hui-Ming Huang T, Lai HC. Distinct methylation profile of mucinous ovarian carcinoma reveals susceptibility to proteasome inhibitors. Int J Cancer 2018; 143:355-367. [PMID: 29451304 PMCID: PMC6001480 DOI: 10.1002/ijc.31324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/14/2018] [Accepted: 02/05/2018] [Indexed: 12/23/2022]
Abstract
Mucinous type of epithelial ovarian cancer (MuOC) is a unique subtype with a poor survival outcome in recurrent and advanced stages. The role of type-specific epigenomics and its clinical significance remains uncertain. We analyzed the methylomic profiles of 6 benign mucinous adenomas, 24 MuOCs, 103 serous type of epithelial ovarian cancers (SeOCs) and 337 nonepithelial ovarian cancers. MuOC and SeOC exhibited distinct DNA methylation profiles comprising 101 genes, 81 of which exhibited low methylation in MuOC and were associated with the response to glucocorticoid, ATP hydrolysis-coupled proton transport, proteolysis involved in the cellular protein catabolic process and ion transmembrane transport. Hierarchical clustering analysis showed that the profiles of MuOC were similar to colorectal adenocarcinoma and stomach adenocarcinoma. Genetic interaction network analysis of differentially methylated genes in MuOC showed a dominant network module is the proteasome subunit beta (PSMB) family. Combined functional module and methylation analysis identified PSMB8 as a candidate marker for MuOC. Immunohistochemical staining of PSMB8 used to validate in 94 samples of ovarian tumors (mucinous adenoma, MuOC or SeOC) and 62 samples of gastrointestinal cancer. PSMB8 was commonly expressed in MuOC and gastrointestinal cancer samples, predominantly as strong cytoplasmic and occasionally weak nuclei staining, but was not expressed in SeOC samples. Carfilzomib, a second-generation proteasome inhibitor, suppressed MuOC cell growth in vitro. This study unveiled a mucinous-type-specific methylation profile and suggests the potential use of a proteasome inhibitor to treat MuOC.
Collapse
Affiliation(s)
- Phui-Ly Liew
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Rui-Lan Huang
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Translational epigenetic center, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Yu-Chun Weng
- Translational epigenetic center, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tim Hui-Ming Huang
- Department of Molecular Medicine/Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Translational epigenetic center, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China
| |
Collapse
|
46
|
Uesato T, Ogihara T, Hara A, Iida H, Miyatsuka T, Fujitani Y, Takeda S, Watada H. Enhanced Expression of the Key Mitosis Regulator Cyclin B1 Is Mediated by PDZ-Binding Kinase in Islets of Pregnant Mice. J Endocr Soc 2018; 2:207-219. [PMID: 29594255 PMCID: PMC5841183 DOI: 10.1210/js.2017-00338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 01/25/2018] [Indexed: 12/16/2022] Open
Abstract
The proliferation of pancreatic β cells is enhanced to enable an increase in β-cell mass and to compensate for insulin resistance during pregnancy. To elucidate the mechanisms involved, we previously investigated islets from pregnant and nonpregnant mice by gene expression profiling and found that the expression of postsynaptic density-95/Discs large/zonula occludens-1 (PDZ)–binding kinase (Pbk), a member of the mitogen-activated protein kinase kinase family, is increased in pregnant mouse islets compared with control mouse islets. Among the pregnancy hormones, treatment with estradiol upregulated Pbk expression. Inhibition of Pbk expression using a small interfering RNA for Pbk reduced bromodeoxyuridine incorporation in mouse insulinoma 6 cells, which was accompanied by a decreased expression of Ccnb1, a regulatory gene involved in mitosis. Ccnb1 expression was augmented in mouse islets during pregnancy. The forced expression of Pbk using an adenovirus system in isolated mouse islets increased Ccnb1 expression, and the Pbk inhibitor HI-TOPK-032 suppressed Ccnb1 expression in islets isolated from pregnant mice. Our results suggest that Pbk contributes to the expansion of islets during pregnancy and that Ccnb1 may assist Pbk in its role in β-cell proliferation.
Collapse
Affiliation(s)
- Tadayoshi Uesato
- Department of Obstetrics and Gynecology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takeshi Ogihara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Akemi Hara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Hitoshi Iida
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takeshi Miyatsuka
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yoshio Fujitani
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan.,AMED-JST-CREST Program, Tokyo 100-0004, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Center for Molecular Diabetology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
47
|
Wang CY, Li CY, Hsu HP, Cho CY, Yen MC, Weng TY, Chen WC, Hung YH, Lee KT, Hung JH, Chen YL, Lai MD. PSMB5 plays a dual role in cancer development and immunosuppression. Am J Cancer Res 2017; 7:2103-2120. [PMID: 29218236 PMCID: PMC5714741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 06/07/2023] Open
Abstract
Tumor progression and metastasis are dependent on the intrinsic properties of tumor cells and the influence of microenvironment including the immune system. It would be important to identify target drug that can inhibit cancer cell and activate immune cells. Proteasome β subunits (PSMB) family, one component of the ubiquitin-proteasome system, has been demonstrated to play an important role in tumor cells and immune cells. Therefore, we used a bioinformatics approach to examine the potential role of PSMB family. Analysis of breast TCGA and METABRIC database revealed that high expression of PSMB5 was observed in breast cancer tissue and that high expression of PSMB5 predicted worse survival. In addition, high expression of PSMB5 was observed in M2 macrophages. Based on our bioinformatics analysis, we hypothesized that PSMB5 contained immunosuppressive and oncogenic characteristics. To study the effects of PSMB5 on the cancer cell and macrophage in vitro, we silenced PSMB5 expression with shRNA in THP-1 monocytes and MDA-MB-231 cells respectively. Knockdown of PSMB5 promoted human THP-1 monocyte differentiation into M1 macrophage. On the other hand, knockdown PSMB5 gene expression inhibited MDA-MB-231 cell growth and migration by colony formation assay and boyden chamber. Collectively, our data demonstrated that delivery of PSMB5 shRNA suppressed cell growth and activated defensive M1 macrophages in vitro. Furthermore, lentiviral delivery of PSMB5 shRNA significantly decreased tumor growth in a subcutaneous mouse model. In conclusion, our bioinformatics study and functional experiments revealed that PSMB5 served as novel cancer therapeutic targets. These results also demonstrated a novel translational approach to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Chung-Yen Li
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Hui-Ping Hsu
- Department of Surgery, College of Medicine, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Chien-Yu Cho
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiung, Taiwan, R.O.C
| | - Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Wei-Ching Chen
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Yu-Hsuan Hung
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Kuo-Ting Lee
- Department of Surgery, College of Medicine, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| | - Jui-Hsiang Hung
- Department of Biotechnology, Chia Nan University of Pharmacy and ScienceTainan, Taiwan, R.O.C
| | - Yi-Ling Chen
- Department of Senior Citizen Service Management, Chia Nan University of Pharmacy and ScienceTainan, Taiwan, R.O.C
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, National Cheng Kung UniversityTainan, Taiwan, R.O.C
- Institute of Basic Medical Sciences, National Cheng Kung UniversityTainan, Taiwan, R.O.C
| |
Collapse
|
48
|
Berg A, Gulati A, Ytre-Hauge S, Fasmer KE, Mauland KK, Hoivik EA, Husby JA, Tangen IL, Trovik J, Halle MK, Stefansson I, Akslen LA, Woie K, Bjørge L, Salvesen HB, Salvesen ØO, Werner HM, Haldorsen IS, Krakstad C. Preoperative imaging markers and PDZ-binding kinase tissue expression predict low-risk disease in endometrial hyperplasias and low grade cancers. Oncotarget 2017; 8:68530-68541. [PMID: 28978135 PMCID: PMC5620275 DOI: 10.18632/oncotarget.19708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/19/2017] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Distinguishing complex atypical hyperplasia (CAH) from grade 1 endometrioid endometrial cancer (EECG1) preoperatively may be valuable in order to prevent surgical overtreatment, particularly in patients wishing preserved fertility or in patients carrying increased risk of perioperative complications. MATERIAL AND METHODS Preoperative histological diagnosis and radiological findings were compared to final histological diagnosis in patients diagnosed with CAH and EECG1. Imaging characteristics at preoperative magnetic resonance imaging (MRI) and fluorodeoxyglucose positron emission tomography/computer tomography (FDG-PET/CT) were compared with tumor DNA oligonucleotide microarray data, immunohistochemistry findings and clinicopathological annotations. RESULTS MRI assessed tumor volume was higher in EECG1 than in CAH (p=0.004) whereas tumor apparent diffusion coefficient value was lower in EECG1 (p=0.005). EECG1 exhibited increased metabolism with higher maximum and mean standard uptake values (SUV) than CAH (p≤0.002). Unsupervised clustering of EECG1 and CAH revealed differentially expressed genes within the clusters, and identified PDZ-binding kinase (PBK) as a potential marker for selecting endometrial lesions with less aggressive biological behavior. CONCLUSION Both PBK expression and preoperative imaging yield promising biomarkers that may aid in the differentiation between EECG1 and CAH preoperatively, and these markers should be further explored in larger patient series.
Collapse
Affiliation(s)
- Anna Berg
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Ankush Gulati
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Sigmund Ytre-Hauge
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Norway
| | | | - Karen K. Mauland
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Erling A. Hoivik
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Jenny A. Husby
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Norway
| | - Ingvild L. Tangen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Jone Trovik
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Mari K. Halle
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Ingunn Stefansson
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, Centre for Cancer Biomarkers, Bergen, Norway
| | - Lars A. Akslen
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, Centre for Cancer Biomarkers, Bergen, Norway
| | - Kathrine Woie
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Helga B. Salvesen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Øyvind O. Salvesen
- Unit for Applied Clinical Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Henrica M.J. Werner
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Ingfrid S. Haldorsen
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
49
|
Fernandez-Retana J, Zamudio-Meza H, Rodriguez-Morales M, Pedroza-Torres A, Isla-Ortiz D, Herrera L, Jacobo-Herrera N, Peralta-Zaragoza O, López-Camarillo C, Morales-Gonzalez F, Cantu de Leon D, Pérez-Plasencia C. Gene signature based on degradome-related genes can predict distal metastasis in cervical cancer patients. Tumour Biol 2017. [PMID: 28639897 DOI: 10.1177/1010428317711895] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cervical cancer is one of the leading causes of death in women worldwide, which mainly affects developing countries. The patients who suffer a recurrence and/or progression disease have a higher risk of developing distal metastases. Proteases comprising the degradome given its ability to promote cell growth, migration, and invasion of tissues play an important role during tumor development and progression. In this study, we used high-density microarrays and quantitative reverse transcriptase polymerase chain reaction to evaluate the degradome profile and their inhibitors in 112 samples of patients diagnosed with locally advanced cervical cancer. Clinical follow-up was done during a period of 3 years. Using a correlation analysis between the response to treatment and the development of metastasis, we established a molecular signature comprising eight degradome-related genes (FAM111B, FAM111A, CFB, PSMB8, PSMB9, CASP7, PRSS16, and CD74) with the ability to discriminate patients at risk of distal metastases. In conclusion, present results show that molecular signature obtained from degradome genes can predict the possibility of metastasis in patients with locally advanced cervical cancer.
Collapse
Affiliation(s)
| | | | | | | | - David Isla-Ortiz
- 3 Unit of Cancer Biomedics Research, National Cancer Institute, Mexico City, Mexico
| | - Luis Herrera
- 3 Unit of Cancer Biomedics Research, National Cancer Institute, Mexico City, Mexico
| | - Nadia Jacobo-Herrera
- 4 Biochemistry Unit, National Nutrition Institute of Mexico "Salvador Zubiran," Mexico City, Mexico
| | - Oscar Peralta-Zaragoza
- 5 Direction of Chronic Infections and Cancer, Research Center for Infectious Diseases, National Institute of Public Health, Morelos, México
| | - César López-Camarillo
- 6 Center for Genomic Sciences, National Autonomous University of México, Mexico City, Mexico
| | | | | | - Carlos Pérez-Plasencia
- 1 FES Iztacala, UBIMED, UNAM, Tlalnepantla, Mexico.,2 Genomics Laboratory, National Cancer Institute, Mexico City, Mexico
| |
Collapse
|
50
|
Ha YJ, Tak KH, Kim CW, Roh SA, Choi EK, Cho DH, Kim JH, Kim SK, Kim SY, Kim YS, Kim JC. PSMB8 as a Candidate Marker of Responsiveness to Preoperative Radiation Therapy in Rectal Cancer Patients. Int J Radiat Oncol Biol Phys 2017; 98:1164-1173. [PMID: 28721901 DOI: 10.1016/j.ijrobp.2017.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/06/2017] [Accepted: 03/14/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE The ability to predict individual responsiveness to cancer therapy is urgently needed. This is particularly true for patients with locally advanced rectal cancer (LARC) because a large proportion are resistant to preoperative chemoradiation therapy (CRT). In this study, we sought to identify markers that could predict response by comparing the gene expression profiles of the tumors of patients who received preoperative CRT. METHODS AND MATERIALS The basal gene expression profiles of tumors from 22 LARC patients who were responders (n=9) and nonresponders (n=13) to preoperative CRT were analyzed using RNA sequencing (RNA-Seq). To validate the RNA-Seq findings, real-time reverse transcriptase polymerase chain reaction (RT-PCR) was performed on tumor samples from an additional 40 LARC patients (n=20 responders; n=20 nonresponders). Candidate genes were stably overexpressed or knocked down in colorectal cancer (CRC) cell lines, and the effect on response to radiation was tested in vitro and also in vivo in a mouse xenograft model. RESULTS Eight differentially expressed (>16-fold) genes (B3GALT4, HSPA1B, KRBOX1, PPBP, PPP1R18, PSMB8, SLC39A7, and TAP2) associated with the preoperative CRT response were identified (P<.0005). Among these genes, real-time RT-PCR showed that PSMB8 and SLC39A7 were upregulated in the responsive group of the additional 40 LARC patients. In CRC cell lines, PSMB8 overexpression significantly reduced colony formation and increased the apoptosis-inducing molecules cleaved caspase-3 and cleaved PARP after 6-Gy irradiation. PSMB8 knockdown increased colony formation and decreased caspase-3 activation and cleaved PARP levels after irradiation. SLC39A7 overexpression had no significant effects on irradiated CSC cells. After irradiation of the xenografted mice, tumors that arose from CRC cell line HCT116 overexpressing PSMB8 grew more slowly than did those from HCT116 with vector alone. CONCLUSION These results suggest that PSMB8 is a predictive marker of preoperative radiosensitivity in LARC patients. Clinical validation in a larger cohort is now required.
Collapse
Affiliation(s)
- Ye Jin Ha
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Ka Hee Tak
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Chan Wook Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea
| | - Seon Ae Roh
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Eun Kyung Choi
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea; Department of Radiation Oncology, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong Hyung Cho
- Graduate School of East-West Medical Science, Kyung Hee University, Gyeonggi-Do, Korea
| | - Jeong Hwan Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Seon Kyu Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Seon Young Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yong Sung Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Jin Cheon Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea.
| |
Collapse
|