1
|
Chin FW, Chan SC, Chau DM, Ong TA, Abdul Razack AH, Yusoff K, Veerakumarasivam A. HOXA13 promotes immune evasion in bladder cancer by suppressing antigen processing and presentation, and phagosome pathways. Funct Integr Genomics 2025; 25:44. [PMID: 39994130 DOI: 10.1007/s10142-025-01553-w] [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: 10/11/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Homebox A13 (HOXA13) and homeobox B13 (HOXB13) expression dysregulation have been previously reported in bladder cancer. However, their roles in bladder carcinogenesis remain unclear. This study characterizes the distinct transcriptomic profile and pathway enrichment of HOXA13 and HOXB13 knockdown in bladder cancer cells. Separate in vitro knockdown models for HOXA13 and HOXB13 were established using small interfering RNAs (siRNAs), and knockdown efficiency was validated through reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Transcriptomic profiling was conducted using RNA sequencing, followed by differential gene expression analysis, and Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analysis. HOXA13 knockdown significantly enriched pathways that are associated with immune evasion (i.e. antigen processing and presentation pathway, and phagosome pathway) through the upregulation of major histocompatibility complex (MHC) class I and II genes. These findings highlight the pivotal role of HOXA13 in promoting immune evasion in bladder cancer. Meanwhile, HOXB13 knockdown significantly enriched estrogen signaling pathway and PI3K-Akt signaling pathway, which are critical for cell proliferation and survival. While the role of HOXB13 in bladder cancer progression requires further delineation, the primary focus of this study is on HOXA13 due to its involvement in immune evasion mechanisms. This study provides novel insights into the potential therapeutic strategies for targeting HOXA13 in bladder cancer, and highlights the distinct roles of HOXA13 and HOXB13 in bladder carcinogenesis.
Collapse
Affiliation(s)
- Fee-Wai Chin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Soon-Choy Chan
- School of Medical and Life Sciences, Sunway University, 47500 Bandar Sunway, Selangor, Malaysia.
- Centre for Pre-University and General Studies, Perdana University, 50490 Kuala Lumpur, Malaysia.
- National Institutes of Biotechnology Malaysia, Malaysia Genome and Vaccine Institute, 43000, 43000 Kajang, Selangor, Malaysia.
| | - De-Ming Chau
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Teng-Aik Ong
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Khatijah Yusoff
- National Institutes of Biotechnology Malaysia, Malaysia Genome and Vaccine Institute, 43000, 43000 Kajang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Abhi Veerakumarasivam
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- School of Medical and Life Sciences, Sunway University, 47500 Bandar Sunway, Selangor, Malaysia.
- National Institutes of Biotechnology Malaysia, Malaysia Genome and Vaccine Institute, 43000, 43000 Kajang, Selangor, Malaysia.
| |
Collapse
|
2
|
Vij P, Hussain MS, Satapathy SK, Cobos E, Tripathi MK. The Emerging Role of Long Noncoding RNAs in Sorafenib Resistance Within Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:3904. [PMID: 39682093 PMCID: PMC11639815 DOI: 10.3390/cancers16233904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC), a liver cancer originating from hepatocytes, is a major health concern and among the most common malignancies worldwide. Sorafenib, approved by the U.S. F.D.A., is the primary first-line treatment for patients with advanced HCC. While the preferred first-line systemic regimen for HCC is immunotherapy with Atezolizumab plus bevacizumab or Tremelimumab-actl + durvalumab, Sorafenib is still an alternative recommended regimen. While some patients with advanced HCC may benefit from Sorafenib treatment, most eventually develop resistance, leading to poor prognosis. Long noncoding RNAs (lncRNAs) have been found to play a critical role in tumorigenesis and the development of HCC, as well as other cancers. They are also key players in tumor drug resistance, though the mechanisms of lncRNAs in Sorafenib resistance in HCC remain poorly understood. This review summarizes the molecular mechanisms contributing to Sorafenib resistance in HCC with their potential correlation with lncRNAs, including the roles of transporters, receptors, cell death regulation, and other influencing factors.
Collapse
Affiliation(s)
- Puneet Vij
- Department of Pharmaceutical Sciences, St. John’s University, Queens, NY 11439, USA;
| | - Mohammad Shabir Hussain
- Medicine and Oncology ISU, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (M.S.H.); (E.C.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Sanjaya K. Satapathy
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health Center for Liver Diseases & Transplantation, Northshore University Hospital, Manhasset, NY 11030, USA;
| | - Everardo Cobos
- Medicine and Oncology ISU, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (M.S.H.); (E.C.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Manish K. Tripathi
- Medicine and Oncology ISU, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (M.S.H.); (E.C.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
3
|
Kasai H, Yamashita A, Akaike Y, Tanaka T, Matsuura Y, Moriishi K. HCV infection activates the proteasome via PA28γ acetylation and heptamerization to facilitate the degradation of RNF2, a catalytic component of polycomb repressive complex 1. mBio 2024; 15:e0169124. [PMID: 39329491 PMCID: PMC11559043 DOI: 10.1128/mbio.01691-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
We previously reported that hepatitis C virus (HCV) infection or HCV core protein expression induces HOX gene expression by impairing histone H2A monoubiquitination via a proteasome-dependent reduction in the level of RNF2, a key catalytic component of polycomb repressive complex 1 (H. Kasai, K. Mochizuki, T. Tanaka, A. Yamashita, et al., J Virol 95:e01784-20, 2021, https://doi.org/10.1128/jvi.01784-20). In this study, we aimed to investigate the mechanism by which HCV infection accelerates RNF2 degradation. Yeast two-hybrid screening and an immunoprecipitation assay revealed that RNF2 is a PA28γ-binding protein. The proteasome activator PA28γ destabilized the RNF2 protein in a proteasome-dependent manner, since RNF2 degradation was impaired by PA28γ knockout or MG132 treatment. HCV infection or core protein expression reduced the levels of RNF2 and histone H2A K119 monoubiquitination and induced the expression of HOX genes in the presence of PA28γ, while PA28γ knockout reversed these changes. Treatment with a lysine acetyltransferase inhibitor inhibited the acetylation of PA28γ at K195 and the degradation of the RNF2 protein, while treatment with a lysine deacetylase inhibitor accelerated these events in a PA28γ-dependent manner. RNF2 protein degradation was increased by expression of the acetylation mimetic PA28γ mutant but not by expression of the acetylation-defective mutant or the proteasome activation-defective mutant. Furthermore, HCV infection or core protein expression facilitated the interaction between PA28γ and the lysine acetyltransferase CBP/p300 and then accelerated PA28γ acetylation and heptazmerization to promote RNF2 degradation. These data suggest that HCV infection accelerates the acetylation-dependent heptamerization of PA28γ to increase the proteasomal targeting of RNF2.IMPORTANCEHCV is a causative agent of HCV-related liver diseases, including hepatic steatosis, cirrhosis, and hepatocellular carcinoma. PA28γ, which, in heptameric form, activates the 20S core proteasome for the degradation of PA28γ-binding proteins, is responsible for HCV-related liver diseases. HCV core protein expression or HCV infection accelerates RNF2 degradation, leading to the induction of HOX gene expression via a decrease in the level of H2Aub on HOX gene promoters. However, the mechanism of RNF2 degradation in HCV-infected cells has not been clarified. The data presented in this study suggest that PA28γ acetylation and heptamerization are promoted by HCV infection or by core protein expression to activate the proteasome for the degradation of RNF2 and are responsible for HCV propagation. This study provides novel insights valuable for the development of therapies targeting both HCV propagation and HCV-related diseases.
Collapse
Affiliation(s)
- Hirotake Kasai
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Atsuya Yamashita
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Yasunori Akaike
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Tomohisa Tanaka
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
- Division of Hepatitis Virology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka, Japan
- Center for Infectious Diseases Education and Research (CiDER), Osaka University, Osaka, Japan
| | - Kohji Moriishi
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
- Division of Hepatitis Virology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
4
|
Chen ZX, Mu MY, Yang G, Qi H, Fu XB, Wang GS, Jiang WW, Huang BJ, Gao F. Hypoxia-induced DTL promotes the proliferation, metastasis, and sorafenib resistance of hepatocellular carcinoma through ubiquitin-mediated degradation of SLTM and subsequent Notch pathway activation. Cell Death Dis 2024; 15:734. [PMID: 39384740 PMCID: PMC11464529 DOI: 10.1038/s41419-024-07089-4] [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: 12/01/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024]
Abstract
Denticleless E3 ubiquitin protein ligase homolog (DTL), the substrate receptor of the CRL4A complex, plays a central role in genome stability. Even though the oncogenic function of DTL has been investigated in several cancers, its specific role in hepatocellular carcinoma (HCC) still needs further elucidation. Data from a clinical cohort (n = 209), RNA-sequencing, and public database (TCGA and GEO) were analyzed, indicating that DTL is closely related to patient prognosis and could serve as a promising prognostic indicator in HCC. Functionally, DTL promoted the proliferation, metastasis, and sorafenib resistance of HCC in vitro. In the orthotopic tumor transplantation and tail vein injection model, DTL promoted the growth and metastasis of HCC in vivo. Mechanically, we revealed for the first time that DTL was transcriptionally activated by hypoxia-inducible factor 1α (HIF-1α) under hypoxia and functioned as a downstream effector molecule of HIF-1α. DTL promotes the ubiquitination of SAFB-like transcription modulator (SLTM) and subsequently relieves the transcriptional repression of Notch1. These results suggested that DTL may be a potential biomarker and therapeutic target for HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Humans
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Cell Proliferation/drug effects
- Animals
- Drug Resistance, Neoplasm/drug effects
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- Mice
- Signal Transduction/drug effects
- Mice, Nude
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Cell Line, Tumor
- Ubiquitination
- Neoplasm Metastasis
- Ubiquitin/metabolism
- Receptors, Notch/metabolism
- Mice, Inbred BALB C
- Male
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Hypoxia
Collapse
Affiliation(s)
- Zi-Xiong Chen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Mao-Yuan Mu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Guang Yang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Han Qi
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiao-Bo Fu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Gui-Song Wang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wei-Wei Jiang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Fei Gao
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
5
|
Qin L, Zhong Y, Li Y, Yang Y. TCM targets ferroptosis: potential treatments for cancer. Front Pharmacol 2024; 15:1360030. [PMID: 38738174 PMCID: PMC11082647 DOI: 10.3389/fphar.2024.1360030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Ferroptosis is caused by the accumulation of cellular reactive oxygen species that exceed the antioxidant load that glutathione (GSH) and phospholipid hydroperoxidases with GSH-based substrates can carry When the antioxidant capacity of cells is reduced, lipid reactive oxygen species accumulate, which can cause oxidative death. Ferroptosis, an iron-dependent regulatory necrosis pathway, has emerged as a new modality of cell death that is strongly associated with cancer. Surgery, chemotherapy and radiotherapy are the main methods of cancer treatment. However, resistance to these mainstream anticancer drugs and strong toxic side effects have forced the development of alternative treatments with high efficiency and low toxicity. In recent years, an increasing number of studies have shown that traditional Chinese medicines (TCMs), especially herbs or herbal extracts, can inhibit tumor cell growth and metastasis by inducing ferroptosis, suggesting that they could be promising agents for cancer treatment. This article reviews the current research progress on the antitumor effects of TCMs through the induction of ferroptosis. The aim of these studies was to elucidate the potential mechanisms of targeting ferroptosis in cancer, and the findings could lead to new directions and reference values for developing better cancer treatment strategies.
Collapse
Affiliation(s)
- Liwen Qin
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yuhan Zhong
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yongfeng Yang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Mazziotta C, Iaquinta MR, Tramarin ML, Badiale G, Cervellera CF, Tonnini G, Patergnani S, Pinton P, Lanza G, Gafà R, Tognon M, Martini F, De Mattei M, Rotondo JC. Hsa-microRNA-1249-3p/Homeobox A13 axis modulates the expression of β-catenin gene in human epithelial cells. Sci Rep 2023; 13:22872. [PMID: 38129477 PMCID: PMC10739948 DOI: 10.1038/s41598-023-49837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Intercellular adhesion is a key function for epithelial cells. The fundamental mechanisms relying on epithelial cell adhesion have been partially uncovered. Hsa-microRNA-1249-3p (hsa-miR-1249-3p) plays a role in the epithelial mesenchymal transition in carcinoma cells, but its physiological function in epithelial cells is unknown. We aimed to investigate the role and molecular mechanisms of hsa-miR-1249-3p on epithelial cell functions. Hsa-miR-1249-3p was overexpressed in human epithelial cells and uterine cervical tissues, compared to cervical carcinoma cells and precancerous tissues, respectively. Hsa-miR-1249-3p was analyzed to verify its regulatory function on Homeobox A13 (HOXA13) target gene and its downstream cell adhesion gene β-catenin. Functional experiments indicated that hsa-miR-1249-3p inhibition prompted the mRNA and protein overexpression of HOXA13 which, in turn, led to the β-catenin protein expression. Moreover, hsa-miR-1249-3p inhibition induced a strong colony forming ability in epithelial cells, suggesting the miR involvement in cell adhesion machinery. These data indicate that hsa-miR-1249-3p regulates the expression of HOXA13 and its downstream cell adhesion gene β-catenin, possible resulting in cell adhesion modification in epithelial cells. This study will allow the set-up of further investigations aimed at exploring the relationship between the hsa-miR-1249-3p/HOXA13 axis and downstream cell adhesion genes.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Maria Letizia Tramarin
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giada Badiale
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Christian Felice Cervellera
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giulia Tonnini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Monica De Mattei
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy.
| |
Collapse
|
7
|
Wang F, Sun Z, Zhang Q, Yang H, Yang G, Yang Q, Zhu Y, Wu W, Xu W, Wu X. Curdione induces ferroptosis mediated by m6A methylation via METTL14 and YTHDF2 in colorectal cancer. Chin Med 2023; 18:122. [PMID: 37735401 PMCID: PMC10512537 DOI: 10.1186/s13020-023-00820-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Curdione is a sesquiterpene isolated from Curcumae Rhizoma that possesses high biological activity and extensive pharmacological effects. As a traditional Chinese medicine, Curcumae Rhizoma can inhibit the development of many types of cancer, especially colorectal cancer. However, the anti-colorectal mechanism of its monomer curdione remains unclear. METHODS Colorectal cancer (CRC) cells were treated with curdione at doses of 12.5 μM, 25 μM, and 50 μM, and then the cells' activity was measured with methyl thiazolyl tetrazolium (MTT). Nude mice were administered different doses of curdione subcutaneously and oxaliplatin by tail vein injection, and then hematoxylin-eosin (HE) staining was adopted to examine tumor histology. Moreover, flow cytometry was applied to detect reactive oxygen species in cells and tissues. Kits were employed to detect the levels of iron ions, malondialdehyde, lipid hydroperoxide, and glutathione. Polymerase chain reaction (PCR) and Western blotting were adopted to detect ferroptosis and m6A modification-related factors. A methylation spot hybridization assay was performed to measure changes in overall methylation. SLC7A11 and HOXA13 were measured by MeRIP-qPCR. The shRNA-METTL14 plasmid was constructed to verify the inhibitory effect of curdione on CRC. RESULTS A dose-dependent decrease in activity was observed in curdione-treated cells. Curdione increased the accumulation of reactive oxygen species in CRC cells and tumor tissues, greatly enhanced the levels of malondialdehyde, lipid hydroperoxide and Fe2+, and lowered the activity of glutathione. According to the qPCR and Western blot results, curdione promoted the expression of METTL14 and YTHDF2 in CRC cells and tissues, respectively, and decreased the expression of SLC7A11, SLC3A2, HOXA13, and glutathione peroxidase 4. Additionally, in animal experiments, the curdione-treated group showed severe necrosis of tumor cells, as displayed by HE staining. Furthermore, compared with the control group, levels of m6A modifying factors (namely, SLC7A11 and HOXA13) were increased in the tissues after drug intervention. METTL14 knockdown was followed by an increase in CRC cell activity and glutathione levels. However, the levels of reactive oxygen species, malondialdehyde, and iron ions decreased. The expression levels of SLC7A11, SLC3A2, HOXA13, and GPX4 were all increased after METTL14 knockdown. CONCLUSION The results suggest that curdione induces ferroptosis in CRC by virtue of m6A methylation.
Collapse
Affiliation(s)
- Fang Wang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Zheng Sun
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Qunyao Zhang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Hao Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Gang Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Qi Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Yimiao Zhu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Wenya Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Wenwen Xu
- Department of Gynecology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Xiaoyu Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
8
|
Liu J, Feng H, Wang D, Wang Y, Luo J, Xu S, Zhao F, Qin G. HOXA13 promotes the proliferation, migration, and invasion of nasopharyngeal carcinoma HNE1 cells by upregulating the expression of Snail and MMP-2. Sci Rep 2023; 13:12978. [PMID: 37563232 PMCID: PMC10415404 DOI: 10.1038/s41598-023-40041-8] [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: 04/17/2022] [Accepted: 08/03/2023] [Indexed: 08/12/2023] Open
Abstract
Homeobox A13 (HOXA13) has been verified as an oncogen in some malignancies. However, its role in nasopharyngeal carcinoma (NPC) is still unclear. This study aims to explore the role of HOXA13 in NPC and its underlying mechanism. The mRNA expression of HOXA13 in NPC was obtained from the GSE53819 and GSE64634 datasets in the Gene Expression Omnibus (GEO) database. MTT, colony formation and transwell assays and xenograft tumour models were used to investigate the effects of HOXA13 on NPC HNE1 cells in vitro and in vivo. The expression of HOXA13, epithelial-mesenchymal transition-transcription factor (EMT-TF) Snail and matrix metalloproteinase 2 (MMP-2) was detected by immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The results showed that HOXA13 was upregulated in NPC. Silencing HOXA13 suppressed the proliferation, migration, and invasion of HNE1 cells, which inhibited tumour growth, while overexpression of HOXA13 induced the opposite effects. In addition, the expression of Snail and MMP-2 at the transcriptional and protein levels was associated with the expression of HOXA13. In summary, our results suggest that HOXA13 plays a role as a cancer-promoting gene in NPC. The underlying mechanism may be related to the upregulation of Snail and MMP-2.
Collapse
Affiliation(s)
- Jinping Liu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Huajun Feng
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Dingting Wang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Yuanyuan Wang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Jian Luo
- Department of Otolaryngology Head and Neck Surgery, The First People's Hospital of Yibin, Yibin, 644000, China
| | - Shengen Xu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Feipeng Zhao
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, NO: 25, Taiping Street, Jiangyang District, Luzhou, 646000, China.
| |
Collapse
|
9
|
Chin FW, Hussin H, Chau DM, Ong TA, Yunus R, Abdul Razack AH, Yusoff K, Chan SC, Veerakumarasivam A. Differential Protein Expression Patterns of HOXA13 and HOXB13 Are Associated with Bladder Cancer Progression. Diagnostics (Basel) 2023; 13:2636. [PMID: 37627895 PMCID: PMC10453033 DOI: 10.3390/diagnostics13162636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 08/27/2023] Open
Abstract
Bladder cancer is a common urological cancer and has the highest recurrence rate of any cancer. The aim of our study was to profile and characterize the protein expression of homeobox A13 (HOXA13) and homeobox B13 (HOXB13) genes in Malaysian bladder cancer patients. The protein expression of HOXA13 and HOXB13 in formalin-fixed paraffin-embedded (FFPE) bladder cancer tissues was determined by immunohistochemistry (IHC) analysis. The association between HOXA13/HOXB13 protein expression and demographic/clinicopathological characteristics of the bladder cancer patients was determined by chi-square analysis. Approximately 63.6% of the bladder cancer tissues harbored high HOXA13 expression. High HOXA13 expression was significantly associated with non-muscle invasive bladder cancer, lower tumor grade, higher number of lymph node metastases, and recurrence risk. In contrast, low HOXB13 expression (including those with negative expression) was observed in 71.6% of the bladder cancer tissues analyzed. Low HOXB13 expression was significantly associated with muscle-invasive bladder cancer, higher tumor stage, tumor grade, and metastatic risk. Both HOXA13 and HOXB13 protein expression were found to be associated with bladder tumorigenesis. The putative oncogenic and tumor suppressive roles of HOXA13 and HOXB13, respectively, suggest their potential utility as biomarkers in bladder cancer.
Collapse
Affiliation(s)
- Fee-Wai Chin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Huzlinda Hussin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - De-Ming Chau
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Teng-Aik Ong
- Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Rosna Yunus
- Department of Pathology, Hospital Kuala Lumpur, Kuala Lumpur 50586, Malaysia
| | | | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Kajang 43000, Selangor, Malaysia
| | - Soon-Choy Chan
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Kajang 43000, Selangor, Malaysia
- School of Liberal Arts, Science and Technology, Perdana University, Kuala Lumpur 50490, Malaysia
| | - Abhi Veerakumarasivam
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Kajang 43000, Selangor, Malaysia
- School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Selangor, Malaysia
| |
Collapse
|
10
|
Feng H, Zhao F, Luo J, Xu S, Liang Z, Xu W, Bao Y, Qin G. Long non-coding RNA HOTTIP exerts an oncogenic function by regulating HOXA13 in nasopharyngeal carcinoma. Mol Biol Rep 2023; 50:6807-6818. [PMID: 37392284 PMCID: PMC10374758 DOI: 10.1007/s11033-023-08598-9] [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: 05/06/2023] [Accepted: 06/15/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND The long non-coding RNA HOXA transcript at the distal tip (HOTTIP) and homeobox A13 (HOXA13) have been identified as oncogenes that play a pivotal role in tumorigenesis. However, their specific mechanisms of action in nasopharyngeal carcinoma (NPC) progression remain unclear. METHODS AND RESULTS In the present study, RT-qPCR was employed to quantify RNA expression in NPC cells and tissues. Flow cytometry, MTT, CCK8 and colony formation assays were utilized to assess cell apoptosis and proliferation. Transwell assay was conducted to evaluate migration and invasion while Western blotting was performed for protein expression analysis. Our findings revealed that the expression of HOTTIP was significantly upregulated in NPC cell lines. Inhibition of HOTTIP could induce apoptosis and suppress proliferation, clonogenicity, invasion and metastasis in NPC cells. Knockdown of HOTTIP led to downregulation of HOXA13 expression, which subsequently inhibited the proliferation and metastasis in NPC cells. The inhibitory effects on cell proliferation and metastasis caused by HOTTIP silencing were rescued by HOXA13 overexpression. Additionally, there was a significant positive correlation between HOTTIP and HOXA13, which were found to be elevated in NPC tissues compared to normal tissues. CONCLUSIONS We have determined that LncRNA HOTTIP facilitates tumorigenesis by modulating the expression of HOXA13 in NPC cells. Targeting HOTTIP/HOXA13 may be a promising therapeutic strategy for NPC.
Collapse
Affiliation(s)
- Huajun Feng
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Feipeng Zhao
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Jian Luo
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Shengen Xu
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Zhuoping Liang
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Wei Xu
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Yilin Bao
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
11
|
Ruiz C, Alborelli I, Manzo M, Calgua B, Keller E, Vuaroqueaux V, Quagliata L, Rentsch CA, Spagnoli GC, Diener PA, Bubendorf L, Morant R, on behalf of the former members of the Urology Team in St. Gallen **, Eppenberger-Castori S. Critical Evaluation of Transcripts and Long Noncoding RNA Expression Levels in Prostate Cancer Following Radical Prostatectomy. Pathobiology 2023; 90:400-408. [PMID: 37463569 PMCID: PMC10733933 DOI: 10.1159/000531175] [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: 11/23/2022] [Accepted: 05/18/2023] [Indexed: 07/20/2023] Open
Abstract
INTRODUCTION The clinical course of prostate cancer (PCa) is highly variable, ranging from indolent behavior to rapid metastatic progression. The Gleason score is widely accepted as the primary histologic assessment tool with significant prognostic value. However, additional biomarkers are required to better stratify patients, particularly those at intermediate risk. METHODS In this study, we analyzed the expression of 86 cancer hallmark genes in 171 patients with PCa who underwent radical prostatectomy and focused on the outcome of the 137 patients with postoperative R0-PSA0 status. RESULTS Low expression of the IGF1 and SRD52A, and high expression of TIMP2, PLAUR, S100A2, and CANX genes were associated with biochemical recurrence (BR), defined as an increase of prostate-specific antigen above 0.2 ng/mL. Furthermore, the analysis of the expression of 462 noncoding RNAs (ncRNA) in a sub-cohort of 39 patients with Gleason score 7 tumors revealed that high levels of expression of the ncRNAs LINC00624, LINC00593, LINC00482, and cd27-AS1 were significantly associated with BR. Our findings provide further evidence for tumor-promoting roles of ncRNAs in PCa patients at intermediate risk. The strong correlation between expression of LINC00624 and KRT8 gene, encoding a well-known cell surface protein present in PCa, further supports a potential contribution of this ncRNA to PCa progression. CONCLUSION While larger and further studies are needed to define the role of these genes/ncRNA in PCa, our findings pave the way toward the identification of a subgroup of patients at intermediate risk who may benefit from adjuvant treatments and new therapeutic agents.
Collapse
Affiliation(s)
- Christian Ruiz
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ilaria Alborelli
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Massimiliano Manzo
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Byron Calgua
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Eveline Keller
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Luca Quagliata
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- Medical Affairs Team, Genetic Sciences Group, Thermo Fisher Scientific, Monza, Italy
| | - Cyrill A. Rentsch
- Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Giulio C. Spagnoli
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | | | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Rudolf Morant
- Tumor-und Brustzentrum ZeTuP, St. Gallen, Switzerland
| | - on behalf of the former members of the Urology Team in St. Gallen**
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- 4HF Biotec, Freiburg, Germany
- Medical Affairs Team, Genetic Sciences Group, Thermo Fisher Scientific, Monza, Italy
- Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
- Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Tumor-und Brustzentrum ZeTuP, St. Gallen, Switzerland
| | | |
Collapse
|
12
|
Zheng ZQ, Yuan GQ, Zhang GG, Nie QQ, Wang Z. Development and validation of a predictive model in diagnosis and prognosis of primary glioblastoma patients based on Homeobox A family. Discov Oncol 2023; 14:108. [PMID: 37351805 DOI: 10.1007/s12672-023-00726-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Homeobox A (HOXA) family is involved in the development of malignancies as either tumor suppressors or oncogenes. However, their roles in glioblastoma (GBM) and clinical significance have not been fully elucidated. METHODS HOXA mutation and expressions in pan-cancers were investigated using GSCA and Oncomine, which in GBM were validated by cBioPortal, Chinese Glioma Genome Atlas (CGGA), and The Cancer Genome Atlas (TCGA) datasets. Kaplan-Meier analyses were conducted to determine prognostic values of HOXAs at genetic and mRNA levels. Diagnostic roles of HOXAs in tumor classification were explored by GlioVis and R software. Independent prognostic HOXAs were identified using Cox survival analyses, the least absolute shrinkage and selection operator (LASSO) regression, quantitative real-time PCR, and immunohistochemical staining. A HOXAs-based nomogram survival prediction model was developed and evaluated using Kaplan-Meier analysis, time-dependent Area Under Curve, calibration plots, and Decision Curve Analysis in training and validation cohorts. RESULTS HOXAs were highly mutated and overexpressed in pan-cancers, especially in CGGA and TCGA GBM datasets. Genetic alteration and mRNA expression of HOXAs were both found to be prognostic. Specific HOXAs could distinguish IDH mutation (HOXA1-7, HOXA9, HOXA13) and molecular GBM subtypes (HOXA1-2, HOXA9-11, HOXA13). HOXA1/2/3/10 were confirmed to be independent prognostic members, with high expressions validated in clinical GBM tissues. The HOXAs-based nomogram model exhibited good prediction performance and net benefits for patients in training and validation cohorts. CONCLUSION HOXA family has diagnostic values, and the HOXAs-based nomogram model is effective in survival prediction, providing a novel approach to support the treatment of GBM patients.
Collapse
Affiliation(s)
- Zong-Qing Zheng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Gui-Qiang Yuan
- Beijing Neurosurgical Institute & Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Capital Medical University, Beijing, China
| | - Guo-Guo Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Qian-Qian Nie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
13
|
Farzaneh M, Masoodi T, Ghaedrahmati F, Radoszkiewicz K, Anbiyaiee A, Sheykhi-Sabzehpoush M, Rad NK, Uddin S, Jooybari SPM, Khoshnam SE, Azizidoost S. An updated review of contribution of long noncoding RNA-NEAT1 to the progression of human cancers. Pathol Res Pract 2023; 245:154380. [PMID: 37043964 DOI: 10.1016/j.prp.2023.154380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Abstract
Long non-coding RNAs (lncRNAs) present pivotal roles in cancer tumorigenesis and progression. Recently, nuclear paraspeckle assembly transcript 1 (NEAT1) as a lncRNA has been shown to mediate cell proliferation, migration, and EMT in tumor cells. NEAT1 by targeting several miRNAs/mRNA axes could regulate cancer cell behavior. Therefore, NEAT1 may function as a potent biomarker for the prediction and treatment of some human cancers. In this review, we summarized various NEAT1-related signaling pathways that are critical in cancer initiation and progression.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Poland
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Niloofar Khoshdel Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Seyedeh Pardis Motiee Jooybari
- Department of Biology, Faculty of Basic Sciences and Engineering, University of Gonbad Kavous, Gonbad Kavus, Golestan, Iran
| | - Seyed Esmaeil Khoshnam
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
14
|
Feng Q, Wang D, Guo P, Zhang Z, Feng J. Apatinib Functioned as Tumor Suppressor of Synovial Sarcoma through Regulating miR-34a-5p/HOXA13 Axis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7214904. [PMID: 36276991 PMCID: PMC9581677 DOI: 10.1155/2022/7214904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022]
Abstract
Objective Synovial sarcoma is a rare malignant tumor. The role of apatinib in synovial sarcoma remains unclear. In this study, we aimed to determine the biological functions and the potential molecular mechanism of action of apatinib in synovial sarcoma. Methods SW982 cells were stimulated with apatinib. The relative expression of the genes was determined by performing qPCR. Protein levels were evaluated by western blot and immunohistochemistry assays. Proliferation, apoptosis, migration, and invasion of SW982 cells were determined by the CCK-8 assay, clone formation assay, flow cytometry, wound healing, and the transwell assay, respectively. Additionally, SW982 cells were injected into mice to induce synovial sarcoma. Results Apatinib decreased the proliferation, migration, and invasion but increased the apoptosis of SW982 cells. Apatinib repressed tumor growth in vivo and elevated miR-34a-5p in SW982 cells. The inhibition of miR-34a-5p repressed the reduction of proliferation, migration, and invasion and also the elevation of apoptosis in apatinib-treated SW982 cells. The luciferase activity decreased after cotransfection of the miR-34a-5p mimic and the wild-type HOXA13 vector. Additionally, an increase in miR-34a-5p repressed the levels of HOXA13 mRNA and protein. Moreover, HOXA13 reversed these patterns caused by the inhibition of miR-34a-5p in apatinib-treated SW982 cells. Conclusion Apatinib elevated miR-34a-5p and reduced HOXA13, leading to a significant decrease in proliferation, migration, and invasion, along with an enhancement of apoptosis in SW982 cells. Apatinib suppressed tumorigenesis and tumor growth in SW982 cells in vivo.
Collapse
Affiliation(s)
- Qi Feng
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei Province, China
| | - Donglai Wang
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei Province, China
| | - Peng Guo
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei Province, China
| | - Zibo Zhang
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei Province, China
| | - Jiangang Feng
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei Province, China
| |
Collapse
|
15
|
Shenoy US, Adiga D, Kabekkodu SP, Hunter KD, Radhakrishnan R. Molecular implications of HOX genes targeting multiple signaling pathways in cancer. Cell Biol Toxicol 2022; 38:1-30. [PMID: 34617205 PMCID: PMC8789642 DOI: 10.1007/s10565-021-09657-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022]
Abstract
Homeobox (HOX) genes encode highly conserved homeotic transcription factors that play a crucial role in organogenesis and tissue homeostasis. Their deregulation impacts the function of several regulatory molecules contributing to tumor initiation and progression. A functional bridge exists between altered gene expression of individual HOX genes and tumorigenesis. This review focuses on how deregulation in the HOX-associated signaling pathways contributes to the metastatic progression in cancer. We discuss their functional significance, clinical implications and ascertain their role as a diagnostic and prognostic biomarker in the various cancer types. Besides, the mechanism of understanding the theoretical underpinning that affects HOX-mediated therapy resistance in cancers has been outlined. The knowledge gained shall pave the way for newer insights into the treatment of cancer.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Keith D Hunter
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, UK
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
16
|
Zhang K, Liu H, Yu M, Zhao H, Yang N, Bi X, Sun L, Lin R, Lü G. Upregulated LINC01667 Expression Is Correlated With Poor Prognosis in Hepatocellular Carcinoma. Front Oncol 2021; 11:650173. [PMID: 34458133 PMCID: PMC8397520 DOI: 10.3389/fonc.2021.650173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022] Open
Abstract
The development of hepatocellular carcinoma (HCC) is a complex pathological process. Long intergenic non–protein-coding RNA 1667 (LINC01667, also known as MGC38584) plays an oncogenic role in several human cancers; however, its functional role in HCC tumorigenesis remains unknown. Here, we first evaluated the gene expression levels of LINC01667 in HCC using data from The Cancer Genome Atlas and Gene Expression Profiling Interactive Analysis (GEPIA) databases. We then elucidated the association between LINC01667 gene expression levels and the survival rates of patients with HCC. We detected the effect of LINC01667 on the malignant phenotypes (cell proliferation, migration, invasion and apoptosis etc.) and the MAPK and PI3K/AKT/mTOR signaling pathways of HepG2, SMMC-7721 and HUH7 cells. We also analyzed the sensitivity of HepG2, SMMC-7721 and HUH7 with different expression levels of LINC01667 to anti-HCC drugs in vitro. Based on data from the aforementioned databases and our experiments in vitro, we found that LINC01667 was overexpressed in HCC, and that patients with high LINC01667 levels had a remarkably poor overall survival rate. In addition, inhibition of LINC01667 expression suppressed the proliferation, migration and invasion of HepG2 and SMMC-7721 cells and promoted their apoptosis in vitro. In contrast, overexpression of LINC01667 promoted the proliferation, migration and invasion of HUH7 cells and suppressed their apoptosis in vitro. ChIRP-seq (chromatin isolation by RNA purification) showed that LINC01667 bound to MEG3, and downregulated the expression of MEG3. In addition, western blotting showed that LINC01667 could activate the NF-κB pathway to promote cancer progression. In conclusion, we report that LINC01667 is an important oncogene in HCC and may be used as a potential diagnostic and prognostic biomarker of HCC.
Collapse
Affiliation(s)
- Kainan Zhang
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Graduate Academy, Xinjiang Medical University, Urumqi, China
| | - Hui Liu
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Mengsi Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hui Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ning Yang
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaojuan Bi
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Sun
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Renyong Lin
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Guodong Lü
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,College of Pharmacy, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
17
|
Zeng Z, Lu Q, Liu Y, Zhao J, Zhang Q, Hu L, Shi Z, Tu Y, Xiao Z, Xu Q, Huang D. Effect of the Hypoxia Inducible Factor on Sorafenib Resistance of Hepatocellular Carcinoma. Front Oncol 2021; 11:641522. [PMID: 34307125 PMCID: PMC8292964 DOI: 10.3389/fonc.2021.641522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
Sorafenib a multi-target tyrosine kinase inhibitor, is the first-line drug for treating advanced hepatocellular carcinoma (HCC). Mechanistically, it suppresses tumor angiogenesis, cell proliferation and promotes apoptosis. Although sorafenib effectively prolongs median survival rates of patients with advanced HCC, its efficacy is limited by drug resistance in some patients. In HCC, this resistance is attributed to multiple complex mechanisms. Previous clinical data has shown that HIFs expression is a predictor of poor prognosis, with further evidence demonstrating that a combination of sorafenib and HIFs-targeted therapy or HIFs inhibitors can overcome HCC sorafenib resistance. Here, we describe the molecular mechanism underlying sorafenib resistance in HCC patients, and highlight the impact of hypoxia microenvironment on sorafenib resistance.
Collapse
Affiliation(s)
- Zhi Zeng
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Qiliang Lu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Yang Liu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Junjun Zhao
- Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China.,Graduate Department, Bengbu Medical College, Bengbu, China
| | - Qian Zhang
- The Medical College of Qingdao University, Qingdao, China
| | - Linjun Hu
- The Medical College of Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Zhan Shi
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yifeng Tu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zunqiang Xiao
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| | - Dongsheng Huang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
18
|
Lin X, Xiang X, Feng B, Zhou H, Wang T, Chu X, Wang R. Targeting Long Non-Coding RNAs in Hepatocellular Carcinoma: Progress and Prospects. Front Oncol 2021; 11:670838. [PMID: 34249710 PMCID: PMC8267409 DOI: 10.3389/fonc.2021.670838] [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: 02/22/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma is the fifth-ranked cancer worldwide with a relatively low five-year survival rate. Long non-coding RNAs are a group of RNAs with remarkable aberrant expression which could act on multiple bioprocesses and ultimately impact upon tumor proliferation, invasion, migration, metastasis, apoptosis, and therapy resistance in cancer cells including hepatocellular carcinoma cells. In recent years, long non-coding RNAs have been reported to be indispensable targets in clinical target therapy to stop the growth of cancer and prolong the lifespan of patients with hepatocellular carcinoma. In this review, we enumerate the signaling pathways and life activities affected by long non-coding RNAs in hepatocellular carcinoma cells to illustrate the role of long non-coding RNAs in the development and therapy resistance of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xinrong Lin
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaosong Xiang
- Affiliated Jingling Hospital Research Institution of General Surgery, School of Medicine, Nanjing University, Nanjing, China
| | - Bing Feng
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hao Zhou
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ting Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rui Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
19
|
Janmaat VT, Nesteruk K, Spaander MCW, Verhaar AP, Yu B, Silva RA, Phillips WA, Magierowski M, van de Winkel A, Stadler HS, Sandoval-Guzmán T, van der Laan LJW, Kuipers EJ, Smits R, Bruno MJ, Fuhler GM, Clemons NJ, Peppelenbosch MP. HOXA13 in etiology and oncogenic potential of Barrett's esophagus. Nat Commun 2021; 12:3354. [PMID: 34099670 PMCID: PMC8184780 DOI: 10.1038/s41467-021-23641-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Barrett's esophagus in gastrointestinal reflux patients constitutes a columnar epithelium with distal characteristics, prone to progress to esophageal adenocarcinoma. HOX genes are known mediators of position-dependent morphology. Here we show HOX collinearity in the adult gut while Barrett's esophagus shows high HOXA13 expression in stem cells and their progeny. HOXA13 overexpression appears sufficient to explain both the phenotype (through downregulation of the epidermal differentiation complex) and the oncogenic potential of Barrett's esophagus. Intriguingly, employing a mouse model that contains a reporter coupled to the HOXA13 promotor we identify single HOXA13-positive cells distally from the physiological esophagus, which is mirrored in human physiology, but increased in Barrett's esophagus. Additionally, we observe that HOXA13 expression confers a competitive advantage to cells. We thus propose that Barrett's esophagus and associated esophageal adenocarcinoma is the consequence of expansion of this gastro-esophageal HOXA13-expressing compartment following epithelial injury.
Collapse
Affiliation(s)
- Vincent T Janmaat
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kateryna Nesteruk
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Auke P Verhaar
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bingting Yu
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rodrigo A Silva
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wayne A Phillips
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Surgery (St. Vincent's Hospital), The University of Melbourne, Melbourne, VIC, Australia
| | - Marcin Magierowski
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Anouk van de Winkel
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - H Scott Stadler
- Department of Skeletal Biology, Shriners Hospital for Children, Portland, OR, USA
| | | | - Luc J W van der Laan
- Department of Surgery, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ernst J Kuipers
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicholas J Clemons
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Chen Z, Qin Z, Li L, Wo Q, Chen X. HOXA13, Negatively Regulated by miR-139-5p, Decreases the Sensitivity of Gastric Cancer to 5-Fluorouracil Possibly by Targeting ABCC4. Front Oncol 2021; 11:645979. [PMID: 34094932 PMCID: PMC8175971 DOI: 10.3389/fonc.2021.645979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/19/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose Chemoresistance remains a major challenge in the therapy of gastric cancer (GC). The homeobox (HOX) gene family has gained attention in carcinogenesis and chemoresistance. Here, this study aimed to explore the mechanism of HOXA13 in GC chemoresistance. Methods Quantitative real-time PCR (qRT-PCR) and Western blot were used to evaluate the expression of HOXA13 in GC tissues. The Kaplan–Meier plotter database was mined for prognosis analysis of GC patients with different HOXA13 expression receiving 5-Fluorouracil (5-FU) therapy. The effects of HOXA13 on sensitivity of GC cells to 5-FU were investigated by Cell Counting Kit-8 (CCK-8), 5-Ethynyl-2’-deoxyuridine (EdU) incorporation, flow cytometry and experiment in vivo. RNA-Sequencing analysis was performed to explore the underlying mechanism of HOXA13-mediated 5-FU resistance in GC. Chromatin immunoprecipitation (ChIP) and rescue experiments were applied to determine the relationship between HOXA13 and ABCC4. Luciferase reporter assay was performed to assess interaction of miR-139-5p and HOXA13. Results HOXA13 was upregulated in GC and its high expression was associated with poor prognosis of GC patients with 5-FU treatment. Overexpression of HOXA13 impaired the inhibitory effects of 5-FU on GC cells proliferation in vitro and vivo, and knockdown of HOXA13 exacerbated 5-FU-induced GC cells apoptosis. Mechanistically, HOXA13, directly targeted by miR-139-5p in GC, might upregulate ABCC4 expression, thereby accentuating 5-FU resistance of GC cells. Conclusion Our study suggests that HOXA13 attenuates 5-FU sensitivity of GC possibly by upregulating ABCC4. Thus, targeting HOXA13 would provide a novel prospective into the potential therapeutic strategy for reversing chemoresistance.
Collapse
Affiliation(s)
- Zhengqian Chen
- Department of Breast Surgical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, China.,Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwei Qin
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Chen
- Department of Breast Surgical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
21
|
Ma L, Zhang X, Yu K, Xu X, Chen T, Shi Y, Wang Y, Qiu S, Guo S, Cui J, Miao Y, Tian X, Du L, Yu Y, Xia J, Wang J. Targeting SLC3A2 subunit of system X C- is essential for m 6A reader YTHDC2 to be an endogenous ferroptosis inducer in lung adenocarcinoma. Free Radic Biol Med 2021; 168:25-43. [PMID: 33785413 DOI: 10.1016/j.freeradbiomed.2021.03.023] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 12/16/2022]
Abstract
The m6A reader YT521-B homology containing 2 (YTHDC2) has been identified to inhibit lung adenocarcinoma (LUAD) tumorigenesis by suppressing solute carrier 7A11 (SLC7A11)-dependent antioxidant function. SLC7A11 is a major functional subunit of system XC-. Inhibition of system XC- can induce ferroptosis. However, whether suppressing SLC7A11 is sufficient for YTHDC2 to be an endogenous ferroptosis inducer in LUAD is unknown. Here, we found that induction of YTHDC2 to a high level can induce ferroptosis in LUAD cells but not in lung and bronchus epithelial cells. In addition to SLC7A11, solute carrier 3A2 (SLC3A2), another subunit of system XC- was equally important for YTHDC2-induced ferroptosis. YTHDC2 m6A-dependently destabilized Homeo box A13 (HOXA13) mRNA because a potential m6A recognition site was identified within its 3' untranslated region (3'UTR). Interestingly, HOXA13 acted as a transcription factor to stimulate SLC3A2 expression. Thereby, YTHDC2 suppressed SLC3A2 via inhibiting HOXA13 in an m6A-indirect manner. Mouse experiments further confirmed the associations among YTHDC2, SLC3A2 and HOXA13, and demonstrated that SLC3A2 and SLC7A11 were both important for YTHDC2-impaired tumor growth and -induced lipid peroxidation in vivo. Moreover, higher expression of SLC7A11, SLC3A2 and HOXA13 indicate poorer clinical outcome in YTHDC2-suppressed LUAD patients. In conclusion, YTHDC2 is believed to be a powerful endogenous ferroptosis inducer and targeting SLC3A2 subunit of system XC- is essential for this process. Increasing YTHDC2 is an alternative ferroptosis-based therapy to treat LUAD.
Collapse
Affiliation(s)
- Lifang Ma
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Keke Yu
- Department of Bio-bank, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xin Xu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tianxiang Chen
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yi Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorder, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yikun Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shiyu Qiu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Susu Guo
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Jiangtao Cui
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250033, Shandong province, China
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Jinjing Xia
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Jiayi Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China; Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
| |
Collapse
|
22
|
Leistico JR, Saini P, Futtner CR, Hejna M, Omura Y, Soni PN, Sandlesh P, Milad M, Wei JJ, Bulun S, Parker JB, Barish GD, Song JS, Chakravarti D. Epigenomic tensor predicts disease subtypes and reveals constrained tumor evolution. Cell Rep 2021; 34:108927. [PMID: 33789109 PMCID: PMC8111960 DOI: 10.1016/j.celrep.2021.108927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 10/23/2020] [Accepted: 03/09/2021] [Indexed: 12/02/2022] Open
Abstract
Understanding the epigenomic evolution and specificity of disease subtypes from complex patient data remains a major biomedical problem. We here present DeCET (decomposition and classification of epigenomic tensors), an integrative computational approach for simultaneously analyzing hierarchical heterogeneous data, to identify robust epigenomic differences among tissue types, differentiation states, and disease subtypes. Applying DeCET to our own data from 21 uterine benign tumor (leiomyoma) patients identifies distinct epigenomic features discriminating normal myometrium and leiomyoma subtypes. Leiomyomas possess preponderant alterations in distal enhancers and long-range histone modifications confined to chromatin contact domains that constrain the evolution of pathological epigenomes. Moreover, we demonstrate the power and advantage of DeCET on multiple publicly available epigenomic datasets representing different cancers and cellular states. Epigenomic features extracted by DeCET can thus help improve our understanding of disease states, cellular development, and differentiation, thereby facilitating future therapeutic, diagnostic, and prognostic strategies.
Collapse
Affiliation(s)
- Jacob R Leistico
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Priyanka Saini
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christopher R Futtner
- Department of Medicine, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Miroslav Hejna
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yasuhiro Omura
- Department of Medicine, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Pritin N Soni
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Poorva Sandlesh
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Magdy Milad
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Serdar Bulun
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - J Brandon Parker
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Grant D Barish
- Department of Medicine, Northwestern University, Chicago, IL, USA; Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jun S Song
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Department of Pharmacology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
23
|
Zhao X, Fan H, Chen X, Zhao X, Wang X, Feng Y, Liu M, Li S, Tang H. Hepatitis B Virus DNA Polymerase Restrains Viral Replication Through the CREB1/HOXA Distal Transcript Antisense RNA Homeobox A13 Axis. Hepatology 2021; 73:503-519. [PMID: 32314410 DOI: 10.1002/hep.31284] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 02/20/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Long noncoding RNAs (lncRNAs) have been associated with infection and hepatitis B virus (HBV)-related diseases, though the underlying mechanisms remain unclear. APPROACH AND RESULTS We obtained HBV-HCC lncRNA profiles by deep sequencing and found HOXA distal transcript antisense RNA (HOTTIP) to be significantly up-regulated. RT-qPCR indicated that HOTTIP is highly expressed in HBV-positive hepatoma tissue and induced by HBV in vitro. Virological experiments showed that HOTTIP significantly suppresses the generation of hepatitis B viral surface antigen, hepatitis B viral e antigen and HBV replication. Homeobox A13 (HOXA13), a downstream factor of HOTTIP, was found to bind to HBV enhancer I and X promotor to repress the production of HBV pregenome RNA (pgRNA) and total RNA as well as HBV replication, suggesting that HOXA13 mediates HOTTIP-induced suppression of HBV replication. More interestingly, HBV DNA polymerase (DNA pol) binds to and stabilizes cAMP-responsive element-binding protein 1 (CREB1) mRNA to facilitate translation of the protein, which, in turn, binds to the regulatory element of HOTTIP to promote its expression. CONCLUSIONS Our findings demonstrate that HBV DNA pol attenuates HBV replication through activation of the CREB1-HOTTIP-HOXA13 axis. These findings shed light on the mechanism by which HBV restrains replication to contribute to persistent infection.
Collapse
Affiliation(s)
- Xiaopei Zhao
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Hongxia Fan
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Xi Chen
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Xiaoqing Zhao
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Xu Wang
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Yujie Feng
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Min Liu
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Shengping Li
- State Key Laboratory of Oncology in Southern ChinaDepartment of Hepatobiliary OncologySun Yat-sen UniversityCancer CenterGuangzhouChina
| | - Hua Tang
- Tianjin Life Science Research CenterTianjin Key Laboratory of Inflammation BiologyCollaborative Innovation Center of Tianjin for Medical EpigeneticsDepartment of Pathogen BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| |
Collapse
|
24
|
Chen BW, Zhou Y, Wei T, Wen L, Zhang YB, Shen SC, Zhang J, Ma T, Chen W, Ni L, Wang Y, Bai XL, Liang TB. lncRNA-POIR promotes epithelial-mesenchymal transition and suppresses sorafenib sensitivity simultaneously in hepatocellular carcinoma by sponging miR-182-5p. J Cell Biochem 2020; 122:130-142. [PMID: 32951268 DOI: 10.1002/jcb.29844] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/31/2022]
Abstract
Sorafenib (SOR) resistance remains a major obstacle in the effective treatment of hepatocellular carcinoma (HCC). A number of long noncoding RNAs (lncRNAs) are responsible for this chemoresistance. This study aimed to reveal the essential function of a recently defined lncRNA, lncRNA-POIR, in the epithelial-mesenchymal transition (EMT) and SOR sensitivity of HCC cells. SOR-induced cytotoxicity was analyzed via cell counting kit-8 and ethynyl-2'-deoxyuridine incorporation assays, whereas immunoblotting and confocal immunofluorescence were used to determine the expression levels of EMT markers. Furthermore, loss- or gain-of-function approaches were used to demonstrate the role of lncRNA-POIR/miR-182-5p on EMT and SOR sensitivity in HCC. The direct interaction between lncRNA-POIR and miR-182-5p was verified using a luciferase reporter assay. We found that knockdown of lncRNA-POIR sensitized HCC cells to SOR and simultaneously reversed EMT. As expected, miR-182-5p was confirmed as the downstream target of lncRNA-POIR. Moreover, miR-182-5p overexpression clearly reversed EMT and promoted SOR-induced cytotoxicity in representative HCC cells, whereas miR-182-5p downregulation played a contrasting role; miR-182-5p knockdown abolished the modulatory effects of lncRNA-POIR siRNA on EMT and SOR sensitivity. Together, these pieces of data suggest that lncRNA-POIR promotes EMT progression and suppresses SOR sensitivity simultaneously by sponging miR-182-5p. Thus, we proposed a compelling rationale for the use of lncRNA-POIR as a promising predictor of SOR response and as a potential therapeutic target for HCC treatment in the future.
Collapse
Affiliation(s)
- Bryan Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tao Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Liang Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yi-Bo Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Shi-Chao Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Wen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Lei Ni
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Ting-Bo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| |
Collapse
|
25
|
Jin X, Dai L, Ma Y, Wang J, Yan H, Jin Y, Zhu X, Liu Z. Homeobox proteins are potential biomarkers and therapeutic targets in gastric cancer: a systematic review and meta-analysis. BMC Cancer 2020; 20:866. [PMID: 32907552 PMCID: PMC7487678 DOI: 10.1186/s12885-020-07346-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND An increasing number of studies have described the aberrant expression of homeobox (HOX) proteins in gastric cancer (GC), which is critically associated with the prognosis and clinicopathological characteristics of GC. This study was conducted to investigate the clinical value and action mechanisms of HOX proteins in GC. METHODS A comprehensive search of PubMed, Embase, Web of Science and Cochrane Library was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) statement. The pooled hazard ratio (HR) with its 95% confidence interval (95% CI) and the pooled odds ratio (OR) with its 95% CI were used to assess the effect of HOX protein expression on the prognosis and clinicopathological features of GC, respectively. RESULTS Nineteen studies containing 3775 patients were selected for this study. Heterogeneity among HRs of overall survival (OS) was markedly high (I2 = 90.5%, p = 0.000). According to the subgroup analysis, increased expression of HOX protein in the downregulated subgroup was associated with a good prognosis for patients with GC (pooled HR: 0.46, 95% CI: 0.36-0.59, I2 = 3.1%, p = 0.377), while overexpression of HOX protein in the upregulated subgroup was correlated with a reduced OS (pooled HR: 2.59, 95% CI: 1.79-3.74, I2 = 73.5%, p = 0.000). The aberrant expression of HOX protein was crucially related to the TNM stage, depth of tumour invasion, tumour size, lymph node metastasis, distant metastasis, vascular invasion, histological differentiation and Lauren classification in patients with GC. In addition, the molecular mechanisms by which HOX proteins regulate tumorigenesis and development of GC were also explored. CONCLUSIONS HOX proteins play vital roles in GC progression, which might serve as prognostic markers and therapeutic targets for GC.
Collapse
Affiliation(s)
- Xiao Jin
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Lu Dai
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Yilan Ma
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Jiayan Wang
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Haihao Yan
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Ye Jin
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Xiaojuan Zhu
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China
| | - Zheng Liu
- Institute of Digestive Endoscopy and Medical Centre for Digestive Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 210011, People's Republic of China.
| |
Collapse
|
26
|
Gu Y, Gu J, Shen K, Zhou H, Hao J, Li F, Yu H, Chen Y, Li J, Li Y, Liang H, Dong Y. HOXA13 promotes colon cancer progression through β-catenin-dependent WNT pathway. Exp Cell Res 2020; 395:112238. [PMID: 32822724 DOI: 10.1016/j.yexcr.2020.112238] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/29/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022]
Abstract
Human class I homeobox A13 (HOXA13) was initially identified as a transcription factor and has an important role in embryonic development and malignant transformation. However, the clinical significance and the molecular mechanisms of HOXA13 in colon cancer development and progression are still unknown. In this study, we found that HOXA13 was highly expressed in colon cancer tissues, and its expression was associated with histological grade, T stage, N stage and tumour size. In vitro studies showed that HOXA13 promoted colon cancer cell proliferation, migration and invasion. Bioinformatics analysis revealed that HOXA13 expression was positively correlated with the WNT signalling pathway. In vitro studies showed that HOXA13 promoted the malignant phenotype of colon cancer cells by facilitating the nuclear translocation of β-Catenin. Moreover, XAV939, an inhibitor of β-Catenin, reversed the HOXA13-mediated effects on invasion and proliferation of colon cancer cells. In vivo studies further verified that HOXA13 promoted tumour formation through the Wnt/β-Catenin pathway. Collectively, these results suggest that HOXA13 is a potential oncogene that functions by promoting the nuclear translocation of β-Catenin, thereby maintaining the proliferation and metastasis of colon cancer.
Collapse
Affiliation(s)
- Yan Gu
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Jun Gu
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Kaicheng Shen
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Hongxu Zhou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Jie Hao
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Fu Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Hua Yu
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, 610072, China
| | - Yueqi Chen
- Department of Orthopaedic, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Yifei Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China.
| | - Yan Dong
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
27
|
The mechanisms of sorafenib resistance in hepatocellular carcinoma: theoretical basis and therapeutic aspects. Signal Transduct Target Ther 2020; 5:87. [PMID: 32532960 PMCID: PMC7292831 DOI: 10.1038/s41392-020-0187-x] [Citation(s) in RCA: 660] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/14/2020] [Accepted: 04/26/2020] [Indexed: 02/07/2023] Open
Abstract
Sorafenib is a multikinase inhibitor capable of facilitating apoptosis, mitigating angiogenesis and suppressing tumor cell proliferation. In late-stage hepatocellular carcinoma (HCC), sorafenib is currently an effective first-line therapy. Unfortunately, the development of drug resistance to sorafenib is becoming increasingly common. This study aims to identify factors contributing to resistance and ways to mitigate resistance. Recent studies have shown that epigenetics, transport processes, regulated cell death, and the tumor microenvironment are involved in the development of sorafenib resistance in HCC and subsequent HCC progression. This study summarizes discoveries achieved recently in terms of the principles of sorafenib resistance and outlines approaches suitable for improving therapeutic outcomes for HCC patients.
Collapse
|
28
|
Cheng Z, Wei-Qi J, Jin D. New insights on sorafenib resistance in liver cancer with correlation of individualized therapy. Biochim Biophys Acta Rev Cancer 2020; 1874:188382. [PMID: 32522600 DOI: 10.1016/j.bbcan.2020.188382] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
Liver cancer is highly malignant and insensitive to cytotoxic chemotherapy and is associated with very poor patient prognosis. In 2007, the small-molecule targeted drug sorafenib was approved for the treatment of advanced liver cancer. In the subsequent ten years, sorafenib has been the only first-line therapeutic targeted drug for advanced hepatocellular carcinoma (HCC). However, a number of clinical studies show that a considerable percentage of patients with liver cancer are insensitive to sorafenib. The number of patients who actually benefit significantly from sorafenib treatment is very limited, and the overall efficacy of sorafenib is far from satisfactory, which has attracted the attention of researchers. Based on previous studies and reports, this article reviews the potential mechanisms of sorafenib resistance (SR) and summarizes the biomarkers and clinicopathological indicators that might be used for predicting sorafenib response and developing personalized therapy.
Collapse
Affiliation(s)
- Zhang Cheng
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China
| | - Jiang Wei-Qi
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China
| | - Ding Jin
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China.
| |
Collapse
|
29
|
Niu Y, Tang G, Wu X, Wu C. LncRNA NEAT1 modulates sorafenib resistance in hepatocellular carcinoma through regulating the miR-149-5p/AKT1 axis. Saudi J Gastroenterol 2020; 26:285018. [PMID: 32461380 PMCID: PMC7580733 DOI: 10.4103/sjg.sjg_4_20] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/31/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS The purpose of this study is to explore the expression characteristics of lncRNA NEAT1 in hepatocellular carcinoma (HCC) and the molecular mechanism of its regulation on sorafenib resistance. MATERIALS AND METHODS This experimental study was performed from June 2013 to June 2019. The level of NEAT1 was determined using RT-PCR in HCC and matched adjacent tissues from 79 HCC patients in Linyi central hospital. The patients were divided into two groups to compare their prognosis based on the median NEAT1 expressions as a cutoff value. HCC cell line HepG2 negative control (HepG2-NC), sorafenib-resistant HepG2 cells (HepG2-SR) were transfected with or without NEAT1 siRNA, followed by subsequent molecular analysis, to determine the function of NEAT1 on sorafenib resistance in HCC cells. The cell transcripts were determined by RNA-sequencing analysis. The binding site of the NEAT1 and microRNA-149-5p (miR-149-5p) was verified by luciferase assay. RESULTS We found that NEAT1 was significantly increased in HCC tissues. Furthermore, NEAT1 expressions were significantly associated with HCC prognosis and chemoresistance patterns against sorafenib. Subsequently, the sorafenib-resistant HCC cell lines, together with the controls, were used to determine the regulatory effect of NEAT1 on HCC cells' progression and sorafenib resistance. NEAT1 targets the miR-149-5p, and therefore, decrease the activity of sorafenib against HCC cells. NEAT1 functions were demonstrated to be triggered by the regulation of miR-149-5p/AKT1 axis. CONCLUSIONS NEAT1/miR-149-5p/AKT1 pathway-based therapy might be a potential clinical application for HCC patients.
Collapse
Affiliation(s)
- Yuexiang Niu
- Department of Infectious Diseases, Linyi Central Hospital, Linyi, Shandong, P.R. China
| | - Gongen Tang
- Department of Infectious Diseases, Linyi Central Hospital, Linyi, Shandong, P.R. China
| | - Xiuli Wu
- Department of Respiration, Yishui people's Hospital, Linyi, Shandong, P.R. China
| | - Chaoyu Wu
- Department of Infectious Diseases, Linyi Central Hospital, Linyi, Shandong, P.R. China
| |
Collapse
|
30
|
The Underlying Mechanisms of Noncoding RNAs in the Chemoresistance of Hepatocellular Carcinoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:13-27. [PMID: 32505000 PMCID: PMC7270498 DOI: 10.1016/j.omtn.2020.05.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/15/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal human malignancies. Chemotherapeutic agents, such as sorafenib and lenvatinib, can improve the outcomes of HCC patients. Nevertheless, chemoresistance has become a major hurdle in the effective treatment of HCC. Noncoding RNAs (ncRNAs), including mircoRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), have been demonstrated to participate in the onset and progression of HCC. Moreover, multiple lines of evidence have indicated that ncRNAs also play a pivotal role in HCC drug resistance. ncRNAs can regulate drug efflux and metabolism, glucose metabolism, cellular death pathways, and malignant characteristics in HCC. A deeper understanding of the molecular mechanisms responsible for ncRNA-mediated drug resistance in HCC will provide new opportunities for improving the treatment of HCC. In this review, we summarize recent findings on the molecular mechanisms by which ncRNAs regulate HCC chemoresistance, as well as their potential clinical implications in overcoming HCC chemoresistance.
Collapse
|
31
|
Ramos Caetano BF, Baptista Tablas M, Ribeiro Romualdo G, Marchesan Rodrigues MA, Barbisan LF. Early molecular events associated with liver and colon sub-acute responses to 1,2-dimethylhydrazine: Potential implications on preneoplastic and neoplastic lesion development. Toxicol Lett 2020; 329:67-79. [PMID: 32387197 DOI: 10.1016/j.toxlet.2020.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
This study unveiled the early cellular and molecular events induced by 1,2-dimethylhydrazine (DMH) in the colon and liver and their implications on pre- and neoplastic lesion burden in a late timepoint. Male Wistar rats received four DMH injections (40 mg/kg body weight) for 2 weeks and were sacrificed 24 h (short-term study) or 22 (medium-term study) weeks after the last DMH administration. In the short-term study, DMH led to increased leukocyte (comet assay) and colon (H2AX) genotoxicity, enhanced proliferation (Ki-67) and apoptosis (caspase-3) indexes in both liver and colon. Furthermore, the expression of mRNA (Cat, Gsta1, Gsta2, Gpx1, Gstm1, Sod1, Sod2 and Sod3) and the activity of antioxidant agents were diminished in the colon and liver of DMH-induced rats, eliciting an environment of oxidative stress featuring elevated lipid hydroperoxide levels. Apoptosis effectors were upregulated in the liver (Bax, Casp3 and Fas), and developmental genes were downregulated in both colon and liver (Foxa1, Foxa2, Smad2 and Smad4). In the medium-term study, DMH led to a high number of preneoplastic colonic aberrant crypt foci and tumors (adenomas and invasive adenocarcinomas) but few preneoplastic hepatic glutathione S-transferase (GST-P)-positive foci. Our novel gene expression data highlights overlooked mechanisms in the liver (main metabolizing organ) and colon (main target organ) on toxicity and carcinogenesis induced by repeated doses of DMH, as both organs should be considered in further interventions on the initiation stage of colon carcinogenesis.
Collapse
Affiliation(s)
| | - Mariana Baptista Tablas
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Guilherme Ribeiro Romualdo
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
32
|
Shi S, Zhang J, Liu M, Dong H, Li N. Ras-ERK signalling represses H1.4 phosphorylation at serine 36 to promote non-small-cell lung carcinoma cells growth and migration. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2343-2351. [PMID: 31184227 DOI: 10.1080/21691401.2019.1624558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent papers suggest that oncogenic Ras participate in regulating tumour cells proliferation and metastasis. This work linked Ras with H1.4 modification in non-small-cell lung carcinoma (NSCLC), to better understand the oncogenic effects of Ras. A plasmid for expressing Ras mutated at G13D and T35S was transfected into NCI-H2126 and A549 cells. Phosphorylation of H1.4S36 was determined by immunoblotting. Effects of phosphorylation of H1.4 at serine (S) 36 (H1.4S36ph) on NCI-H2126 and A549 cells were tested by MTT assay, soft-agar colony formation assay, flow cytometry and transwell assay. Chromatin-immunoprecipitation (ChIP) and RT-qPCR were conducted to measure the effects of H1.4S36ph on Ras downstream genes. The catalyzing enzymes participate in H1.4S36 phosphorylation were further studied. We found that Ras-ERK signalling repressed the phosphorylation of H1.4 at S36. H1.4S36ph functioned as a tumour suppressor, as its overexpression repressed NCI-H2126 and A549 cells viability, colony formation, S-phase arrest, migration and invasion. H1.4S36ph was able to mediate the transcription of Ras downstream genes. Ras-ERK signalling repressed H1.4S36ph through degradation of PKA, and the degradation was mediated by MDM2. In conclusion, Ras-ERK signalling repressed H1.4 phosphorylation at S36 to participate in NSCLC cells growth, migration and invasion. Ras-ERK signalling repressed H1.4S36ph through MDM2-dependent degradation of PKA. This study provides a novel explanation for Ras-ERK's tumour-promoting function. Highlights: H1.4S36 phosphorylation is repressed by Ras-ERK activation; H1.4S36ph inhibits the phenotype of NSCLC cells; H1.4S36ph regulates the transcription of Ras downstream genes; Ras-ERK represses H1.4S36ph by MDM2-dependent degradation of PKA.
Collapse
Affiliation(s)
- Shaomin Shi
- a Department of Respiratory, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Jingzhe Zhang
- b Department of Orthopedics, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Meihan Liu
- c Department of Ultrasound, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Hang Dong
- b Department of Orthopedics, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Ning Li
- a Department of Respiratory, China-Japan Union Hospital of Jilin University , Changchun , China
| |
Collapse
|
33
|
Zhang H, Chen X, Zhang J, Wang X, Chen H, Liu L, Liu S. Long non‑coding RNAs in HBV‑related hepatocellular carcinoma (Review). Int J Oncol 2019; 56:18-32. [PMID: 31746420 DOI: 10.3892/ijo.2019.4909] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022] Open
Abstract
Hepatitis B virus (HBV)‑related hepatocellular carcinoma (HCC) is a global health problem that accounts for more than half of total liver cancer cases in developing countries. Despite the growing number of researches conducted, the molecular mechanism underlying the development of HCC remains elusive. Long non‑coding RNAs (lncRNAs), which are non‑coding RNAs >200 nt in length that were previously considered to be transcriptional noise, have been found to be dysregulated in HBV‑related HCC with the help of high‑throughput omics techniques. Subsequent investigations revealed that aberrant expression of lncRNAs may affect the risk of HBV‑related HCC through diverse mechanisms, including epigenetic silencing of transcriptional activation, alternative splicing, molecular sponging, modulating protein stability, and by serving as precursors of miRNAs. Although the sensitivity and specificity of lncRNAs must be further validated, a number of circulating lncRNAs have been identified as useful biomarkers for HBV‑related HCC. In addition to these findings, recent studies also unveiled that certain genetic polymorphisms in lncRNAs may affect the occurrence and prognosis of HBV‑related HCC. The aim of the present review was to provide an overview of the mechanisms underlying the involvement of lncRNAs in HBV‑related HCC. Subsequently, lncRNAs found to be dysregulated in HBV‑related HCC were focused on and current findings on circulating lncRNAs and their genetic polymorphisms were discussed.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Sichuan 610000, P.R. China
| | - Xuebing Chen
- Department of Infectious Diseases, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Jian Zhang
- Department of Pathology, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Xianwei Wang
- Department of Pathology, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Huijuan Chen
- Department of Pathology, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Lin Liu
- Department of Pathology, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Shanling Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Sichuan 610000, P.R. China
| |
Collapse
|
34
|
HOXD9 promotes the growth, invasion and metastasis of gastric cancer cells by transcriptional activation of RUFY3. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:412. [PMID: 31547840 PMCID: PMC6755711 DOI: 10.1186/s13046-019-1399-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Abstract
Background The transcription factor HOXD9 is one of the members of the HOX family, which plays an important role in neoplastic processes. However, the role of HOXD9 in the growth and metastasis of gastric cancer (GC) remains to be elucidated. Methods In vitro functional role of HOXD9 and RURY3 in GC cells was determined using the TMA-based immunohistochemistry, western blot, EdU incorporation, gelatin zymography, luciferase, chromatin Immunoprecipitation (ChIP) and cell invasion assays. In vivo tumor growth and metastasis were conducted in nude mice. Results HOXD9 is overexpressed in GC cells and tissues. The high expression of HOXD9 was correlated with poor survival in GC patients. Functionally, HOXD9 expression significantly promoted the proliferation, invasion and migration of GC cells. Mechanically, HOXD9 directly associated with the RUFY3 promoter to increase the transcriptional activity of RUFY3. Inhibition of RUFY3 attenuated the proliferation, migration and invasiveness of HOXD9-overexpressing GC cells in vitro and in vivo. Moreover, both HOXD9 and RUFY3 were highly expressed in cancer cells but not in normal gastric tissues, with their expressions being positively correlated. Conclusions The evidence presented here suggests that the HOXD9-RUFY3 axis promotes the development and progression of human GC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1399-1) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Lee YJ, Oh H, Kim E, Ahn B, Lee JH, Lee Y, Chae YS, Kang SG, Kim CH. Long noncoding RNA HOTTIP overexpression: A potential prognostic biomarker in prostate cancer. Pathol Res Pract 2019; 215:152649. [PMID: 31570281 DOI: 10.1016/j.prp.2019.152649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/06/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022]
Abstract
HOXA transcript at the distal tip (HOTTIP) is a long noncoding RNA (lncRNA), which is >200 nucleotides in length. HOTTIP expression has been demonstrated to play a crucial oncogenic role in cancer pathogenesis, and is said to be associated with poor human cancer prognosis. In prostate cancer, HOTTIP has been identified as an oncogene, but its clinicopathologic significance remains unclear. Array-based qRT-PCR was used to investigate lncRNA levels in 10 pairs of prostate cancer tissues and non-neoplastic parenchyma. Tissue microarray (TMA) was constructed using a total of 70 surgically resected prostatic adenocarcinoma tissues obtained from the Korea University Anam Hospital from 2009 to 2013. HOTTIP expression was determined by RNA in situ hybridization(ISH) and was correlated with clinicopathologic features. Increased HOTTIP expression was observed in all available prostate cancer tissue specimens compared with that in paired normal tissue. High HOTTIP expression was positively associated with bad clinicopathologic features, including higher pathologic T stage (p < 0.001), presence of extraprostatic extension (p < 0.001), seminal vesicle invasion (p < 0.001), perineural invasion (p < 0.001), and the tumor involvement of resection margin (p = 0.044). In particular, significantly increased HOTTIP expression was observed in specimens from patients in the high or very high-risk group, according to the 2018 National Comprehensive Cancer Network (NCCN) guidelines (p < 0.001). Also, patients with high HOTTIP expression showed poorer overall survival than those with low expression. In conclusion, we analytically validated the poor prognostic significance of HOTTIP overexpression and its association with bad clinicopathologic features, and present HOTTIP as a potential prognostic biomarker in prostate cancer.
Collapse
Affiliation(s)
- Yoo Jin Lee
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Harim Oh
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Eojin Kim
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Bokyung Ahn
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Jeong Hyeon Lee
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Youngseok Lee
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Yang Seok Chae
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Sung Gu Kang
- Department of Urology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Chul Hwan Kim
- Department of Pathology, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| |
Collapse
|
36
|
Yang P, Kang W, Pan Y, Zhao X, Duan L. Overexpression of HOXC6 promotes cell proliferation and migration via MAPK signaling and predicts a poor prognosis in glioblastoma. Cancer Manag Res 2019; 11:8167-8179. [PMID: 31564976 PMCID: PMC6731974 DOI: 10.2147/cmar.s209904] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/10/2019] [Indexed: 12/27/2022] Open
Abstract
Background Homeobox (HOX) genes encode transcription factors that are critical to morphogenesis and cell differentiation. Although the dysregulation of several HOX genes in glioblastoma (GBM) has been reported, little is known about HOXC6 expression in GBM. Therefore, in this study, we investigated the expression levels of the HOXC6 in GBM and explored the regulatory mechanism underlying the role of HOXC6 in GBM progression. Methods The ONCOMINE and Oncolnc databases were used to predict the expression level of HOXC6 mRNA and its prognostic value in GBM. The expressions of HOXC6 mRNA in GBM tissues and adjacent brain tissues were detected using qRT-PCR and Western blot. Immunohistochemistry was performed to verify the HOXC6 protein expression in 107 GBM tissues. Kaplan–Meier and Cox analyses were performed to validate the correlation between HOXC6 expression and GBM prognosis. Lentivirus-mediated HOXC6 mRNA overexpression and interference system were established and transfected into U251 and U87 cell lines. CCK-8, colony formation, wound healing and transwell assay were utilized to evaluate the effects of HOXC6 on proliferation and migration of human GBM cells. Results High expression of HOXC6 was observed in GBM tissues and GBM cells lines, and it correlated with a decreased overall survival and disease-free survival. Overexpression of HOXC6 promoted the GBM cell proliferation and migration, whereas depletion of HOXC6 reduced GBM cell proliferation and migration. Mechanistic study showed that upregulation of HOXC6 significantly increased the phosphorylation of Jun amino-terminal kinase, ERK and P38, as well as the expression of mitogen-activated protein kinase (MAPK) signaling–related genes, including c-myc, c-jun and p53. Inversely, silencing HOXC6 showed the opposite results. Conclusion HOXC6 promoted proliferation and migration of GBM cells via the activation of MAPK pathway.
Collapse
Affiliation(s)
- PengYu Yang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Wei Kang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - YaWen Pan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - XianJun Zhao
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Lei Duan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| |
Collapse
|
37
|
Jiang Y, Han QJ, Zhang J. Hepatocellular carcinoma: Mechanisms of progression and immunotherapy. World J Gastroenterol 2019; 25:3151-3167. [PMID: 31333308 PMCID: PMC6626719 DOI: 10.3748/wjg.v25.i25.3151] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 05/18/2019] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is one of the most common malignancies, and various pathogenic factors can lead to its occurrence and development. Among all primary liver cancers, hepatocellular carcinoma (HCC) is the most common. With extensive studies, an increasing number of molecular mechanisms that promote HCC are being discovered. Surgical resection is still the most effective treatment for patients with early HCC. However, early detection and treatment are difficult for most HCC patients, and the postoperative recurrence rate is high, resulting in poor clinical prognosis of HCC. Although immunotherapy takes longer than conventional chemotherapy to produce therapeutic effects, it persists for longer. In recent years, the emergence of many new immunotherapies, such as immune checkpoint blockade and chimeric antigen receptor T cell therapies, has given new hope for the treatment of HCC.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Antineoplastic Agents, Immunological/therapeutic use
- Cancer Vaccines/therapeutic use
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Disease Progression
- Humans
- Immunotherapy, Adoptive/methods
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/prevention & control
- Receptors, Chimeric Antigen/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- Yu Jiang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Qiu-Ju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
38
|
Paralogous HOX13 Genes in Human Cancers. Cancers (Basel) 2019; 11:cancers11050699. [PMID: 31137568 PMCID: PMC6562813 DOI: 10.3390/cancers11050699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/17/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
Hox genes (HOX in humans), an evolutionary preserved gene family, are key determinants of embryonic development and cell memory gene program. Hox genes are organized in four clusters on four chromosomal loci aligned in 13 paralogous groups based on sequence homology (Hox gene network). During development Hox genes are transcribed, according to the rule of “spatio-temporal collinearity”, with early regulators of anterior body regions located at the 3’ end of each Hox cluster and the later regulators of posterior body regions placed at the distal 5’ end. The onset of 3’ Hox gene activation is determined by Wingless-type MMTV integration site family (Wnt) signaling, whereas 5’ Hox activation is due to paralogous group 13 genes, which act as posterior-inhibitors of more anterior Hox proteins (posterior prevalence). Deregulation of HOX genes is associated with developmental abnormalities and different human diseases. Paralogous HOX13 genes (HOX A13, HOX B13, HOX C13 and HOX D13) also play a relevant role in tumor development and progression. In this review, we will discuss the role of paralogous HOX13 genes regarding their regulatory mechanisms during carcinogenesis and tumor progression and their use as biomarkers for cancer diagnosis and treatment.
Collapse
|
39
|
Homeobox Genes and Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11050621. [PMID: 31058850 PMCID: PMC6562709 DOI: 10.3390/cancers11050621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/27/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common type of cancer, and is the third leading cause of cancer-related deaths each year. It involves a multi-step progression and is strongly associated with chronic inflammation induced by the intake of environmental toxins and/or viral infections (i.e., hepatitis B and C viruses). Although several genetic dysregulations are considered to be involved in disease progression, the detailed regulatory mechanisms are not well defined. Homeobox genes that encode transcription factors with homeodomains control cell growth, differentiation, and morphogenesis in embryonic development. Recently, more aberrant expressions of Homeobox genes were found in a wide variety of human cancer, including HCC. In this review, we summarize the currently available evidence related to the role of Homeobox genes in the development of HCC. The objective is to determine the roles of this conserved transcription factor family and its potential use as a therapeutic target in future investigations.
Collapse
|
40
|
Ji S, Wang S, Zhao X, Lv L. Long noncoding RNA NEAT1 regulates the development of osteosarcoma through sponging miR-34a-5p to mediate HOXA13 expression as a competitive endogenous RNA. Mol Genet Genomic Med 2019; 7:e673. [PMID: 31044561 PMCID: PMC6565592 DOI: 10.1002/mgg3.673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/09/2019] [Accepted: 03/13/2019] [Indexed: 12/27/2022] Open
Abstract
Background Long noncoding RNA (lncRNA) exerts a potential regulatory role in tumorigenesis. LncRNA NEAT1 expression remains high in osteosarcoma tissues. However, its biological mechanism in osteosarcoma remains unknown. Methods In this study, NEAT1 expression in osteosarcoma cells was detected by qRT‐PCR. Proliferative and apoptosis potentials of osteosarcoma cells were determined by CCK‐8 assay and Flow Cytometry, respectively. We identified the potential target of NEAT1 through bioinformatics and dual‐luciferase reporter gene assay. Furthermore, their interaction and functions in regulating the development of osteosarcoma were clarified by Western blot and RIP assay. Results Our results demonstrated a high expression of NEAT1 in osteosarcoma tissues and cells. Overexpression of NEAT1 markedly accelerated proliferative and reduced apoptosis potentials of osteosarcoma cells. Besides, NEAT1 could positively regulate the expression of HOXA13 by competing with miR‐34a‐5p. Conclusion These results indicated that NEAT1 participated in the development of osteosarcoma as a ceRNA to competitively bind to miR‐34a‐5p and thus mediate HOXA13 expression.
Collapse
Affiliation(s)
- Shaolin Ji
- Hand Foot Surgery, Yidu Central Hospital of Weifang City, Qingzhou, China
| | - Shunsheng Wang
- Anorectal Surgery, Yidu Central Hospital of Weifang City, Qingzhou, China
| | - Xiaodan Zhao
- Thoracic Surgery, The First Hospital of Xingtai City, Xingtai, China
| | - Li Lv
- Hand Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
41
|
Long noncoding RNA LUCAT1 promotes malignancy of ovarian cancer through regulation of miR-612/HOXA13 pathway. Biochem Biophys Res Commun 2018; 503:2095-2100. [DOI: 10.1016/j.bbrc.2018.07.165] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 07/31/2018] [Indexed: 01/04/2023]
|
42
|
Shi Q, Shen L, Dong B, Fu H, Kang X, Dai L, Yang Y, Yan W, Chen K. Downregulation of HOXA13 sensitizes human esophageal squamous cell carcinoma to chemotherapy. Thorac Cancer 2018; 9:836-846. [PMID: 29757528 PMCID: PMC6026615 DOI: 10.1111/1759-7714.12758] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/02/2018] [Accepted: 04/06/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chemoresistance often develops in esophageal squamous cell carcinoma (ESCC), leading to poor prognosis. HOX genes play a crucial role in embryonic development and cell differentiation. Studies have recently linked HOX with chemoresistance, thus we explored whether HOXA13 is involved in ESCC chemoresistance. METHODS One hundred thirty-one ESCC patients who received neoadjuvant chemotherapy were enrolled. HOXA13 expression was examined by immunohistochemistry. RNA interference was used to knock down the HOXA13 expression in KYSE70 and transfected HOXA13 plasmid to overexpress HOXA13 in KYSE510 cells. We examined half-maximal inhibitory concentration of cisplatin, apoptosis, and epithelial-to-mesenchymal transition (EMT) in ESCC cell lines with different HOXA13 expression levels by cell counting kit-8, flow cytometry, and transwell analysis. RESULTS The median survival of patients with high HOXA13 expression was significantly shorter than those with low expression (P = 0.027). HOXA13 was associated with worse tumor regression grade (P = 0.009). Low HOXA13 expressed cells decreased the half-maximal inhibitory concentration of cisplatin (P < 0.05), increased cisplatin-induced apoptosis (P < 0.05), and decreased EMT (P < 0.05) compared with high HOXA13 expressed cells. In low HOXA13 expressed cells, cleaved caspase-3 and cleaved PARP expression induced by cisplatin increased, while expression of E-cadherin and Snail protein, markers of EMT, was upregulated and downregulated, respectively. EMT decreased in low HOXA13 expressed cells. CONCLUSION High HOXA13 expression was associated with inferior tumor regression grade and poor overall survival in ESCC patients treated with neoadjuvant chemotherapy. HOXA13 increased cisplatin-resistance and promoted EMT in ESCC cells.
Collapse
Affiliation(s)
- Qi Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Luyan Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Bin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Hao Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Xiaozheng Kang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Liang Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Yongbo Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Wanpu Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| | - Ke‐Neng Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery IPeking University Cancer Hospital & InstituteBeijingChina
| |
Collapse
|
43
|
Peng L, Yuan XQ, Zhang CY, Peng JY, Zhang YQ, Pan X, Li GC. The emergence of long non-coding RNAs in hepatocellular carcinoma: an update. J Cancer 2018; 9:2549-2558. [PMID: 30026854 PMCID: PMC6036883 DOI: 10.7150/jca.24560] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounting for roughly 90% of all primary liver neoplasms is the sixth most frequent neoplasm and the second prominent reason of tumor fatality worldwide. As regulators of diverse biological processes, long non-coding RNAs (lncRNAs) are involved in onset and development of neoplasms. With the continuous booming of well-featured lncRNAs in HCC from 2016 to now, we reviewed the newly-presented comprehension about the relationship between lncRNAs and HCC in this study. To be specific, we summarized the overview function and study tools of lncRNAs, elaborated the roles of lncRNAs in HCC, and sketched the molecule mechanisms of lncRNAs in HCC. In addition, the application of lncRNAs serving as biomarkers in early diagnosis and outcome prediction of HCC patients was highlighted.
Collapse
Affiliation(s)
- Li Peng
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, P.R. China; Cancer Research Institute, Central South University, Changsha 410078, P.R. China
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Xiao-Qing Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Chao-Yang Zhang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, P.R. China; Cancer Research Institute, Central South University, Changsha 410078, P.R. China
| | - Jiang-Yun Peng
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Ya-Qin Zhang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, P.R. China; Cancer Research Institute, Central South University, Changsha 410078, P.R. China
| | - Xi Pan
- Department of Oncology, the third Xiangya Hospital, Central South University, Changsha 410013, P.R. China
| | - Guan-Cheng Li
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, P.R. China; Cancer Research Institute, Central South University, Changsha 410078, P.R. China
| |
Collapse
|