1
|
Yay F, Yıldırım HÇ, Kuş F, Yalçın Ş. Dynamine 3 as a diagnostic and prognostic biomarker in pancreatic cancer: Implications for early detection and targeted therapy. Biomarkers 2025:1-20. [PMID: 39847517 DOI: 10.1080/1354750x.2025.2458104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Dynamins are defined as a group of molecules with GTPase activity. Among them, DNM3 has gained recognition in oncology for its tumor suppressor role. Based on this, the aim of this study is to investigate the effects of the DNM3 gene in patients diagnosed with pancreatic cancer using bioinformatics databases. MATERIALS AND METHODS For differential gene expression analysis, TCGA TARGET GTEx study on the UCSC Xena and GEO datasets were utilized; for the analysis of changes in gene expression according to clinical and pathological characteristics, UALCAN was employed; for Overall Survival (OS) analysis, Kaplan-Meier Plotter was used; for gene alteration analysis, cBioPortal was utilized; for immune cell infiltration analysis, Tumor Immune Estimation Resource (TIMER) and TIMER2.0 were employed; for enrichment analyses Enrichr was used; for Gene Set Correlation Enrichment Analysis Gscore was used on GSE15471; for essentiality of DNM3 gene in pancratic cancer cell lines DepMap was used; and for the detection of miRNAs, miRDB was utilized; ENCORI was used for gene-miRNA correlation and miRNA prognosis analyses. RESULTS In the pancreatic adenocarcinoma (PAAD) cohort, DNM3 gene expression was higher in tumor samples, and there was no significant difference in expression among cancer stages. High levels of DNM3 gene expression were associated with longer OS in PAAD. A weak positive correlation was observed between DNM3 gene expression and B-Cell and CD4+ T Cell infiltrations, while a moderate positive correlation was found with CD8+ T Cell, Macrophage, Neutrophil, and Dendritic Cell infiltrations in TIMER. NK cell by QUANTISEQ, CD 4+ T Cell by TIMER, T cell regulatory (Tregs) by CIBERSORT-ABS infiltrations were positively associated with DNM3 gene expression and decreased risk in prognosis. Common lymphoid progenitor by XCELL and MDSC by TIDE infiltrations were negatively associated with DNM3 gene expression and increased risk of prognosis. Macrophage M1 by QUANTISEQ was positively associated with DNM3 gene expression and increased risk in prognosis. DNM3 gene appears to be associated with various pathways related to inflammation and the immune system. Amplification of the DNM3 gene was detected in 5 out of 175 patients. Enrichment was observed in pathways such as bacterial invasion of epithelial cells, endocytosis, endocrine and other factor-regulated calcium reabsorption, synaptic vesicle cycle, and phospholipase D signaling pathway. According to Gscore, DNM3 gene was associated with Fc epsilon RI signaling pathway, HALLMARK MTORC1 SIGNALING, HALLMARK EPITHELIAL MESENCHYMAL TRANSITION gene sets. According to ENCORI, DNM3 gene was negatively correlated with hsa-miR-203a-3p and increased expression of this miRNA was associated with adverse prognosis in PAAD. CONCLUSIONS The DNM3 gene may play a tumor suppressor role in pancreatic cancer, similar to its role in other malignancies. The contribution of immune cells may also be significant in this effect. However, in vitro studies are needed to elucidate the mechanisms triggered in pancreatic cancer.
Collapse
Affiliation(s)
- Fatih Yay
- Clinical Biochemistry Laboratory, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Hasan Çağrı Yıldırım
- Department of Medical Oncology, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Fatih Kuş
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| | - Şuayib Yalçın
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
2
|
Liu WY, Xu D, Meng HH, Wang CY, Feng X, Wang JS. Metabolic fate of the natural anticancer agent cucurbitacin B: an LC-MS/MS-enabled profiling of its major phase I and II conjugates in vivo. Anal Bioanal Chem 2024; 416:7043-7062. [PMID: 39441433 DOI: 10.1007/s00216-024-05608-y] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Cucurbitacin B (CuB) is a natural triterpenoid with diverse pharmacological effects including potent anticancer activity. However, its oral bioavailability is hampered by limited metabolism in vivo. We characterized CuB's in vivo metabolism in rats to uncover bioactive metabolites retaining therapeutic potential, using a robust UHPLC-Q-TOF-MS/MS workflow. This workflow combined molecular networking, fragmentation filtering, and mass defect filtering to identify CuB metabolites in rat urine, plasma, and feces following oral administration. Thirteen metabolites were identified and seven were confirmed. Major phase I transformations involved hydrolysis, reduction, epoxidation, and amination. Phase II conjugation included cysteine, glutathione, glucuronide, and gluconic acid conjugates. Notably, one of the main metabolites formed was the cysteine conjugate CuB-Cys. CuB-Cys maintained similar in vitro antiproliferative activity to CuB on HepG2, MCF-7, and PANC-1 cancer cell lines. However, it demonstrated lower cytotoxicity towards non-cancerous L02 cells, highlighting improved therapeutic selectivity. Mechanistically, CuB-Cys induced greater apoptotic signaling in HepG2 cells than CuB via enhanced caspase activation and disrupted BAX-Bcl-2 balance. This represents the first systematic characterization of CuB's in vivo metabolic pathway. The identification and confirmation of CuB-Cys provide insight for drug development efforts aiming to maintain therapeutic efficacy while reducing toxicity, via metabolite-based approaches. Our findings shed light on strategies for improving CuB's clinical potential.
Collapse
Affiliation(s)
- Wen-Ya Liu
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Di Xu
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Hui-Hui Meng
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Cheng-Yao Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Xin Feng
- Tibetan Medicine Institute, China Tibetology Research Center, Beijing, 100101, China
| | - Jun-Song Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China.
| |
Collapse
|
3
|
Wang C, Tan J, Jin Y, Li Z, Yang J, Jia Y, Xia Y, Gong B, Dong Q, Zhao Q. A mitochondria-related genes associated neuroblastoma signature - based on bulk and single-cell transcriptome sequencing data analysis, and experimental validation. Front Immunol 2024; 15:1415736. [PMID: 38962012 PMCID: PMC11220120 DOI: 10.3389/fimmu.2024.1415736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Background Neuroblastoma (NB), characterized by its marked heterogeneity, is the most common extracranial solid tumor in children. The status and functionality of mitochondria are crucial in regulating NB cell behavior. While the significance of mitochondria-related genes (MRGs) in NB is still missing in key knowledge. Materials and methods This study leverages consensus clustering and machine learning algorithms to construct and validate an MRGs-related signature in NB. Single-cell data analysis and experimental validation were employed to characterize the pivotal role of FEN1 within NB cells. Results MRGs facilitated the classification of NB patients into 2 distinct clusters with considerable differences. The constructed MRGs-related signature and its quantitative indicators, mtScore and mtRisk, effectively characterize the MRGs-related patient clusters. Notably, the MRGs-related signature outperformed MYCN in predicting NB patient prognosis and was adept at representing the tumor microenvironment (TME), tumor cell stemness, and sensitivity to the chemotherapeutic agents Cisplatin, Topotecan, and Irinotecan. FEN1, identified as the most contributory gene within the MRGs-related signature, was found to play a crucial role in the communication between NB cells and the TME, and in the developmental trajectory of NB cells. Experimental validations confirmed FEN1's significant influence on NB cell proliferation, apoptosis, cell cycle, and invasiveness. Conclusion The MRGs-related signature developed in this study offers a novel predictive tool for assessing NB patient prognosis, immune infiltration, stemness, and chemotherapeutic sensitivity. Our findings unveil the critical function of FEN1 in NB, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxiong Tan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zongyang Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yubin Jia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuren Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiuping Dong
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
4
|
Thorne JW, Redden R, Bowdridge SA, Becker GM, Stegemiller MR, Murdoch BM. Genome-Wide Analysis of Sheep Artificially or Naturally Infected with Gastrointestinal Nematodes. Genes (Basel) 2023; 14:1342. [PMID: 37510248 PMCID: PMC10379027 DOI: 10.3390/genes14071342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
The anthelmintic resistance of gastrointestinal nematodes (GINs) poses a significant threat to sheep worldwide, but genomic selection can serve as an alternative to the use of chemical treatment as a solution for parasitic infection. The objective of this study is to conduct genome-wide association studies (GWASs) to identify single nucleotide polymorphisms (SNPs) in Rambouillet (RA) and Dorper × White Dorper (DWD) lambs associated with the biological response to a GIN infection. All lambs were genotyped with a medium-density genomic panel with 40,598 markers used for analysis. Separate GWASs were conducted using fecal egg counts (FECs) from lambs (<1 year of age) that acquired their artificial infections via an oral inoculation of 10,000 Haemonchus contortus larvae (n = 145) or naturally while grazing on pasture (n = 184). A GWAS was also performed for packed cell volume (PCV) in artificially GIN-challenged lambs. A total of 26 SNPs exceeded significance and 21 SNPs were in or within 20 kb of genes such as SCUBE1, GALNT6, IGF1R, CAPZB and PTK2B. The ontology analysis of candidate genes signifies the importance of immune cell development, mucin production and cellular signaling for coagulation and wound healing following epithelial damage in the abomasal gastric pits via H. contortus during GIN infection in lambs. These results add to a growing body of the literature that promotes the use of genomic selection for increased sheep resistance to GINs.
Collapse
Affiliation(s)
- Jacob W Thorne
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA
- Texas A&M AgriLife Research and Extension, San Angelo, TX 76901, USA
| | - Reid Redden
- Texas A&M AgriLife Research and Extension, San Angelo, TX 76901, USA
| | - Scott A Bowdridge
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV 26506, USA
| | - Gabrielle M Becker
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Morgan R Stegemiller
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Brenda M Murdoch
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
5
|
Wu F, Tian F, Qin C, Qin X, Zeng W, Liu X, Chen C, Lin Y. Peroxiredoxin2 regulates trophoblast proliferation and migration through SPIB-HDAC2 pathway. Exp Cell Res 2023; 422:113428. [PMID: 36400181 DOI: 10.1016/j.yexcr.2022.113428] [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: 05/12/2022] [Revised: 10/20/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
Adequate proliferation and migration of placental trophoblasts is the prerequisite of a successful pregnancy. Peroxiredoxin2 (Prdx2) is a multi-functional gene involved in various signal events to maintain essential biological functions and normal cellular homeostasis. In this study, substantially lower Prdx2 levels were found in the first trimester cytotrophoblasts of women who suffered from recurrent miscarriage (RM). Prdx2 downregulation inhibited trophoblast proliferation and migration. We demonstrated that histone deacetylase2 (HDAC2) acts downstream of Prdx2 in regulating trophoblast proliferation and migration. HDAC2 deacetylates histone-3-lysine-9 in E-cadherin (E-cad) promoter and reduces the transcription of E-cad epigenetically, whereas it promotes the expression of Slug and Snail genes. These molecular changes may contribute to the trophoblast epithelial-mesenchymal transition. We further verified whether Prdx2 modulated the expression of HDAC2 through SPIB. SPIB could bind to the HDAC2 promoter PU-box region and induce HDAC2 expression. In RM, down-regulated Prdx2 suppresses SPIB-HDAC2 pathway, leading to increased E-cad and decreased Slug and Snail, and eventually restrains trophoblast proliferation and migration. Our study unveils the role of Prdx2-regulated SPIB-HDAC2 pathway in the pathology of RM and provides diagnostic and therapeutic targets for RM as well as other "great obstetrical syndromes" including preeclampsia and intrauterine growth restriction.
Collapse
Affiliation(s)
- Fan Wu
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Fuju Tian
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Chuanmei Qin
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Xiaoli Qin
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Weihong Zeng
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Xiaorui Liu
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Diseases, Municipal Key Clinical Speciality, Shanghai, 200030, PR China
| | - Cailian Chen
- Department of Automation, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, 200240, PR China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China.
| |
Collapse
|
6
|
Wong KK, Bateman NW, Ng CW, Tsang YTM, Sun CS, Celestino J, Nguyen TV, Malpica A, Hillman RT, Zhang J, Futreal PA, Rojas C, Conrads KA, Hood BL, Dalgard CL, Wilkerson MD, Phippen NT, Conrads TP, Maxwell GL, Sood AK, Gershenson DM. Integrated multi-omic analysis of low-grade ovarian serous carcinoma collected from short and long-term survivors. J Transl Med 2022; 20:606. [PMID: 36528667 PMCID: PMC9758924 DOI: 10.1186/s12967-022-03820-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Low-grade serous ovarian cancer (LGSOC) is a rare disease that occurs more frequently in younger women than those with high-grade disease. The current treatment is suboptimal and a better understanding of the molecular pathogenesis of this disease is required. In this study, we compared the proteogenomic analyses of LGSOCs from short- and long-term survivors (defined as < 40 and > 60 months, respectively). Our goal was to identify novel mutations, proteins, and mRNA transcripts that are dysregulated in LGSOC, particularly in short-term survivors. METHODS Initially, targeted sequencing of 409 cancer-related genes was performed on 22 LGSOC and 6 serous borderline ovarian tumor samples. Subsequently, whole-genome sequencing analysis was performed on 14 LGSOC samples (7 long-term survivors and 7 short-term survivors) with matched normal tissue samples. RNA sequencing (RNA-seq), quantitative proteomics, and phosphoproteomic analyses were also performed. RESULTS We identified single-nucleotide variants (SNVs) (range: 5688-14,833 per sample), insertion and deletion variants (indels) (range: 880-1065), and regions with copy number variants (CNVs) (range: 62-335) among the 14 LGSOC samples. Among all SNVs and indels, 2637 mutation sites were found in the exonic regions. The allele frequencies of the detected variants were low (median12%). The identified recurrent nonsynonymous missense mutations included KRAS, NRAS, EIF1AX, UBR5, and DNM3 mutations. Mutations in DNM3 and UBR5 have not previously been reported in LGSOC. For the two samples, somatic DNM3 nonsynonymous missense mutations in the exonic region were validated using Sanger sequencing. The third sample contained two missense mutations in the intronic region of DNM3, leading to a frameshift mutation detected in RNA transcripts in the RNA-seq data. Among the 14 LGSOC samples, 7754 proteins and 9733 phosphosites were detected by global proteomic analysis. Some of these proteins and signaling pathways, such as BST1, TBXAS1, MPEG1, HBA1, and phosphorylated ASAP1, are potential therapeutic targets. CONCLUSIONS This is the first study to use whole-genome sequencing to detect somatic mutations in LGSOCs with matched normal tissues. We detected and validated novel mutations in DNM3, which were present in 3 of the 14 samples analyzed. Additionally, we identified novel indels, regions with CNVs, dysregulated mRNA, dysregulated proteins, and phosphosites that are more prevalent in short-term survivors. This integrated proteogenomic analysis can guide research into the pathogenesis and treatment of LGSOC.
Collapse
Affiliation(s)
- Kwong-Kwok Wong
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Nicholas W. Bateman
- grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Chun Wai Ng
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Yvonne T. M. Tsang
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Charlotte S. Sun
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Joseph Celestino
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Tri V. Nguyen
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Anais Malpica
- grid.240145.60000 0001 2291 4776Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - R. Tyler Hillman
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Jianhua Zhang
- grid.240145.60000 0001 2291 4776Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - P. Andrew Futreal
- grid.240145.60000 0001 2291 4776Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Christine Rojas
- grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA
| | - Kelly A. Conrads
- grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Brian L. Hood
- grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Clifton L. Dalgard
- grid.265436.00000 0001 0421 5525Department of Anatomy, Physiology and Genetics and Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD USA
| | - Matthew D. Wilkerson
- grid.265436.00000 0001 0421 5525Department of Anatomy, Physiology and Genetics and Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD USA
| | - Neil T. Phippen
- grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA
| | - Thomas P. Conrads
- grid.414629.c0000 0004 0401 0871Women’s Health Integrated Research Center at Inova Health System, Women’s Service Line, Inova Fairfax Medical Campus, Falls Church, VA USA ,grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA
| | - George L. Maxwell
- grid.414629.c0000 0004 0401 0871Women’s Health Integrated Research Center at Inova Health System, Women’s Service Line, Inova Fairfax Medical Campus, Falls Church, VA USA ,grid.414467.40000 0001 0560 6544Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD USA
| | - Anil K. Sood
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - David M. Gershenson
- grid.240145.60000 0001 2291 4776Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Room T4-3900, Clinical Research Building, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| |
Collapse
|
7
|
Xu C, Song L, Peng H, Yang Y, Liu Y, Pei D, Guo J, Liu N, Liu J, Li X, Li C, Kang Z. Clinical Eosinophil-Associated Genes can Serve as a Reliable Predictor of Bladder Urothelial Cancer. Front Mol Biosci 2022; 9:963455. [PMID: 35936781 PMCID: PMC9353774 DOI: 10.3389/fmolb.2022.963455] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 01/04/2023] Open
Abstract
Background: Numerous studies have shown that infiltrating eosinophils play a key role in the tumor progression of bladder urothelial carcinoma (BLCA). However, the roles of eosinophils and associated hub genes in clinical outcomes and immunotherapy are not well known. Methods: BLCA patient data were extracted from the TCGA database. The tumor immune microenvironment (TIME) was revealed by the CIBERSORT algorithm. Candidate modules and hub genes associated with eosinophils were identified by weighted gene co-expression network analysis (WGCNA). The external GEO database was applied to validate the above results. TIME-related genes with prognostic significance were screened by univariate Cox regression analysis, lasso regression, and multivariate Cox regression analysis. The patient's risk score (RS) was calculated and divided subjects into high-risk group (HRG) and low-risk group (LRG). The nomogram was developed based on the risk signature. Models were validated via receiver operating characteristic (ROC) curves and calibration curves. Differences between HRG and LRG in clinical features and tumor mutational burden (TMB) were compared. The Immune Phenomenon Score (IPS) was calculated to estimate the immunotherapeutic significance of RS. Half-maximal inhibitory concentrations (IC50s) of chemotherapeutic drugs were predicted by the pRRophetic algorithm. Results: 313 eosinophil-related genes were identified by WGCNA. Subsequently, a risk signature containing 9 eosinophil-related genes (AGXT, B3GALT2, CCDC62, CLEC1B, CLEC2D, CYP19A1, DNM3, SLC5A9, SLC26A8) was finally developed via multiplex analysis and screening. Age (p < 0.001), grade (p < 0.001), and RS (p < 0.001) were independent predictors of survival in BLCA patients. Based on the calibration curve, our risk signature nomogram was confirmed as a good predictor of BLCA patients' prognosis at 1, 3, and 5 years. The association analysis of RS and immunotherapy indicated that low-risk patients were more credible for novel immune checkpoint inhibitors (ICI) immunotherapy. The chemotherapeutic drug model suggests that RS has an effect on the drug sensitivity of patients. Conclusions: In conclusion, the eosinophil-based RS can be used as a reliable clinical predictor and provide insights into the precise treatment of BLCA.
Collapse
Affiliation(s)
- Chaojie Xu
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lishan Song
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Hui Peng
- Department of Orthopaedics, Affiliated National Hospital of Guangxi Medical University, Nanning, China
| | - Yubin Yang
- College of Pharmacy, Shantou University School of Medicine, Shantou, China
| | - Yi Liu
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Dongchen Pei
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jianhua Guo
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Nan Liu
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jiabang Liu
- College of Pharmacy, Shantou University School of Medicine, Shantou, China
| | - Xiaoyong Li
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, Berlin, Germany
| | - Zhengjun Kang
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Role of main RNA modifications in cancer: N 6-methyladenosine, 5-methylcytosine, and pseudouridine. Signal Transduct Target Ther 2022; 7:142. [PMID: 35484099 PMCID: PMC9051163 DOI: 10.1038/s41392-022-01003-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the major diseases threatening human life and health worldwide. Epigenetic modification refers to heritable changes in the genetic material without any changes in the nucleic acid sequence and results in heritable phenotypic changes. Epigenetic modifications regulate many biological processes, such as growth, aging, and various diseases, including cancer. With the advancement of next-generation sequencing technology, the role of RNA modifications in cancer progression has become increasingly prominent and is a hot spot in scientific research. This review studied several common RNA modifications, such as N6-methyladenosine, 5-methylcytosine, and pseudouridine. The deposition and roles of these modifications in coding and noncoding RNAs are summarized in detail. Based on the RNA modification background, this review summarized the expression, function, and underlying molecular mechanism of these modifications and their regulators in cancer and further discussed the role of some existing small-molecule inhibitors. More in-depth studies on RNA modification and cancer are needed to broaden the understanding of epigenetics and cancer diagnosis, treatment, and prognosis.
Collapse
|
9
|
Lu Q, Ni Y, Wang W, Wang L, Jiang T, Shang L. Dynamin 3 Inhibits the Proliferation of Non-small-Cell Lung Cancer Cells by Suppressing c-MET-GBR2-STAT3 Complex Formation. Front Cell Dev Biol 2021; 9:641403. [PMID: 34490234 PMCID: PMC8416685 DOI: 10.3389/fcell.2021.641403] [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: 12/14/2020] [Accepted: 07/20/2021] [Indexed: 01/05/2023] Open
Abstract
Dynamin 3 (DNM3) has gained increased attention ever since its potential as a tumor suppressor was reported. However, its action in lung cancer (LC) is undefined. In this study, the role of DNM3 in LC development was investigated. DNM3 expression was found to be downregulated in tumors of patients with LC, especially those with metastasis. The DNM3 downregulation enhanced the proliferative and metastatic ability of LC cells, whereas its upregulation had the opposite effects. In vivo xenograft experiments confirmed that lung tumors with lower DNM3 expression had higher growth and metastatic abilities. Mechanistic studies revealed that DNM3 interacts with growth factor receptor-bound protein 2 (GBR2), thereby interrupting tyrosine-protein kinase Met (c-MET)-GBR2-signal transducer and activator of transcription 3 (STAT3) complex formation, which suppressed STAT3 activation. Therefore, the absence of DNM3 frees GBR2 to activate STAT3, which regulates the expression of genes related to LC proliferation and metastasis (e.g., cyclin D1 and Snail family transcriptional repressor 1). Additionally, the c-MET inhibitor crizotinib effectively suppressed LC cell proliferation and migration in vitro and in vivo, even with DNM3 depleted. Therefore, our study has demonstrated the antitumor effect of DNM3 in LC and suggests that the inhibition of c-MET might be a promising strategy for treating those LC patients with low DNM3 expression.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Yunfeng Ni
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Wuping Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Lei Shang
- The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Department of Health Statistics, School of Public Health, The Air Force Military Medical University, Xi'an, China
| |
Collapse
|
10
|
JuknytĖ G, LaurinaitytĖ I, VilkeviČiŪtĖ A, GedvilaitĖ G, GlebauskienĖ B, KriauČiŪnienĖ L, LiutkeviČienĖ R. TBX15 rs98422, DNM3 rs1011731, RAD51B rs8017304, and rs2588809 Gene Polymorphisms and Associations With Pituitary Adenoma. In Vivo 2021; 35:815-826. [PMID: 33622874 DOI: 10.21873/invivo.12322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Pituitary adenoma (PA) is a benign tumor of parenchymal cells in the adenohypophysis, and it's development is strongly associated with genetic factors.This study aim was to find whether TBX15 rs98422, DNM3 rs1011731, RAD51B rs8017304, and rs2588809 single nucleotide polymorphisms can be associated with pituitary adenoma. While the TBX15 gene belongs to the T-box family of genes and is a transcription factor involved in many developmental processes, the DNM3 encodes a protein that is a member of the dynamin family with mechanochemical properties involved in actin-membrane processes, predominantly in membrane budding, and the RAD51B gene plays a significant role in homologous recombination in DNA repair for genome stability. MATERIALS AND METHODS The study enrolled 113 patients with pituitary adenoma and 283 healthy control subjects. DNA samples were extracted and purified from peripheral blood leukocytes. Genotyping was carried out using real-time polymerase chain reaction. The results were assessed using binomial logistic regression. RESULTS Our study revealed that RAD51B rs2588809 TT genotype could be associated with PA development in the co-dominant (OR=6.833; 95% CI=2.557-18.262; p<0.001) and recessive (OR=7.066; 95% CI=2.667-18.722; p<0.001) models. The same results were observed in females but not in males and PA without recurrence, while in PA with recurrence, no statistically significant results were obtained. CONCLUSION RAD51B rs2588809 TT genotype may increase the odds of PA development in women; it may also be associated with non-recurrent PA development.
Collapse
Affiliation(s)
- Gabija JuknytĖ
- Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Inga LaurinaitytĖ
- Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alvita VilkeviČiŪtĖ
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Greta GedvilaitĖ
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Brigita GlebauskienĖ
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Loresa KriauČiŪnienĖ
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa LiutkeviČienĖ
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
11
|
Fa J. Dynamin 3 overexpression suppresses the proliferation, migration and invasion of cervical cancer cells. Oncol Lett 2021; 22:524. [PMID: 34025791 PMCID: PMC8130055 DOI: 10.3892/ol.2021.12785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Dynamin 3 (DNM3) functions as a tumor suppressor in various malignancies. However, the underlying mechanism of DNM3 in cervical cancer remains to be elucidated. The present study aimed to indicate the function of DNM3 in cervical cancer. The expression of DNM3 in cervical tissues and cells was measured using bioinformatics analysis, immunohistochemistry and reverse transcription-quantitative PCR. The pcDNA3.1 plasmid was used to overexpress DNM3 in SiHa and C33A cells. The effects of DNM3 overexpression on cell proliferation, migration, invasion and apoptosis was detected by the CCK-8, clone formation, Transwell, flow cytometry and western blotting assays. In the present study, it was revealed that DNM3 was expressed at significantly low levels in cervical cancer tissues and cell lines compared with normal cervical tissues and cell lines. In addition, the low expression of DNM3 was significantly associated with high pathological grading of cervical cancer. The overall survival rate of patients with low DNM3 expression was significantly improved compared with patients with high DNM3 expression. In addition, the overexpression of DNM3 significantly inhibited the proliferation, migration and invasion of cervical carcinoma cells and induced cell apoptosis. The findings of the present study further revealed that the overexpression of DNM3 may inhibit cell migration and invasion by inactivating the epithelial mesenchymal transition process. In summary, the present study demonstrated that DNM3 was a tumor suppressor in cervical cancer progression and that it may serve as a potential prognostic biomarker for patients with cervical carcinoma.
Collapse
Affiliation(s)
- Jing Fa
- Department of Obstetrics and Gynecology, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
12
|
Li W, Yan P, Meng X, Zhang J, Yang Y. The microRNA cluster miR-214/miR-3120 prevents tumor cell switching from an epithelial to a mesenchymal-like phenotype and inhibits autophagy in gallbladder cancer. Cell Signal 2020; 80:109887. [PMID: 33340658 DOI: 10.1016/j.cellsig.2020.109887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Tumor cells switch from an epithelial to a mesenchymal-like phenotype, which represents a key hallmark of human cancer metastasis, including gallbladder cancer (GBC). A large set of microRNAs (miRNAs/miRs) have been studied to elucidate their functions in initiating or inhibiting this phenotypic switching in GBC cells. In this paper, we attempted to identify the expression pattern of the miR-214/-3120 cluster and its mode of action in the context of GBC, with a specific focus being placed on their effects on EMT and autophagy in GBC cells. Human GBC cells GBC-SD were assayed for their migration, invasion, and autophagy using the Transwell chamber system, MDC staining, and transmission electron microscopy. The tumorigenicity and metastatic behavior of GBC-SD cells were tested in nude mice. The expression of EMT- and autophagy-specific markers (E-cadherin, N-cadherin, vimentin, ATG5, LC3II/LC3I, and Beclin1) was analyzed in cultured GBC-SD cells and in human GBC-SD xenografts. The E2F3 luciferase reporter activity in the presence of miR-214/-3120 was evaluated by a dual luciferase assay. The miR-214/-3120 was downregulated in GBC. Exogenous miR-214/-3120 inhibited the phenotypic switching of GBC cells from epithelial to mesenchymal, prevented autophagy, and suppressed the tumorigenicity and metastatic behavior of GBC-SD cells in vitro and in vivo. E2F3 was demonstrated to be the target gene of miR-214/-3120, and its knockdown in part mimicked the effect of miR-214/-3120 on the EMT, autophagy, tumorigenicity, and metastatic behavior of GBC-SD cells. These results demonstrated that the miR-214/-3120 cluster blocks the process of EMT and autophagy to limit GBC metastasis by repressing E2F3 expression.
Collapse
Affiliation(s)
- Wujun Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, PR China
| | - Pu Yan
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, PR China
| | - Xiaofen Meng
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Jinpei Zhang
- Department of Encephalopathy, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, PR China.
| | - Yi Yang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China.
| |
Collapse
|
13
|
PRDX2 Promotes the Proliferation and Metastasis of Non-Small Cell Lung Cancer In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8359860. [PMID: 32908916 PMCID: PMC7474358 DOI: 10.1155/2020/8359860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
Purpose Previous studies have reported that the levels of PRDX2 were correlated with tumorigenicity, recurrence, and prognosis of patients with different cancers. We investigated the association between PRDX2 levels and the prognosis of lung cancer patients. We also measured PRDX2 expression of non-small cell lung cancer (NSCLC) cells and examined its roles in the proliferation and migration in vitro and in vivo. Methods We used the Kaplan–Meier plotter to analyze the survival of different levels of PRDX2 in lung cancer patients. The expression of PRDX2 in normal bronchial epithelial cell line and NSCLC cell lines was measured by qRT-PCR and western blot assays. Biological functions of NSCLC cells were detected by CCK8 and Transwell assays. We constructed tumor growth model using subcutaneously injection of nude mice and metastasis model by tail vein injection in vivo. The protein levels of proliferation related markers were measured by immunohistochemistry assay. Immunofluorescence method was used to detected EMT-related proteins. Results The high levels of PRDX2 were associated with bad prognosis in lung cancer patients, especially in patients with adenocarcinoma. The expression of PRDX2 in NSCLC cell lines was higher than normal bronchial epithelial cells. Knockdown of PRDX2 inhibited the proliferation, migration, and invasion in A549 cells, while overexpression of PRDX2 promoted the malignancy in NCI-H1299 cells in vitro. Silencing PRDX2 restrained tumor growth and repressed lung metastasis by EMT in vivo. Conclusion Our data indicates that PRDX2 functions as a protumor regulator and is involved in tumorigenesis and tumor progression of lung cancer.
Collapse
|
14
|
Lu C, Xie T, Guo X, Wu D, Li S, Li X, Lu Y, Wang X. LncRNA DSCAM-AS1 Promotes Colon Cancer Cells Proliferation and Migration via Regulating the miR-204/SOX4 Axis. Cancer Manag Res 2020; 12:4347-4356. [PMID: 32606930 PMCID: PMC7293419 DOI: 10.2147/cmar.s250670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Long non-coding RNA (lncRNA) DSCAM-AS1 was reported to be aberrantly expressed and play pivotal roles in various human cancers. The aim of the present study was to investigate the expression and roles of DSCAM-AS1 in colon cancer (CC). Methods Quantitative real-time PCR (qRT-PCR) was used to detect the expression of DSCAM-AS1, miR-204 and the mRNA level of SOX4. Cell proliferation and cell cycle were analyzed by MTT assay and flow cytometry, respectively. Transwell assay was used for migration capacity detection. Luciferase activity assay was conducted to verify the direct binding of DSCAM-AS1 and miR-204 or miR-204 and SOX4. The protein expression of SOX4 was determined by Western blot. Kaplan–Meier curves were calculated and the Log rank test was performed for the survival data analysis. Results DSCAM-AS1 was significantly upregulated in CC and high expression of DSCAM-AS1 was associated with poor prognosis in patients with colon cancer. Knockdown of DSCAM-AS1 significantly suppressed CC cells proliferation and migration. In addition, DSCAM-AS acted as a molecular sponge for miR-204 and SOX4 was identified as a direct target of miR-204 in CC. Moreover, the rescue assay revealed that miR-204 inhibition partly abolished the effects of DSCAM-AS1 knockdown on CC cells proliferation, migration and SOX4 expression. Discussion The present study demonstrated that DSCAM-AS1 acted as an oncogenic lncRNA in CC progression by regulating miR-204/SOX4 axis and DSCAM-AS1 may serve as a novel therapeutic target in the treatment of colon cancer.
Collapse
Affiliation(s)
- Canrong Lu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Tianyu Xie
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Xin Guo
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Di Wu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Shuo Li
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Xiongguang Li
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Yixun Lu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| | - Xinxin Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, People's Republic of China
| |
Collapse
|
15
|
Silencing of PRDX2 Inhibits the Proliferation and Invasion of Non-Small Cell Lung Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1276328. [PMID: 32337219 PMCID: PMC7157786 DOI: 10.1155/2020/1276328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022]
Abstract
Peroxiredoxin 2 (PRDX2), a member of the peroxiredoxin family of antioxidant enzymes, has been revealed to be an important player in cancer progression. However, the biological role of PRDX2 in the progression of non-small cell lung cancer (NSCLC) is poor reported. In the present study, the loss-of-function experiments were performed to investigate the specific role of PRDX2 in the growth and invasion of NSCLC. The results revealed that knockdown of PRDX2 by siRNA interference significantly suppressed the proliferation, migration, and invasion of A549 and H1299 cells, as well as diminished the activity of MMP9. Additionally, the decrease in PRDX2 expression significantly promoted apoptosis in NSCLC cells by downregulating expression of Bcl-2 and upregulating the expression of Bax, cleaved caspase 3 and cleaved caspase 9, but had no significant effect on the apoptosis of normal lung epithelial cells BEAS-2B. Moreover, PRDX2 inhibitor also inhibited the proliferation, migration, and invasion of A549 cells and promoted apoptosis. Further, our data demonstrated that silencing of PRDX2 markedly reduced the phosphorylation of Akt and mTOR and expression of downstream proteins Cyclin D1 and p70S6k. In conclusion, our findings indicate that PRDX2 exerts a prooncogenic role in the progression of NSCLC and might be a potential therapeutic target for NSCLC treatment.
Collapse
|
16
|
Systematic Characterization of Prognostic Values of Peroxiredoxin Family in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3948183. [PMID: 32382548 PMCID: PMC7199545 DOI: 10.1155/2020/3948183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/06/2019] [Indexed: 12/24/2022]
Abstract
The peroxiredoxin (PRDX) gene family has been reported to participate in regulating occurrence and development of cancerous diseases, but its exact prognostic values in gastric cancer (GC) remain largely elusive. In the current research, we evaluated the prognostic value in predicting overall survival (OS) of each individual PRDX mRNA expression based on patients' cohorts from the Kaplan–Meier (KM) plotter database, which contains clinical information and gene expression data obtained from a total of 876 GC patients. Our results revealed that mRNA expressions of PRDX1, PRDX2, PRDX3, and PRDX4 were significantly associated with worse OS in GC patients, whereas PRDX5 and PRDX6 mRNA expressions were not associated with OS in GC patients. In addition, the prognostic values of PRDXs in the different clinicopathological features according to clinical stages, Lauren classifications, HER2 expression status, differentiation degree, and treatment strategies of GC patients were further evaluated in the KM plotter database. As a result, more potential beneficiaries who may benefit from prognostic assessment using PRDX mRNA expressions were identified. Our results elucidated the exact values of PRDXs in assessing GC prognosis and might provide primary evidence for further study on the mechanism of PRDXs participating in occurrence and development of GC.
Collapse
|