1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Zou F, Rao T, Chen W, Song T, Li T, Hu W, Li L, Yu W, Cheng F. DUSP2 affects bladder cancer prognosis by down-regulating MEK/ERK and P38 MAPK signaling pathways through PTPN7. Cell Signal 2023; 112:110893. [PMID: 37739277 DOI: 10.1016/j.cellsig.2023.110893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND As one of the leading causes of cancer death worldwide, bladder cancer (BCa) ranks 12th in incidence rate. Dual Specific Phosphatase 2 (DUSP2) is a member of the bispecific protein phosphatase subfamily. DUSP2 is closely related to the prognosis of cancer, but the role of DUSP2 in bladder cancer is still unclear. This study aims to explore how DUSP2 affects the prognosis of bladder cancer and clarify the important mechanism in bladder cancer. METHODS Bioinformatics and experiments have detected the anti-tumor effect of DUSP2. Construct a DUSP2 overexpression cell model, and then use protein blotting experiments to verify the efficiency of transfection. The effects of DUSP2 on proliferation, metastasis, apoptosis, epithelial mesenchymal transition (EMT) and immune invasion of bladder cancer cells were detected in vitro or in vivo. In addition, the mechanism of DUSP2 regulating MEK/ERK through PTPN7 pathway and P38 MAPK inhibiting the progression of bladder cancer was also discussed. RESULTS The expression of DUSP2 was down regulated in bladder cancer samples and cell lines. The overexpression of DUSP2 inhibits the proliferation, metastasis and immune microenvironment of bladder cancer cells. In addition, we confirmed that DUSP2 regulates MEK/ERK and P38 MAPK through PTPN7 pathway to inhibit the progression of bladder cancer. CONCLUSION DUSP2 inhibits the progression of bladder cancer by regulating PTPN7. These results suggest that DUSP2/PTPN7/MEK/ERK pathway may become a new therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Fan Zou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianbao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tongjie Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430026, China
| | - Weimin Hu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Linzhi Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
3
|
Nakagawa-Saito Y, Mitobe Y, Suzuki S, Togashi K, Sugai A, Kitanaka C, Okada M. Domatinostat Targets the FOXM1-Survivin Axis to Reduce the Viability of Ovarian Cancer Cells Alone and in Combination with Chemotherapeutic Agents. Int J Mol Sci 2023; 24:10817. [PMID: 37445993 DOI: 10.3390/ijms241310817] [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: 05/29/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The deregulation of the FOXM1 transcription factor is a key molecular alteration in ovarian cancer, contributing to the development and progression of ovarian cancer via activation of the target genes. As such, FOXM1 is a highly attractive therapeutic target in the treatment of ovarian cancer, but there has been no clinically tested FOXM1 inhibitor to date. We investigated in this study the effects of domatinostat, a class I-selective HDAC inhibitor currently in the clinical stage of development as a cancer therapeutic, on the expression of FOXM1 and viability of ovarian cancer cells. Cell viability, as well as protein and mRNA expression of FOXM1 and its transcriptional target survivin, was examined after domatinostat treatment of TOV21G and SKOV3 ovarian cancer cell lines in the absence or presence of cisplatin and paclitaxel. The effect of FOXM1 knockdown on survivin expression and those of genetic and pharmacological inhibition of survivin alone or in combination with the chemotherapeutic agents on cell viability were also examined. Domatinostat reduced the protein and mRNA expression of FOXM1 and survivin and also the viability of ovarian cancer cells alone and in combination with cisplatin or paclitaxel at clinically relevant concentrations. Knockdown experiments showed survivin expression was dependent on FOXM1 in ovarian cancer cells. Survivin inhibition was sufficient to reduce the viability of ovarian cancer cells alone and in combination with the chemotherapeutic agents. Our findings suggest that domatinostat, which effectively targets the FOXM1-survivin axis required for the viability of ovarian cancer cells, is a promising option for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yurika Nakagawa-Saito
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yuta Mitobe
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
- Department of Neurosurgery, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Shuhei Suzuki
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
- Department of Clinical Oncology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Keita Togashi
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
- Department of Ophthalmology and Visual Sciences, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Asuka Sugai
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
- Research Institute for Promotion of Medical Sciences, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Masashi Okada
- Department of Molecular Cancer Science, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| |
Collapse
|
4
|
Güler S, Yalçın A. Expression of dual-specificity phosphatases in TGFß1-induced EMT in SKOV3 cells. Turk J Med Sci 2023; 53:640-646. [PMID: 37476896 PMCID: PMC10387886 DOI: 10.55730/1300-0144.5626] [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: 09/16/2022] [Accepted: 02/21/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND The study aims to profile the dual-specificity phosphatases (DUSP) expression in response to Transforming growth factor β1 (TGFβ1)-induced epithelial- mesenchymal transition (EMT) in ovarian adenocarcinoma cells. METHODS The ovarian adenocarcinoma cell line SKOV3 was used as a TGFβ1-induced EMT model. Cells were incubated with 5 ng/mL TGFβ1 to induce EMT. EMT was confirmed with real-time qPCR, western blot, and immunofluorescence analyses of various EMT markers. Western blot was used to analyze phospho- and total MAPK protein levels. Typical and atypical DUSPs mRNA expression profile was determined by real-time qPCR. RESULTS The epithelial marker E-cadherin expressions were decreased and mesenchymal EMT markers Snail and Slug expression levelswere increased after TGFβ1 induction. Phosphorylation of ERK1/2 and p38 MAPK were enhanced in response to TGFβ1 treatment. The expression of DUSP2, DUSP6, DUSP8, DUSP10, and DUSP13 were decreased while DUSP7, DUSP16, DUSP18, DUSP21, and DUSP27 were increased by TGFβ1. DISCUSSION TGFβ1 induced EMT which was accompanied by increased activity of MAPKs, and led to marked changes in expressions of several DUSPs in SKOV3 cells.
Collapse
Affiliation(s)
- Sabire Güler
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Abdullah Yalçın
- Department of Biochemistry, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
5
|
Sanders BE, Yamamoto TM, McMellen A, Woodruff ER, Berning A, Post MD, Bitler BG. Targeting DUSP Activity as a Treatment for High-Grade Serous Ovarian Carcinoma. Mol Cancer Ther 2022; 21:1285-1295. [PMID: 35587258 PMCID: PMC9357222 DOI: 10.1158/1535-7163.mct-21-0682] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/21/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023]
Abstract
Identifying novel, durable treatments for high-grade serous ovarian cancer (HGSOC) is paramount to extend both progression-free survival (PFS) and overall survival (OS) in patients afflicted with this disease. Dual-specificity phosphatase 1 (DUSP1) was identified as one of seven genes that may significantly affect prognosis in patients with HGSOC; however, the role of DUSP inhibition (DUSPi) in the treatment of HGSOC remains largely unknown. In this study, we show that DUSP1 is highly expressed in HGSOC and confers worse PFS and OS. Further, we corroborate data that show DUSP1 expression is directly associated with therapy resistance. Using a tissue microarray of 137 different serous ovarian carcinomas, we demonstrate the high expression of DUSP1 in primary and recurrent serous ovarian cancer. In both acquired and de novo therapy HGSOC-resistant models, DUSPi both inhibited cellular proliferation and promoted cell death. RPPA analysis of HGSOC cells revealed DUSPi led to the differential regulation of several pathways, including AMPK and mTORC. Further, in a patient-derived xenograft HGSOC model, DUSPi significantly inhibited tumor progression.
Collapse
Affiliation(s)
- Brooke E. Sanders
- Department of Obstetrics & Gynecology, Division of
Gynecologic Oncology, The University of Colorado Anschutz Medical Campus, Aurora, CO
80045, USA
- University of Colorado Comprehensive Cancer Center, Aurora,
CO 80045, USA
| | - Tomomi M. Yamamoto
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Alexandra McMellen
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Elizabeth R. Woodruff
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Amber Berning
- Department of Pathology, The University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Miriam D. Post
- Department of Pathology, The University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin G. Bitler
- University of Colorado Comprehensive Cancer Center, Aurora,
CO 80045, USA
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| |
Collapse
|
6
|
Song J, Xu X, He S, Wang N, Bai Y, Li B, Zhang S. Exosomal hsa_circ_0017252 attenuates the development of gastric cancer via inhibiting macrophage M2 polarization. Hum Cell 2022; 35:1499-1511. [PMID: 35796939 DOI: 10.1007/s13577-022-00739-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/15/2022] [Indexed: 01/07/2023]
Abstract
Gastric cancer (GC) is an aggressive malignant tumor of the digestive system, with high morbidity rates. We previously demonstrated that miR-17-5p can modify tumorigenesis in GC. In addition, other studies have shown that circRNAs can regulate GC progression by sponging various miRNAs. However, the association between circRNAs and miR-17-5p in GC has not yet been explored. Hence, this study aimed to explore the possible interactions between various circRNAs and miR-17-5p using a dual-luciferase assay. CCK-8 was used to determine cell viability, and a Transwell assay was used to measure cell invasion and migration. Gene expression was assessed using quantitative reverse transcription PCR (RT-qPCR), and exosomes were identified using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Annexin V/PI staining was also used to detect cell apoptosis. These investigations collectively revealed that miR-17-5p is a target of the circRNA hsa_circ_0017252 and hsa_circ_0017252 is significantly downregulated in GC tissues. In addition, the overexpression of hsa_circ_0017252 inhibited GC cell migration by sponging of miR-17-5p, and GC cell-secreted exosomal hsa_circ_0017252 effectively inhibited macrophage M2-like polarization, which in turn suppressed GC cell invasion. Notably, exosomes containing hsa_circ_0017252 also suppressed GC tumor growth in vivo. Thus, our data suggest that the overexpression of hsa_circ_0017252 suppresses GC malignancy by sponging miR-17-5p. In addition, exosomal hsa_circ_0017252 excreted from GC cells attenuated GC progression by suppressing macrophage M2-like polarization. These findings improve our basic understanding of GC and open a novel avenue for developing more effective GC treatments.
Collapse
Affiliation(s)
- Jin Song
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.,Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.,Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.,Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Ning Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.,Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Yunjing Bai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.,Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Bo Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China. .,Beijing Institute of Chinese Medicine, Beijing, 100010, China.
| | - Shengsheng Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23 Back Street, Art Museum, Beijing, 100010, China.
| |
Collapse
|
7
|
Zhang F, Yin Y, Xu W, Zhou Z, Sun X, Li P. Apatinib combined with Keytruda treatment induces apoptosis of gastric carcinoma cells through CES4/miR-616-5p/DUSP2 axis. Basic Clin Pharmacol Toxicol 2021; 129:345-356. [PMID: 34365722 DOI: 10.1111/bcpt.13641] [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: 02/23/2021] [Revised: 07/05/2021] [Accepted: 08/02/2021] [Indexed: 01/02/2023]
Abstract
Gastric carcinoma (GC) is a highly malignant and heterogeneous tumour. Long non-coding RNA CES4 is down-regulated in GC. However, whether CES4 can participate in GC remains unclear; we have carried out research on this topic. GC cells (HGC-27 and MKN-7) were treated with anti-tumour drugs: apatinib combined with Keytruda. Cell viability and apoptosis were detected by CCK-8 assay and flow cytometry. Gene and protein expression were examined by quantitative real-time PCR and western blot. Luciferase reporter assay was performed to verify the relationship among CES4, miR-616-5p and dual-specificity phosphatase-2 (DUSP2). CES4 was highly expressed in the apatinib combined with Keytruda-treated HGC-27 and MKN-7 cells. Apatinib combined with Keytruda treatment repressed cell viability and promoted apoptosis of HGC-27 and MKN-7 cells, which was abrogated by CES4 knockdown. Furthermore, CES4 promoted DUSP2 expression by sponging miR-616-5p in HGC-27 and MKN-7 cells. CES4 knockdown promoted cell viability and inhibited apoptosis of drug-treated HGC-27 and MKN-7 cells by regulating miR-616-5p/DUSP2 axis. In conclusion, these data demonstrate that apatinib combined with Keytruda treatment induces apoptosis of GC cells through CES4/miR-616-5p/DUSP2 axis. Thus, this work provides the experimental basis for the combination of apatinib and Keytruda as a treatment for GC.
Collapse
Affiliation(s)
- Fengli Zhang
- Department of Traditional Chinese and Western Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanfen Yin
- Department of Oncology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Wenwen Xu
- The Graduate School, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Zhou Zhou
- Department of Traditional Chinese and Western Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xin Sun
- Department of Traditional Chinese and Western Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ping Li
- Department of Traditional Chinese and Western Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
8
|
Liu S, Wu M, Wang F. Research Progress in Prognostic Factors and Biomarkers of Ovarian Cancer. J Cancer 2021; 12:3976-3996. [PMID: 34093804 PMCID: PMC8176232 DOI: 10.7150/jca.47695] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is a serious threat to women's health; its early diagnosis rate is low and prone to metastasis and recurrence. The current conventional treatment for ovarian cancer is a combination of platinum and paclitaxel chemotherapy based on surgery. The recurrence and progression of ovarian cancer with poor prognosis is a major challenge in treatment. With rapid advances in technology, understanding of the molecular pathways involved in ovarian cancer recurrence and progression has increased, biomarker-guided treatment options can greatly improve the prognosis of patients. This review systematically discusses and summarizes existing and new information on prognostic factors and biomarkers of ovarian cancer, which is expected to improve the clinical management of patients and lead to effective personalized treatment.
Collapse
Affiliation(s)
- Shuna Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Ming Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| |
Collapse
|