1
|
Karthikeyan MC, Srinivasan C, Prabhakar K, Manogar P, Jayaprakash A, Arockiam AJV. Doxorubicin downregulates cell cycle regulatory hub genes in breast cancer cells. Med Oncol 2024; 41:220. [PMID: 39115587 DOI: 10.1007/s12032-024-02468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/03/2024]
Abstract
Breast cancer (BC) is the leading commonly diagnosed cancer in the world, with complex mechanisms underlying its development. There is an urgent need to enlighten key genes as potential therapeutic targets crucial to advancing BC treatment. This study sought to investigate the influence of doxorubicin (DOX) on identified key genes consistent across numerous BC datasets obtained through bioinformatic analysis. To date, a meta-analysis of publicly available coding datasets for expression profiling by array from the Gene Expression Omnibus (GEO) has been carried out. Differentially Expressed Genes (DEGs) identified using GEO2R revealed a total of 23 common DEGs, including nine upregulated genes and 14 downregulated genes among the datasets of three platforms (GPL570, GPL6244, and GPL17586), and the commonly upregulated DEGs, showed significant enrichment in the cell cycle in KEGG analysis. The top nine genes, NUSAP1, CENPF, TPX2, PRC1, ANLN, BUB1B, AURKA, CCNB2, and CDK-1, with higher degree values and MCODE scores in the cytoscape program, were regarded as hub genes. The hub genes were activated in disease states commonly across all the subclasses of BC and correlated with the unfavorable overall survival of BC patients, as verified by the GEPIA and UALCAN databases. qRT-PCR confirmed that DOX treatment resulted in reduced expression of these genes in BC cell lines, which reinforces the evidence that DOX remains an effective drug for BC and suggests that developing modified formulations of doxorubicin to reduce toxicity and resistance, could enhance its efficacy as an effective therapeutic option for BC.
Collapse
Affiliation(s)
- Mano Chitra Karthikeyan
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Chandhru Srinivasan
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Kowsika Prabhakar
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Priyadharshini Manogar
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Abirami Jayaprakash
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Antony Joseph Velanganni Arockiam
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
2
|
Hao L, Li S, Chen G, Nie A, Zeng L, Xiao Z, Hu X. Study on the mechanism of quercetin in Sini Decoction Plus Ginseng Soup to inhibit liver cancer and HBV virus replication through CDK1. Chem Biol Drug Des 2024; 103:e14567. [PMID: 38858165 DOI: 10.1111/cbdd.14567] [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: 11/29/2023] [Revised: 03/18/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND To explore the anti-tumor and anti-virus key active ingredients of Sini Decoction Plus Ginseng Soup (SNRS) and their mechanisms. METHODS The main ingredients of SNRS were analyzed by network pharmacology, and quercetin was identified as the key active ingredient. Then, we obtained the targets of quercetin by using Drugbank, PharmMapper, and SwissTargetPrediction databases. Then, the targets of HBV-related hepatocellular carcinoma (HBV-related HCC) were obtained by using Genecards database. In addition, using the gene expression profiles of HBV-related HCC patients in GEO database and the genes with the greatest survival difference in GEPIA 2 database identified the potential targets of quercetin. In addition, the mechanism of potential genes was studied through GO, KEGG analysis, and PPI network. Using AUC and survival analysis to evaluate the diagnostic and prognostic value of cyclin-dependent kinase 1 (CDK1) and CCNB1. Finally, the effects of quercetin on proliferation of Hep3B and HepG2215 cells and the level of CDK1 and CCNB1 were verified in vitro. ELISA was used to measure the expression levels of hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) after the intervention by quercetin for 24 h and 48 h in HepG2215 cell. RESULTS The first 10 key ingredients of SNRS were identified, and quercetin was the most key ingredient. The 101 potential quercetin targets were identified for the treatment of HBV-related HCC. GO and KEGG showed that 101 potential target enrichment in cancer and cell cycle regulation. By Venn analysis, CDK1 and CCNB1 were intersection targets, which could be used as potential targets for the action of quercetin on HBV-related HCC. Moreover, the expression of CDK1 and CCNB1 was highly expressed in the high-risk group, while the OS rate was low. The 1-year, 3-year and 5-year area under the curve (AUC) curves of CDK1 and CCNB1 were 0.724, 0.676, 0.622 and 0.745, 0.678, 0.634, respectively. Moreover, experimental results also showed that quercetin inhibited cell proliferation and reduced CDK1 expression in Hep3B and HepG2215 cells. The expressions of HBsAg and HBeAg in HepG2215 cell supernatant and cell gradually decreased with the increase of intervention time of quercetin and CDK1 inhibitor. CONCLUSIONS Quercetin is a key ingredient of anti-HBV-related HCC activity and inhibits HBV replication in SNRS by inhibiting CDK1.
Collapse
Affiliation(s)
- Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, P.R. China
| | - Guo Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Aiyu Nie
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Liang Zeng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Zhonghui Xiao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| |
Collapse
|
3
|
Masoudi E, Soleimani M, Zarinfard G, Homayoun M, Bakhtiari M. The effects of chitosan-loaded JQ1 nanoparticles on OVCAR-3 cell cycle and apoptosis-related gene expression. Res Pharm Sci 2024; 19:53-63. [PMID: 39006975 PMCID: PMC11244706 DOI: 10.4103/1735-5362.394820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/14/2023] [Accepted: 01/02/2024] [Indexed: 07/16/2024] Open
Abstract
Background and purpose Ovarian cancer is the deadliest gynecological cancer. Bromodomain and extra terminal domain (BET) proteins play major roles in the regulation of gene expression at the epigenetic level. Jun Qi (JQ1) is a potent inhibitor of BET proteins. Regarding the short half-life and poor pharmacokinetic profile, JQ1 was loaded into newly developed nano-carriers. Chitosan nanoparticles are one of the best and potential polymers in cancer treatment. The present study aimed to build chitosan-JQl nanoparticles (Ch-J-NPs), treat OVCAR-3 cells with Ch-J-NPs, and evaluate the effects of these nanoparticles on cell cycle and apoptosis-associated genes. Experimental approach Ch-J-NPs were synthesized and characterized. The size and morphology of Ch-J-NPs were defined by DLS and FE-SEM techniques. OVCAR-3 cells were cultured and treated with Ch-J-NPs. Then, IC50 was measured using MTT assay. The groups were defined and cells were treated with IC50 concentration of Ch-J-NPs, for 48 h. Finally, cells in different groups were assessed for the expression of genes of interest using quantitative RT-PCR. Findings/Results IC50 values for Ch-J-NPs were 5.625 μg/mL. RT-PCR results demonstrated that the expression of genes associated with cell cycle activity (c-MYC, hTERT, CDK1, CDK4, and CDK6) was significantly decreased following treatment of cancer cells with Ch-J-NPs. Conversely, the expression of caspase-3, and caspase-9 significantly increased. BAX (pro-apoptotic) to BCL2 (anti-apoptotic) expression ratio, also increased significantly after treatment of cells with Ch-J-NPs. Conclusion and implications Ch-J-NPs showed significant anti-cell cyclic and apoptotic effects on OVCAR-3 cells.
Collapse
Affiliation(s)
- Ehsan Masoudi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mitra Soleimani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Giti Zarinfard
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Homayoun
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bakhtiari
- Department of Anatomical Sciences, School of Medicine, Behbahan University of Medical Sciences, Behbahan, Iran
| |
Collapse
|
4
|
Jacob TV, Doshi GM. A Mini-review on Helicobacter pylori with Gastric Cancer and Available Treatments. Endocr Metab Immune Disord Drug Targets 2024; 24:277-290. [PMID: 37622707 DOI: 10.2174/1871530323666230824161901] [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: 05/30/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Helicobacter pylori (H. pylori) is the most thoroughly researched etiological component for stomach inflammation and malignancies. Even though there are conventional recommendations and treatment regimens for eradicating H. pylori, failure rates continue to climb. Antibiotic resistance contributes significantly to misdiagnoses, false positive results, and clinical failures, all of which raise the chance of infection recurrence. This review aims to explore the molecular mechanisms underlying drug resistance in H. pylori and discuss novel approaches for detecting genotypic resistance. Modulation of drug uptake/ efflux, biofilm, and coccoid development. Newer genome sequencing approaches capable of detecting H. pylori genotypic resistance are presented. Prolonged infection in the stomach causes major problems such as gastric cancer. The review discusses how H. pylori causes stomach cancer, recent biomarkers such as miRNAs, molecular pathways in the development of gastric cancer, and diagnostic methods and clinical trials for the disease. Efforts have been made to summarize the recent advancements made toward early diagnosis and novel therapeutic approaches for H. pylori-induced gastric cancer.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
5
|
Frederick MI, Hovey OFJ, Kakadia JH, Shepherd TG, Li SSC, Heinemann IU. Proteomic and Phosphoproteomic Reprogramming in Epithelial Ovarian Cancer Metastasis. Mol Cell Proteomics 2023; 22:100660. [PMID: 37820923 PMCID: PMC10652129 DOI: 10.1016/j.mcpro.2023.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is a high-risk cancer presenting with heterogeneous tumors. The high incidence of EOC metastasis from primary tumors to nearby tissues and organs is a major driver of EOC lethality. We used cellular models of spheroid formation and readherence to investigate cellular signaling dynamics in each step toward EOC metastasis. In our system, adherent cells model primary tumors, spheroid formation represents the initiation of metastatic spread, and readherent spheroid cells represent secondary tumors. Proteomic and phosphoproteomic analyses show that spheroid cells are hypoxic and show markers for cell cycle arrest. Aurora kinase B abundance and downstream substrate phosphorylation are significantly reduced in spheroids and readherent cells, explaining their cell cycle arrest phenotype. The proteome of readherent cells is most similar to spheroids, yet greater changes in the phosphoproteome show that spheroid cells stimulate Rho-associated kinase 1 (ROCK1)-mediated signaling, which controls cytoskeletal organization. In spheroids, we found significant phosphorylation of ROCK1 substrates that were reduced in both adherent and readherent cells. Application of the ROCK1-specific inhibitor Y-27632 to spheroids increased the rate of readherence and altered spheroid density. The data suggest ROCK1 inhibition increases EOC metastatic potential. We identified novel pathways controlled by Aurora kinase B and ROCK1 as major drivers of metastatic behavior in EOC cells. Our data show that phosphoproteomic reprogramming precedes proteomic changes that characterize spheroid readherence in EOC metastasis.
Collapse
Affiliation(s)
- Mallory I Frederick
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Owen F J Hovey
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jenica H Kakadia
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Trevor G Shepherd
- Department of Obstetrics & Gynaecology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Shawn S C Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| | - Ilka U Heinemann
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
6
|
Yang F, Yuan C. KNTC1 knockdown inhibits proliferation and metastases of liver cancer. 3 Biotech 2023; 13:309. [PMID: 37621322 PMCID: PMC10444909 DOI: 10.1007/s13205-023-03722-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/29/2023] [Indexed: 08/26/2023] Open
Abstract
To investigate the mechanism of kinetochore-associated protein 1 (KNTC1) in hepatocellular carcinoma. To query the TCGA database for KNTC1 expression in hepatocellular carcinoma. Detection of protein and mRNA levels of KNTC1 in hepatocellular carcinoma cell lines SK-Hep-1, Huh7, HepG2 and SNU449. Cell proliferation, migration and invasion ability were examined after KNTC1 knockdown in SK-Hep-1 and Huh7. Proteins related to KNTC1 were identified through protein interregulation, and their role in hepatocellular carcinoma was investigated. Our results showed that KNTC1 was significantly upregulated in hepatocellular carcinoma tissues and was associated with poorer prognostic survival. The expression of KNTC1 in hepatocellular carcinoma cell lines SK-Hep-1, Huh7, HepG2 and SNU449 was significantly higher than that in normal hepatocyte line L02. Knockdown of KNTC1 in SK-Hep-1 and Huh7 significantly inhibited cell viability, migration ability and invasion ability. KNTC1 is involved in the regulation of hepatocellular carcinoma through its interaction with cyclin-dependent kinase 1 (CDK1). Knockdown of KNTC1 inhibited CDK1 expression, while CDK1 overexpression was able to rescue the regulation of KNTC1 on the viability, migration and invasive ability of hepatocellular carcinoma cell lines. Knockdown of KNTC1 was found to resulted a cell cycle arrest at the S-phase, potentially through the modulation of CDK1, leading to decreased migration and invasion of hepatocellular carcinoma cells. Moreover, knockdown of KNTC1 in mouse transplanted tumors significantly inhibits tumor growth. Inhibition of high expression of KNTC1 in hepatocellular carcinoma was effective in suppressing the progression of hepatocellular carcinoma cells after knockdown. It may be a potential target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Fan Yang
- Department of Integrated Traditional Chinese and Western Medicine Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No.26 Shengli Street, Jiangan District, Wuhan, 430014 Hubei China
| | - Changjin Yuan
- Department of Integrated Traditional Chinese and Western Medicine Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No.26 Shengli Street, Jiangan District, Wuhan, 430014 Hubei China
| |
Collapse
|
7
|
Huang Y, Fan Y, Zhao Z, Zhang X, Tucker K, Staley A, Suo H, Sun W, Shen X, Deng B, Pierce SR, West L, Yin Y, Emanuele MJ, Zhou C, Bae-Jump V. Inhibition of CDK1 by RO-3306 Exhibits Anti-Tumorigenic Effects in Ovarian Cancer Cells and a Transgenic Mouse Model of Ovarian Cancer. Int J Mol Sci 2023; 24:12375. [PMID: 37569750 PMCID: PMC10418904 DOI: 10.3390/ijms241512375] [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: 06/26/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological malignancy of the reproductive organs in the United States. Cyclin-dependent kinase 1 (CDK1) is an important cell cycle regulatory protein that specifically controls the G2/M phase transition of the cell cycle. RO-3306 is a selective, ATP-competitive, and cell-permeable CDK1 inhibitor that shows potent anti-tumor activity in multiple pre-clinical models. In this study, we investigated the effect of CDK1 expression on the prognosis of patients with ovarian cancer and the anti-tumorigenic effect of RO-3306 in both ovarian cancer cell lines and a genetically engineered mouse model of high-grade serous ovarian cancer (KpB model). In 147 patients with epithelial ovarian cancer, the overexpression of CDK1 was significantly associated with poor prognosis compared with a low expression group. RO-3306 significantly inhibited cellular proliferation, induced apoptosis, caused cellular stress, and reduced cell migration. The treatment of KpB mice with RO-3306 for four weeks showed a significant decrease in tumor weight under obese and lean conditions without obvious side effects. Overall, our results demonstrate that the inhibition of CDK1 activity by RO-3306 effectively reduces cell proliferation and tumor growth, providing biological evidence for future clinical trials of CDK1 inhibitors in ovarian cancer.
Collapse
Affiliation(s)
- Yu Huang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing 400044, China;
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Yali Fan
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Xin Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Katherine Tucker
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Allison Staley
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Hongyan Suo
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Xiaochang Shen
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Boer Deng
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100054, China
| | - Stuart R. Pierce
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Lindsay West
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
| | - Michael J. Emanuele
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.F.); (Z.Z.); (X.Z.); (K.T.); (A.S.); (H.S.); (W.S.); (X.S.); (B.D.); (S.R.P.); (L.W.); (Y.Y.)
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Song Y, Huang T, Pan H, Du A, Wu T, Lan J, Zhou X, Lv Y, Xue S, Yuan K. The influence of COVID-19 on colorectal cancer was investigated using bioinformatics and systems biology techniques. Front Med (Lausanne) 2023; 10:1169562. [PMID: 37457582 PMCID: PMC10348756 DOI: 10.3389/fmed.2023.1169562] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) is a global pandemic and highly contagious, posing a serious threat to human health. Colorectal cancer (CRC) is a risk factor for COVID-19 infection. Therefore, it is vital to investigate the intrinsic link between these two diseases. Methods In this work, bioinformatics and systems biology techniques were used to detect the mutual pathways, molecular biomarkers, and potential drugs between COVID-19 and CRC. Results A total of 161 common differentially expressed genes (DEGs) were identified based on the RNA sequencing datasets of the two diseases. Functional analysis was performed using ontology keywords, and pathway analysis was also performed. The common DEGs were further utilized to create a protein-protein interaction (PPI) network and to identify hub genes and key modules. The datasets revealed transcription factors-gene interactions, co-regulatory networks with DEGs-miRNAs of common DEGs, and predicted possible drugs as well. The ten predicted drugs include troglitazone, estradiol, progesterone, calcitriol, genistein, dexamethasone, lucanthone, resveratrol, retinoic acid, phorbol 12-myristate 13-acetate, some of which have been investigated as potential CRC and COVID-19 therapies. Discussion By clarifying the relationship between COVID-19 and CRC, we hope to provide novel clues and promising therapeutic drugs to treat these two illnesses.
Collapse
Affiliation(s)
- Yujia Song
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyuan Pan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ao Du
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Wu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiang Lan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhou
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Lv
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai Xue
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Zhou C, Zhu S, Li H. miR-195-5p Targets CDK1 To Regulate New DNA Synthesis and Inhibit the Proliferation of Hepatocellular Carcinoma Cells. Appl Biochem Biotechnol 2023; 195:3477-3490. [PMID: 36607481 DOI: 10.1007/s12010-022-04279-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
In cell biological functions and viability, cyclin-dependent kinase 1 (CDK1) takes an essential part. miR-195-5p is pivotal in pathogenesis and development of hepatocellular carcinoma (HCC). But in HCC, whether there is a connection between CDK1 and miR-195-5p remains an unanswered question. In view of this, this study focuses on exploring the mechanism of miR-195-5p/CDK1 in the progression of HCC. The bioinformatics method was applied to predict target mRNA and upstream miRNAs, and further analyzes the signal enrichment pathway of target mRNA. We utilized qRT-PCR and Western blot for detecting expression of genes, as well as their corresponding protein levels. Cell cycle was assayed through flow cytometry. As for the examination of DNA replication, the EDU staining was employed. Cell proliferation was determined via plate colony formation assay. The combined application of bioinformatics analysis and dual-luciferase gene assay assisted in figuring out the binding relationship between miR-195-5p and CDK1. DNA damage was marked by immunofluorescence staining. CDK1 was overexpressed in HCC cells, and enriched in cell cycle and DNA replication pathway. Silencing CDK1 modulated cell cycle of HCC cells and inhibited DNA replication and proliferation. In HCC cells, miR-195-5p targeted and reduced CDK1 expression, inhibited the G1 phase-to-S phase transition, induced DNA damage response, and inhibited DNA replication and proliferation. miR-195-5p targeted CDK1 and repressed synthesis of new DNA in HCC cells, thus restraining HCC cell proliferation.
Collapse
Affiliation(s)
- Chunhui Zhou
- Radiological Intervention Center, Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Kai Fu District Changsha City, Hunan Province, 410008, China
| | - Sujuan Zhu
- The Tumor Hospital of SUMC, Cancer Hospital of Shantou University Medical College, Shantou City, Guangdong Province, 515000, China
| | - Haiping Li
- Radiological Intervention Center, Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Kai Fu District Changsha City, Hunan Province, 410008, China.
| |
Collapse
|
10
|
Xiong Z, Wang M, Wu J, Shi X. Tceal7 Regulates Skeletal Muscle Development through Its Interaction with Cdk1. Int J Mol Sci 2023; 24:ijms24076264. [PMID: 37047236 PMCID: PMC10094454 DOI: 10.3390/ijms24076264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
We have previously reported Tceal7 as a muscle-specific gene that represses myoblast proliferation and promotes myogenic differentiation. The regulatory mechanism of Tceal7 gene expression has been well clarified recently. However, the underlying mechanism of Tceal7 function in skeletal muscle development remains to be elucidated. In the present study, we have generated an MCK 6.5 kb-HA-Tceal7 transgenic model. The transgenic mice are born normally, while they have displayed defects in the growth of body weight and skeletal muscle myofiber during postnatal development. Although four RxL motifs have been identified in the Tceal7 protein sequence, we have not detected any direct protein-protein interaction between Tceal7 and Cyclin A2, Cyclin B1, Cylin D1, or Cyclin E1. Further analysis has revealed the interaction between Tceal7 and Cdk1 instead of Cdk2, Cdk4, or Cdk6. Transgenic overexpression of Tceal7 reduces phosphorylation of 4E-BP1 Ser65, p70S6K1 Thr389, and Cdk substrates in skeletal muscle. In summary, these studies have revealed a novel mechanism of Tceal7 in skeletal muscle development.
Collapse
|
11
|
Zhu S, Al-Mathkour M, Cao L, Khalafi S, Chen Z, Poveda J, Peng D, Lu H, Soutto M, Hu T, McDonald OG, Zaika A, El-Rifai W. CDK1 bridges NF-κB and β-catenin signaling in response to H. pylori infection in gastric tumorigenesis. Cell Rep 2023; 42:112005. [PMID: 36681899 PMCID: PMC9973518 DOI: 10.1016/j.celrep.2023.112005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/31/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Infection with Helicobacter pylori (H. pylori) is the main risk factor for gastric cancer, a leading cause of cancer-related death worldwide. The oncogenic functions of cyclin-dependent kinase 1 (CDK1) are not fully understood in gastric tumorigenesis. Using public datasets, quantitative real-time PCR, western blot, and immunohistochemical (IHC) analyses, we detect high levels of CDK1 in human and mouse gastric tumors. H. pylori infection induces activation of nuclear factor κB (NF-κB) with a significant increase in CDK1 in in vitro and in vivo models (p < 0.01). We confirm active NF-κB binding sites on the CDK1 promoter sequence. CDK1 phosphorylates and inhibits GSK-3β activity through direct binding with subsequent accumulation and activation of β-catenin. CDK1 silencing or pharmacologic inhibition reverses these effects and impairs tumor organoids and spheroid formation. IHC analysis demonstrates a positive correlation between CDK1 and β-catenin. The results demonstrate a mechanistic link between infection, inflammation, and gastric tumorigenesis where CDK1 plays a critical role.
Collapse
Affiliation(s)
- Shoumin Zhu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Marwah Al-Mathkour
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Longlong Cao
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shayan Khalafi
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Zheng Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Julio Poveda
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dunfa Peng
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mohammed Soutto
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Tianling Hu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Oliver G McDonald
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.
| |
Collapse
|
12
|
Wu M, Zhu C, Yang J, Cheng S, Yang X, Gu S, Xu S, Wu Y, Shen W, Huang S, Wang Y. Exploring prognostic indicators in the pathological images of ovarian cancer based on a deep survival network. Front Genet 2023; 13:1069673. [PMID: 36685892 PMCID: PMC9846244 DOI: 10.3389/fgene.2022.1069673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Tumor pathology can assess patient prognosis based on a morphological deviation of tumor tissue from normal. Digitizing whole slide images (WSIs) of tissue enables the use of deep learning (DL) techniques in pathology, which may shed light on prognostic indicators of cancers, and avoid biases introduced by human experience. Purpose: We aim to explore new prognostic indicators of ovarian cancer (OC) patients using the DL framework on WSIs, and provide a valuable approach for OC risk stratification. Methods: We obtained the TCGA-OV dataset from the NIH Genomic Data Commons Data Portal database. The preprocessing of the dataset was comprised of three stages: 1) The WSIs and corresponding clinical data were paired and filtered based on a unique patient ID; 2) a weakly-supervised CLAM WSI-analysis tool was exploited to segment regions of interest; 3) the pre-trained model ResNet50 on ImageNet was employed to extract feature tensors. We proposed an attention-based network to predict a hazard score for each case. Furthermore, all cases were divided into a high-risk score group and a low-risk one according to the median as the threshold value. The multi-omics data of OC patients were used to assess the potential applications of the risk score. Finally, a nomogram based on risk scores and age features was established. Results: A total of 90 WSIs were processed, extracted, and fed into the attention-based network. The mean value of the resulting C-index was 0.5789 (0.5096-0.6053), and the resulting p-value was 0.00845. Moreover, the risk score showed a better prediction ability in the HRD + subgroup. Conclusion: Our deep learning framework is a promising method for searching WSIs, and providing a valuable clinical means for prognosis.
Collapse
Affiliation(s)
- Meixuan Wu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chengguang Zhu
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jiani Yang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaokang Yang
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shilin Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongsong Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Shen
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| | - Shan Huang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| | - Yu Wang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Yu Wang, ; Shan Huang, ; Wei Shen,
| |
Collapse
|
13
|
Zhang X, Hong S, Yang J, Liu J, Wang Y, Peng J, Wang H, Hong L. Purvalanol A induces apoptosis and reverses cisplatin resistance in ovarian cancer. Anticancer Drugs 2023; 34:29-43. [PMID: 35946506 PMCID: PMC9760476 DOI: 10.1097/cad.0000000000001339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Cisplatin (DDP) resistance limits therapeutic efficacy in patients diagnosed with ovarian cancer. Purvalanol A (Pur) is a novel cyclin-dependent kinase (CDK) inhibitor that has been demonstrated to induce apoptosis in various cancer cells. The present study investigated the effect of the combination treatment of Pur and DDP, and the potential anticancer mechanisms in epithelial ovarian cancer (EOC) cells in vitro and in vivo . We found that Pur enhanced the anti-tumor efficacy of cisplatin in EOC cells. The combination of Pur and DDP had more significant effects on apoptosis induction in EOC cells compared with the individual-treatment groups and the control group. We further demonstrated that the combination of Pur and DDP may trigger apoptosis and autophagy in EOC cells by inducing reactive oxygen species (ROS). And the ROS/Akt/mammalian target of rapamycin signaling pathway as a potential mechanism for the initiation of autophagy induced by combination therapy. Similar results were observed in vivo . These results demonstrated that Pur sensitized the response of EOC cells to cisplatin in vitro and in vivo , reversing the resistance to cisplatin in ovarian cancer.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Shasha Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Jiang Yang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Jingchun Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Ying Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Jiaxin Peng
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Haoyu Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| |
Collapse
|
14
|
Bradbury M, Borràs E, Vilar M, Castellví J, Sánchez-Iglesias JL, Pérez-Benavente A, Gil-Moreno A, Santamaria A, Sabidó E. A combination of molecular and clinical parameters provides a new strategy for high-grade serous ovarian cancer patient management. J Transl Med 2022; 20:611. [PMID: 36544142 PMCID: PMC9773449 DOI: 10.1186/s12967-022-03816-7] [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: 08/30/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High-grade serous carcinoma (HGSC) is the most common and deadly subtype of ovarian cancer. Although most patients will initially respond to first-line treatment with a combination of surgery and platinum-based chemotherapy, up to a quarter will be resistant to treatment. We aimed to identify a new strategy to improve HGSC patient management at the time of cancer diagnosis (HGSC-1LTR). METHODS A total of 109 ready-available formalin-fixed paraffin-embedded HGSC tissues obtained at the time of HGSC diagnosis were selected for proteomic analysis. Clinical data, treatment approach and outcomes were collected for all patients. An initial discovery cohort (n = 21) were divided into chemoresistant and chemosensitive groups and evaluated using discovery mass-spectrometry (MS)-based proteomics. Proteins showing differential abundance between groups were verified in a verification cohort (n = 88) using targeted MS-based proteomics. A logistic regression model was used to select those proteins able to correctly classify patients into chemoresistant and chemosensitive. The classification performance of the protein and clinical data combinations were assessed through the generation of receiver operating characteristic (ROC) curves. RESULTS Using the HGSC-1LTR strategy we have identified a molecular signature (TKT, LAMC1 and FUCO) that combined with ready available clinical data (patients' age, menopausal status, serum CA125 levels, and treatment approach) is able to predict patient response to first-line treatment with an AUC: 0.82 (95% CI 0.72-0.92). CONCLUSIONS We have established a new strategy that combines molecular and clinical parameters to predict the response to first-line treatment in HGSC patients (HGSC-1LTR). This strategy can allow the identification of chemoresistance at the time of diagnosis providing the optimization of therapeutic decision making and the evaluation of alternative treatment strategies. Thus, advancing towards the improvement of patient outcome and the individualization of HGSC patients' care.
Collapse
Affiliation(s)
- Melissa Bradbury
- grid.473715.30000 0004 6475 7299Centre de Regulació Genòmica, Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Dr Aiguader 88, 08003 Barcelona, Spain ,grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.411083.f0000 0001 0675 8654Department of Gynecology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Eva Borràs
- grid.473715.30000 0004 6475 7299Centre de Regulació Genòmica, Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Dr Aiguader 88, 08003 Barcelona, Spain
| | - Marta Vilar
- grid.473715.30000 0004 6475 7299Centre de Regulació Genòmica, Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain ,grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Josep Castellví
- grid.411083.f0000 0001 0675 8654Department of Pathology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - José Luis Sánchez-Iglesias
- grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.411083.f0000 0001 0675 8654Department of Gynecology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Assumpció Pérez-Benavente
- grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.411083.f0000 0001 0675 8654Department of Gynecology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Antonio Gil-Moreno
- grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.411083.f0000 0001 0675 8654Department of Gynecology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, Avenida de Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Anna Santamaria
- grid.7080.f0000 0001 2296 0625Biomedical Research Group in Gynecology, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Vall, d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain ,grid.7080.f0000 0001 2296 0625Cell Cycle and Cancer Laboratory, Biomedical Research Group in Urology, Vall Hebron Institut de Recerca, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Eduard Sabidó
- grid.473715.30000 0004 6475 7299Centre de Regulació Genòmica, Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Dr Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
15
|
Dastjerdi S, Haghparast A, Amroabadi JM, Dolatabadi NF, Mirzaei S, Zamani A, Hashemi M, Mahdevar M, Ghaedi K. Elevated CDK5R1 expression associated with poor prognosis, proliferation, and drug resistance in colorectal and breast malignancies: CDK5R1 as an oncogene in cancers. Chem Biol Interact 2022; 368:110190. [PMID: 36162454 DOI: 10.1016/j.cbi.2022.110190] [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: 08/23/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Studies have shown that the CDK5R1 gene could have a part in some types of cancer. This study sought to examine the relationship between CDK5R1 expression and prognosis and medication resistance in 13 commonly occurring cancers. METHOD The cancer genome atlas data and clinical data were utilized to assess the role of CDK5R1 in malignancies. The expression data of 13 cancers were also integrated and used for the co-expression network. The relationship between CDK5R1 expression and drug resistance and sensitivity was evaluated using pharmacogenomics data. The colorectal cancer (CRC) and breast cancer (BC) were used to confirm the results through the RT-qPCR method. RESULTS With the exception of gastric cancer, all common malignancies showed an increase in CDK5R1 expression. Also, outcomes of sensitivity and specificity showed that CDK5R1 level could be a really good potential biomarker. Additionally, CDK5R1 expression was higher in CRC and BC samples compared to adjacent normal, according to RT-qPCR data. In six types of tumors and combined data, a poor prognosis was associated with increased CDK5R1 expression. The CDK5R1-associated genes were connected to the primary oncogenic pathways in cancer cells, according to the co-expression network. Also, CDK5R1 level was significantly linked to the resistance and sensitivity of several chemotherapy drugs and caused the highest resistance to cyclophosphamide. CONCLUSION CDK5R1 expression is upregulated in 12 prevalent cancers and can play an oncogenic role. Also, this gene's expression could be used as a biomarker to predict patient survival and medication resistance.
Collapse
Affiliation(s)
- Shaghayegh Dastjerdi
- Department of Cellular and Molecular Sciences, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | | | | | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Atefeh Zamani
- Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
16
|
Yang L, Gilbertsen A, Smith K, Xia H, Higgins L, Guerrero C, Henke CA. Proteomic analysis of the IPF mesenchymal progenitor cell nuclear proteome identifies abnormalities in key nodal proteins that underlie their fibrogenic phenotype. Proteomics 2022; 22:e2200018. [PMID: 35633524 PMCID: PMC9541064 DOI: 10.1002/pmic.202200018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
IPF is a progressive fibrotic lung disease whose pathogenesis remains incompletely understood. We have previously discovered pathologic mesenchymal progenitor cells (MPCs) in the lungs of IPF patients. IPF MPCs display a distinct transcriptome and create sustained interstitial fibrosis in immune deficient mice. However, the precise pathologic alterations responsible for this fibrotic phenotype remain to be uncovered. Quantitative mass spectrometry and interactomics is a powerful tool that can define protein alterations in specific subcellular compartments that can be implemented to understand disease pathogenesis. We employed quantitative mass spectrometry and interactomics to define protein alterations in the nuclear compartment of IPF MPCs compared to control MPCs. We identified increased nuclear levels of PARP1, CDK1, and BACH1. Interactomics implicated PARP1, CDK1, and BACH1 as key hub proteins in the DNA damage/repair, differentiation, and apoptosis signaling pathways respectively. Loss of function and inhibitor studies demonstrated important roles for PARP1 in DNA damage/repair, CDK1 in regulating IPF MPC stemness and self-renewal, and BACH1 in regulating IPF MPC viability. Our quantitative mass spectrometry studies combined with interactomic analysis uncovered key roles for nuclear PARP1, CDK1, and BACH1 in regulating IPF MPC fibrogenicity.
Collapse
Affiliation(s)
- Libang Yang
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Adam Gilbertsen
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Karen Smith
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Hong Xia
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - LeeAnn Higgins
- Center for Mass Spectrometry and ProteomicsUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Candace Guerrero
- Center for Mass Spectrometry and ProteomicsUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Craig A. Henke
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
17
|
Heo J, Lee J, Nam YJ, Kim Y, Yun H, Lee S, Ju H, Ryu CM, Jeong SM, Lee J, Lim J, Cho YM, Jeong EM, Hong B, Son J, Shin DM. The CDK1/TFCP2L1/ID2 cascade offers a novel combination therapy strategy in a preclinical model of bladder cancer. Exp Mol Med 2022; 54:801-811. [PMID: 35729325 PMCID: PMC9256744 DOI: 10.1038/s12276-022-00786-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 01/03/2023] Open
Abstract
Aberrant activation of embryogenesis-related molecular programs in urothelial bladder cancer (BC) is associated with stemness features related to oncogenic dedifferentiation and tumor metastasis. Recently, we reported that overexpression of transcription factor CP2-like protein-1 (TFCP2L1) and its phosphorylation at Thr177 by cyclin-dependent kinase-1 (CDK1) play key roles in regulating bladder carcinogenesis. However, the clinical relevance and therapeutic potential of this novel CDK1-TFCP2L1 molecular network remain elusive. Here, we demonstrated that inhibitor of DNA binding-2 (ID2) functions as a crucial mediator by acting as a direct repressive target of TFCP2L1 to modulate the stemness features and survival of BC cells. Low ID2 and high CDK1 expression were significantly associated with unfavorable clinical characteristics. TFCP2L1 downregulated ID2 by directly binding to its promoter region. Consistent with these findings, ectopic expression of ID2 or treatment with apigenin, a chemical activator of ID2, triggered apoptosis and impaired the proliferation, suppressed the stemness features, and reduced the invasive capacity of BC cells. Combination treatment with the specific CDK1 inhibitor RO-3306 and apigenin significantly suppressed tumor growth in an orthotopic BC xenograft animal model. This study demonstrates the biological role and clinical utility of ID2 as a direct target of the CDK1-TFCP2L1 pathway for modulating the stemness features of BC cells. Combination therapy with apigenin, a powerful antioxidant found in plants such as parsley and camomile, and a drug that inhibits the cell cycle protein CDK1 shows promise for developing therapies for bladder cancer (BC). Switching on genes usually activated in stem cells can cause cancer, including BC. Although CDK1 was known to activate one of these genes in BC cells, no way to suppress the activation had been identified. Jinbeom Heo at University of Ulsan College of Medicine, South Korea, and coworkers investigated CDK1’s role in BC. They found that the transcription factor activated by CDK1 suppressed a protein, ID2, that suppressed stem cell-like characteristics. Simultaneously suppressing CDK1 and boosting ID2 with apigenin strongly repressed tumor growth in a mouse model. These results help point the way to developing new treatment options for BC patients.
Collapse
Affiliation(s)
- Jinbeom Heo
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinyoung Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yun Ji Nam
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - YongHwan Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - HongDuck Yun
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seungun Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyein Ju
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chae-Min Ryu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Center for Cell Therapy, Asan Medical Center, Seoul, Korea
| | - Seon Min Jeong
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinwon Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jisun Lim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju, Korea.,Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Bio-Health Materials Core-Facility Center and Practical Translational Research Center, Jeju National University, Jeju, Korea
| | - Bumsik Hong
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jaekyoung Son
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Dong-Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Center for Cell Therapy, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
18
|
Teo T, Kasirzadeh S, Albrecht H, Sykes MJ, Yang Y, Wang S. An Overview of CDK3 in Cancer: Clinical Significance and Pharmacological Implications. Pharmacol Res 2022; 180:106249. [DOI: 10.1016/j.phrs.2022.106249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
|
19
|
Li W, Feng SS, Wu H, Deng J, Zhou WY, Jia MX, Shi Y, Ma L, Zeng XX, Zuberi Z, Fu D, Liu X, Chen Z. Comprehensive Analysis of CDK1-Associated ceRNA Network Revealing the Key Pathways LINC00460/LINC00525-Hsa-Mir-338-FAM111/ZWINT as Prognostic Biomarkers in Lung Adenocarcinoma Combined with Experiments. Cells 2022; 11:cells11071220. [PMID: 35406786 PMCID: PMC8997540 DOI: 10.3390/cells11071220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the leading cause of cancer deaths worldwide, and effective biomarkers are still lacking for early detection and prognosis prediction. Here, based on gene expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA), 806 long non-coding RNAs (lncRNAs), 122 microRNAs (miRNAs) and 1269 mRNAs associated with CDK1 were identified. The regulatory axis of LINC00460/LINC00525-hsa-mir-338-FAM111B/ZWINT was determined according to the correlation between gene expression and patient prognosis. The abnormal up-regulation of FAM111B/ZWINT in LUAD was related to hypomethylation. Furthermore, immune infiltration analysis suggested FAM111B/ZWINT could affect the development and prognosis of cancer by regulating the LUAD immune microenvironment. EMT feature analysis suggested that FAM111B/ZWINT promoted tumor spread through the EMT process. Functional analysis showed FAM111B/ZWINT was involved in cell cycle events such as DNA replication and chromosome separation. We analyzed the HERB and GSCALite databases to identify potential target medicines that may play a role in the treatment of LUAD. Finally, the expression of LINC00460/LINC00525-hsa-mir-338-FAM111B/ZWINT axis was verified in LUAD cells by RT-qPCR, and these results were consistent with bioinformatics analysis. Overall, we constructed a CDK1-related ceRNA network and revealed the LINC00460/LINC00525-hsa-mir-338-FAM111/ZWINT pathways as potential diagnostic biomarkers or therapeutic targets of LUAD.
Collapse
Affiliation(s)
- Wen Li
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Shan-Shan Feng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Hao Wu
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Jing Deng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Wang-Yan Zhou
- Department of Medical Record, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang 421001, China;
| | - Ming-Xi Jia
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Yi Shi
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Liang Ma
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Xiao-Xi Zeng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Zavuga Zuberi
- Department of Science and Laboratory Technology, Dar es Salaam Institute of Technology, Dar es Salaam P.O. Box 2958, Tanzania;
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China;
| | - Xiang Liu
- Department of Thoracic Surgery, Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang 421001, China
- Correspondence: (X.L.); (Z.C.); Tel.: +86-0734-889-9990 (X.L.); +86-158-6971-6968 (Z.C.)
| | - Zhu Chen
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
- Correspondence: (X.L.); (Z.C.); Tel.: +86-0734-889-9990 (X.L.); +86-158-6971-6968 (Z.C.)
| |
Collapse
|
20
|
Bhattacharyya N, Gupta S, Sharma S, Soni A, Bagabir SA, Bhattacharyya M, Mukherjee A, Almalki AH, Alkhanani MF, Haque S, Ray AK, Malik MZ. CDK1 and HSP90AA1 Appear as the Novel Regulatory Genes in Non-Small Cell Lung Cancer: A Bioinformatics Approach. J Pers Med 2022; 12:jpm12030393. [PMID: 35330393 PMCID: PMC8955443 DOI: 10.3390/jpm12030393] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/08/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is one of the most invasive cancers affecting over a million of the population. Non-small cell lung cancer (NSCLC) constitutes up to 85% of all lung cancer cases, and therefore, it is essential to identify predictive biomarkers of NSCLC for therapeutic purposes. Here we use a network theoretical approach to investigate the complex behavior of the NSCLC gene-regulatory interactions. We have used eight NSCLC microarray datasets GSE19188, GSE118370, GSE10072, GSE101929, GSE7670, GSE33532, GSE31547, and GSE31210 and meta-analyzed them to find differentially expressed genes (DEGs) and further constructed a protein–protein interaction (PPI) network. We analyzed its topological properties and identified significant modules of the PPI network using cytoscape network analyzer and MCODE plug-in. From the PPI network, top ten genes of each of the six topological properties like closeness centrality, maximal clique centrality (MCC), Maximum Neighborhood Component (MNC), radiality, EPC (Edge Percolated Component) and bottleneck were considered for key regulator identification. We further compared them with top ten hub genes (those with the highest degrees) to find key regulator (KR) genes. We found that two genes, CDK1 and HSP90AA1, were common in the analysis suggesting a significant regulatory role of CDK1 and HSP90AA1 in non-small cell lung cancer. Our study using a network theoretical approach, as a summary, suggests CDK1 and HSP90AA1 as key regulator genes in complex NSCLC network.
Collapse
Affiliation(s)
| | - Samriddhi Gupta
- Department of Biochemistry, University of Hyderabad, Hyderabad 500046, India;
| | - Shubham Sharma
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.S.); (A.S.)
| | - Aman Soni
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.S.); (A.S.)
| | - Sali Abubaker Bagabir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia;
| | - Malini Bhattacharyya
- Department of Environmental Plant Biology, Hemvati Nandan Bahuguna, Garhwal Central University, Srinagar 246174, India;
| | - Atreyee Mukherjee
- Department of Life Sciences, Presidency University, Kolkata 700073, India;
| | - Atiah H. Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
- Addiction and Neuroscience Research Unit, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Mustfa F. Alkhanani
- Emergency Service Department, College of Applied Sciences, Al Maarefa University, Riyadh 11597, Saudi Arabia;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Faculty of Medicine, Bursa Uludağ University, Görükle Campus, Bursa 16059, Turkey
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University Delhi, New Delhi 110007, India
- Correspondence: (A.K.R.); (M.Z.M.)
| | - Md. Zubbair Malik
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.S.); (A.S.)
- Correspondence: (A.K.R.); (M.Z.M.)
| |
Collapse
|
21
|
Elgiushy HR, Mohamed SH, Taha H, Sawaf H, Hassan Z, Abou-Taleb NA, El-labbad EM, Hassan AS, Abouzid KA, Hammad SF. Identification of a promising hit from a new series of pyrazolo[1,5-a]pyrimidine based compounds as a potential anticancer agent with potent CDK1 inhibitory and pro-apoptotic properties through a multistep in vitro assessment. Bioorg Chem 2022; 120:105646. [DOI: 10.1016/j.bioorg.2022.105646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022]
|
22
|
Gouda G, Gupta MK, Donde R, Behera L, Vadde R. Metabolic pathway-based target therapy to hepatocellular carcinoma: a computational approach. THERANOSTICS AND PRECISION MEDICINE FOR THE MANAGEMENT OF HEPATOCELLULAR CARCINOMA, VOLUME 2 2022:83-103. [DOI: 10.1016/b978-0-323-98807-0.00003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
23
|
Kong S, Liu J, Zhang B, Lv F, Yu Y, Qin T. MicroRNA-337-3p impedes breast cancer progression by targeting cyclin-dependent kinase 1. Oncol Lett 2021; 23:15. [PMID: 34820014 PMCID: PMC8607341 DOI: 10.3892/ol.2021.13133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/19/2021] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) function as key regulators in breast cancer (BC). The present study aimed to verify the function and molecular regulation of miR-337-3p in BC cells. Bioinformatics analysis was performed to screen key genes and miRNAs associated with BC. Reverse transcription-quantitative PCR and western blot analyses were performed to detect RNA and protein expression levels. Cell Counting Kit-8, BrdU and cell adhesion assays, and flow cytometric analysis were performed to assess the biological behaviors of BC cells. The dual-luciferase reporter, RNA pull-down assays, and Pearson's correlation analysis were performed to determine the association between miRNAs and mRNAs. Bioinformatics analysis revealed that miR-337-3p and cyclin-dependent kinase 1 (CDK1) acted as key regulators in BC. In addition, miR-337-3p was expressed at low levels in BC cells and tissues, which suppressed BC progression. CDK1 expression was upregulated in BC cells and tissues, which was associated with increased cell proliferation and adhesion, as well as decreased apoptosis in BC. Notably, miR-337-3p targeted CDK1 to inhibit BC cell progression. Taken together, the results of the present study suggest that miR-337-3p plays a tumor-suppressive role in BC by targeting CDK1.
Collapse
Affiliation(s)
- Shuxin Kong
- Department of Breast Surgery, The People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jianyang Liu
- Department of Aortic Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People's Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Bin Zhang
- Department of Breast Surgery, The People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Feng Lv
- Department of Breast Surgery, The People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yang Yu
- Department of Breast Surgery, The People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Tao Qin
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
24
|
Chen X, Xia Z, Wan Y, Huang P. Identification of hub genes and candidate drugs in hepatocellular carcinoma by integrated bioinformatics analysis. Medicine (Baltimore) 2021; 100:e27117. [PMID: 34596112 PMCID: PMC8483840 DOI: 10.1097/md.0000000000027117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 08/14/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third cancer-related cause of death in the world. Until now, the involved mechanisms during the development of HCC are largely unknown. This study aims to explore the driven genes and potential drugs in HCC. METHODS Three mRNA expression datasets were used to analyze the differentially expressed genes (DEGs) in HCC. The bioinformatics approaches include identification of DEGs and hub genes, Gene Ontology terms analysis and Kyoto encyclopedia of genes and genomes enrichment analysis, construction of protein-protein interaction network. The expression levels of hub genes were validated based on The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis, and the Human Protein Atlas. Moreover, overall survival and disease-free survival analysis of HCC patients were further conducted by Kaplan-Meier plotter and Gene Expression Profiling Interactive Analysis. DGIdb database was performed to search the candidate drugs for HCC. RESULTS A total of 197 DEGs were identified. The protein-protein interaction network was constructed using Search Tool for the Retrieval of Interacting Genes software, 10 genes were selected by Cytoscape plugin cytoHubba and served as hub genes. These 10 genes were all closely related to the survival of HCC patients. DGIdb database predicted 29 small molecules as the possible drugs for treating HCC. CONCLUSION Our study provides some new insights into HCC pathogenesis and treatments. The candidate drugs may improve the efficiency of HCC therapy in the future.
Collapse
Affiliation(s)
- Xiaolong Chen
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhixiong Xia
- Department of Pathology, The Center Hospital of Wuhan, Hubei, China
| | - Yafeng Wan
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Huang
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Qiang R, Zhao Z, Tang L, Wang Q, Wang Y, Huang Q. Identification of 5 Hub Genes Related to the Early Diagnosis, Tumour Stage, and Poor Outcomes of Hepatitis B Virus-Related Hepatocellular Carcinoma by Bioinformatics Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9991255. [PMID: 34603487 PMCID: PMC8483908 DOI: 10.1155/2021/9991255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/25/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The majority of primary liver cancers in adults worldwide are hepatocellular carcinomas (HCCs, or hepatomas). Thus, a deep understanding of the underlying mechanisms for the pathogenesis and carcinogenesis of HCC at the molecular level could facilitate the development of novel early diagnostic and therapeutic treatments to improve the approaches and prognosis for HCC patients. Our study elucidates the underlying molecular mechanisms of HBV-HCC development and progression and identifies important genes related to the early diagnosis, tumour stage, and poor outcomes of HCC. METHODS GSE55092 and GSE121248 gene expression profiling data were downloaded from the Gene Expression Omnibus (GEO) database. There were 119 HCC samples and 128 nontumour tissue samples. GEO2R was used to screen for differentially expressed genes (DEGs). Volcano plots and Venn diagrams were drawn by using the ggplot2 package in R. A heat map was generated by using Heatmapper. By using the clusterProfiler R package, KEGG and GO enrichment analyses of DEGs were conducted. Through PPI network construction using the STRING database, key hub genes were identified by cytoHubba. Finally, KM survival curves and ROC curves were generated to validate hub gene expression. RESULTS By GO enrichment analysis, 694 DEGs were enriched in the following GO terms: organic acid catabolic process, carboxylic acid catabolic process, carboxylic acid biosynthetic process, collagen-containing extracellular matrix, blood microparticle, condensed chromosome kinetochore, arachidonic acid epoxygenase activity, arachidonic acid monooxygenase activity, and monooxygenase activity. In the KEGG pathway enrichment analysis, DEGs were enriched in arachidonic acid epoxygenase activity, arachidonic acid monooxygenase activity, and monooxygenase activity. By PPI network construction and analysis of hub genes, we selected the top 10 genes, including CDK1, CCNB2, CDC20, BUB1, BUB1B, CCNB1, NDC80, CENPF, MAD2L1, and NUF2. By using TCGA and THPA databases, we found five genes, CDK1, CDC20, CCNB1, CENPF, and MAD2L1, that were related to the early diagnosis, tumour stage, and poor outcomes of HBV-HCC. CONCLUSIONS Five abnormally expressed hub genes of HBV-HCC are informative for early diagnosis, tumour stage determination, and poor outcome prediction.
Collapse
Affiliation(s)
- Rui Qiang
- Department of Infectious Diseases, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100053, China
| | - Zitong Zhao
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Lu Tang
- Department of Traditional Chinese Medicine, Kunming Second People's Hospital, Kunming, 650000 Yunnan, China
| | - Qian Wang
- Department of Basic Medicine, Yunnan University of Business Management, Kunming, 650000 Yunnan, China
| | - Yanhong Wang
- Department of Second Internal Medicine, Chongming Branch of Yueyang Integrated Hospital of Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Chongming, 202150 Shanghai, China
| | - Qian Huang
- Department of Oncology, Shanghai Xinhua Hospital Chongming Branch Affiliated to Shanghai Jiaotong University School of Medicine, 25 Nanmen Road, Chengqiao Town, Chongming District, 200000 Shanghai, China
| |
Collapse
|
26
|
Li DF, Tulahong A, Uddin MN, Zhao H, Zhang H. Meta-analysis identifying epithelial-derived transcriptomes predicts poor clinical outcome and immune infiltrations in ovarian cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:6527-6551. [PMID: 34517544 DOI: 10.3934/mbe.2021324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Previous studies revealed that the epithelial component is associated with the modulation of the ovarian tumor microenvironment (TME). However, the identification of key transcriptional signatures of laser capture microdissected human ovarian cancer epithelia remains lacking. METHODS We identified the differentially expressed transcriptional signatures of human ovarian cancer epithelia by meta-analysis of GSE14407, GSE2765, GSE38666, GSE40595, and GSE54388. Then we investigated the enrichment of KEGG pathways that are associated with epithelia-derived transcriptomes. Finally, we investigated the correlation of key epithelia-hub genes with the survival prognosis and immune infiltrations. Finally, we investigated the genetic alterations of key prognostic hub genes and their diagnostic efficacy in ovarian cancer epithelia. RESULTS We identified 1339 differentially expressed genes (DEGs) in ovarian cancer epithelia including 541upregulated and 798 downregulated genes. We identified 21 (such as E2F4, FOXM1, TFDP1, E2F1, and SIN3A) and 11 (such as JUN, DDX4, FOSL1, NOC2L, and HMGA1) master transcriptional regulators (MTRs) that are interacted with upregulated and the downregulated genes in ovarian tumor epithelium, respectively. The STRING-based analysis identified hub genes (such as CDK1, CCNB1, AURKA, CDC20, and CCNA2) in ovarian cancer epithelia. The significant clusters of identified hub genes are associated with the enrichment of KEGG pathways including cell cycle, DNA replication, cytokine-cytokine receptor interaction, pathways in cancer, and focal adhesion. The upregulation of SCNN1A and CDCA3 and the downregulation of SOX6 are correlated with a shorter survival prognosis in ovarian cancer (OV). The expression level of SOX6 is negatively correlated with immune score and positively correlated with tumor purity in OV. Moreover, SOX6 is negatively correlated with the infiltration of TILs, CD8+ T cells, CD4+ Regulatory T cells, cytolytic activity, T cell activation, pDC, neutrophils, and macrophages in OV. Also, SOX6 is negatively correlated with various immune markers including CD8A, PRF1, GZMA, GZMB, NKG7, CCL3, and CCL4, indicating the immune regulatory efficiency of SOX6 in the TME of OV. Furthermore, SCNN1A, CDCA3, and SOX6 genes are genetically altered in OV and the expression levels of SCNN1A and SOX6 genes showed diagnostic efficacy in ovarian cancer epithelia. CONCLUSIONS The identified ovarian cancer epithelial-derived key transcriptional signatures are significantly correlated with survival prognosis and immune infiltrations, and may provide new insight into the diagnosis and treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Dong-Feng Li
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Aisikeer Tulahong
- Department of Oncology, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Md Nazim Uddin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Huan Zhao
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Hua Zhang
- Department of Oncology, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| |
Collapse
|
27
|
Hao L, Li S, Peng Q, Guo Y, Ji J, Zhang Z, Xue Y, Liu Y, Shi X. Anti-malarial drug dihydroartemisinin downregulates the expression levels of CDK1 and CCNB1 in liver cancer. Oncol Lett 2021; 22:653. [PMID: 34386075 PMCID: PMC8299009 DOI: 10.3892/ol.2021.12914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Liver cancer is the third leading cause of cancer-associated mortality worldwide. By the time liver cancer is diagnosed, it is already in the advanced stage. Therefore, novel therapeutic strategies need to be identified to improve the prognosis of patients with liver cancer. In the present study, the profiles of GSE84402, GSE19665 and GSE121248 were used to screen differentially expressed genes (DEGs). Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses for DEGs were conducted using the Database for Annotation, Visualization and Integrated Discovery. The protein-protein interaction network was established to screen the hub genes associated with liver cancer. Additionally, the expression levels of hub genes were validated using the Gene Expression Profiling Interactive Analysis and Oncomine databases. In addition, the prognostic value of hub genes in patients with liver cancer was analyzed using Kaplan-Meier Plotter. It was demonstrated that 132 and 246 genes were upregulated and downregulated, respectively, in patients with liver cancer. Among these DEGs, 10 hub genes with high connected node values were identified, which were AURKA, BIRC5, BUB1B, CCNA2, CCNB1, CCNB2, CDC20, CDK1, DLGAP5 and MAD2L1. CDK1 and CCNB1 had the most connection nodes and the highest score and were therefore, the most significantly expressed. In addition, it was demonstrated that high expression levels of CDK1 and CCNB1 were associated with poor overall survival time of patients with liver cancer. Dihydroartemisinin (DHA) is a Food and Drug Administration-approved drug, which is derived from the traditional Chinese medicine Artemisia annua Linn. DHA inhibits cell proliferation in numerous cancer types, including liver cancer. In our previous study, it was revealed that DHA inhibited the proliferation of HepG2215 cells. In the present study, it was further demonstrated that DHA reduced the expression levels of CDK1 and CCNB1 in liver cancer. Overall, CDK1 and CCNB1 were the potential therapeutic targets of liver cancer, and DHA reduced the expression levels of CDK1 and CCNB1, and inhibited the proliferation of liver cancer cells.
Collapse
Affiliation(s)
- Liyuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Shenghao Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jingmin Ji
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Zhiqin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yu Xue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
28
|
Khedkar HN, Wang YC, Yadav VK, Srivastava P, Lawal B, Mokgautsi N, Sumitra MR, Wu ATH, Huang HS. In-Silico Evaluation of Genetic Alterations in Ovarian Carcinoma and Therapeutic Efficacy of NSC777201, as a Novel Multi-Target Agent for TTK, NEK2, and CDK1. Int J Mol Sci 2021; 22:ijms22115895. [PMID: 34072728 PMCID: PMC8198179 DOI: 10.3390/ijms22115895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is often detected at the advanced stages at the time of initial diagnosis. Early-stage diagnosis is difficult due to its asymptomatic nature, where less than 30% of 5-year survival has been noticed. The underlying molecular events associated with the disease’s pathogenesis have yet to be fully elucidated. Thus, the identification of prognostic biomarkers as well as developing novel therapeutic agents for targeting these markers become relevant. Herein, we identified 264 differentially expressed genes (DEGs) common in four ovarian cancer datasets (GSE14407, GSE18520, GSE26712, GSE54388), respectively. We constructed a protein-protein interaction (PPI) interaction network with the overexpressed genes (72 genes) and performed gene enrichment analysis. In the PPI networks, three proteins; TTK Protein Kinase (TTK), NIMA Related Kinase 2 (NEK2), and cyclin-dependent kinase (CDK1) with higher node degrees were further evaluated as therapeutic targets for our novel multi-target small molecule NSC777201. We found that the upregulated DEGs were enriched in KEGG and gene ontologies associated with ovarian cancer progression, female gamete association, otic vesicle development, regulation of chromosome segregation, and therapeutic failure. In addition to the PPI network, ingenuity pathway analysis also implicate TTK, NEK2, and CDK1 in the elevated salvage pyrimidine and pyridoxal pathways in ovarian cancer. The TTK, NEK2, and CDK1 are over-expressed, demonstrating a high frequency of genetic alterations, and are associated with poor prognosis of ovarian cancer cohorts. Interestingly, NSC777201 demonstrated anti-proliferative and cytotoxic activities (GI50 = 1.6 µM~1.82 µM and TGI50 = 3.5 µM~3.63 µM) against the NCI panels of ovarian cancer cell lines and exhibited a robust interaction with stronger affinities for TTK, NEK2, and CDK1, than do the standard drug, paclitaxel. NSC777201 displayed desirable properties of a drug-like candidate and thus could be considered as a novel small molecule for treating ovarian carcinoma.
Collapse
Affiliation(s)
- Harshita Nivrutti Khedkar
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.N.K.); (B.L.); (N.M.); (M.R.S.)
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chi Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Vijesh Kumar Yadav
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (V.K.Y.); (P.S.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 23561, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Prateeti Srivastava
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (V.K.Y.); (P.S.)
| | - Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.N.K.); (B.L.); (N.M.); (M.R.S.)
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ntlotlang Mokgautsi
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.N.K.); (B.L.); (N.M.); (M.R.S.)
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Maryam Rachmawati Sumitra
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.N.K.); (B.L.); (N.M.); (M.R.S.)
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander T. H. Wu
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (V.K.Y.); (P.S.)
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: (A.T.H.W.); (H.-S.H.)
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Academia Sinica, Taipei 11031, Taiwan; (H.N.K.); (B.L.); (N.M.); (M.R.S.)
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- National Defense Medical Center, School of Pharmacy, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (A.T.H.W.); (H.-S.H.)
| |
Collapse
|
29
|
Zhou D, Wang M, Zhang Y, Wang K, Zhao M, Wang Y, Wang X, Yu R, Zhou X. Screening and identification of LMNB1 and DLGAP5, two key biomarkers in gliomas. Biosci Rep 2021; 41:BSR20210231. [PMID: 33956061 PMCID: PMC8144940 DOI: 10.1042/bsr20210231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/27/2021] [Accepted: 05/04/2021] [Indexed: 01/03/2023] Open
Abstract
Glioma is the most common primary cancer in the central nervous system. Despite advances in surgery, radiotherapy and chemotherapy over the past decades, the prognosis of glioblastoma patients remains poor. We aim to identify robust gene signatures to better understand the complex molecular mechanisms and to discover potential novel molecular biomarkers for glioma. By exploring GSE16011, GSE4290 and GSE50161 data in Gene Expression Omnibus (GEO) database, we screened out 380 differentially expressed genes between non-tumor and glioma tissues, and further selected 30 hub genes through the Molecular Complex Detection (MCODE) plug-in in Cytoscape. In addition, LMNB1 and DLGAP5 were selected for further analyses due to their high expression in gliomas and were verified by using our cohort. Our study confirmed that LMNB1 and DLGAP5 were up-regulated in gliomas, and patients with high expression of LMNB1 or DLGAP5 had poor survival rate. Furthermore, silence of LMNB1 and DLGAP5 inhibited the proliferation of glioma cells. Together, LMNB1 and DLGAP5 were two potentially novel molecular biomarkers for diagnosis and prognosis of glioma.
Collapse
Affiliation(s)
- Ding Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Mengmeng Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Kai Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Min Zhao
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yan Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
30
|
Tong Y, Huang Y, Zhang Y, Zeng X, Yan M, Xia Z, Lai D. DPP3/CDK1 contributes to the progression of colorectal cancer through regulating cell proliferation, cell apoptosis, and cell migration. Cell Death Dis 2021; 12:529. [PMID: 34023852 PMCID: PMC8141054 DOI: 10.1038/s41419-021-03796-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023]
Abstract
At present, colorectal cancer (CRC) has become a serious threat to human health in the world. Dipeptidyl peptidase 3 (DPP3) is a zinc-dependent hydrolase that may be involved in several physiological processes. However, whether DPP3 affects the development and progression of CRC remains a mystery. This study is the first to demonstrate the role of DPP3 in CRC. Firstly, the results of immunohistochemistry analysis showed the upregulation of DPP3 in CRC tissues compared with normal tissues, which is statistically analyzed to be positively correlated with lymphatic metastasis, pathological stage, positive number of lymph nodes. Moreover, the high expression of DPP3 predicts poor prognosis in CRC patients. In addition, the results of cell dysfunction experiments clarified that the downregulation of DPP3 significantly inhibited cell proliferation, colony formation, cell migration, and promoted apoptosis in vitro. DPP3 depletion could induce cell apoptosis by upregulating the expression of BID, BIM, Caspase3, Caspase8, HSP60, p21, p27, p53, and SMAC. In addition, downregulation of DPP3 can reduce tumorigenicity of CRC cells in vivo. Furthermore, CDK1 is determined to be a downstream target of DPP3-mediated regulation of CRC by RNA-seq, qPCR, and WB. The interaction between DPP3 and CDK1 shows mutual regulation. Specifically, downregulation of DPP3 can accentuate the effects of CDK1 knockdown on the function of CRC cells. Overexpression of CDK1 alleviates the inhibitory effects of DPP3 knockdown in CRC cells. In summary, DPP3 has oncogene-like functions in the development and progression of CRC by targeting CDK1, which may be an effective molecular target for the prognosis and treatment of CRC.
Collapse
Affiliation(s)
- Yixin Tong
- Department of Tongji Medical College of Huazhong University, 1095 Jiefang Dadao, Wuhan, Hubei Province, China
| | - Yuan Huang
- Department of Endoscopy Center, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Xuhui, Shanghai, China
| | - Yuchao Zhang
- Department of Gastrointestinal Surgery, Sun Yat-sen memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, Guangdong Province, China
| | - Xiangtai Zeng
- Department of The First Affiliated Hospital, Gannan Medical University, 23 Qingnian Road, Zhanggong District, Ganzhou, Jiangxi Province, China
| | - Mei Yan
- Department of Gastrointestinal Surgery, Sun Yat-sen memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, Guangdong Province, China
| | - Zhongsheng Xia
- Department of Gastrointestinal Surgery, Sun Yat-sen memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, Guangdong Province, China.
| | - Dongming Lai
- Department of Gastrointestinal Surgery, Sun Yat-sen memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, Guangdong Province, China.
| |
Collapse
|
31
|
Palma F, Affinito A, Nuzzo S, Roscigno G, Scognamiglio I, Ingenito F, Martinez L, Franzese M, Zanfardino M, Soricelli A, Fiorelli A, Condorelli G, Quintavalle C. miR-34c-3p targets CDK1 a synthetic lethality partner of KRAS in non-small cell lung cancer. Cancer Gene Ther 2021; 28:413-426. [PMID: 32948832 PMCID: PMC8119240 DOI: 10.1038/s41417-020-00224-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/29/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022]
Abstract
Lung cancer is still the leading cause of death by cancer worldwide despite advances both in its detection and therapy. Multiple oncogenic driver alterations have been discovered, opening the prospective for new potential therapeutic targets. Among them, KRAS mutations represent the most frequent oncogene aberrations in non-small cell lung cancer (NSCLC) patients with a negative prognostic impact, but effective therapies targeting KRAS are not well characterized yet. Here, we demonstrate that the microRNA miR-34c-3p is a positive prognostic factor in KRAS-mutated NSCLC patients. Firstly, looking at the TGCA dataset, we found that high miR-34c-3p expression correlated with longer survival of KRAS-mutated NSCLC patients. In vitro assays on immortalized and patient-derived primary NSCLC cells revealed that miR-34c-3p overexpression increased apoptosis and lowered proliferation rate in KRASmut cells. Computational analysis and in vitro assays identified CDK1, one of the most promising lethal targets for KRAS-mutant cancer, as a target of miR-34c-3p. Moreover, the combination of CDK1 inhibition (mediated by RO3306) and miR-34c-3p overexpression resulted in an additive effect on the viability of KRASmut-expressing cells. Altogether, our findings demonstrate that miR-34c-3p is a novel biomarker that may allow tailored treatment for KRAS-mutated NSCLC patients.
Collapse
Affiliation(s)
- Francesco Palma
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
- Percuros BV, Zernikedreef 8, 2333 CL, Leiden, The Netherlands
| | | | | | - Giuseppina Roscigno
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Iolanda Scognamiglio
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Francesco Ingenito
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
- Percuros BV, Zernikedreef 8, 2333 CL, Leiden, The Netherlands
| | - Lola Martinez
- Flow Cytometry Core Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), E-28029, Madrid, Spain
| | | | | | | | - Alfonso Fiorelli
- Thoracic Surgery Unit, Università degli Studi della Campania "Luigi Vanvitelli,", Naples, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy.
- Institute of Experimental Endocrinology and Oncology (IEOS) G. Salvatore, CNR, Naples, Italy.
| | - Cristina Quintavalle
- Institute of Experimental Endocrinology and Oncology (IEOS) G. Salvatore, CNR, Naples, Italy.
| |
Collapse
|
32
|
Jiang P, Zhang M, Gui L, Zhang K. Expression patterns and prognostic values of the cyclin-dependent kinase 1 and cyclin A2 gene cluster in pancreatic adenocarcinoma. J Int Med Res 2021; 48:300060520930113. [PMID: 33290118 PMCID: PMC7727076 DOI: 10.1177/0300060520930113] [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] [Indexed: 01/05/2023] Open
Abstract
Objective Pancreatic adenocarcinoma (PAAD) is one of the most lethal malignant tumors worldwide. Various studies based on cell lines, preclinical mouse models, and human tissue samples have shown that cell cycle-associated proteins are involved in the tumorigenesis and progression of PAAD. Methods Herein, we analyzed the relationships between CDK1 and CCNA2 gene expression and prognosis in patients with pancreatic cancer, using information from the Oncomine, cBioportal, Kaplan–Meier Plotter, and GEPIA databases. Results Expression levels of CDK1 and CCNA2 were significantly higher in PAAD compared with control tissues, and were associated with more advanced tumor stage. Survival analyses using the Kaplan–Meier Plotter database further confirmed that increased expression levels of CDK1 and CCNA2 were associated with a poor prognosis in patients with pancreatic cancer. Conclusions The results of this study suggest that CDK1 and CCNA2 may be potential therapeutic targets and prognostic biomarkers in patients with PAAD.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Gastroenterology, The Central Hospital of Weihai, Weihai, Shandong, China
| | - Ming Zhang
- Hepatobiliary Surgery Department, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Liangliang Gui
- Department of Gastroenterology, The Central Hospital of Weihai, Weihai, Shandong, China
| | - Kai Zhang
- Hepatobiliary Surgery Department, Shandong Provincial Third Hospital, Jinan, Shandong, China
| |
Collapse
|
33
|
Construction of a novel prognostic-predicting model correlated to ovarian cancer. Biosci Rep 2021; 40:225895. [PMID: 32716025 PMCID: PMC7414523 DOI: 10.1042/bsr20201261] [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: 04/23/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Ovarian cancer (OC) is one of the most lethal gynecological cancers worldwide. The pathogenesis of the disease and outcomes prediction of OC patients remain largely unclear. The present study aimed to explore the key genes and biological pathways in ovarian carcinoma development, as well as construct a prognostic model to predict patients’ overall survival (OS). Results: We identified 164 up-regulated and 80 down-regulated differentially expressed genes (DEGs) associated with OC. Gene Ontology (GO) term enrichment showed DEGs mainly correlated with spindle microtubes. For Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, cell cycle was mostly enriched for the DEGs. The protein–protein interaction (PPI) network yielded 238 nodes and 1284 edges. Top three modules and ten hub genes were further filtered and analyzed. Three candidiate drugs targeting for therapy were also selected. Thirteen OS-related genes were selected and an eight-mRNA model was present to stratify patients into high- and low-risk groups with significantly different survival. Conclusions: The identified DEGs and biological pathways may provide new perspective on the pathogenesis and treatments of OC. The identified eight-mRNA signature has significant clinical implication for outcome prediction and tailored therapy guidance for OC patients.
Collapse
|
34
|
Fadaka AO, Samantha Sibuyi NR, Bakare OO, Klein A, Madiehe AM, Meyer M. Expression of cyclin-dependent kinases and their clinical significance with immune infiltrates could predict prognosis in colorectal cancer. ACTA ACUST UNITED AC 2021; 29:e00602. [PMID: 33732631 PMCID: PMC7937668 DOI: 10.1016/j.btre.2021.e00602] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/26/2021] [Accepted: 02/20/2021] [Indexed: 12/15/2022]
Abstract
The expression and prognostic values of AURKA and RB1 may also be significant to CRC diagnosis than previously studies. The association of CDKs with immune infiltrates may serve as target molecules for immunotherapy in CRC. The expression of CDK is significant among CRC subtypes and therefore, it can be inferred as a potential biomarker in the cancer subtype. An increase in tumor purity was positively correlated with the expression of CDK-1 in COAD due to CD4+ cells and CDK-4 in COAD and READ resulting from a fraction of immune cells.
Introduction Colorectal cancer (CRC) is one of the most cancer-related mortalities worldwide and remains a major public health issue. Despite several attempts to develop promising therapies for CRC, its survival rate decreases with metastasis. Cyclin-dependent kinases (CDKs) are a family of protein kinases with various regulatory activities including cell cycle, mRNA expression, transcription, and differentiation. Aside from their role in cell proliferation when mutated, abnormal expression of these genes has been reported in some human cancer subtypes. This study explored the roles and therapeutic potentials of CDK 1 and 4 as prognostic biomarkers in CRC. Methods Bioinformatics analyses were carried out to demonstrate the expression and prognostic values of CDK-1 and CDK-4 with immune infiltrate in CRC. Discussion CDK levels in CRC were remarkably higher than those in normal tissues (p < 0.05), and overexpression in CRC tissues was significantly related to nodal metastatic status (p < 0.05) and histological subtypes. Kaplan-Meier analyses showed that patients with CRC who exhibited CDK-1 overexpression had worse overall survival (OS) as against patients with CDK-4 overexpression. The alteration observed was a mutation while the mutation hotspots include E163* and R24A/C/H/L respectively for CDK-1 and CDK-4 on the Pkinase domain. Of the associated genes, AURKA and RB1 were predominantly altered. Furthermore, CDK-4 is positively correlated with tumor purity in both COAD and READ while CDK-1is only positively correlated in COAD. CDK-1 overexpression was significantly associated with poor prognosis as opposed to CDK-4. Conclusion The expression and prognostic values of AURKA and RB1 may also be significant to CRC diagnosis. CDKs together with the co-expressed genes and their association with immune infiltrates may serve as target molecules for immunotherapy in CRC.
Collapse
Affiliation(s)
- Adewale Oluwaseun Fadaka
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, South Africa
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, South Africa
| | - Olalekan Olanrewaju Bakare
- Bioinformatics Research Group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Ashwil Klein
- Plant Omics Laboratory, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Abram Madimabe Madiehe
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, South Africa.,Nanobiotechnology Research Group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
35
|
Ying X, Che X, Wang J, Zou G, Yu Q, Zhang X. CDK1 serves as a novel therapeutic target for endometrioid endometrial cancer. J Cancer 2021; 12:2206-2215. [PMID: 33758599 PMCID: PMC7974891 DOI: 10.7150/jca.51139] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Endometrial cancer (EC) is one of the most common and prevalent gynecologic malignancies worldwide. The aim of this study was to identify a novel therapeutic target for endometrioid endometrial cancer. Materials and Methods: Bioinformatic analysis was performed and CDK1 was screen out as one of the hub genes in the pathogenesis of EC. Immunohistochemistry was used to verify the expression of CDK1 in endometrial cancer tissue. Cell viability and colony formation were used to study the effects of CDK1 on the proliferation and colony formation of endometrial cancer cells in vitro. Apoptosis and cell cycle assays were used to elucidate the mechanism of CDK1 affecting cell proliferation. Tumor xenograft transplantation assay was performed to show the effects of CDK1 on the growth of endometrial cancer cells in vivo. Results: CDK1 was over expressed in endometrioid endometrial cancer, and accumulation of cytoplasmic CDK1 was associated with histological grade of EC. CDK1 promoted endometrial cancer cell growth and colony formation in vitro. The inhibition of CDK1 activity induced cell apoptosis and caused G2/M phase arrest of cell cycle in endometrial cancer cells. The inhibition of CDK1 activity also inhibited endometrial cancer growth in xenograft models. Conclusion: CDK1 was involved in the pathogenesis of endometrioid endometrial cancer and provided a novel therapeutic target for endometrioid endometrial cancer.
Collapse
Affiliation(s)
- Xue Ying
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Xuan Che
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006.,Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, P.R. China, 314000
| | - Jianzhang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Gen Zou
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Qin Yu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Xinmei Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| |
Collapse
|
36
|
Li M, He F, Zhang Z, Xiang Z, Hu D. CDK1 serves as a potential prognostic biomarker and target for lung cancer. J Int Med Res 2020; 48:300060519897508. [PMID: 32020821 PMCID: PMC7111107 DOI: 10.1177/0300060519897508] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Mingyao Li
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Fenyi He
- Department of Special Examination, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Zhanchun Zhang
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Zhenfei Xiang
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Danfei Hu
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
37
|
Dong B, Chai M, Chen H, Feng Q, Jin R, Hu S. Screening and verifying key genes with poor prognosis in colon cancer through bioinformatics analysis. Transl Cancer Res 2020; 9:6720-6732. [PMID: 35117282 PMCID: PMC8797306 DOI: 10.21037/tcr-20-2309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022]
Abstract
Background Colon cancer (CC) is one of the tumors with high morbidity and mortality in the world, and has a trend of younger generation. The molecular level of CC has not been fully elaborated. The purpose of this study is to screen and identify important genes with poor prognosis and their mechanisms at different levels. Methods GSE74602 and GSE10972 gene expression profiles were downloaded from the Gene Expression Omnibus (GEO) database. There were 58 normal tissues and 58 CC tissues. Differentially expressed genes (DEGs) were screened out by using the GEO2R tool and Venn diagram. Then, the DAVID online database was used to perform the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Six hub genes with the highest correlation were screened out after the modular analysis of the protein-protein interaction (PPI) network by using Cytoscape’s MCODE plug-in. Finally, the overall survival of key hub genes and potential pathways were verified in GEPIA and UALCAN database. Results A total of 78 up-regulated DEGs were enriched in the mitotic nuclear division, cell division, cell proliferation, anaphase-promoting complex-dependent catabolic process and G2/M transition of the mitotic cell cycle. In total, 130 down-regulated DEGs were enriched in muscle contraction, bicarbonate transport, cellular response to zinc ion, negative regulation of growth, negative regulation of leukocyte apoptotic process and one-carbon metabolic process. CDK1, CCNB1, CDC20, AURKA, CCNA2 and TOP2A were the top six hub genes, mainly enriched in cell cycle pathways. Among them, CCNB1, CDK1, CDC20, CCNA2 were enriched in the G2/M phase. GEPIA and UALCAN database confirmed that CCNA2 and CCNB1 had a significant relationship with the poor prognosis of CC patients. Meanwhile, there was a positive correlation between the two. Conclusions Screening out genes with abnormal expression in CC help understand the initiation and progression of CC at the molecular level and explore candidate biomarkers for diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Buyuan Dong
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengyu Chai
- Department of Respiratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Chen
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qian Feng
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rong Jin
- Department of Epidemiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
Downregulation of miRNA-205 Expression and Biological Mechanism in Prostate Cancer Tumorigenesis and Bone Metastasis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6037434. [PMID: 33178832 PMCID: PMC7646560 DOI: 10.1155/2020/6037434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/22/2020] [Accepted: 10/15/2020] [Indexed: 12/23/2022]
Abstract
Background The expression and mechanism of microRNA-205 (miRNA-205) in prostate cancer (PCa) and its bone metastasis remain controversial. Materials and Methods The expression and discriminating capability of miRNA-205 were assessed by drawing a forest plot and a summarized receiver operating characteristic (SROC) curve, using data available from 27 miRNA-array and miRNA-sequencing datasets. The miRNA-205 target genes were acquired from online prediction tools, differentially upregulated genes in PCa, and differentially expressed genes (DEGs) after miRNA-205 transfection into PCa cell lines. Functional enrichment analysis was conducted to explore the biological mechanism of miRNA-205 targets. Immunohistochemistry (IHC) was applied to verify the protein level of the hub gene. Results The expression of miRNA-205 in the PCa group (1,461 samples) was significantly lower than that in the noncancer group (510 samples), and the downregulation of miRNA-205 showed excellent sensitivity and specificity in differentiating between the two groups. In bone metastatic PCa, the miRNA-205 level was further reduced than in nonbone metastatic PCa, and it showed a good capability in distinguishing between the two groups. In total, 153 miRNA-205 targets were screened through the three aforementioned methods. Based on the results of functional enrichment analysis, the targets of miRNA-205 were mainly enriched during chromosome segregation and phospholipid-translocating ATPase activity and in the spindle microtubule and the p53 signaling pathway. CDK1 had the highest connectivity in the PPI network analysis and was screened as one of the hub genes. A statistically significant negative correlation between miRNA-205 and CDK1 was observed. The expression of CDK1 in PCa samples was pronouncedly upregulated in terms of both the mRNA level and the protein level when compared with noncancer samples. Conclusion miRNA-205 may play a vital role in PCa tumorigenesis and bone metastasis by targeting CDK1.
Collapse
|
39
|
Wang B, Hua P, Zhao B, Li J, Zhang Y. Circular RNA circDLGAP4 is involved in lung cancer development through modulating microRNA-143/CDK1 axis. Cell Cycle 2020; 19:2007-2017. [PMID: 32646340 DOI: 10.1080/15384101.2020.1786649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To investigate the role of circular RNA DLGAP4 (circDLGAP4) in lung cancer. circDLGAP4 expression was detected in lung cancer tissues and cell lines by PCR. The correlation between circDLGAP4 and clinicopathological characteristics of lung cancer patients was investigated. Moreover, the influences of depression of circDLGAP4 on the biological processes biological processes of lung cancer cells were explored in vitro. In addition, whether circDLGAP4 regulated lung cancer cell biological processes by sponging microRNA-143 (miR-143) to regulate cyclin-dependent kinase 1 (CDK1) expression was explored and verified in another lung cell line. CircDLGAP4 expression was remarkably elevated in lung cancer tissues and was significantly corrected with TNM stage and tumor metastasis. Suppression of circDLGAP4 inhibited the biological performances of lung cancer cells. Also, there was a negative regulatory relationship between circDLGAP4 and miR-143. Inhibition of miR-143 alleviated the influences of circDLGAP4 depression on lung cancer cell biological processes. Moreover, CDK1 was discovered as a target of miR-143, and miR-143 was involved in the process of lung cancer cell biological processes through targeting CDK1. Our findings reveal that circular RNA circDLGAP4 is involved in lung cancer development through modulating microRNA-143/CDK1 axis. circDLGAP4 may serve as a potential biomarker for the diagnosis or treatment of lung cancer.
Collapse
Affiliation(s)
- Bin Wang
- Department of Thoracic Surgery, The Second Hospital of Jilin University , Changchun, Jilin, China
| | - Peiyan Hua
- Department of Thoracic Surgery, The Second Hospital of Jilin University , Changchun, Jilin, China
| | - Bin Zhao
- Department of Neurosurgery, The Second Hospital of Jilin University , Changchun, Jilin, China
| | - Jindong Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University , Changchun, Jilin, China
| | - Yan Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University , Changchun, Jilin, China
| |
Collapse
|
40
|
Repo H, Löyttyniemi E, Kurki S, Kallio L, Kuopio T, Talvinen K, Kronqvist P. A prognostic model based on cell-cycle control predicts outcome of breast cancer patients. BMC Cancer 2020; 20:558. [PMID: 32546141 PMCID: PMC7296704 DOI: 10.1186/s12885-020-07045-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Background A prognostic model combining biomarkers of metaphase-anaphase transition of the cell cycle was developed for invasive breast cancer. The prognostic value and clinical applicability of the model was evaluated in comparison with the routine prognosticators of invasive breast carcinoma. Methods The study comprised 1135 breast cancer patients with complete clinical data and up to 22-year follow-up. Regulators of metaphase-anaphase transition were detected immunohistochemically and the biomarkers with the strongest prognostic impacts were combined into a prognostic model. The prognostic value of the model was tested and evaluated in separate patient materials originating from two Finnish breast cancer centers. Results The designed model comprising immunoexpressions of Securin, Separase and Cdk1 identified 8.4-fold increased risk of breast cancer mortality (p < 0.0001). A survival difference exceeding 15 years was observed between the majority (> 75%) of patients resulting with favorable as opposed to unfavorable outcome of the model. Along with nodal status, the model showed independent prognostic impact for all breast carcinomas and for subgroups of luminal, N+ and N- disease. Conclusions The impact of the proposed prognostic model in predicting breast cancer survival was comparable to nodal status. However, the model provided additional information in N- breast carcinoma in identifying patients with aggressive course of disease, potentially in need of adjuvant treatments. Concerning N+, in turn, the model could provide evidence for withholding chemotherapy from patients with favorable outcome.
Collapse
Affiliation(s)
- Heli Repo
- Institute of Biomedicine, University of Turku, Turku, Finland.,Central Hospital of Central Finland, Jyväskylä, Finland
| | | | - Samu Kurki
- Turku University Hospital, Turku, Finland
| | | | - Teijo Kuopio
- Central Hospital of Central Finland, Jyväskylä, Finland
| | - Kati Talvinen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pauliina Kronqvist
- Institute of Biomedicine, University of Turku, Turku, Finland. .,Department of Pathology, University of Turku, Kiinamyllynkatu 10/MedD5A, 20500, Turku, Finland.
| |
Collapse
|
41
|
Liping X, Jia L, Qi C, Liang Y, Dongen L, Jianshuai J. Cell Cycle Genes Are Potential Diagnostic and Prognostic Biomarkers in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6206157. [PMID: 32596342 PMCID: PMC7298261 DOI: 10.1155/2020/6206157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/26/2020] [Accepted: 05/02/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND The cell cycle pathway genes are comprised of 113 members which are critical to the maintenance of cell cycle and survival of tumor cells. This study was performed to investigate the diagnostic and prognostic values of cell cycle gene expression in hepatocellular carcinoma (HCC) patients. METHODS Clinical features and cell cycle pathway gene expression data were obtained from the Gene Expression Omnibus and The Cancer Genome Atlas databases. Differentially expressed genes (DEGs) were determined by the student t-test between HCC and noncancerous samples. Kaplan-Meier survival, univariate, and multivariate survival analyses and validation analysis were performed to characterize the associations between cell cycle gene expression and patients' overall survival and recurrence-free survival. RESULTS 47 and 5 genes were significantly upregulated and downregulated genes in HCC samples, respectively. The high expression of BUB3, CDK1, and CHEK1 was associated with increased mortality (adjusted P value = 0.04, odds ratio (OR): 1.89 (95% confidence interval (CI): 1.04-3.46); adjusted P value = 0.02, OR: 2.06 (95% CI:1.15-3.75); and adjusted P value = 0.04, OR: 1.84 (%95 CI: 1.03-3.32), respectively). The expression of PTTG2 and RAD21 was significantly associated with cancer recurrence (adjusted P value = 0.01, OR: 2.17 (95% CI: 1.24-3.86); adjusted P value = 0.03, OR: 1.88[95% CI:1.08-3.28], respectively), while the low expression of MAD1L1 was associated with cancer recurrence (adjusted P value = 0.03, OR: 0.53 (%95 CI: 0.3-0.93)). CONCLUSIONS The present study demonstrated that BUB3, CDK1, and CHEK1 may serve as a prognostic biomarker for HCC patients. PTTG2, RAD21, and MAD1L1 expression is a major factor affecting the recurrence of HCC patients.
Collapse
Affiliation(s)
- Xu Liping
- Department of Hepatobiliary Pancreatic Surgery, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Li Jia
- Department of Breast and Thyroid, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Qi
- Department of Hepatobiliary Pancreatic Surgery, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Yang Liang
- Department of Hepatobiliary Pancreatic Surgery, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Li Dongen
- Department of Hepatobiliary Pancreatic Surgery, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Jiang Jianshuai
- Department of Hepatobiliary Pancreatic Surgery, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| |
Collapse
|
42
|
Yang W, Shin HY, Cho H, Chung JY, Lee EJ, Kim JH, Kang ES. TOM40 Inhibits Ovarian Cancer Cell Growth by Modulating Mitochondrial Function Including Intracellular ATP and ROS Levels. Cancers (Basel) 2020; 12:cancers12051329. [PMID: 32456076 PMCID: PMC7281007 DOI: 10.3390/cancers12051329] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 12/17/2022] Open
Abstract
TOM40 is a channel-forming subunit of translocase, which is essential for the movement of proteins into the mitochondria. We found that TOM40 was highly expressed in epithelial ovarian cancer (EOC) cells at both the transcriptional and translational levels; its expression increased significantly during the transformation from normal ovarian epithelial cells to EOC (p < 0.001), and TOM40 expression negatively correlated with disease-free survival (Hazard ratio = 1.79, 95% Confidence inerval 1.16–2.78, p = 0.009). TOM40 knockdown decreased proliferation in several EOC cell lines and reduced tumor burden in an in vivo xenograft mouse model. TOM40 expression positively correlated with intracellular adenosine triphosphate (ATP) levels. The low ATP and high reactive oxygen species (ROS) levels increased the activity of AMP-activated protein kinase (AMPK) in TOM40 knockdown EOC cells. However, AMPK activity did not correlate with declined cell growth in TOM40 knockdown EOC cells. We found that metformin, first-line therapy for type 2 diabetes, effectively inhibited the growth of EOC cell lines in an AMPK-independent manner by inhibiting mitochondria complex I. In conclusion, TOM40 positively correlated with mitochondrial activities, and its association enhances the proliferation of ovarian cancer. Also, metformin is an effective therapeutic option in TOM40 overexpressed ovarian cancer than normal ovarian epithelium.
Collapse
Affiliation(s)
- Wookyeom Yang
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (W.Y.); (H.-Y.S.); (H.C.); (E.-j.L.)
| | - Ha-Yeon Shin
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (W.Y.); (H.-Y.S.); (H.C.); (E.-j.L.)
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (W.Y.); (H.-Y.S.); (H.C.); (E.-j.L.)
| | - Joon-Yong Chung
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Eun-ju Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (W.Y.); (H.-Y.S.); (H.C.); (E.-j.L.)
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (W.Y.); (H.-Y.S.); (H.C.); (E.-j.L.)
- Correspondence: (J.-H.K.); (E.-S.K.); Tel.:+82-2-2019-3430 (J.-H.K.); +82-2-3410-2703 (E.-S.K.); Fax: +82-2-3462-8209 (J.-H.K.); +82-2-3410-2719 (E.-S.K.)
| | - Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Correspondence: (J.-H.K.); (E.-S.K.); Tel.:+82-2-2019-3430 (J.-H.K.); +82-2-3410-2703 (E.-S.K.); Fax: +82-2-3462-8209 (J.-H.K.); +82-2-3410-2719 (E.-S.K.)
| |
Collapse
|
43
|
Jin J, Xu H, Li W, Xu X, Liu H, Wei F. LINC00346 Acts as a Competing Endogenous RNA Regulating Development of Hepatocellular Carcinoma via Modulating CDK1/CCNB1 Axis. Front Bioeng Biotechnol 2020; 8:54. [PMID: 32133348 PMCID: PMC7039823 DOI: 10.3389/fbioe.2020.00054] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/22/2020] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the important types of liver cancer. LncRNA is an important regulatory factor that regulates many biological processes such as tumor cells during tumorigenesis and metastasis. LINC00346 has been associated with various types of liver cancer, but its role and regulatory mechanism in HCC remain unclear. In our study, we found the LINC00356-miR-199a-3p-CDK1/CCNB1 axis through bioinformatics analysis. The expressions of LINC00356, miR-199a-3p, CDK1, and CCNB1 in HCC and normal hepatocytes were determined by qRT-PCR and WB. The results showed that LINC00356, CDK1 and CCNB1 were highly expressed in HCC, while miR-199a-3p was lowly expressed. Dual luciferase reporter gene assay, RIP and RNA-pull down assays demonstrated the targeted binding relationship of LINC00346-miR-199a-3p-CDK1/CCNB1. Overexpressing LINC00460 and silencing miR-199a-3p promoted cell invasion, inhibited apoptosis of HCC, and arrested the cell cycle in S phase while opposite results were obtained when silencing LINC00346, CDK1, and CCNB1. LINC00346 indirectly affects liver cancer by promoting the expression of CDK1/CCNB1 through competitive adsorption of miR-199a-3p. In addition, the study also demonstrated that overexpression of LINC00346 indirectly inhibited the expression of p53 and p21 proteins by promoting CDK1/CCNB1 expressions, thereby blocking the p53 signaling pathway. These results proved that LINC00346 could regulate the expression of CDK1/CCNB1 through the competitive adsorption of miR-199a-3p, thereby affecting the p53 signaling pathway and finally regulating the apoptosis, invasion and cell cycle of HCC cells. In conclusion, LINC00346 can be used as a tumor promoter and potential therapeutic target for HCC metastasis and prognosis.
Collapse
Affiliation(s)
- Jinglan Jin
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Hongqin Xu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Wanyu Li
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Xiaotong Xu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
44
|
Identification of Key Genes and Prognostic Value Analysis in Hepatocellular Carcinoma by Integrated Bioinformatics Analysis. Int J Genomics 2019; 2019:3518378. [PMID: 31886163 PMCID: PMC6893264 DOI: 10.1155/2019/3518378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023] Open
Abstract
Emerging evidence indicates that various functional genes with altered expression are involved in the tumor progression of human cancers. This study is aimed at identifying novel key genes that may be used for hepatocellular carcinoma (HCC) diagnosis, prognosis, and targeted therapy. This study included 3 expression profiles (GSE45267, GSE74656, and GSE84402), which were obtained from the Gene Expression Omnibus (GEO). GEO2R was used to analyze the differentially expressed genes (DEGs) between HCC and normal samples. The functional and pathway enrichment analysis was performed by the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction (PPI) network of the identified DEGs was constructed using the Search Tool for the Retrieval of Interacting Gene, and hub genes were identified. ONCOMINE and CCLE databases were used to verify the expression of the hub genes in HCC tissues and cells. Kaplan-Meier plotter was used to assess the effects of the hub genes on the overall survival of HCC patients. A total of 99 DEGs were identified from the 3 expression profiles. These DEGs were enriched with functional processes and pathways related to HCC pathogenesis. From the PPI network, 5 hub genes were identified. The expression of the 5 hub genes was all upregulated in HCC tissues and cells compared with the control tissues and cells. Kaplan-Meier survival curves indicated that high expression of cyclin-dependent kinase (CDK1), cyclin B1 (CCNB1), cyclin B2 (CCNB2), MAD2 mitotic arrest deficient-like 1 (MAD2L1), and topoisomerase IIα (TOP2A) predicted poor overall survival in HCC patients (all log-rank P < 0.01). These results revealed that the DEGs may serve as candidate key genes during HCC pathogenesis. The 5 hub genes, including CDK1, CCNB1, CCNB2, MAD2L1, and TOP2A, may serve as promising prognostic biomarkers in HCC.
Collapse
|
45
|
Zhang YB, Jiang Y, Wang J, Ma J, Han S. Evaluation of core serous epithelial ovarian cancer genes as potential prognostic markers and indicators of the underlying molecular mechanisms using an integrated bioinformatics analysis. Oncol Lett 2019; 18:5508-5522. [PMID: 31612059 PMCID: PMC6781641 DOI: 10.3892/ol.2019.10884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/23/2019] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is a major cause of mortality in women. However, the molecular events underlying the pathogenesis of the disease are yet to be fully elucidated. In the present study, an integrated bioinformatics analysis was performed to identify core genes involved in serous epithelial ovarian cancer. A total of three expression datasets were downloaded from the Gene Expression Omnibus database, and included 46 serous epithelial ovarian cancer and 30 ovarian surface epithelium samples. The three datasets were merged, and batch normalization was performed. The normalized merged data were subsequently analyzed for differentially expressed genes (DEGs). In total, 2,212 DEGs were identified, including 1,300 upregulated and 912 downregulated genes. Gene Ontology analysis revealed that these DEGs were primarily involved in ‘regulation of cell cycle’, ‘mitosis’, ‘DNA packaging’ and ‘nucleosome assembly’. The main cellular components included ‘extracellular region part’, ‘chromosome’, ‘extracellular matrix’ and ‘condensed chromosome kinetochore’, whereas the molecular functions included ‘Calcium ion binding’, ‘polysaccharide binding’, ‘enzyme inhibitor activity’, ‘growth factor activity’, ‘cyclin-dependent protein kinase regulator activity’, ‘microtubule motor activity’ and ‘Wnt receptor activity’. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that these DEGs were predominantly involved in ‘Wnt signaling pathway’, ‘pathways in cancer’, ‘PI3K-Akt signaling pathway’, ‘cell cycle’, ‘ECM-receptor interaction’, ‘p53 signaling pathway’ and ‘focal adhesion’. The 20 most significant DEGs were identified from the protein-protein interaction network, and Oncomine analysis of these core genes revealed that 13 were upregulated and two were downregulated in serous epithelial ovarian cancer. Survival analysis revealed that cyclin B1, polo like kinase 1, G protein subunit γ transducin 1 and G protein subunit γ 12 are key molecules that may be involved in the prognosis of serous epithelial ovarian cancer. These core genes may provide novel treatment targets, although their roles in the carcinogenesis and prognosis of serous epithelial ovarian cancer require further study.
Collapse
Affiliation(s)
- Yu-Bo Zhang
- Department of Gynecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yuhan Jiang
- Department of Gynecology, The Affiliated Hospital of Jining Medical College, Jining, Shandong 272000, P.R. China
| | - Jiao Wang
- Department of Gynecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jing Ma
- Department of Gynecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Shiyu Han
- Department of Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
46
|
Inchiosa MA. Further investigation of the potential anti-neoplastic, anti-inflammatory and immunomodulatory actions of phenoxybenzamine using the Broad Institute CLUE platform.. [DOI: 10.1101/767392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractPrevious clinical studies with the FDA-approved alpha-adrenergic antagonist, phenoxybenzamine, showed apparent efficacy to reverse the symptoms and disabilities of the neuropathic condition, Complex Regional Pain Syndrome; also, the anatomic spread and intensity of this syndrome has a proliferative character and it was proposed that phenoxybenzamine may have an anti-inflammatory, immunomodulatory mode of action. A previous study gave evidence that phenoxybenzamine had anti-proliferative activity in suppression of growth in several human tumor cell cultures. The same report demonstrated that the drug possessed significant histone deacetylase inhibitory activity. Utilizing the Harvard/Massachusetts Institute of Technology Broad Institute genomic database, CLUE, the present study suggests that the gene expression signature of phenoxybenzamine in malignant cell lines is consistent with anti-inflammatory/immunomodulatory activity and suppression of tumor expansion by several possible mechanisms of action. Of particular note, phenoxybenzamine demonstrated signatures that were highly similar to those with glucocorticoid agonist activity. Also, gene expression signatures of phenoxbenzamine were consistent with several agents in each case that were known to suppress tumor proliferation, notably, protein kinase C inhibitors, Heat Shock Protein inhibitors, epidermal growth factor receptor inhibitors, and glycogen synthase kinase inhibitors. Searches in CLUE also confirmed the earlier observations of strong similarities between gene expression signatures of phenoxybenzamine and several histone deacetylase inhibitors.
Collapse
|
47
|
Li L, Zhang Z, Yang Q, Ning M. Lycorine inhibited the cell growth of non-small cell lung cancer by modulating the miR-186/CDK1 axis. Life Sci 2019; 231:116528. [PMID: 31176784 DOI: 10.1016/j.lfs.2019.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 12/24/2022]
Abstract
AIMS Lycorine is a kind of natural alkaloid with anti-cancer potential. It has been demonstrated that lycorine processes high activity and specificity against the progression of cancers. However, the underlying molecular mechanisms by which lycorine regulates the formation and development of non-small cell lung cancer (NSCLC) remain largely unknown. MAIN METHODS The effects of lycorine on the growth of NSCLC cells were determined by the cell counting kit-8 (CCK-8) assay, colony formation and flow cytometry analysis. RT-qPCR was performed to detect the expression of microRNA with lycorine treatment. The binding of miRNA and target genes was confirmed by luciferase reporter assay. KEY FINDINGS Lycorine significantly inhibited the proliferation and induced apoptosis of NSCLC cells. Mechanistically, lycorine up-regulated the expression of microRNA-186 in NSCLC cells. Depletion of miR-186 significantly reversed the suppressive effect of lycorine on the proliferation of NSCLC cells. Furthermore, the cyclin dependent kinase 1 (CDK1) was identified as one of the binding candidates of miR-186. Experimental analysis showed that miR-186 bound the 3'-untranslated region (3'-UTR) of CDK1 and suppressed the level of CDK1 in NSCLC cells. Consistently, exposure of lycorine significantly decreased the expression of CDK1. Restoration of CDK1 remarkably attenuated the inhibition of lycorine on the proliferation of NSCLC cells. SIGNIFICANCE Our results uncovered the novel molecular mechanism of lycorine in suppressing the progression of NSCLC partially via regulating the miR-186/CDK1 axis.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Zao Zhang
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Qian Yang
- Traditional Chinese Medicine Department, Cangzhou Central Hospital, Cangzhou, Hebei, China, 061001
| | - Meiying Ning
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China.
| |
Collapse
|
48
|
Liu J, Li S, Liang J, Jiang Y, Wan Y, Zhou S, Cheng W. ITLNI identified by comprehensive bioinformatic analysis as a hub candidate biological target in human epithelial ovarian cancer. Cancer Manag Res 2019; 11:2379-2392. [PMID: 30988639 PMCID: PMC6438265 DOI: 10.2147/cmar.s189784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) is a female malignant tumor. Bioinformatics has been widely utilized to analyze genes related to cancer progression. Targeted therapy for specific biological factors has become more valuable. Materials and methods Gene expression profiles of GSE18520 and GSE27651 were downloaded from Gene Expression Omnibus. We used the “limma” package to screen differentially expressed genes (DEGs) between EOC and normal ovarian tissue samples and then used Clusterprofiler to do functional and pathway enrichment analyses. We utilized Search Tool for the Retrieval of Interacting Genes Database to assess protein–protein interaction (PPI) information and the plug-in Molecular Complex Detection to screen hub modules of PPI network in Cytoscape, and then performed functional analysis on the genes in the hub module. Next, we utilized the Weighted Gene Expression Network Analysis package to establish a co-expression network. Validation of the key genes in databases and Gene Expression Profiling Interactive Analysis (GEPIA) were completed. Finally, we used quantitative real-time PCR to validate hub gene expression in clinical tissue samples. Results We analyzed the DEGs (96 samples of EOC tissue and 16 samples of normal ovarian tissue) for functional analysis, which showed that upregulated DEGs were strikingly enriched in phosphate ion binding and the downregulated DEGs were significantly enriched in glycosaminoglycan binding. In the PPI network, CDK1 was screened as the most relevant protein. In the co-expression network, one EOC-related module was identified. For survival analysis, database and clinical sample validation of genes in the turquoise module, we found that ITLN1 was positively correlated with EOC prognosis and had lower level in EOC than in normal tissues, which was consistent with the results predicted in GEPIA. Conclusion In this study, we exhibited the key genes and pathways involved in EOC and speculated that ITLN1 was a tumor suppressor which could be used as a potential biomarker for treating EOC, Gene Expression Omnibus, prognosis.
Collapse
Affiliation(s)
- JinHui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| | - SiYue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| | - JunYa Liang
- Hypertension Research Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu, China
| | - Yi Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| | - YiCong Wan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| | - ShuLin Zhou
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| | - WenJun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China,
| |
Collapse
|
49
|
High expression of CDK1 and BUB1 predicts poor prognosis of pancreatic ductal adenocarcinoma. Gene 2019; 701:15-22. [PMID: 30898709 DOI: 10.1016/j.gene.2019.02.081] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most common causes of cancer-related death. Increasing evidence suggests that cell cycle dysregulation is one of the hallmarks of cancer. In this study, by using the GEO database, we predicted the cell cycle-related protein CDK1 and BUB1 to be significantly overexpressed in PDAC tissues. Thus, this study aimed to investigate the clinical pathological significance of CDK1 and BUB1 in PDAC. METHODS To explore the role of CDK1 and BUB1 in PDAC progression and evaluate their prognostic value, we investigated the expression patterns of CDK1 and BUB1 by using immunohistochemical staining in 99 PDAC and 71 normal pancreatic tissues with complete pathological parameters and survival data. RESULTS CDK1 and BUB1 were significantly overexpressed in PDAC tissues. The expression of CDK1 was correlated with tumor size and histological grade, and the expression of BUB1 was correlated with the tumor size of PDAC. With regard to survival, a high expression of either CDK1 or BUB1 was correlated with a short survival of PDAC patients. Additionally, PDAC patients with a concurrent high expression of CDK1 and BUB1 showed the shortest survival. CONCLUSIONS Our study demonstrated that CDK1 and BUB1 may play a role in PDAC progression and could be prognostic biomarkers for PDAC patients.
Collapse
|
50
|
Overexpression of BUB1B, CCNA2, CDC20, and CDK1 in tumor tissues predicts poor survival in pancreatic ductal adenocarcinoma. Biosci Rep 2019; 39:BSR20182306. [PMID: 30765611 PMCID: PMC6390130 DOI: 10.1042/bsr20182306] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/28/2023] Open
Abstract
Overexpressed genes in tumors usually contributed to aggressiveness in pancreatic ductal adenocarcinoma (PDAC). Using Gene Expression Omnibus (GEO) profiles including GSE46234, GSE71989, and GSE107610, we detected overexpressed genes in tumors with R program, which were enriched by Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene ontology (GO), and Reactome pathway databases. Then, we performed a survival analysis of enriched genes based on TCGA profile. Our results revealed that high BUB1B, CCNA2, CDC20, and CDK1 expression in tumors was significantly associated with worse overall survival (OS) (Log rank P=0.00338, P=0.0447, P=0.00965, and P=0.00479, respectively), which was validated using a Kaplan–Meier plotter with a median cutoff (Log rank P=0.028, P=0.0035, P=0.039, and P=0.0033, respectively). Moreover, overexpression of BUB1B, CCNA2, CDC20, and CDK1 in tumor tissues was significantly associated with disease-free survival (DFS) in PDAC patients (Log rank P=0.00565, P=0.0357, P=0.00104, and P=0.00121, respectively). BUB1B, CCNA2, CDC20, and CDK1 were significantly overexpressed in deceased PDAC patients (all P<0.01) and in patients with recurrence/disease progression (all P<0.05). In addition, PDAC patients with neoplasms of histologic grade G3-4 had significantly higher BUB1B, CCNA2 and CDC20 levels (all P<0.05). In conclusion, the up-regulation of BUB1B, CCNA2, CDC20, CDK1, and WEE1 in tumor tissues are associated with worse OS and DFS in PDAC and is correlated with advanced tumor stage and tumor development.
Collapse
|