1
|
Comprehensive Analysis of HOX Family Members as Novel Diagnostic and Prognostic Markers for Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5758601. [PMID: 35251173 PMCID: PMC8890896 DOI: 10.1155/2022/5758601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022]
Abstract
Background. The homeobox (HOX) gene family has been found to be involved in human cancers. However, its involvement in hepatocellular carcinoma (HCC) has not been well documented. Here, we comprehensively evaluated the role of HOXs in HCC. Methods. RNA sequencing profile of TCGA-LIHC and LIRI-JP were obtained from the Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), respectively. Data of TCGA-LIHC methylation were downloaded from UCSC Xena. Genetic alteration data for the TCGA samples was obtained from cBioPortal and GSCA. The diagnostic efficiency was assessed using ROC curves. The prognostic significance was evaluated by the Kaplan–Meier method and Cox regression analysis. Subsequent functional analysis was performed through the clusterProfiler package. ssGSEA, ESTIMATE, and TIDE algorithms were employed to explore the relationship between HOXs and the HCC microenvironment. Finally, pRRophetic package and NCI-60 cancerous cell lines were applied to estimate anticancer drug sensitivity. Results. The mRNA levels of HOXs in HCC tissues were higher than those of noncancerous tissues and were correlated with poor overall survival (OS). HOXA6, C6, D9, D10, and D13 could serve as independent risk factors for OS. Further functional analysis revealed that these five HOXs regulate the cell proliferation, cell cycle, immune response, and microenvironment composition of HCC. In addition, the aberrant expression and methylation of HOXs is of great value in the diagnosis of HCC. Conclusion. HOXs play crucial roles in HCC and could serve as potential markers for HCC diagnosis and prognosis.
Collapse
|
2
|
Wang L, Gao Y, Tong D, Wang X, Guo C, Guo B, Yang Y, Zhao L, Zhang J, Yang J, Qin Y, Liu L, Huang C. MeCP2 drives hepatocellular carcinoma progression via enforcing HOXD3 promoter methylation and expression through the HB-EGF/EGFR pathway. Mol Oncol 2021; 15:3147-3163. [PMID: 34028973 PMCID: PMC8564637 DOI: 10.1002/1878-0261.13019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/21/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
Homeobox D3 (HOXD3), a member of the homeobox family, was described to regulate tumorigenesis, invasion, metastasis, and angiogenesis in various tumor types. However, the molecular mechanisms regulating HOXD3 during hepatocellular carcinoma (HCC) migration, invasion, and angiogenesis remain elusive. In this study, we demonstrated that HOXD3 expression is enhanced by the binding of methyl-CpG-binding protein 2 (MeCP2), a methyl-CpG binding protein, together with CREB1to the hypermethylated promoter of HOXD3. Inhibition of HOXD3 eliminated the tumorigenic effects of MeCP2 on HCC cells. Furthermore, HOXD3 directly targeted the promoter region of heparin-binding epidermal growth factor (HB-EGF) via the EGFR-ERK1/2 cell signaling pathway and promoted invasion, metastasis, and angiogenesis of HCC in vitro and in vivo. Additionally, elevated expression of MeCP2, CREB1, and HB-EGF in HCC correlated with a poor survival rate. Our findings reveal the function of the MeCP2/HOXD3/HB-EGF regulatory axis in HCC, rendering it an attractive candidate for the development of targeted therapeutics and as a potential biomarker in patients with HCC.
Collapse
Affiliation(s)
- Lumin Wang
- Department of Digestive Diseases in Precision Medicine Institutethe Second Affiliated Hospital of Xi'an Jiaotong UniversityChina
| | - Yi Gao
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Yan'an Key Laboratory of Chronic Disease Prevention and ResearchChina
| | - Dongdong Tong
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Xiaofei Wang
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Chen Guo
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Bo Guo
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Yang Yang
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Lingyu Zhao
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Jing Zhang
- Yan'an Key Laboratory of Chronic Disease Prevention and ResearchChina
| | - Juan Yang
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Yannan Qin
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Liying Liu
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
| | - Chen Huang
- Department of cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterChina
- Institute of Genetics and Developmental BiologyTranslational Medicine InstituteSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterChina
- Cardiovascular Research CenterXi'an Jiaotong University Health Science CenterChina
| |
Collapse
|
3
|
Zhang G, Gao X, Zhao X, Wu H, Yan M, Li Y, Zeng H, Ji Z, Guo X. Decitabine inhibits the proliferation of human T-cell acute lymphoblastic leukemia molt4 cells and promotes apoptosis partly by regulating the PI3K/AKT/mTOR pathway. Oncol Lett 2021; 21:340. [PMID: 33747197 PMCID: PMC7967925 DOI: 10.3892/ol.2021.12601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/11/2020] [Indexed: 11/30/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is a highly aggressive hematological cancer; however, there is a lack of effective chemotherapeutic or targeted drugs for the treatment of T-ALL. Decitabine is a DNA demethylation agent but it has not been used for T-ALL treatment. Therefore, the present study aimed to assess the inhibitory effect of decitabine on T-ALL molt4 cells and determine its regulatory role in the PI3K/AKT/mTOR pathway. Molt4 cells were stimulated with decitabine in vitro, after which cell proliferation, apoptosis and cell cycle analyses were performed to assess cell viability. Subcellular morphology was observed using transmission electron microscopy. Expression levels of phosphate and tension homology (PTEN), genes involved in the PI3K/AKT/mTOR pathway and the corresponding downstream genes were analyzed using reverse transcription-quantitative PCR and western blotting. The results showed that decitabine induced apoptosis, inhibited proliferation and arrested molt4 cells in the G2 phase. Following decitabine intervention, an increase in the number of lipid droplets, autophagosomes and mitochondrial damage was observed. At concentrations of 1 and 10 µM, decitabine downregulated the expression of PI3K, AKT, mTOR, P70S6 and eukaryotic initiating factor 4E-binding protein 1, which in turn upregulated PTEN expression; however, 50 µM decitabine downregulated PTEN levels. Overall, these results demonstrated that decitabine reduced the viability of molt4 cells partly by inhibiting the PI3K/AKT/mTOR pathway via PTEN, especially at low decitabine concentrations.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaohui Gao
- Department of Pediatrics, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaoyan Zhao
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Haibing Wu
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Minchao Yan
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Yuan Li
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Hui Zeng
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Zhaoning Ji
- Department of Medical Oncology, The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Xiaojun Guo
- Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
4
|
Paço A, de Bessa Garcia SA, Freitas R. Methylation in HOX Clusters and Its Applications in Cancer Therapy. Cells 2020; 9:cells9071613. [PMID: 32635388 PMCID: PMC7408435 DOI: 10.3390/cells9071613] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023] Open
Abstract
HOX genes are commonly known for their role in embryonic development, defining the positional identity of most structures along the anterior–posterior axis. In postembryonic life, HOX gene aberrant expression can affect several processes involved in tumorigenesis such as proliferation, apoptosis, migration and invasion. Epigenetic modifications are implicated in gene expression deregulation, and it is accepted that methylation events affecting HOX gene expression play crucial roles in tumorigenesis. In fact, specific methylation profiles in the HOX gene sequence or in HOX-associated histones are recognized as potential biomarkers in several cancers, helping in the prediction of disease outcomes and adding information for decisions regarding the patient’s treatment. The methylation of some HOX genes can be associated with chemotherapy resistance, and its identification may suggest the use of other treatment options. The use of epigenetic drugs affecting generalized or specific DNA methylation profiles, an approach that now deserves much attention, seems likely to be a promising weapon in cancer therapy in the near future. In this review, we summarize these topics, focusing particularly on how the regulation of epigenetic processes may be used in cancer therapy.
Collapse
Affiliation(s)
- Ana Paço
- Centre Bio: Bioindustries, Biorefineries and Bioproducts, BLC3 Association—Technology and Innovation Campus, 3405-169 Oliveira do Hospital, Portugal;
| | | | - Renata Freitas
- I3S—Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal;
- ICBAS—Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Correspondence:
| |
Collapse
|
5
|
Sanaei M, Kavoosi F, Arabloo M. Effect of Curcumin in Comparison with Trichostatin A on the Reactivation of Estrogen Receptor Alpha gene Expression, Cell Growth Inhibition and Apoptosis Induction in Hepatocellular Carcinoma Hepa 1-6 Cell lLine. Asian Pac J Cancer Prev 2020; 21:1045-1050. [PMID: 32334468 PMCID: PMC7445996 DOI: 10.31557/apjcp.2020.21.4.1045] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: A multistep process with an accumulation of epigenetic alterations of tumor suppressor genes (TSGs) can induce cancer. Abnormal regional hypermethylation and histone deacetylation of several TSGs has been observed in hepatocellular carcinoma (HCC). Acetylation and deacetylation of histone are carried out by histone acetyltransferase (HAT) and histone deacetylase (HDAC) respectively. Besides, DNA methylation is carried out by DNA methyltransferases (DNMTs). Previously, we evaluated the effect of DNA demethylating agents and histone deacetylase inhibitors on HCC and colon cancer. This study aimed to evaluate the effect of curcumin (CUR) in comparison with trichostatin A (TSA) on estrogen receptor alpha (ERα) reactivation, apoptotic induction, and cell growth inhibition in HCC. Methods: the cells were cultured and treated with various concentrations of CUR and TSA and the MTT assay, flow cytometry assay and Real-Time RT-PCR were achieved to determine cell viability, cell apoptosis, and ERα gene expression respectively. Results: CUR indicated dose and time-dependent antiproliferative effects (P < 0.035). A similar antiproliferative effect was observed by TSA (P < 0.001). Both compounds indicated significant apoptotic effects in all different periods (P < 0.001), CUR indicated a more significant apoptotic effect than TSA (P < 0.001). The ERα gene expression quantity was increased significantly by treatment with CUR and TSA (P <0.012). Conclusion: CUR and TSA play important roles in restoring the ERα resulting in cell growth inhibition and apoptosis induction. Therefore, ERα may be a potential target for therapeutic intervention in the treatment of HCC.
Collapse
Affiliation(s)
- Masumeh Sanaei
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Fraidoon Kavoosi
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mehrnoosh Arabloo
- Student of Research Committee, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
6
|
Meucci S, Keilholz U, Heim D, Klauschen F, Cacciatore S. Somatic genome alterations in relation to age in lung squamous cell carcinoma. Oncotarget 2018; 9:32161-32172. [PMID: 30181806 PMCID: PMC6114948 DOI: 10.18632/oncotarget.25848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/12/2018] [Indexed: 12/31/2022] Open
Abstract
Lung squamous cell carcinoma (LUSC) is the most common cause of global cancer-related mortality and the major risk factors is smoking consumption. By analyzing ∼500 LUSC samples from The Cancer Genome Atlas, we detected a higher mutational burden as well as a higher level of methylation changes in younger patients. The SNPs mutational profiling showed enrichments of smoking-related signature 4 and defective DNA mismatch repair (MMR)-related signature 6 in younger patients, while the defective DNA MMR signature 26 was enriched among older patients. Furthermore, gene set enrichment analysis was performed in order to explore functional effect of somatic alterations in relation to patient age. Extracellular Matrix-Receptor Interaction, Nucleotide Excision Repair and Axon Guidance seem crucial disrupted pathways in younger patients. We hypothesize that a higher sensitivity to smoking-related damages and the enrichment of defective DNA MMR related mutations may contribute to the higher mutational burden of younger patients. The two distinct age-related defective DNA MMR signatures 6 and 26 might be crucial mutational patterns in LUSC tumorigenesis which may develop distinct phenotypes. Our study provides indications of age-dependent differences in mutational backgrounds (SNPs and CNVs) as well as epigenetic patterns that might be relevant for age adjusted treatment approaches.
Collapse
Affiliation(s)
- Stefano Meucci
- Charité Comprehensive Cancer Center, Charité University Hospital, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité University Hospital, Berlin, Germany
| | - Daniel Heim
- Institut für Pathologie, Charité University Hospital, Berlin, Germany
| | | | - Stefano Cacciatore
- Imperial College Parturition Research Group, Division of the Institute of Reproductive and Developmental Biology, Imperial College London, London, England, UK
- International Centre for Genetic Engineering and Biotechnology, Cancer Genomics Group, Cape Town, South Africa
| |
Collapse
|