1
|
Dong SY, Ding S, Meng Z, Zou B. The clinicopathological and prognostic significance of PSMD14 in cancers based on bioinformatics and meta-analysis. Future Sci OA 2024; 10:2409054. [PMID: 39392083 PMCID: PMC11486200 DOI: 10.1080/20565623.2024.2409054] [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: 11/26/2023] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Aim: To evaluate the clinic-pathological features and prognostic value regarding PSMD14 in cancers.Materials & methods: Literature was gathered from public databases until 22 June 2023 to analyze data on survival rates and clinicopathological characteristics associated with PSMD14. TCGA and GEO data were also utilized for validation.Results: Eight reports on seven types of tumors showed that high PSMD14 expression was linked to poorer overall survival and disease-free survival. PSMD14 expression also correlated with larger tumor size, differentiation and metastasis, as well as the effectiveness of various chemotherapy drugs.Conclusion: PSMD14 could serve as a potential biomarker of poor prognosis in cancers, including lung cancer, head and neck squamous cell carcinoma, ovarian cancer, breast cancer and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Shu-Yi Dong
- Department of Oral & Maxillofacial Surgery, Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, Shandong, P. R. China
- Key Laboratory of Oral Maxillofacial-Head & Neck medical biology of Shandong Province, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| | - Shuxin Ding
- Department of Oral & Maxillofacial Surgery, Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, Shandong, P. R. China
- Key Laboratory of Oral Maxillofacial-Head & Neck medical biology of Shandong Province, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| | - Zhen Meng
- Department of Oral & Maxillofacial Surgery, Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, Shandong, P. R. China
- Key Laboratory of Oral Maxillofacial-Head & Neck medical biology of Shandong Province, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| | - Bo Zou
- Department of Oral & Maxillofacial Surgery, Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, Shandong, P. R. China
- Key Laboratory of Oral Maxillofacial-Head & Neck medical biology of Shandong Province, Liaocheng People's Hospital, Liaocheng, Shandong, P. R. China
| |
Collapse
|
2
|
Tang ZQ, Tang YL, Qin K, Li Q, Chen G, Huang YB, Li JJ. Overexpression of proteasome 26S subunit non-ATPase 6 protein and its clinicopathological significance in intrahepatic cholangiocarcinoma. World J Hepatol 2024; 16:1282-1289. [DOI: 10.4254/wjh.v16.i11.1282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/23/2024] [Accepted: 07/23/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Currently, intrahepatic cholangiocarcinoma (ICC) poses a continuing, significant health challenge, but the relationship has yet to be established between ICC and the proteasome 26S subunit non-ATPase 6 (PSMD6).
AIM To investigate the protein expression and clinicopathological significance of PSMD6 in ICC.
METHODS The potential impact of the PSMD6 gene on the growth of ICC cell lines was analyzed using clustered regularly interspaced short palindromic repeat knockout screening technology. Forty-two paired specimens of ICC and adjacent non-cancerous tissues were collected. PSMD6 protein expression was determined by immunohistochemistry. Receiver operating characteristic curve analysis was performed to validate PSMD6 expression level, and its association with ICC patients’ various clinicopathological characteristics was investigated.
RESULTS The PSMD6 gene was found to be essential for the growth of ICC cell lines. PSMD6 protein was significantly overexpressed in ICC tissues (P < 0.001), but showed no significant association with patient age, gender, pathological grade, or tumor-node-metastasis stage (P > 0.05).
CONCLUSION PSMD6 can promote the growth of ICC cells, thus playing a pro-oncogenic role.
Collapse
Affiliation(s)
- Zhong-Qing Tang
- Department of Pathology, Wuzhou Gongren Hospital/The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou 543000, Guangxi Zhuang Autonomous Region, China
| | - Yu-Lu Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Kai Qin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Qi Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Bin Huang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Jian-Jun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
3
|
Zhuang YZ, Tong LQ, Sun XY. Is 26S proteasome non-ATPase regulatory subunit 6 a potential molecular target for intrahepatic cholangiocarcinoma? World J Hepatol 2024; 16:1219-1224. [DOI: 10.4254/wjh.v16.i11.1219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/29/2024] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
In this editorial we comment on the article by Tang et al published in the recent issue of World Journal of Hepatology. Drug therapy of intrahepatic cholangiocarcinoma (iCCA) poses an enormous challenge since only a small proportion of patients demonstrate beneficial responses to therapeutic agents. Thus, there has been a sustained search for novel molecular targets for iCCA. The study by Tang et al evaluated the role of 26S proteasome non-ATPase regulatory subunit 6 (PSMD6), a 19S regulatory subunit of the proteasome, in human iCCA cells and specimens. The authors employed clustered regularly interspaced short palindromic repeat (CRISPR) knockout screening technology integrated with the computational CERES algorithm, and analyzed the human protein atlas (THPA) database and tissue microarrays. The results show that PSMD6 is a gene essential for the proliferation of 17 iCCA cell lines, and PSMD6 protein was overexpressed in iCCA tissues without a significant correlation with the clinicopathological parameters. The authors conclude that PSMD6 may play a promoting role in iCCA. The major limitations and defects of this study are the lack of detailed information of CRISPR knockout screening, in vivo experiments, and a discussion of plausible mechanistic cues, which, therefore, dampen the significance of the results. Further studies are required to verify PSMD6 as a molecular target for developing novel therapeutics for iCCA. In addition, the editorial article summarizes the latest advances in molecular targeted drugs and recently emerging immunotherapy in the clinical management of iCCA, development of proteasome inhibitors for cancer therapy, and advantages of CRISPR screening technology, computational methods, and THPA database as experimental tools for fighting cancer. We hope that these comments may provide some clues for those engaged in the field of basic and clinical research into iCCA.
Collapse
Affiliation(s)
- Yong-Zhi Zhuang
- Department of Oncology, The Fifth Affiliated Hospital of Harbin Medical University, Daqing 163316, Heilongjiang Province, China
| | - Li-Quan Tong
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing 163316, Heilongjiang Province, China
| | - Xue-Ying Sun
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
4
|
Huang Q, Tian R, Yu J, Du W. Identification of PSMD11 as a novel cuproptosis- and immune-related prognostic biomarker promoting lung adenocarcinoma progression. Cancer Med 2024; 13:e7379. [PMID: 38859698 PMCID: PMC11165170 DOI: 10.1002/cam4.7379] [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: 11/25/2023] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Due to the unfavorable prognosis associated with lung adenocarcinoma (LUAD), the development of targeted therapies and immunotherapies is essential. Cuproptosis, an emerging form of regulated cell death, is implicated in mitochondrial metabolism and is induced by copper ions. This study aimed to explore the prognostic value of cuproptosis- and immune-related genes (CIRGs) in LUAD. METHODS We used The Cancer Genome Atlas database to develop a prognostic prediction model for LUAD patients based on eight CIRGs. Using Cox regression analysis, we determined that the CIRG signature is a reliable, independent prognostic factor. We further identified PSMD11 as a critical CIRG and performed immunohistochemistry to study the protein expression levels of PSMD11 in LUAD tissues. We also investigated the impact of PSMD11 on the biological behavior of lung cancer cell lines. RESULTS We found that patients with low PSMD11 expression levels displayed an improved prognosis compared with those with high PSMD11 expression levels. Overexpression of PSMD11 enhanced proliferation, migration, invasion, and tumor growth of lung carcinoma cell line A549, while PSMD11 knockdown diminished proliferation, migration, invasion, and tumor growth of lung carcinoma cell line PC9. Additionally, we discovered that PSMD11 expression was positively correlated with the infiltration of myeloid-derived suppressor cells and the increased expression of immunosuppressive molecules. CONCLUSION These findings suggest that PSMD11 may serve as a valuable prognostic biomarker and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Qiumin Huang
- Department of Immunology, Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin, China
- Department of Laboratory and Diagnosis, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Ran Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Jinxi Yu
- Department of Immunology, Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin, China
| | - Wei Du
- Department of Immunology, Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin, China
| |
Collapse
|
5
|
Li Y, Liu X, Zhao F, Zhao Z, Li X, Wang J, Huang B, Chen A. Comprehensive analysis of PSMD family members and validation of PSMD9 as a potential therapeutic target in human glioblastoma. CNS Neurosci Ther 2024; 30:e14366. [PMID: 37485655 PMCID: PMC10848081 DOI: 10.1111/cns.14366] [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: 02/20/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/25/2023] Open
Abstract
AIMS PSMD family members, as important components of the 26S proteasome, are well known to be involved in protein degradation. However, their role in glioblastoma (GBM) has not been rigorously investigated. We aimed to perform systematic analysis of the expression signature, prognostic significance and functions of PSMD family genes in GBM to reveal potential prognostic markers and new therapeutic targets among PSMD family members. METHODS In this study, we systemically analyzed PSMD family members in terms of their expression profiles, prognostic implications, DNA methylation levels, and genetic alterations; the relationships between their expression levels and immune infiltration and drug sensitivity; and their potential functional enrichment in GBM through bioinformatics assessment. Moreover, in vitro and in vivo experiments were used to validate the biological functions of PSMD9 and its targeted therapeutic effect in GBM. RESULTS The mRNA levels of PSMD5/8/9/10/11/13/14 were higher in GBM than in normal brain tissues, and the mRNA levels of PSMD1/4/5/8/9/11/12 were higher in high-grade glioma (WHO grade III & IV) than in low-grade glioma (WHO grade II). High mRNA expression of PSMD2/6/8/9/12/13/14 and low mRNA expression of PSMD7 were associated with poor overall survival (OS). Multivariate Cox regression analysis identified PSMD2/5/6/8/9/10/11/12 as independent prognostic factors for OS prediction. In addition, the protein-protein interaction network and gene set enrichment analysis results suggested that PSMD family members and their interacting molecules were involved in the regulation of the cell cycle, cell invasion and migration, and other biological processes in GBM. In addition, knockdown of PSMD9 inhibited cell proliferation, invasion and migration and induced G2/M cell cycle arrest in LN229 and A172 GBM cells. Moreover, PSMD9 promoted the malignant progression of GBM in vivo. GBM cell lines with high PSMD9 expression were more resistant to panobinostat, a potent deacetylase inhibitor, than those with low PSMD9 expression. In vitro and in vivo experiments further validated that PSMD9 overexpression rescued the GBM inhibitory effect of panobinostat. CONCLUSION This study provides new insights into the value of the PSMD family in human GBM diagnosis and prognosis evaluation, and we further identified PSMD9 as a potential therapeutic target. These findings may lead to the development of effective therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Yaquan Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Xuemeng Liu
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Feihu Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Zhimin Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Xingang Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Jian Wang
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| | - Bin Huang
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Anjing Chen
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| |
Collapse
|
6
|
Wu X, Zhang X, Liu P, Wang Y. Involvement of Ataxin-3 (ATXN3) in the malignant progression of pancreatic cancer via deubiquitinating HDAC6. Pancreatology 2023; 23:630-641. [PMID: 37460341 DOI: 10.1016/j.pan.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND Pancreatic cancer is a common digestive system cancer and one of the most lethal malignancies worldwide. Ataxin-3 (ATXN3) protein is a deubiquitinating enzyme implicated in the occurrence of diverse human cancers. The potential role of ATXN3 in pancreatic cancer still remains unclear. METHODS ATXN3 was screened from differentially-upregulated genes of GSE71989, GSE27890 and GSE40098 datasets. The mRNA and protein levels of ATXN3 was evaluated in pancreatic cancer samples and cell lines. Through the gain- and loss-of-function experiments, the effects of ATXN3 on cell proliferation, migration and invasion were evaluated using cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU) staining, wound healing and Transwell assays. Subsequently, the interaction between ATXN3 and HDAC6 was confirmed using double immunofluorescence staining, co-immunoprecipitation (co-IP) and proximity ligation assay (PLA). The underlying mechanism of ATXN3 was determined by knockdown of HDAC6 in ATXN3-upregulated pancreatic cancer cells. The function of ATXN3 in vivo was verified through xenograft assay. RESULTS High expression of ATXN3 was found in pancreatic cancer tissues. Increased ATXN3 expression dramatically promoted cell proliferation, migration, and invasion. The malignant phenotypes were suppressed in ATXN3-silenced pancreatic cancer cells. ATXN3 was proved to interact with HDAC6 and regulate its degradation through deubiquitination. Downregulation of HDAC6 inhibited ATXN3-induced development of pancreatic cancer cells through regulating the expression of PCNA, vimentin and E-cadherin. ATXN3 facilitated tumor growth of pancreatic cancer and increased HDAC6 expression in vivo. CONCLUSIONS This study confirmed that ATXN3 facilitated malignant phenotypes of pancreatic cancer via reducing the ubiquitination of HDAC6.
Collapse
Affiliation(s)
- Xin Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, PR China
| | - Xin Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, PR China
| | - Peng Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, PR China
| | - Yao Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, PR China.
| |
Collapse
|
7
|
Ma J, Zhou W, Yuan Y, Wang B, Meng X. PSMD12 interacts with CDKN3 and facilitates pancreatic cancer progression. Cancer Gene Ther 2023; 30:1072-1083. [PMID: 37037907 DOI: 10.1038/s41417-023-00609-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 04/12/2023]
Abstract
Proteasome 26S subunit, non-ATPase 12 (PSMD12) genes have been implicated in several types of malignancies but the role of PSMD12 in pancreatic cancer (PC) remains elusive. Bioinformatics analysis showed that PSMD12 was highly expressed in PC patients and was associated with shorter overall survival. PSMD12 was also shown to be highly expressed in PC tissues and cell lines. Upregulated PSMD12 showed enhanced cell viability, increased colony formation rate and upregulated levels of PCNA and c-Myc, while the inhibition of PSMD12 abated these levels. PSMD12 knockdown promoted cell apoptosis. The results of xenografts in nude mice confirmed that PSMD12 promoted PC tumor growth in vivo. Protein‒protein interaction network and functional enrichment analyses implied that PSMD12 may have a connection with cyclin-dependent kinase inhibitor 3 (CDKN3). Co‑immunoprecipitation and western blot results confirmed that PSMD12 could interact with and abate the ubiquitination level of CDKN3, thus stabilizing the CDKN3 protein. Rescue assays showed that PSMD12 overexpression caused cell proliferation and that knockdown-induced cell apoptosis could be reversed by CDKN3 regulation. This work reveals the essential roles of PSMD12 in the proliferation and apoptosis of PC development. PSMD12 may regulate CDKN3 expression by interacting with and abating the ubiquitination level of CDKN3, thereby participating in the malignant behavior of PC.
Collapse
Affiliation(s)
- Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wenyang Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yifeng Yuan
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Baosheng Wang
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiangpeng Meng
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
8
|
Li X, Li X, Hu Y, Liu O, Wang Y, Li S, Yang Q, Lin B. PSMD8 can serve as potential biomarker and therapeutic target of the PSMD family in ovarian cancer: based on bioinformatics analysis and in vitro validation. BMC Cancer 2023; 23:573. [PMID: 37349676 DOI: 10.1186/s12885-023-11017-8] [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: 09/28/2022] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND The ubiquity-proteasome system is an indispensable mechanism for regulating intracellular protein degradation, thereby affecting human antigen processing, signal transduction, and cell cycle regulation. We used bioinformatics database to predict the expression and related roles of all members of the PSMD family in ovarian cancer. Our findings may provide a theoretical basis for early diagnosis, prognostic assessment, and targeted therapy of ovarian cancer. METHODS GEPIA, cBioPortal, and Kaplan-Meier Plotter databases were used to analyze the mRNA expression levels, gene variation, and prognostic value of PSMD family members in ovarian cancer. PSMD8 was identified as the member with the best prognostic value. The TISIDB database was used to analyze the correlation between PSMD8 and immunity, and the role of PSMD8 in ovarian cancer tissue was verified by immunohistochemical experiments. The relationship of PSMD8 expression with clinicopathological parameters and survival outcomes of ovarian cancer patients was analyzed. The effects of PSMD8 on malignant biological behaviors of invasion, migration, and proliferation of ovarian cancer cells were studied by in vitro experiments. RESULTS The expression levels of PSMD8/14 mRNA in ovarian cancer tissues were significantly higher than those in normal ovarian tissues, and the expression levels of PSMD2/3/4/5/8/11/12/14 mRNA were associated with prognosis. Up-regulation of PSMD4/8/14 mRNA expression was associated with poor OS, and the up-regulation of PSMD2/3/5/8 mRNA expression was associated with poor PFS in patients with ovarian serous carcinomas. Gene function and enrichment analysis showed that PSMD8 is mainly involved in biological processes such as energy metabolism, DNA replication, and protein synthesis. Immunohistochemical experiments showed that PSMD8 was mainly expressed in the cytoplasm and the expression level was correlated with FIGO stage. Patients with high PSMD8 expression had poor prognosis. Overexpression of PSMD8 significantly enhanced the proliferation, migration, and invasion abilities in ovarian cancer cells. CONCLUSION We observed different degrees of abnormal expression of members of PSMD family in ovarian cancer. Among these, PSMD8 was significantly overexpressed in ovarian malignant tissue, and was associated with poor prognosis. PSMDs, especially PSMD8, can serve as potential diagnostic and prognostic biomarkers and therapeutic targets in ovarian cancer.
Collapse
Affiliation(s)
- Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Xinru Li
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Yuexin Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Ouxuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Yuxuan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Siting Li
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
- Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, China.
| |
Collapse
|
9
|
Adler J, Oren R, Shaul Y. Depleting the 19S proteasome regulatory PSMD1 subunit as a cancer therapy strategy. Cancer Med 2023; 12:10781-10790. [PMID: 36934426 DOI: 10.1002/cam4.5775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/18/2022] [Accepted: 02/24/2023] [Indexed: 03/20/2023] Open
Abstract
BACKGROUND Proteasome inhibitors are in use in treating certain types of cancers. These drugs inhibit the catalytic activity of the 20S proteasome, shared by all the different proteasome complexes. Inhibitors of the 26S-associated deubiquitinating activity explicitly inhibit the 26S proteasomal degradation of ubiquitinylated substrates. We have previously reported an alternative strategy that is based on reducing the 26S/20S ratio by depleting PSMD1, 6, and 11, the subunits of the 19S proteasome regulatory complex. Given the addiction of the many cancer types to a high 26S/20S ratio, the depletion strategy is highly effective in killing many aggressive cancer cell lines but not mouse and human immortalized and normal cells. METHODS We used two aggressive cell lines, MDA-MB-231, a triple-negative breast tumor cell line, and OVCAR8, a high-grade ovary adenocarcinoma. Cell culture, mouse MDA-MB-231, OVCAR8 xenografts, and patient-derived ovarian cancer xenograft (PDX) models were transduced with lentivectors expressing PSMD1 shRNA. Tumor size was measured to follow treatment efficacy. RESULTS Using different experimental strategies of expressing shRNA, we found that PSMD1 depletion, either by expressing PSMD1 shRNA in an inducible manner or in a constitutive manner, robustly inhibited MDA-MB-231, and OVCAR8 xenograft tumor growth. Furthermore, the PSMD1 depletion strategy compromised the growth of the PDX of primary ovarian cancer. CONCLUSION Our results suggest that reducing the 26S/20S ratio might be a valuable strategy for treating drug-resistant aggressive types of cancers.
Collapse
Affiliation(s)
- Julia Adler
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Roni Oren
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
10
|
Bustamante HA, Albornoz N, Morselli E, Soza A, Burgos PV. Novel insights into the non-canonical roles of PSMD14/POH1/Rpn11 in proteostasis and in the modulation of cancer progression. Cell Signal 2023; 101:110490. [PMID: 36241058 DOI: 10.1016/j.cellsig.2022.110490] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
PSMD14/POH1/Rpn11 plays a crucial role in cellular homeostasis. PSMD14 is a structural subunit of the lid subcomplex of the proteasome 19S regulatory particle with constitutive deubiquitinase activity. Canonically, PSMD14 removes the full ubiquitin chains with K48-linkages by hydrolyzing the isopeptide bond between the substrate and the C-terminus of the first ubiquitin, a crucial step for the entry of substrates into the catalytic barrel of the 20S proteasome and their subsequent degradation, all in context of the 26S proteasome. However, more recent discoveries indicate PSMD14 DUB activity is not only coupled to the translocation of substrates into the core of 20S proteasome. During the assembly of the lid, activity of PSMD14 has been detected in the context of the heterodimer with PSMD7. Additionally, assembly of the lid subcomplex occurs as an independent event of the base subcomplex and 20S proteasome. This feature opens the possibility that the regulatory particle, free lid subcomplex or the heterodimer PSMD14-PSMD7 might play other physiological roles including a positive function on protein stability through deubiquitination. Here we discuss scenarios that could enhance this PSMD14 non-canonical pathway, the potential impact in preventing degradation of substrates by autophagy highlighting the main findings that support this hypothesis. Finally, we discuss why this information should be investigated in biomedicine specifically with focus on cancer progression to design new therapeutic strategies against the lid subcomplex and the heterodimer PSMD14-PSMD7, highlighting PSMD14 as a druggable target for cancer therapy.
Collapse
Affiliation(s)
- Hianara A Bustamante
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile.
| | - Nicolás Albornoz
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile; Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile.
| | - Eugenia Morselli
- Departamento de Ciencias Básicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile; Centro de Investigación en Autofagia, Santiago, Chile.
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile; Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago 8331150, Chile; Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile.
| | - Patricia V Burgos
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile; Centro de Investigación en Autofagia, Santiago, Chile; Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago 8331150, Chile; Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile.
| |
Collapse
|
11
|
Panels of mRNAs and miRNAs for decoding molecular mechanisms of Renal Cell Carcinoma (RCC) subtypes utilizing Artificial Intelligence approaches. Sci Rep 2022; 12:16393. [PMID: 36180558 PMCID: PMC9525704 DOI: 10.1038/s41598-022-20783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/19/2022] [Indexed: 11/12/2022] Open
Abstract
Renal Cell Carcinoma (RCC) encompasses three histological subtypes, including clear cell RCC (KIRC), papillary RCC (KIRP), and chromophobe RCC (KICH) each of which has different clinical courses, genetic/epigenetic drivers, and therapeutic responses. This study aimed to identify the significant mRNAs and microRNA panels involved in the pathogenesis of RCC subtypes. The mRNA and microRNA transcripts profile were obtained from The Cancer Genome Atlas (TCGA), which were included 611 ccRCC patients, 321 pRCC patients, and 89 chRCC patients for mRNA data and 616 patients in the ccRCC subtype, 326 patients in the pRCC subtype, and 91 patients in the chRCC for miRNA data, respectively. To identify mRNAs and miRNAs, feature selection based on filter and graph algorithms was applied. Then, a deep model was used to classify the subtypes of the RCC. Finally, an association rule mining algorithm was used to disclose features with significant roles to trigger molecular mechanisms to cause RCC subtypes. Panels of 77 mRNAs and 73 miRNAs could discriminate the KIRC, KIRP, and KICH subtypes from each other with 92% (F1-score ≥ 0.9, AUC ≥ 0.89) and 95% accuracy (F1-score ≥ 0.93, AUC ≥ 0.95), respectively. The Association Rule Mining analysis could identify miR-28 (repeat count = 2642) and CSN7A (repeat count = 5794) along with the miR-125a (repeat count = 2591) and NMD3 (repeat count = 2306) with the highest repeat counts, in the KIRC and KIRP rules, respectively. This study found new panels of mRNAs and miRNAs to distinguish among RCC subtypes, which were able to provide new insights into the underlying responsible mechanisms for the initiation and progression of KIRC and KIRP. The proposed mRNA and miRNA panels have a high potential to be as biomarkers of RCC subtypes and should be examined in future clinical studies.
Collapse
|
12
|
Lu L, Wang H, Fang J, Zheng J, Liu B, Xia L, Li D. Overexpression of OAS1 Is Correlated With Poor Prognosis in Pancreatic Cancer. Front Oncol 2022; 12:944194. [PMID: 35898870 PMCID: PMC9309611 DOI: 10.3389/fonc.2022.944194] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background OAS1 expression in pancreatic cancer has been confirmed by many studies. However, the prognostic value and mechanism of OAS1 in pancreatic cancer have not been analyzed. Methods The RNA-seq in pancreatic cancer were obtained by UCSC XENA and GEO database. In addition, immunohistochemical validation and analysis were performed using samples from the 900th hospital. The prognosis of OAS1 was evaluated by timeROC package, Cox regression analysis, and Kaplan-Meier survival curves. Then, the main functional and biological signaling pathways enrichment and its relationship with the abundance of immune cells were analyzed by bioinformatics. Results OAS1 was highly expressed in pancreatic cancer compared with normal pancreatic tissue. High OAS1 expression was associated with poor overall survival (p<0.05). The OAS1 was significantly correlated to TNM staging (p=0.014). The timeROC analysis showed that the AUC of OAS1 was 0.734 for 3-year OS. In addition, the expression of OAS1 was significantly correlated with the abundance of a variety of immune markers. GSEA showed that enhanced signaling pathways associated with OAS1 include Apoptosis, Notch signaling pathway, and P53 signaling pathway. Conclusions OAS1 is a valuable prognostic factor in pancreatic cancer. Moreover, it may be a potential immunotherapeutic target.
Collapse
Affiliation(s)
- Lingling Lu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Huaxiang Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, China
| | - Jian Fang
- Department of Hepatobiliary Medicine, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiaolong Zheng
- Department of Hepatobiliary Disease, The 900th Hospital of the People’s Liberation Army Joint Logistics Support Force, Fuzhou, China
| | - Bang Liu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lei Xia
- Department of Hepatobiliary Medicine, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dongliang Li
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary Disease, The 900th Hospital of the People’s Liberation Army Joint Logistics Support Force, Fuzhou, China
- *Correspondence: Dongliang Li,
| |
Collapse
|
13
|
Barbosa-Silva A, Magalhães M, Da Silva GF, Da Silva FAB, Carneiro FRG, Carels N. A Data Science Approach for the Identification of Molecular Signatures of Aggressive Cancers. Cancers (Basel) 2022; 14:2325. [PMID: 35565454 PMCID: PMC9103663 DOI: 10.3390/cancers14092325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 02/05/2023] Open
Abstract
The main hallmarks of cancer include sustaining proliferative signaling and resisting cell death. We analyzed the genes of the WNT pathway and seven cross-linked pathways that may explain the differences in aggressiveness among cancer types. We divided six cancer types (liver, lung, stomach, kidney, prostate, and thyroid) into classes of high (H) and low (L) aggressiveness considering the TCGA data, and their correlations between Shannon entropy and 5-year overall survival (OS). Then, we used principal component analysis (PCA), a random forest classifier (RFC), and protein-protein interactions (PPI) to find the genes that correlated with aggressiveness. Using PCA, we found GRB2, CTNNB1, SKP1, CSNK2A1, PRKDC, HDAC1, YWHAZ, YWHAB, and PSMD2. Except for PSMD2, the RFC analysis showed a different list, which was CAD, PSMD14, APH1A, PSMD2, SHC1, TMEFF2, PSMD11, H2AFZ, PSMB5, and NOTCH1. Both methods use different algorithmic approaches and have different purposes, which explains the discrepancy between the two gene lists. The key genes of aggressiveness found by PCA were those that maximized the separation of H and L classes according to its third component, which represented 19% of the total variance. By contrast, RFC classified whether the RNA-seq of a tumor sample was of the H or L type. Interestingly, PPIs showed that the genes of PCA and RFC lists were connected neighbors in the PPI signaling network of WNT and cross-linked pathways.
Collapse
Affiliation(s)
- Adriano Barbosa-Silva
- Center for Medical Statistics, Informatics and Intelligent Systems, Institute for Artificial Intelligence, Medical University of Vienna, 1090 Vienna, Austria
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London E14NS, UK
- ITTM S.A.-Information Technology for Translational Medicine, Esch-sur-Alzette, 4354 Luxembourg, Luxembourg
| | - Milena Magalhães
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Gilberto Ferreira Da Silva
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Fabricio Alves Barbosa Da Silva
- Laboratório de Modelagem Computacional de Sistemas Biológicos, Scientific Computing Program, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Flávia Raquel Gonçalves Carneiro
- Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231050, Brazil
| | - Nicolas Carels
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| |
Collapse
|
14
|
Salah Fararjeh A, Al-Khader A, Al-Saleem M, Abu Qauod R. The Prognostic Significance of Proteasome 26S Subunit, Non-ATPase (PSMD) Genes for Bladder Urothelial Carcinoma Patients. Cancer Inform 2022; 20:11769351211067692. [PMID: 34992336 PMCID: PMC8725213 DOI: 10.1177/11769351211067692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023] Open
Abstract
Proteasome a highly sophisticated systems that alter protein structure and
function. Proteasome 26S Subunit, Non-ATPase (PSMD) genes have been implicated
in several types of malignancies. This is the first study to look at how
proteasomal subunits are expressed in patients with bladder urothelial carcinoma
(BLCA). BLCA was used to evaluate the predictive value of PSMD genes (PSMD1 to
PSMD12) in relation to clinicopathological characteristics. PSMD genes’
expression patterns at the mRNA level were analyzed using a variety of
bioinformatics methods, including gene expression profile integrative analysis
(GEPIA), Oncomine, TCGA, and Gene expression Omnibus (GEO) databases. The GEPIA
and TCGA dataset survival plot functions were used to assess the prognostic
significance of PSMD genes. PSMD2, PSMD3, PSMD4, PSMD8, and PSMD11 genes were
significantly overexpressed in BLCA compared with normal bladder tissues. PSMD2
and PSMD8 were significantly overexpressed in BLCA more than other types of
cancer. High level of PSMD2 and PSMD8 predicted shorter overall (OS) and
progression free survival (PFS) in BLCA patients. High level of PSMD2 was
significantly associated with elder age
(P < .001), female gender
(P = .014), tumor grade
(P < .001), and metastasis
(P = .003). PSMD2 has been shown to be an independent
predictor for OS in BLCA patients based on univariate and multivariate analysis
(P < .001). Overall, according to this
study, PSMD2 and PSMD8 could be served as a prognostic biomarker for BLCA
patients.
Collapse
Affiliation(s)
- AbdulFattah Salah Fararjeh
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ali Al-Khader
- Department of Pathology and Forensic Medicine, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan.,Department of Pathology, Al-Hussein Salt Hospital, Al-Salt, Jordan
| | - Malak Al-Saleem
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Rinad Abu Qauod
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
15
|
Xuan DTM, Wu CC, Kao TJ, Ta HDK, Anuraga G, Andriani V, Athoillah M, Chiao CC, Wu YF, Lee KH, Wang CY, Chuang JY. Prognostic and immune infiltration signatures of proteasome 26S subunit, non-ATPase (PSMD) family genes in breast cancer patients. Aging (Albany NY) 2021; 13:24882-24913. [PMID: 34839279 PMCID: PMC8660617 DOI: 10.18632/aging.203722] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022]
Abstract
The complexity of breast cancer includes many interacting biological processes that make it difficult to find appropriate therapeutic treatments. Therefore, identifying potential diagnostic and prognostic biomarkers is urgently needed. Previous studies demonstrated that 26S proteasome delta subunit, non-ATPase (PSMD) family members significantly contribute to the degradation of damaged, misfolded, abnormal, and foreign proteins. However, transcriptional expressions of PSMD family genes in breast cancer still remain largely unexplored. Consequently, we used a holistic bioinformatics approach to explore PSMD genes involved in breast cancer patients by integrating several high-throughput databases, including The Cancer Genome Atlas (TCGA), cBioPortal, Oncomine, and Kaplan-Meier plotter. These data demonstrated that PSMD1, PSMD2, PSMD3, PSMD7, PSMD10, PSMD12, and PSMD14 were expressed at significantly higher levels in breast cancer tissue compared to normal tissues. Notably, the increased expressions of PSMD family genes were correlated with poor prognoses of breast cancer patients, which suggests their roles in tumorigenesis. Meanwhile, network and pathway analyses also indicated that PSMD family genes were positively correlated with ubiquinone metabolism, immune system, and cell-cycle regulatory pathways. Collectively, this study revealed that PSMD family members are potential prognostic biomarkers for breast cancer progression and possible promising clinical therapeutic targets.
Collapse
Affiliation(s)
- Do Thi Minh Xuan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chung-Che Wu
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.,Division of Neurosurgery, Department of Surgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Tzu-Jen Kao
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hoang Dang Khoa Ta
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | - Gangga Anuraga
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan.,Department of Statistics, Faculty of Science and Technology, PGRI Adi Buana University, Surabaya 60234, East Java, Indonesia
| | - Vivin Andriani
- Department of Biological Science, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, East Java, Indonesia
| | - Muhammad Athoillah
- Department of Statistics, Faculty of Science and Technology, PGRI Adi Buana University, Surabaya 60234, East Java, Indonesia
| | - Chung-Chieh Chiao
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | - Yung-Fu Wu
- Department of Medical Research, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan.,Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Yang Wang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | - Jian-Ying Chuang
- The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 11031, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|