1
|
Moon DO. Advancing Cancer Therapy: The Role of KIF20A as a Target for Inhibitor Development and Immunotherapy. Cancers (Basel) 2024; 16:2958. [PMID: 39272816 PMCID: PMC11393963 DOI: 10.3390/cancers16172958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
The analysis begins with a detailed examination of the gene expression and protein structure of KIF20A, highlighting its interaction with critical cellular components that influence key processes such as Golgi membrane transport and mitotic spindle assembly. The primary focus is on the development of specific KIF20A inhibitors, detailing their roles and the challenges encountered in enhancing their efficacy, such as achieving specificity, overcoming tumor resistance, and optimizing delivery systems. Additionally, it delves into the prognostic value of KIF20A across multiple cancer types, emphasizing its role as a novel tumor-associated antigen, which lays the groundwork for the development of targeted peptide vaccines. The therapeutic efficacy of these vaccines as demonstrated in recent clinical trials is discussed. Future directions are proposed, including the integration of precision medicine strategies to personalize treatments and the use of combination therapies to improve outcomes. By concentrating on the significant potential of KIF20A as both a direct target for inhibitors and an antigen in cancer vaccines, this review sets a foundation for future research aimed at harnessing KIF20A for effective cancer treatment.
Collapse
Affiliation(s)
- Dong Oh Moon
- Department of Biology Education, Daegu University, 201, Daegudae-ro, Gyeongsan-si 38453, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
2
|
Umar SM, Dev AJR, Kashyap A, Rathee M, Chauhan SS, Sharma A, Prasad CP. 7-amino carboxycoumarin 2 inhibits lactate induced epithelial-to-mesenchymal transition via MPC1 in oral and breast cancer cells. Cell Biol Int 2024; 48:1185-1197. [PMID: 38773713 DOI: 10.1002/cbin.12172] [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: 06/27/2023] [Revised: 03/31/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024]
Abstract
Lactate is an oncometabolite that play important role in tumor aggressiveness. Lactate from the tumor microenvironment (TME) is taken up by cancer cells as an energy resource via mitochondrial oxidative phosphorylation (or OXPHOS). In the present study, by using an online meta-analysis tool we demonstrated that in oral squamous cancer cells (OSCCs) glycolytic and OXPHOS governing genes are overexpressed, like in breast cancer. For experimental demonstration, we treated the OSCC cell line (SCC4) and breast cancer cells (MDA-MB-231) with sodium L-lactate and analyzed its effects on changes in EMT and migration. For the therapeutic intervention of lactate metabolism, we used AZD3965 (an MCT1 inhibitor), and 7ACC2 (an MPC inhibitor). Like breast cancer, oral cancer tissues showed increased transcripts of 12 genes that were previously shown to be associated with glycolysis and OXPHOS. We experimentally demonstrated that L-lactate treatment induced mesenchymal markers and migration of cancer cells, which was significantly neutralized by MPC inhibitor that is, 7ACC2. Such an effect on EMT status was not observed with AZD3965. Furthermore, we showed that lactate treatment increases the MPC1 expression in both cancer cells, and this might be the reason why cancer cells in the high lactate environment are more sensitive to 7ACC2. Overall, our present findings demonstrate that extracellular lactate positively regulates the MPC1 protein expression in cancer cells, thereby putting forward the notion of using 7ACC2 as a potential therapeutic alternative to inhibit malignant oxidative cancers. Future preclinical studies are warranted to validate the present findings.
Collapse
Affiliation(s)
- Sheikh Mohammad Umar
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Arundhathi J R Dev
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Akanksha Kashyap
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Meetu Rathee
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Jin Z, Peng F, Zhang C, Tao S, Xu D, Zhu Z. Expression, regulating mechanism and therapeutic target of KIF20A in multiple cancer. Heliyon 2023; 9:e13195. [PMID: 36798768 PMCID: PMC9925975 DOI: 10.1016/j.heliyon.2023.e13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Kinesin family member 20A (KIF20A) is a member of the kinesin family. It transports chromosomes during mitosis, plays a key role in cell division. Recently, studies proved that KIF20A was highly expressed in cancer. High expression of KIF20A was correlated with poor overall survival (OS). In this review, we summarized all the cancer that highly expressed KIF20A, described the role of KIF20A in cancer. We also organized phase I and phase II clinical trials of KIF20A peptides vaccine. All results indicated that KIF20A was a promising therapeutic target for multiple cancer.
Collapse
Key Words
- ATP, adenosine triphosphate
- BTC, biliary tract cancer
- CPC, chromosomal passenger complex
- CTL, cytotoxic T lymphocyte
- Cancer
- Cdk1, cyclin-dependent kinase 1
- DLG5, discs large MAGUK scaffold protein 5
- EMT, epithelial-mesenchymal transition
- Expression
- FoxM1, forkhead box protein M1
- GC, gastric cancer
- GEM, gemcitabine
- Gli2, glioma-associated oncogene 2
- HLA, human leukocyte antigen
- HNMT, head-and-neck malignant tumor
- IRF, interferon regulatory factor
- JAK, Janus kinase
- KIF20A
- KIF20A, kinesin family member 20A
- LP, long peptide
- MHC I, major histocompatibility complex I
- MKlp2, mitotic kinesin-like protein 2
- Mad2, mitotic arrest deficient 2
- OS, overall survival
- PBMC, peripheral blood mononuclear cell
- Plk1, polo-like kinase 1
- Regulating mechanisms
- Therapeutic target
- circRNA, circular RNA
- miRNA, microRNA
Collapse
Affiliation(s)
- Zheng Jin
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Fei Peng
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, Texas, USA
| | - Chao Zhang
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Shuang Tao
- Department of Otorhinolaryngology Head and Neck Surgery, Longgang Central Hospital of Shenzhen, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Damo Xu
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China,State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong Province, China,Corresponding author. Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China.
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China,Corresponding author. Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Han R, Yang H, Ling C, Lu L. Tiliroside suppresses triple-negative breast cancer as a multifunctional CAXII inhibitor. Cancer Cell Int 2022; 22:368. [PMID: 36424626 PMCID: PMC9685933 DOI: 10.1186/s12935-022-02786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by poor prognosis, early recurrence, and the lack of durable chemotherapy responses and specific targeted treatments. In this preclinical study, we examines Tiliroside (TS, C30H26O13), as one of the major compounds of Tribulus terrestris L. which has been used as an alternative therapy in clinic practice of breast cancer treatment, for its therapeutic use in TNBC. The association between CAXII expression level and survival probability of TNBC patients, and the difference of CAXII expression level between TNBC and normal samples were evaluated by using publicly accessible databases. To determine the anticancer efficacy of TS on TNBC cells, cell proliferation, wound healing, cell invasion, and 3D spheroid formation assays were performed and excellent anticancer activities of TS were displayed. Mouse models further demonstrated that TS significantly reduced the tumor burden and improved survival rate. The properties of TS as a novel CAXII inhibitor have also been evaluated by CAXII activity assay, pHi, pHe and lactate level assay. Further RT-PCR and Caspase-3 activity analyses also revealed the positive regulating effects of TS on E2F1,3/Caspase-3 axis in TNBC cells cultured in 2D or 3D systems. The findings indicate that TS suppresses TNBC progression as a potential novel CAXII inhibitor in preclinical experiments, which warrants further investigation on its therapeutic implications.
Collapse
Affiliation(s)
- Rui Han
- grid.73113.370000 0004 0369 1660Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433 People’s Republic of China ,grid.73113.370000 0004 0369 1660Department of Chinese Medicine, Naval Medical University, Shanghai, 200433 People’s Republic of China ,grid.47100.320000000419368710Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT 06510 USA
| | - Hongxing Yang
- grid.412595.eDepartment of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405 Guangdong People’s Republic of China
| | - Changquan Ling
- grid.73113.370000 0004 0369 1660Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433 People’s Republic of China ,grid.73113.370000 0004 0369 1660Department of Chinese Medicine, Naval Medical University, Shanghai, 200433 People’s Republic of China
| | - Lingeng Lu
- grid.47100.320000000419368710Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT 06510 USA ,School of Medicine, Center for Biomedical Data Science, 200 George Street, New Haven, CT 06511 USA ,grid.47100.320000000419368710Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520-8034 USA ,grid.47100.320000000419368710Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, 06520-8034 New Haven, CT USA
| |
Collapse
|
5
|
Ngalim SH, Yusoff N, Johnson RR, Abdul Razak SR, Chen X, Hobbs JK, Lee YY. A review on mechanobiology of cell adhesion networks in different stages of sporadic colorectal cancer to explain its tumorigenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:63-72. [PMID: 36116549 DOI: 10.1016/j.pbiomolbio.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
Sporadic colorectal cancer (CRC) is strongly linked to extraneous factors, like poor diet and lifestyle, but not to inherent factors like familial genetics. The changes at the epigenomics and signalling pathways are known across the sporadic CRC stages. The catch is that temporal information of the onset, the feedback loop, and the crosstalk of signalling and noise are still unclear. This makes it challenging to diagnose and treat colon cancer effectively with no relapse. Various microbial cells and native cells of the colon, contribute to sporadic CRC development. These cells secrete autocrine and paracrine for their bioenergetics and communications with other cell types. Imbalances of the biochemicals affect the epithelial lining of colon. One side of this epithelial lining is interfacing the dense colon tissue, while the other side is exposed to microbiota and excrement from the lumen. Hence, the epithelial lining is prone to tumorigenesis due to the influence of both biochemical and mechanical cues from its complex surrounding. The role of physical transformations in tumorigenesis have been limitedly discussed. In this context, cellular and tissue structures, and force transductions are heavily regulated by cell adhesion networks. These networks include cell anchoring mechanism to the surrounding, cell structural integrity mechanism, and cell effector molecules. This review will focus on the progression of the sporadic CRC stages that are governed by the underlaying cell adhesion networks within the epithelial cells. Additionally, current and potential technologies and therapeutics that target cell adhesion networks for treatments of sporadic CRC will be incorporated.
Collapse
Affiliation(s)
- Siti Hawa Ngalim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Norwahida Yusoff
- School of Mechanical Engineering, Universiti Sains Malaysia (USM) Engineering Campus, 14300 Nibong Tebal, Penang, Malaysia
| | - Rayzel Renitha Johnson
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Siti Razila Abdul Razak
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Xinyue Chen
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Jamie K Hobbs
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia (USM) Kubang Kerian, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
6
|
Ali A, Unnikannan H, Shafarin J, Bajbouj K, Taneera J, Muhammad JS, Hasan H, Salehi A, Awadallah S, Hamad M. Metformin enhances LDL-cholesterol uptake by suppressing the expression of the pro-protein convertase subtilisin/kexin type 9 (PCSK9) in liver cells. Endocrine 2022; 76:543-557. [PMID: 35237909 DOI: 10.1007/s12020-022-03022-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 02/16/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE Metformin (MF) intake associates with reduced levels of circulating low-density lipoprotein-cholesterol (LDL-C). This has been attributed to the activation of AMPK, which differentially regulates the expression of multiple genes involved in cholesterol synthesis and trafficking. However, the exact mechanism underlying the LDL-C lowering effect of MF remains ambiguous. METHODS MF-treated Hep-G2 and HuH7 cells were evaluated for cell viability and the expression status of key lipid metabolism-related genes along with LDL-C uptake efficiency. RESULTS MF treatment resulted in decreased expression and secretion of PCSK9, increased expression of LDLR and enhanced LDL-C uptake in hepatocytes. It also resulted in increased expression of activated AMPK (p-AMPK) and decreased expression of SREBP2 and HNF-1α proteins. Transcriptomic analysis of MF-treated Hep-G2 cells confirmed these findings and showed that other key lipid metabolism-related genes including those that encode apolipoproteins (APOB, APOC2, APOC3 and APOE), MTTP and LIPC are downregulated. Lastly, MF treatment associated with reduced HMG-CoA reductase expression and activity. CONCLUSIONS These findings suggest that MF treatment reduces circulating LDL-C levels by suppressing PCSK9 expression and enhancing LDLR expression; hence the potential therapeutic utility of MF in hypercholesterolemia.
Collapse
Affiliation(s)
- Amjad Ali
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hema Unnikannan
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Jasmin Shafarin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jalal Taneera
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Haydar Hasan
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Albert Salehi
- Department of Clinical science, UMAS, Clinical Research Center, Lund University, Malmö, Sweden
- Department of Neuroscience and Physiology, Metabolic Research Unit, University of Gothenburg, Gothenburg, Sweden
| | - Samir Awadallah
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - Mawieh Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
7
|
Kong J, Yu G, Si W, Li G, Chai J, Liu Y, Liu J. Identification of a glycolysis-related gene signature for predicting prognosis in patients with hepatocellular carcinoma. BMC Cancer 2022; 22:142. [PMID: 35123420 PMCID: PMC8817563 DOI: 10.1186/s12885-022-09209-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary liver cancer in the world. Although great advances in HCC diagnosis and treatment have been achieved, due to the complicated mechanisms in tumor development and progression, the prognosis of HCC is still dismal. Recent studies have revealed that the Warburg effect is related to the development, progression and treatment of various cancers; however, there have been a few explorations of the relationship between glycolysis and HCC prognosis. Methods mRNA expression profiling was downloaded from public databases. Gene set enrichment analysis (GSEA) was used to explore glycolysis-related genes (GRGs), and the LASSO method and Cox regression analysis were used to identify GRGs related to HCC prognosis and to construct predictive models associated with overall survival (OS) and disease-free survival (DFS). The relationship between the predictive model and the tumor mutation burden (TMB) and tumor immune microenvironment (TIME) was explored. Finally, real-time PCR was used to validate the expression levels of the GRGs in clinical samples and different cell lines. Results Five GRGs (ABCB6, ANKZF1, B3GAT3, KIF20A and STC2) were identified and used to construct gene signatures to predict HCC OS and DFS. Using the median value, HCC patients were divided into low- and high-risk groups. Patients in the high-risk group had worse OS/DFS than those in the low-risk group, were related to higher TMB and were associated with a higher rate of CD4+ memory T cells resting and CD4+ memory T cells activated. Finally, real-time PCR suggested that the five GRGs were all dysregulated in HCC samples compared to adjacent normal samples. Conclusions We identified five GRGs associated with HCC prognosis and constructed two GRGs-related gene signatures to predict HCC OS and DFS. The findings in this study may contribute to the prediction of prognosis and promote HCC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09209-9.
Collapse
|
8
|
KIF2C Is a Novel Prognostic Biomarker and Correlated with Immune Infiltration in Endometrial Cancer. Stem Cells Int 2021; 2021:1434856. [PMID: 34650608 PMCID: PMC8510809 DOI: 10.1155/2021/1434856] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/24/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022] Open
Abstract
Endometrial cancer (EC) is commonly diagnosed cancer in women, and the prognosis of advanced types of EC is extremely poor. Kinesin family member 2C (KIF2C) has been reported as an oncogene in cancers. However, its pathophysiological roles and the correlation with tumor-infiltrating lymphocytes in EC remain unclear. The mRNA and protein levels of KIF2C in EC tissues were detected by qRT-PCR, Western blot (WB), and IHC. CCK8, Transwell, and colony formation assay were applied to assess the effects of KIF2C on cell proliferation, migration, and invasion. Cell apoptosis and cell cycle were analyzed by flow cytometry. The antitumor effect was further validated in the nude mouse xenograft cancer model and humanized mouse model. KIF2C expression was higher in EC. Knockdown of KIF2C prolonged the G1 phases and inhibited EC cell proliferation, migration, and invasion in vitro. Bioinformatics analysis indicated that KIF2C is negatively correlated with the infiltration level of CD8+ T cells but positively with the poor prognosis of EC patients. The apoptosis of CD8+ T cell was inhibited after the knockdown of KIF2C and was further inhibited when it is combined with anti-PD1. Conversely, compared to the knockdown of KIF2C expression alone, the combination of anti-PD1 further promoted the apoptosis of Ishikawa and RL95-2 cells. Moreover, the knockdown of KIF2C inhibited the expression of Ki-67 and the growth of tumors in the nude mouse xenograft cancer model. Our study found that the antitumor efficacy was further evaluated by the combination of anti-PD1 and KIF2C knockdown in a humanized mouse model. This study indicated that KIF2C is a novel prognostic biomarker that determines cancer progression and also a target for the therapy of EC and correlated with tumor immune cells infiltration in EC.
Collapse
|
9
|
Ju G, Zhou T, Zhang R, Pan X, Xue B, Miao S. DUSP12 regulates the tumorigenesis and prognosis of hepatocellular carcinoma. PeerJ 2021; 9:e11929. [PMID: 34414037 PMCID: PMC8344690 DOI: 10.7717/peerj.11929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/18/2021] [Indexed: 01/20/2023] Open
Abstract
Background Dual specificity protein phosphatase (DUSP)12 is an atypical member of the protein tyrosine phosphatase family, which are overexpressed in multiple types of malignant tumors. This protein family protect cells from apoptosis and promotes the proliferation and motility of cells. However, the pathological role of DUSP12 in hepatocellular carcinoma (HCC) is incompletely understood. Methods We analyzed mRNA expression of DUSP12 between HCC and normal liver tissues using multiple online databases, and explored the status of DUSP12 mutants using the cBioPortal database. The correlation between DUSP12 expression and tumor-infiltrating immune cells was demonstrated using the Tumor Immune Estimation Resource database and the Tumor and Immune System Interaction Database. Loss of function assay was utilized to evaluate the role of DUSP12 in HCC progression. Results DUSP12 had higher expression along with mRNA amplification in HCC tissues compared with those in normal liver tissues, which suggested that higher DUSP12 expression predicted shorter overall survival. Analyses of functional enrichment of differentially expressed genes suggested that DUSP12 regulated HCC tumorigenesis, and that knockdown of DUSP12 expression by short hairpin (sh)RNA decreased the proliferation and migration of HCC cells. Besides, DUSP12 expression was positively associated with the infiltration of cluster of differentiation (CD)4+ T cells (especially CD4+ regulatory T cells), macrophages, neutrophils and dendritic cells. DUSP12 expression was positively associated with immune-checkpoint moieties, and was downregulated in a C3 immune-subgroup of HCC (which had the longest survival). Conclusion These data suggest that DUSP12 may have a critical role in the tumorigenesis, infiltration of immune cells, and prognosis of HCC.
Collapse
Affiliation(s)
- Gaoda Ju
- Department of Medical Oncology, Beijing Cancer Hospital, Peking University, Beijing, China
| | - Tianhao Zhou
- Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xiaozao Pan
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Bing Xue
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
10
|
Song W, He X, Gong P, Yang Y, Huang S, Zeng Y, Wei L, Zhang J. Glycolysis-Related Gene Expression Profiling Screen for Prognostic Risk Signature of Pancreatic Ductal Adenocarcinoma. Front Genet 2021; 12:639246. [PMID: 34249078 PMCID: PMC8261051 DOI: 10.3389/fgene.2021.639246] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022] Open
Abstract
Objective: Pancreatic ductal adenocarcinoma (PDAC) is highly lethal. Although progress has been made in the treatment of PDAC, its prognosis remains unsatisfactory. This study aimed to develop novel prognostic genes related to glycolysis in PDAC and to apply these genes to new risk stratification. Methods: In this study, based on the Cancer Genome Atlas (TCGA) PAAD cohort, the expression level of glycolysis-related gene at mRNA level in PAAD and its relationship with prognosis were analyzed. Non-negative matrix decomposition (NMF) clustering was used to cluster PDAC patients according to glycolytic genes. Prognostic glycolytic genes, screened by univariate Cox analysis and LASSO regression analysis were established to calculate risk scores. The differentially expressed genes (DEGs) in the high-risk group and the low-risk group were analyzed, and the signal pathway was further enriched to analyze the correlation between glycolysis genes. In addition, based on RNA-seq data, CIBERSORT was used to evaluate the infiltration degree of immune cells in PDAC samples, and ESTIMATE was used to calculate the immune score of the samples. Results: A total of 319 glycolysis-related genes were retrieved, and all PDAC samples were divided into two clusters by NMF cluster analysis. Survival analysis showed that PDAC patients in cluster 1 had shorter survival time and worse prognosis compared with cluster 2 samples (P < 0.001). A risk prediction model based on 11 glycolysis genes was constructed, according to which patients were divided into two groups, with significantly poorer prognosis in high-risk group than in low-risk group (P < 0.001). Both internal validation and external dataset validation demonstrate good predictive ability of the model (AUC = 0.805, P < 0.001; AUC = 0.763, P < 0.001). Gene aggregation analysis showed that DEGs highly expressed in high-risk group were mainly concentrated in the glycolysis level, immune status, and tumor cell proliferation, etc. In addition, the samples in high-risk group showed immunosuppressed status and infiltrated by relatively more macrophages and less CD8+T cell. Conclusions: These findings suggested that the gene signature based on glycolysis-related genes had potential diagnostic, therapeutic, and prognostic value for PDAC.
Collapse
Affiliation(s)
- Wenjing Song
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xin He
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Pengju Gong
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yan Yang
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Sirui Huang
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yifan Zeng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jingwei Zhang
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Zhu W, Ru L, Ma Z. Identification of a Novel Four-Gene Signature Correlated With the Prognosis of Patients With Hepatocellular Carcinoma: A Comprehensive Analysis. Front Oncol 2021; 11:626654. [PMID: 33777771 PMCID: PMC7994902 DOI: 10.3389/fonc.2021.626654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a common solid-tumor malignancy with high heterogeneity, and accurate prognostic prediction in HCC remains difficult. This analysis was performed to find a novel prognostic multigene signature. Methods The TCGA-LIHC dataset was analyzed for differentially coexpressed genes through weighted gene coexpression network analysis (WGCNA) and differential gene expression analysis. A protein-protein interaction (PPI) network and univariate Cox regression analysis of overall survival (OS) were utilized to identify their prognostic value. Next, we used least absolute shrinkage and selection operator (LASSO) Cox regression to establish a prognostic module. Subsequently, the ICGC-LIRI-JP dataset was applied for further validation. Based on this module, HCC cases were stratified into high-risk and low-risk groups, and differentially expressed genes (DEGs) were identified. Functional enrichment analyses of these DEGs were conducted. Finally, single-sample gene set enrichment analysis (ssGSEA) was performed to explore the correlation between the prognostic signature and immune status. Results A total of 393 differentially coexpressed genes were obtained. Forty differentially coexpressed hub genes were identified using the CytoHubba plugin, and 38 of them were closely correlated with OS. Afterward, we established the four-gene prognostic signature with an acceptable accuracy (area under the curve [AUC] of 1-year survival: 0.739). The ICGC-LIRI-JP dataset also supported the acceptable accuracy (AUC of 1-year survival:0.752). Compared with low-risk cohort, HCC cases in the high-risk cohort had shorter OS, higher tumor grades, and higher T stages. The risk scores of this signature still act as independent predictors of OS (P<0.001). Functional enrichment analyses suggest that it was mainly organelle fission and nuclear division that were enriched. Finally, ssGSEA revealed that this signature is strongly associated with the immune status of HCC patients. Conclusions The proposed prognostic signature of four differentially coexpressed hub genes has satisfactory prognostic ability, providing important insight into the prediction of HCC prognosis.
Collapse
Affiliation(s)
- Weihua Zhu
- Department of Gastroenterology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Lixin Ru
- Department of Radiation Oncology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Zhenchao Ma
- Department of Radiation Oncology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China.,Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Li X, Huang W, Huang W, Wei T, Zhu W, Chen G, Zhang J. Kinesin family members KIF2C/4A/10/11/14/18B/20A/23 predict poor prognosis and promote cell proliferation in hepatocellular carcinoma. Am J Transl Res 2020; 12:1614-1639. [PMID: 32509165 PMCID: PMC7270015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Kinesin superfamily proteins (KIFs) comprise a family of molecular motors that transport membranous organelles and protein complexes in a microtubule- and ATP-dependent manner, with multiple roles in cancers. Little is known about the function of KIFs in hepatocellular carcinoma (HCC). Here, we investigate the roles of KIFs in the prognosis and progression of HCC. Upregulation of eight KIFs (KIF2C, KIF4A, KIF10, KIF11, KIF14, KIF18B, KIF20A, and KIF23) was found to be significantly associated with the tumor stage and pathological tumor grade of HCC patients. Additionally, a high expression of these eight KIFs was significantly associated with shorter overall survival (OS) and disease-free survival (DFS) in patients with HCC. Cox regression analysis showed the mRNA expression levels of these eight KIF members to be independent prognostic factors for worse outcomes in HCC. Moreover, a risk score model based on the mRNA levels of the eight KIF members effectively predicted the OS rate of patients with HCC. Additional experiments revealed that downregulation of each of the eight KIFs effectively decreased the proliferation and increased the G1 arrest of liver cancer cells in vitro. Taken together, these results indicate that KIF2C/4A/10/11/14/18B/20A/23 may serve as prognostic biomarkers for survival and potential therapeutic targets in HCC patients.
Collapse
Affiliation(s)
- Xishan Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
- Department of Interventional Radiology, Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of TechnologyNo. 1 Panfu Road, Guangzhou 510180, China
| | - Weimei Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
| | - Wenbin Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
| | - Weiliang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
| | - Guodong Chen
- Department of Interventional Radiology, Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of TechnologyNo. 1 Panfu Road, Guangzhou 510180, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University253 Industrial Avenue, Guangzhou 510282, China
| |
Collapse
|
13
|
Pinweha P, Phillips CA, Gregory PA, Li X, Chuayboonya P, Mongkolsiri P, Goodall GJ, Jitrapakdee S. MicroRNA-143-3p targets pyruvate carboxylase expression and controls proliferation and migration of MDA-MB-231 cells. Arch Biochem Biophys 2019; 677:108169. [DOI: 10.1016/j.abb.2019.108169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 01/06/2023]
|
14
|
Cai Y, Mei J, Xiao Z, Xu B, Jiang X, Zhang Y, Zhu Y. Identification of five hub genes as monitoring biomarkers for breast cancer metastasis in silico. Hereditas 2019; 156:20. [PMID: 31285741 PMCID: PMC6588910 DOI: 10.1186/s41065-019-0096-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Breast cancer is one of the most common endocrine cancers among females worldwide. Distant metastasis of breast cancer is causing an increasing number of breast cancer-related deaths. However, the potential mechanisms of metastasis and candidate biomarkers remain to be further explored. RESULTS The gene expression profiles of GSE102484 were downloaded from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was used to screen for the most potent gene modules associated with the metastatic risk of breast cancer, and a total of 12 modules were identified based on the analysis. In the most significant module (R2 = 0.68), 21 network hub genes (MM > 0.90) were retained for further analyses. Next, protein-protein interaction (PPI) networks were used to further explore the biomarkers with the most interactions in gene modules. According to the PPI networks, five hub genes (TPX2, KIF2C, CDCA8, BUB1B, and CCNA2) were identified as key genes associated with breast cancer progression. Furthermore, the prognostic value and differential expression of these genes were validated based on data from The Cancer Genome Atlas (TCGA) and Kaplan-Meier (KM) Plotter. Receiver operating characteristic (ROC) curve analysis revealed that the mRNA expression levels of these five hub genes showed excellent diagnostic value for breast cancer and adjacent tissues. Moreover, these five hub genes were significantly associated with worse distant metastasis-free survival (DMFS) in the patient cohort based on KM Plotter. CONCLUSION Five hub genes (TPX2, KIF2C, CDCA8, BUB1B, and CCNA2) associated with the risk of distant metastasis were extracted for further research, which might be used as biomarkers to predict distant metastasis of breast cancer.
Collapse
Affiliation(s)
- Yun Cai
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166 China
| | - Jie Mei
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Zhuang Xiao
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Bujie Xu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Xiaozheng Jiang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Yongjie Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory for Aging & Diseases of Nanjing Medical University, Nanjing Medical University, Nanjing, 211166 China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| |
Collapse
|
15
|
Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: A route to metastasization and a target for therapy. Eur J Med Chem 2019; 171:343-363. [PMID: 30928707 DOI: 10.1016/j.ejmech.2019.03.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed the peculiarities of metabolic reprogramming in tumour onset and progression, and their relevance in cancer therapy. Also, it has been indicated that the metastatic process may depend on the metabolic rewiring and adaptation of cancer cells to the pressure of tumour microenvironment and limiting nutrient availability. The present review gatherers the existent knowledge on the influence of tumour microenvironment and metabolic routes driving metastasis. A focus will be given to glycolysis, fatty acid metabolism, glutaminolysis, and amino acid handling. In addition, the role of metabolic waste driving metastasization will be explored. Finally, we discuss the status of cancer treatment approaches targeting metabolism. This knowledge revision will highlight the critical metabolic targets in metastasis and the chemicals already used in preclinical studies and clinical trials, providing clues that would be further exploited in medicinal chemistry research.
Collapse
Affiliation(s)
- Tiago Ma Carvalho
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Henrique J Cardoso
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|