1
|
Pang M, Sun X, He T, Liang H, Yang H, Chen J. Development of a prognostic model based on anoikis-related genes for predicting clinical prognosis and immunotherapy of hepatocellular carcinoma. Aging (Albany NY) 2023; 15:10253-10271. [PMID: 37787988 PMCID: PMC10599733 DOI: 10.18632/aging.205073] [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: 04/21/2023] [Accepted: 06/19/2023] [Indexed: 10/04/2023]
Abstract
Hepatocellular Carcinoma (HCC) is the predominant cause of cancer-related mortality worldwide. The majority of HCC patients are diagnosed at advanced stages of the disease, with a high likelihood of metastasis and unfavorable prognosis. Anoikis resistance is a crucial factor contributing to tumor invasion and metastasis, although its specific role in HCC remains unclear. Based on the results of univariate Cox regression and least absolute shrink-age and selection operator (LASSO) analysis, a subset of anoikis-related genes (ARGs) significantly associated with overall survival (OS) was identified. A multivariate Cox regression analysis subsequently identified PDK4, STK11, and TFDP1 as three prognostic ARGs, which were then used to establish a prognostic risk model. Differences in OS caused by risk stratification in HCC patients were demonstrated. The nomogram analysis indicated that the ARGs prognostic signature served as an independent prognostic predictor. In vitro experiments further confirmed the abnormal expression of selected ARGs in HCC. The association between risk scores and OS was further examined through Kaplan-Meier analysis, CIBERSORT analysis, and single-sample gene set enrichment analysis (ssGSEA). This study is a pioneering effort to integrate multiple ARGs and establish a risk-predictive model, providing a unique perspective for the development of personalized and precise therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Mu Pang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Xizhe Sun
- Research Center for Drug Safety Evaluation of Hainan, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ting He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Huichao Liang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Hao Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Jun Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| |
Collapse
|
2
|
Wu M, Ye M. Transcription factor Dp-1 knockdown downregulates thymidine kinase 1 expression to protect against proliferation and epithelial-mesenchymal transition in cervical cancer. Funct Integr Genomics 2023; 23:301. [PMID: 37715794 DOI: 10.1007/s10142-023-01218-6] [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: 07/19/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/18/2023]
Abstract
Thymidine kinase 1 (TK1) level is an independent survival prognostic factor for both premalignant and malignant cervical pathologies. Herein, this study sought to probe the impacts of TK1 on cervical cancer (CC) progression and its underlying mechanism. Transcription factor Dp-1 (TFDP1) and TK1 expression was assessed using qRT-PCR in CC cell lines. After ectopic expression and knockdown experiments, cell counting kit-8 and colony formation assays were adopted to measure cell proliferation, western blot to examine the expression of epithelial-mesenchymal transition (EMT)-related proteins, and Transwell assays to assess cell invasion and migration. The binding of TFDP1 to TK1 was predicted by bioinformatic sites and verified by chromatin immunoprecipitation and dual-luciferase reporter assays. Tumor xenograft experiments in nude mice were performed to validate the influence of TFDP1/TK1 on CC progression in vivo. CC cells had high TK1 and TFDP1 expression. TFDP1 or TK1 knockdown restrained CC cell EMT, invasion, migration, and proliferation. TFDP1 facilitated TK1 expression in CC via transcription. Overexpression of TK1 counteracted the suppressive impacts of TFDP1 knockdown on CC cell malignant behaviors. Moreover, TFDP1 knockdown depressed CC growth in vivo by downregulating TK1. TFDP1 knockdown restricted proliferation and EMT in CC by downregulating TK1 expression.
Collapse
Affiliation(s)
- Mei Wu
- Department of Gynecologic Oncology, Hunan Cancer Hospital, Changsha, Hunan, 410013, People's Republic of China
| | - Mingji Ye
- Department of Urology Surgery, Hunan Cancer Hospital, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
3
|
Song Y, Ma R. Comprehensive Analysis Reveals the Potential Roles of Transcription Factor Dp-1 in Lung Adenocarcinoma. World J Oncol 2023; 14:205-223. [PMID: 37350808 PMCID: PMC10284640 DOI: 10.14740/wjon1595] [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] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Background Transcription factor Dp-1 (TFDP1) was overexpressed and interacted with other genes to impact multiple signaling pathways in various human cancers. However, there is less research about the TFDP1 specific roles in lung adenocarcinoma (LUAD). Methods We first explored TFDP1 expression levels and relative diseases from a pan-cancer perspective using the ONCOMINE, TIMER, and Open Targets Platform databases. Then, we used UALCAN, GEPIA 2, TCGA-LUAD data, and Kaplan-Meier plotter to examine TFDP1 clinicopathological features and prognosis in LUAD patients. Genomic alterations and DNA methylation analysis were performed by cBioPortal and MethSurv, respectively. Then, we used a cancer single-cell state atlas (CancerSEA) to find TFDP1 functions at a single-cell resolution. LinkedOmics was used to find TFDP1 coexpressed genes, biological processes, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Then, Gene Set Cancer Analysis (GSCA) was used to examine the drug resistence of TFDP1 in LUAD. Results We found that TFDP1 was overexpressed in most human cancers and related to various diseases, including LUAD. Moreover, LUAD patients with high TFDP1 expression levels might be significantly associated with individual cancer stages and have a poor prognosis. Multivariate analysis revealed that the American Joint Committee on Cancer (AJCC) pathologic stage, AJCC stage T, and AJCC stage N were the independent prognostic factors. LUAD patients with TFDP1 alterations suggested poor overall survival (OS), and disease-free survival (DFS), while hypermethylation might lead to a good prognosis. TFDP1 and its coexpressed genes were enriched in multiple signaling pathways and biological processes involved in the cell cycle, spliceosome, and DNA replication. Furthermore, TFDP1 was strongly positively related to the half-maximal inhibitory concentration (IC50) values of multiple drugs. Conclusions In summary, TFDP1 was a possible biomarker and potential therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Yipeng Song
- School of Law and Criminal Justice, East China University of Political Science and Law, Songjiang University Town, Shanghai 201620, China
| | - Rongna Ma
- Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai 200051, China
| |
Collapse
|
4
|
Integrated Analysis of miRNA-mRNA Regulatory Networks Associated with Osteonecrosis of the Femoral Head. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8076598. [PMID: 34422080 PMCID: PMC8376457 DOI: 10.1155/2021/8076598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022]
Abstract
Osteonecrosis of the femoral head (ONFH) accounts for as many as 18% of total hip arthroplasties. Knowledge of genetic changes and molecular abnormalities could help identify individuals considered to be at a higher risk of developing ONFH. In this study, we sought to identify differentially expressed miRNAs (DEmiRs) and genes (DEGs) associated with ONFH by integrated bioinformatics analyses as well as to construct the miRNA-mRNA regulatory network involving in the pathogenesis of ONFH. We performed differential expression analysis using a gene expression profile GSE123568 and a miRNA expression profile GSE89587 deposited in the Gene Expression Omnibus and identified 47 DEmiRs (24 upregulated miRNAs and 23 downregulated miRNAs) and 529 DEGs (218 upregulated genes and 311 downregulated genes). Gene Ontology enrichment analyses of DEGs suggested that DEGs were significantly enriched in neutrophil activation, cytosol, and ubiquitin-protein transferase activity. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of DEGs revealed that DEGs were significantly enriched in transcriptional misregulation in cancer. DEGs-based miRNA-mRNA regulatory networks were obtained by searching miRNA-mRNA prediction databases, TargetScan, miTarBase, miRMap, miRDB, and miRanda databases. Then, overlapped miRNAs were selected between these putative miRNAs and DEmiRs between ONFH and non-ONFH, and pairs of the DEmiR-DEG regulatory network were finally depicted. There were 12 nodes and 64 interactions for upDEmiR-downDEG regulatory networks and 6 nodes and 16 interactions for downDEmiR-upDEG regulatory networks. Using the STRING database, we established a protein-protein interaction network based on the overlapped DEGs between ONFH and non-ONFH. C5AR1, CDC27, CDC34, KAT2B, CPPED1, TFDP1, and MX2 were identified as the hub genes. The present study characterizes the miRNA profile, gene profile, and miRNA-mRNA regulatory network in ONFH, which may contribute to the interpretation of the pathogenesis of ONFH and the identification of novel biomarkers and therapeutic targets for ONFH.
Collapse
|
5
|
Erbaba B, Arslan-Ergul A, Adams MM. Effects of caloric restriction on the antagonistic and integrative hallmarks of aging. Ageing Res Rev 2021; 66:101228. [PMID: 33246078 DOI: 10.1016/j.arr.2020.101228] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022]
Abstract
Aging is a significant risk factor for cognitive decline associated with neurodegenerative diseases, which makes understanding what promotes 'healthy brain aging' very important. Studies suggest that caloric restriction (CR) is a non-genetic intervention that reliably extends life- and healthspan. Here, we review the CR literature related to both the subject of aging and alterations in cell cycle machinery, especially surrounding the regulation of the E2F/DP1 complex, to elucidate the cellular protection mechanisms in the brain induced via dietary applications. The alterations extending lifespan via CR appear to exert their effects by promoting survival of individual cells, downregulating cell proliferation, and inducing stem cell quiescence, which results in keeping the stem cell reserve for extreme needs. This survival instinct of cells is believed to cause some molecular adaptations for their maintenance of the system. Avoiding energy waste of proliferation machinery promotes the long term survival of the individual cells and this is due to adaptations to the limited nutrient supply in the environment. Such a protective mechanism induced by diet could be promoted via the downregulation of crucial cell cycle-related transcription activators. This review article aims to bring attention to the importance of molecular adaptations induced by diet that promote healthy brain aging. It will provide insights into alternative targets for new treatments or neuroprotective approaches against neurodegenerative pathophysiologies.
Collapse
Affiliation(s)
- Begun Erbaba
- Interdisciplinary Graduate Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey
| | - Ayca Arslan-Ergul
- National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey
| | - Michelle M Adams
- Interdisciplinary Graduate Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey; Department of Psychology, Bilkent University, Ankara, Turkey.
| |
Collapse
|
6
|
Yang L, Cui Y, Huang T, Sun X, Wang Y. Identification and Validation of MSX1 as a Key Candidate for Progestin Resistance in Endometrial Cancer. Onco Targets Ther 2020; 13:11669-11688. [PMID: 33235459 PMCID: PMC7679365 DOI: 10.2147/ott.s271494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose Progestin resistance is a critical obstacle for endometrial conservative therapy. Therefore, studies to acquire a more comprehensive understanding of the mechanisms are urgent. However, the pivotal molecules are still unexplored. Materials and Methods We downloaded GSE121367 from the GEO database. The “limma” R language package was applied to identify differentially expressed genes (DEGs). We conducted Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA). Protein–protein interaction was constructed by STRING and visualized in Cytoscape. The tumor immune microenvironment was explored by the TISIDB database. Methylation validation and overall survival analysis were conducted by the TCGA database. In addition, the upstream modulators of hub genes were predicted by miRTarBase and Network Analyst databases. The expression levels of candidate genes were validated by quantitative real-time PCR (qRT-PCR), Western blot, and immunohistochemical assay (IHC). Cell growth, clone formation, migration, invasion, and wound healing assays were studied to explore the role of MSX1 in progestin resistance in vitro. Results A total of 3,282 DEGs were identified and they were mostly enriched in the cell adhesion pathway. We screened out ten hub genes whose genomic alteration rates were low based on the current endometrial carcinoma sample sets. Has-miR-335-5p, has-miR-124-3p, MAZ, and TFDP1 were the most prominent upstream regulators. The methylation status of CDH1, JAG1, EPCAM, and MSX1 was decreased, corresponding to their high protein expression, which also predicted better overall survival. The homeobox protein of MSX1 showed significant tissue specificity and better prognostic value and its knockdown inhibited epithelial–mesenchymal transitions (EMT) and enhanced progesterone efficacy. Conclusion Our study identified that the gene of MSX1 promised to be the specific indicator and therapeutic target for progestin resistance. This would shed new light on the underlying biological mechanism to overcome progestin resistance of endometrial cancer.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Municipal Key Clinical Specialty, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, People's Republic of China
| | - Yunxia Cui
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Municipal Key Clinical Specialty, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, People's Republic of China
| | - Ting Huang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Municipal Key Clinical Specialty, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, People's Republic of China
| | - Xiao Sun
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Municipal Key Clinical Specialty, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, People's Republic of China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Municipal Key Clinical Specialty, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, People's Republic of China
| |
Collapse
|
7
|
Kamergorodsky G, Invitti AL, D'Amora P, Parreira RM, Kopelman A, Bonetti TCS, Girão MJBC, Schor E. Progesterone's role in deep infiltrating endometriosis: Progesterone receptor and estrogen metabolism enzymes expression and physiological changes in primary endometrial stromal cell culture. Mol Cell Endocrinol 2020; 505:110743. [PMID: 32004675 DOI: 10.1016/j.mce.2020.110743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 11/22/2022]
Abstract
To study progesterone signaling activation, we measured changes in extracellular pH as a reflection of Na+/H+ exchange (NHE) using a cytosensor microphysiometer and assessed progesterone receptor (PR) and estrogen metabolism enzymes mRNA expression in cultured endometrial cells from women with deep infiltrating endometriosis and healthy controls using real-time quantitative PCR. This study was conducted at a University hospital and included patients with and without deep infiltrating endometriosis (DIE). Primary endometrial stromal cells (ECs) from women with DIE and controls were treated with 17β-estradiol and progesterone prior to microphysiometer measurements and qPCR evaluations. Decreased progesterone responsiveness and decreased total nuclear PR and HSD17B1 mRNA expression were observed in cultured ECs from women with deep infiltrating endometriosis relative to those from control samples before and after hormone treatment. These cells also showed increased 17β-hydroxysteroid dehydrogenases types 2 (HSD17B2) relative to control group and increased expression of aromatase (CYP19) after exposure to progesterone. These physiological and expression patterns observed in ECs cultures from women with DIE reinforces previous findings in the literature supporting the progesterone resistance hypothesis in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Gil Kamergorodsky
- Pelvic Pain and Endometriosis Unit - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil; Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Adriana L Invitti
- Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil.
| | - Paulo D'Amora
- Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Rafael M Parreira
- Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Alexander Kopelman
- Pelvic Pain and Endometriosis Unit - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil; General Gynecology Discipline - Gynecology Department, Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Tatiana C S Bonetti
- Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Manoel J B C Girão
- General Gynecology Discipline - Gynecology Department, Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Eduardo Schor
- Pelvic Pain and Endometriosis Unit - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil; Cellular and Molecular Gynecology Laboratory - Gynecology Department - Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| |
Collapse
|