1
|
Wang C, Ma X. The role of acetylation and deacetylation in cancer metabolism. Clin Transl Med 2025; 15:e70145. [PMID: 39778006 PMCID: PMC11706801 DOI: 10.1002/ctm2.70145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
As a hallmark of cancer, metabolic reprogramming adjusts macromolecular synthesis, energy metabolism and redox homeostasis processes to adapt to and promote the complex biological processes of abnormal growth and proliferation. The complexity of metabolic reprogramming lies in its precise regulation by multiple levels and factors, including the interplay of multiple signalling pathways, precise regulation of transcription factors and dynamic adjustments in metabolic enzyme activity. In this complex regulatory network, acetylation and deacetylation, which are important post-translational modifications, regulate key molecules and processes related to metabolic reprogramming by affecting protein function and stability. Dysregulation of acetylation and deacetylation may alter cancer cell metabolic patterns by affecting signalling pathways, transcription factors and metabolic enzyme activity related to metabolic reprogramming, increasing the susceptibility to rapid proliferation and survival. In this review, we focus on discussing how acetylation and deacetylation regulate cancer metabolism, thereby highlighting the central role of these post-translational modifications in metabolic reprogramming, and hoping to provide strong support for the development of novel cancer treatment strategies. KEY POINTS: Protein acetylation and deacetylation are key regulators of metabolic reprogramming in tumour cells. These modifications influence signalling pathways critical for tumour metabolism. They modulate the activity of transcription factors that drive gene expression changes. Metabolic enzymes are also affected, altering cellular metabolism to support tumour growth.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaoxin Ma
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
2
|
Xu R, Du A, Li J, Yang Q. An anoikis-related gene signature predicts prognosis in patients with acute myeloid leukemia and immunotherapy. Am J Cancer Res 2024; 14:5116-5132. [PMID: 39659934 PMCID: PMC11626272 DOI: 10.62347/mjta2660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/03/2024] [Indexed: 12/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood disorder and the most common type of acute leukemia in adults. Notwithstanding the plethora of therapeutic modalities, a significant cohort of patients fail to respond to treatment and experience relapse. Anoikis, a distinct modality of programmed cell death, has been linked to cancer progression. However, the prognostic significance of anoikis in AML remains unclear. In this study, a non-negative matrix factorization algorithm was utilized to efficiently reduce the dimensions of merged datasets. We used differential analysis, weighted gene co-expression network analysis (WGCNA), univariate Cox regression, and least absolute shrinkage and selection operator (LASSO) regression to identify genes associated with prognosis and develop a risk scoring model. Immunohistochemistry was conducted to assess the expression levels of key genes in clinical samples. The association between risk score and the tumor microenvironment (TME), stemness, clinical characteristics, and immunotherapy was evaluated. We identified 41 AML anoikis-related genes (ANRGs) related to survival, and seven genes were chosen to develop prognostic models. The prognostic risk score combined with the clinical and pathological features of AML was used to develop a nomogram, and decision curve analysis demonstrated the net clinical benefit of the model. Furthermore, analysis of ANRGs revealed that PDGFRB inhibition significantly reduced the proliferation of AML cells, promoted apoptosis, and inhibited AML progression both in vitro and in vivo, indicating that PDGFRB plays a crucial role in AML development.
Collapse
Affiliation(s)
- Rong Xu
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City)Changde 415000, Hunan, China
| | - Ashuai Du
- Department of Infectious Diseases, Guizhou Provincial People’s HospitalGuiyang 550002, Guizhou, China
| | - Jianbo Li
- Department of Pathology, Xiangya Changde HospitalChangde 415000, Hunan, China
| | - Qinglong Yang
- Department of General Surgery, Guizhou Provincial People’s HospitalGuiyang 550002, Guizhou, China
| |
Collapse
|
3
|
Liang L, Huang Y, Chen L, Shi Z, Wang H, Zhang T, Li Z, Mi J, Fan T, Lu Y, Chen F, Huang W, Hu K. Radioprotective efficacy of Astilbin in mitigating radiation-induced lung injury through inhibition of p53 acetylation. ENVIRONMENTAL TOXICOLOGY 2023; 38:2967-2980. [PMID: 37598414 DOI: 10.1002/tox.23931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
Radiation-induced lung injury (RILI) is a common side effect in thoracic tumor patients undergoing radiotherapy. At present, there is no ideal radio-protective agent which is widely used in RILI treatment. Astilbin (AST), a bioactive flavonoid, exhibits various biological effects, including anti-inflammatory, antioxidant, and anti-fibrotic activities, which partly result from reducing oxidative stress and inflammation in various pathogenic conditions. However, the protective efficacy of AST to ameliorate RILI has not been reported. In this study, we employed network pharmacology, RNA sequencing, and experimental evaluation to reveal the effects and pharmacological mechanism of AST to treat RILI in vivo and in vitro. We observed that AST reduced radiation-induced apoptosis, DNA damage, inflammatory reactions, and the reactive oxygen species (ROS) level in human normal lung epithelial cells BEAS-2B. Further study showed that AST treatment significantly ameliorated RILI by reducing the radiation-induced pathology changes and inflammatory reaction of lung tissue in C57BL/6J mice. Mechanistically, the expression of epithelial-mesenchymal transition (EMT) markers and radiation-triggered acetylation of the p53 protein were alleviated by AST treatment. Furthermore, AST alleviated the acetylation of p53 after intervention of Trichostatin A (TSA). Our data indicate that AST can alleviate RILI by inhibiting inflammatory reactions and the EMT process through decreasing the expression of p53 acetylation. In conclusion, our study suggests that AST has great potential to be a new protective and therapeutic compound for RILI.
Collapse
Affiliation(s)
- Lixing Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yaqin Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Liuyin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Housheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhixun Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Jinglin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Ting Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yushuang Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Fuli Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| |
Collapse
|
4
|
Elias MH, Lazim N, Sutaji Z, Abu MA, Abdul Karim AK, Ugusman A, Syafruddin SE, Mokhtar MH, Ahmad MF. HOXA10 DNA Methylation Level in the Endometrium Women with Endometriosis: A Systematic Review. BIOLOGY 2023; 12:biology12030474. [PMID: 36979165 PMCID: PMC10045497 DOI: 10.3390/biology12030474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Endometriosis is an inflammatory chronic systemic disease resulting in pelvic pain and infertility. However, despite a high prevalence of endometriosis, disease identification is still insufficient, and a high percentage of misdiagnosing was observed. Hence, a comprehensive study needs to be done to improve our understanding of the pathogenesis of endometriosis. Aberrant hypermethylation of HOXA10 has been reported to play a role in endometriosis. Thus, a comprehensive literature search was conducted to identify the DNA methylation level of HOXA10 among endometriosis patients across populations. The literature search was done using PubMed, Scopus, EBSCOhost, and Science Direct applying (HOXA10 OR "homeobox A10" OR "HOXA-10" OR HOX1) AND ("DNA methylation" OR methylation) AND (endometriosis OR endometrioma) as keywords. From 491 retrieved studies, five original articles investigating the DNA methylation level of HOXA10 from endometrium tissues among endometriosis women were included. All five included studies were classified as high-quality studies. High HOXA10 DNA methylation level was observed in the endometrium tissue of women with endometriosis in all the included studies. The secretory phase was identified as the best sampling time for HOXA10 DNA methylation study in endometriosis, and the most studied DNA methylation site is the promoter region of the HOXA10. However, more studies are needed to expose the HOXA10 mechanism in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Marjanu Hikmah Elias
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
- Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia, Nilai 71800, Negeri Sembilan, Malaysia
| | - Nurunnajah Lazim
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Zulazmi Sutaji
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
- Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia, Nilai 71800, Negeri Sembilan, Malaysia
| | - Mohammad Azrai Abu
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Abdul Kadir Abdul Karim
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, National Univeristy of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Saiful Effendi Syafruddin
- Medical Molecular Biology Institute, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, National Univeristy of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Mohd Faizal Ahmad
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
5
|
Lin J, Lian X, Xue S, Ouyang L, Zhou L, Lu Y, Xie L. miR-135a inhibits the proliferation of HBV-infected hepatocellular carcinoma cells by targeting HOXA10. Transl Cancer Res 2023; 12:135-149. [PMID: 36760373 PMCID: PMC9906062 DOI: 10.21037/tcr-22-2789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/06/2023] [Indexed: 01/16/2023]
Abstract
Background The incidence of hepatocellular carcinoma (HCC) in patients with hepatitis B virus (HBV) is extremely high. MicroRNAs (miRNAs) are a type of endogenous non-coding small RNA with novel molecular therapeutic mechanisms that plays an important role in the occurrence and development of cancers. This study aimed to explore the regulation mechanism of miR-135a and HOXA10 in the proliferation, invasion, and apoptosis of HCC cells. Methods Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis was used to detect the expression level of miR-135a. Overexpression of miR-135a was used to measure the roles of miR-135a in the proliferation, invasion, and apoptosis of HCC cells. A dual luciferase experiment was performed to assess the relationship between HOXA10 and miR-135a. Western blot was applied to observe the protein levels of p-p38, p-ERK, and p-JNK. Results The expression levels of miR-135a were significantly decreased in HCC tissues and cells. Overexpression of miR-135a inhibited the proliferation and invasion but promoted the apoptosis of HCC cells. Importantly, our results confirmed that HOXA10 was a direct target of miR-135a. Under HBV infection, silencing of HOXA10 significantly blocked the proliferation and invasion and promoted the apoptosis of HCC cells. In addition, miR-135a/HOXA10 regulated the expressions of p-p38, p-ERK, and p-JNK through the miR-135a/HOXA10 axis, thereby inhibiting the activation of the MAPK pathway. Conclusions HBV promoted the proliferation and invasion, and inhibited the apoptosis of HCC cells by regulating the miR-135a/HOXA10 pathway. These findings provide a theoretical basis for improving the treatment of HBV-infected HCC patients.
Collapse
Affiliation(s)
- Jianjun Lin
- Clinical Laboratory Department, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| | - Xiang Lian
- Hepatology Department, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| | - Shihang Xue
- Department of General Surgery, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| | - Lian Ouyang
- Department of Orthopaedic Surgery, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| | - Lihui Zhou
- Department of Orthopaedic Surgery, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| | - Yuyang Lu
- Xiangshan County Center for Disease Control and Prevention, Ningbo, China
| | - Longteng Xie
- Hepatology Department, Xiangshan First People’s Hospital, Ningbo Fourth Hospital, Ningbo, China
| |
Collapse
|
6
|
Jiang L, Yang Q. HOXA10 enhances cell proliferation and suppresses apoptosis in esophageal cancer via activating p38/ERK signaling pathway. Open Med (Wars) 2022; 17:1750-1759. [PMID: 36407869 PMCID: PMC9635270 DOI: 10.1515/med-2022-0558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/17/2022] [Accepted: 07/26/2022] [Indexed: 02/22/2024] Open
Abstract
Esophageal cancer (EC) is an extremely aggressive malignant tumor. Homeobox A10 (HOXA10) is highly expressed and plays an important role in a variety of tumors. However, the function of HOXA10 in EC remains unclear. In this study, HOXA10 was observed to highly express in EC tissues and cells. Interestingly, the CCK-8 assay, flow cytometry, and colony formation assay confirmed that overexpression of HOXA10 promoted proliferation and suppressed cell apoptosis in EC cells. More importantly, the western blot assay indicated that the phosphorylation levels of ERK and p38 were elevated in EC cells overexpressed HOXA10, indicating that overexpression of HOXA10 activated p38/ERK signaling pathway in EC cells. These findings concluded that HOXA10 aggravated EC progression via activating p38/ERK signaling pathway, providing a potential therapeutic target for EC.
Collapse
Affiliation(s)
- Lifeng Jiang
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213003, China
| | - Qixian Yang
- Clinical Laboratory of Diagnostics and Gastroenterology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, No. 68 Gehuzhonglu Road, Wujin District, Changzhou, Jiangsu, 213003, China
| |
Collapse
|
7
|
Mishra A, Ganguli N, Majumdar SS, Modi D. Loss of HOXA10 causes endometrial hyperplasia progressing to endometrial cancer. J Mol Endocrinol 2022; 69:431-444. [PMID: 35917434 DOI: 10.1530/jme-22-0051] [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: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
Endometrial cancer is the fourth most common malignancy in women and the precursor lesion is endometrial hyperplasia. HOXA10 is a transcription factor that plays key roles in endometrial functions such as the endowment of receptivity, embryo implantation, and trophoblast invasion. Herein, using testicular transgenesis, we developed transgenic mice that expressed a shRNA against HOXA10 and there was a nearly 70% reduction in the expression of HOXA10 in these animals. We observed that downregulation of HOXA10 led to the development of endometrial hyperplasia in the young animals (3 months), and as they aged (>1 year), most animals developed well-differentiated endometrial adenocarcinoma. In the endometrium of animals with reduced HOXA10, there was increased proliferation and elevated levels of ERα and ERβ. In parallel, there was increased expression of Wnt4 and β-Catenin, SOX9, and YAP1. We propose that chronic reduction in HOXA10 expression disrupts multiple pathways in the uterus that aids in the development of endometrial hyperplasia which progresses to endometrial cancer with age.
Collapse
Affiliation(s)
- Anuradha Mishra
- National Institute for Research in Reproductive and Child Health, ICMR, Parel, Mumbai, India
| | - Nirmalya Ganguli
- National Institute of Immunology, New Delhi, India
- National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Subeer S Majumdar
- National Institute of Immunology, New Delhi, India
- National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Deepak Modi
- National Institute for Research in Reproductive and Child Health, ICMR, Parel, Mumbai, India
| |
Collapse
|
8
|
Aljabban J, Rohr M, Syed S, Cohen E, Hashi N, Syed S, Khorfan K, Aljabban H, Borkowski V, Segal M, Mukhtar M, Mohammed M, Boateng E, Nemer M, Panahiazar M, Hadley D, Jalil S, Mumtaz K. Dissecting novel mechanisms of hepatitis B virus related hepatocellular carcinoma using meta-analysis of public data. World J Gastrointest Oncol 2022; 14:1856-1873. [PMID: 36187396 PMCID: PMC9516659 DOI: 10.4251/wjgo.v14.i9.1856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/26/2022] [Accepted: 08/07/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is a cause of hepatocellular carcinoma (HCC). Interestingly, this process is not necessarily mediated through cirrhosis and may in fact involve oncogenic processes. Prior studies have suggested specific oncogenic gene expression pathways were affected by viral regulatory proteins. Thus, identifying these genes and associated pathways could highlight predictive factors for HCC transformation and has implications in early diagnosis and treatment.
AIM To elucidate HBV oncogenesis in HCC and identify potential therapeutic targets.
METHODS We employed our Search, Tag, Analyze, Resource platform to conduct a meta-analysis of public data from National Center for Biotechnology Information’s Gene Expression Omnibus. We performed meta-analysis consisting of 155 tumor samples compared against 185 adjacent non-tumor samples and analyzed results with ingenuity pathway analysis.
RESULTS Our analysis revealed liver X receptors/retinoid X receptor (RXR) activation and farnesoid X receptor/RXR activation as top canonical pathways amongst others. Top upstream regulators identified included the Ras family gene rab-like protein 6 (RABL6). The role of RABL6 in oncogenesis is beginning to unfold but its specific role in HBV-related HCC remains undefined. Our causal analysis suggests RABL6 mediates pathogenesis of HBV-related HCC through promotion of genes related to cell division, epigenetic regulation, and Akt signaling. We conducted survival analysis that demonstrated increased mortality with higher RABL6 expression. Additionally, homeobox A10 (HOXA10) was a top upstream regulator and was strongly upregulated in our analysis. HOXA10 has recently been demonstrated to contribute to HCC pathogenesis in vitro. Our causal analysis suggests an in vivo role through downregulation of tumor suppressors and other mechanisms.
CONCLUSION This meta-analysis describes possible roles of RABL6 and HOXA10 in the pathogenesis of HBV-related HCC. RABL6 and HOXA10 represent potential therapeutic targets and warrant further investigation.
Collapse
Affiliation(s)
- Jihad Aljabban
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Michael Rohr
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, United States
| | - Saad Syed
- Department of Medicine, Northwestern Memorial Hospital, Chicago, IL 60611, United States
| | - Eli Cohen
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN 37232, United States
| | - Naima Hashi
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Sharjeel Syed
- Department of Medicine, University of Chicago Hospitals, Chicago, IL 60637, United States
| | - Kamal Khorfan
- Department of Gastroenterology and Hepatology, University of California San Francisco-Fresno, Fresno, CA 93701, United States
| | - Hisham Aljabban
- Department of Medicine, Barry University, Miami, FL 33161, United States
| | - Vincent Borkowski
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Michael Segal
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Mohamed Mukhtar
- Department of Medicine, Michigan State University College of Human Medicine, Lansing, MI 49503, United States
| | - Mohammed Mohammed
- Department of Medicine, Windsor University School of Medicine, Frankfort, IL 60423, United States
| | - Emmanuel Boateng
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN 37232, United States
| | - Mary Nemer
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Maryam Panahiazar
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, United States
| | - Dexter Hadley
- Department of Pathology, University of Central Florida College of Medicine, Orlando, FL 32827, United States
| | - Sajid Jalil
- Department of Gastroenterology and Hepatology, Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| | - Khalid Mumtaz
- Department of Gastroenterology and Hepatology, Ohio State University Wexner Medical Center, Columbus, OH 43210, United States
| |
Collapse
|
9
|
Zhong Q, Wang Z, Kang H, Wu R. Molecular mechanism of FBXW7-mediated ubiquitination modification in nasopharyngeal carcinoma cell proliferation in vitro and in vivo. Pathol Res Pract 2022; 244:154056. [PMID: 36989847 DOI: 10.1016/j.prp.2022.154056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022]
Abstract
OBJECTIVES Nasopharyngeal carcinoma (NPC) is a type of keratinizing squamous cell malignancy. Ubiquitination, a common protein posttranslational modification, participates in cancer development. This study sought to investigate the mechanism of F-box and WD repeat domain containing 7 (FBXW7) in NPC cell proliferation in vivo and in vitro. METHODS FBXW7, Homeobox A10 (HOXA10), and bone morphogenetic protein-2 (BMP2) expression levels in NPC tissues and cells were detected by RT-qPCR and Western blotting. Cell proliferation was assessed by cell counting kit-8 and colony formation assays. The binding of FBXW7 to HOXA10 and HOXA10 ubiquitination level were detected via co-immunoprecipitation and ubiquitination assay. Cells were treated with MG132 (the proteasome inhibitor), followed by the determination of HOXA10 ubiquitination and protein levels. The binding of HOXA10 to BMP2 was testified via dual-luciferase and chromatin immunoprecipitation assays. Collaborative experiments were performed to confirm the role of HOXA10 or BMP2 in FBXW7-mediated NPC cell proliferation. Xenograft tumor assay was performed to confirm the role of FBXW7/HOXA10/BMP2 in vivo. RESULTS FBXW7 was under-expressed, while HOXA10 and BMP2 were up-expressed in NPC tissues and cells. FBXW7 overexpression restricted NPC cell proliferation. Mechanically, FBXW7 bound to HOXA10 to promote ubiquitination-based degradation of HOXA10 and further reduced the binding of HOXA10 to the BMP2 promoter and inhibited BMP2 transcription. Overexpression of HOXA10 or BMP2 attenuated the role of FBXW7 overexpression in inhibiting NPC cell proliferation. FBXW7 overexpression reduced Ki67 positive rate and repressed tumor growth. CONCLUSION FBXW7 overexpression promoted HOXA10 ubiquitination-based degradation and further inhibited BMP2 transcription, consequently restricting NPC cell proliferation in vitro and in vivo.
Collapse
|
10
|
Zhang G, Fan W, Wang H, Wen J, Tan J, Xue M, Li J. Non-Apoptotic Programmed Cell Death-Related Gene Signature Correlates With Stemness and Immune Status and Predicts the Responsiveness of Transarterial Chemoembolization in Hepatocellular Carcinoma. Front Cell Dev Biol 2022; 10:844013. [PMID: 35573678 PMCID: PMC9099410 DOI: 10.3389/fcell.2022.844013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Non-apoptotic programmed cell death, including autophagy, ferroptosis, and pyroptosis, newly discovered in recent years, plays an important role in hepatocellular carcinoma (HCC). So, this study attempted to explore the relationship between non-apoptotic programmed cell death-related genes and the molecular characteristics, tumor microenvironment, and prognosis in HCC patients. Methods: The transcriptomic and clinical data of HCC samples were downloaded from various public datasets, followed by acquiring non-apoptotic programmed cell death-related genes from the database. A gene signature model was then constructed using univariate and multivariate Cox regression analyses and validated in other cohorts as well as our institution sequencing data. Kaplan–Meier survival curves and time-dependent receiver operating characteristic curves were generated to evaluate the model’s predictive capability. Furthermore, the relationships among the gene signature, TP53 mutation, stemness, immune status, and responsiveness of transarterial chemoembolization (TACE) were analyzed. Results: The gene signature model was constructed based on five autophagy-, three ferroptosis-, and two pyroptosis-related differentially expressed genes. The model accurately predicted that patients classified as low risk would have better overall survival than high-risk patients, which was robustly consistent with data from other cohorts as well as our institution sequencing data. The comprehensive results indicated that a high-risk index was correlated with a high TP53 mutation rate, high cancer cell stemness, high infiltration of immunosuppressive cells and low immunophenoscore, and low TACE responsiveness of HCC patients. Conclusion: Collectively, the established non-apoptotic programmed cell death-related gene signature was shown to accurately predict prognosis, associated with the TP53 mutation and liver cancer cell stemness, reflect the tumor immune microenvironment, and predict TACE responsiveness in HCC patients.
Collapse
Affiliation(s)
| | | | | | - Jie Wen
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jizhou Tan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Miao Xue
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jiaping Li,
| |
Collapse
|
11
|
Xiulin J, Wang C, Guo J, Wang C, Pan C, Nie Z. Next-generation sequencing identifies HOXA6 as a novel oncogenic gene in low grade glioma. Aging (Albany NY) 2022; 14:2819-2854. [PMID: 35349479 PMCID: PMC9004573 DOI: 10.18632/aging.203977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/27/2022] [Indexed: 11/25/2022]
Abstract
Background: Low grade glioma is one of the most common lethal cancers in the human nervous system. Emerging evidence has demonstrated that homeobox A cluster (HOXA) gene family plays a critical role in the transcriptional regulation as well as cancer initiation and progression. However, the expression, biological functions and upstream regulatory mechanism of 11 HOXAs in low grade glioma are not yet clear. Methods: In this study, we utilized various public databases and bioinformatics analyzed, including TCGA, CGGA, Rembrandt, HPA, LinkedOmics, cBioPortal, TISDIB, single-sample GSEA (ssGSEA), TIMER, LnCeVar, LASSO regression, Cox regression, Kaplan-Meier plot, and receiver operating, characteristic (ROC) analyses, GDSC and CTRP databases to analyzed the mRNA and protein expression profiles, gene mutation, clinical features, diagnosis, prognosis, signaling pathway, TMB, immune subtype, immune cell infiltration, immune modulator, ceRNA network and drug sensitivity of 11 HOXAs. Growth curve and transwell assays were utilized to study the biological characteristics of HOXA6 in LGG progression. Results: In the present study, we found that 11 HOXAs (HOXA1, HOXA2, HOXA3, HOXA4, HOXA5, HOXA6, HOXA7, HOXA9, HOXA10, HOXA11 and HOXA3) were consistently up-regulated in LGG tissues and GBM tissues. Up-regulated of the HOXAs expression were significantly correlated with higher tumor stage, IDH mutation status, 1p/19q co-deletion, histological type and primary therapy outcome. Survival analyses showed that higher expression of HOXA1, HOXA2, HOXA3, HOXA4, HOXA5, HOXA7, HOXA9, HOXA10, HOXA11 and HOXA13 were correlated with shorter overall survival (OS), disease-specific survival (DSS) and progression-free survival (PFS) in LGG patients. Univariate and multivariate analyses revealed that HOXA1, HOXA6 expression and tumor grade, age, primary therapy outcome and age were independent factors affecting the prognosis of LGG patients. ROC curve analysis of HOXAs showed that HOXAs had a high accuracy (AUC > 0.80) in predicting LGG. Furthermore, gene functional enrichment analysis indicated that HOXAs mainly involved in the inflammatory response and immune regulation signaling pathway. CNV and DNA methylation significantly affect the expression of HOXAs. Finally, we uncover that HOXAs expression are highly correlated with immune cells infiltrate, immune modulator and drug sensitivity. We also uncover that the HOXAs related ceRNA network in LGG. More importantly, we found that HOXA6 was highly expressed in LGG cells lines and significantly affected their proliferation and migration abilities. Conclusions: In conclusion, our data demonstrated that HOXA was correlated with progression and immune infiltration, and could serve as a prognostic biomarker for LGG.
Collapse
Affiliation(s)
- Jiang Xiulin
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming 650223, Yunnan, China
| | - Chunyan Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jishu Guo
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Science, Yunnan University, Kunming 650500, Yunnan, China
| | - Chenyang Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Chenglong Pan
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Zhi Nie
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.,Yunnan Province Clinical Research Center for Neurological Diseases, Kunming 650032, Yunnan, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming 650223, Yunnan, China
| |
Collapse
|
12
|
Wang J, Li Y, Zhang C, Chen X, Zhu L, Luo T. Characterization of diagnostic and prognostic significance of cell cycle-linked genes in hepatocellular carcinoma. Transl Cancer Res 2022; 10:4636-4651. [PMID: 35116320 PMCID: PMC8799204 DOI: 10.21037/tcr-21-1145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022]
Abstract
Background The high degree of heterogeneity of hepatocellular carcinoma (HCC) imposes a significant challenge to predict the prognosis. Currently, increasing evidence has indicated that cell cycle-linked genes are strongly linked to occurrence and progress of HCC. Herein, we purposed to create a prediction model on the basis of cell cycle-linked genes. Methods The transcriptome along with clinicopathological data abstracted from The Cancer Genome Atlas (TCGA) were used as a training cohort. Lasso regression analysis was employed to create a prediction model in TCGA cohort. The data of samples obtained from the International Cancer Genome Consortium (ICGC) data resource were applied in the verification of the model. A series of bioinformatics analyzed the relationship of the risk signature with overall survival (OS), biological function, and clinicopathological features. Results Six cell cycle-linked genes (PLK1, CDC20, HSP90AA1, CHEK1, HDAC1, and NDC80) were chosen to create the prognostic model, demonstrating a good prognostic capacity. Further analyses indicated that the model could independently assess the OS of HCC patients. A single-sample gene set enrichment analysis (ssGSEA) indicated that the risk signature was remarkably linked to immune status. Additionally, there was a remarkable association of the risk signature with TP53 mutation frequency, as well as immune checkpoint molecule expression levels. Conclusions We created a prediction model using six cell cycle-linked genes to predict HCC prognosis. The six genes are expected to be novel markers for HCC diagnosis, as well as treatment.
Collapse
Affiliation(s)
- Jukun Wang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yu Li
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chao Zhang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Chen
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Linzhong Zhu
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Cao M, Wang L, Xu D, Bi X, Guo S, Xu Z, Chen L, Zheng D, Li P, Xu J, Zheng S, Wang H, Wang B, Lu J, Li K. The synergistic interaction landscape of chromatin regulators reveals their epigenetic regulation mechanisms across five cancer cell lines. Comput Struct Biotechnol J 2022; 20:5028-5039. [PMID: 36187922 PMCID: PMC9483781 DOI: 10.1016/j.csbj.2022.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/28/2022] [Accepted: 09/06/2022] [Indexed: 11/03/2022] Open
|
14
|
Cheng Y, Heng X, Feng F. G-protein Coupled Receptor 34 Promotes Gliomagenesis by Inducing Proliferation and Malignant Phenotype via TGF-Beta/Smad Signaling Pathway. Technol Cancer Res Treat 2022; 21:15330338221105733. [PMID: 35770303 PMCID: PMC9252019 DOI: 10.1177/15330338221105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background: G-protein coupled receptor 34 (GPR34) is involved in cell motility, differentiation, and mitosis. GPR34 was reported to be highly expressed and play an oncogenic role in several solid tumors. Here, we investigated the mechanisms underlying how GPR34 promotes glioma progression. Methods: Bioinformatic analysis was performed on RNA-seq and clinical data from the gene expression omnibus (GEO), cancer genome atlas (TCGA), and Genotype-Tissue Expression (GTEx) databases. TIMER database and single-sample GSEA (ssGAEA) method were used to investigate the association between the GPR34 expression and immune infiltration level in glioma. Cox regression analysis was employed to ascertain whether the risk signature was an independent prognostic indicator for glioma. The viability and migratory/invasive potential of glioma cells were assessed using Cell Counting Kit-8, colony formation, wound healing, and Transwell assays. Results: We found that GPR34 expression was positively correlated with immune infiltration level and that high GPR34 level may be associated with poor prognosis in glioma. We further found that GPR34 may serve as an independent prognostic marker and prediction factor for the clinicopathological features of glioma. We showed that knocking down GPR34 attenuated the viability and migratory/invasive capacity of glioma cells (U251 and LN229), while GPR34 overexpression exerted the opposite effects. Additionally, core enrichment in the GSEA analysis indicated that GPR34-mediated gliomagenesis was associated with the cell cycle arrest, epithelial–mesenchymal transition (EMT), and activation of the TGF-β/Smad pathway; furthermore, inhibiting TGF-β/Smad signaling using LY2157299, a TGF-β inhibitor, reversed the oncogenic effects and malignant phenotype associated with GPR34 overexpression. Conclusion: GPR34 enhances the malignancy and carcinogenesis of glioma by promoting an EMT-like process, G1/S phase cell cycle transition, and TGF-β/Smad signaling. Accordingly, GPR34 likely functions as an oncogene in glioma and may represent a potential therapeutic target for this cancer.
Collapse
Affiliation(s)
- Yanhao Cheng
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Institute of Brain Science and Brain-Like Intelligence, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China.,Department of Neurosurgery, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China
| | - Xueyuan Heng
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Institute of Brain Science and Brain-Like Intelligence, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China.,Department of Neurosurgery, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China
| | - Fan Feng
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Institute of Brain Science and Brain-Like Intelligence, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China.,Department of Neurosurgery, 529858Linyi People's Hospital, Linyi, Shandong, People's Republic of China
| |
Collapse
|
15
|
Gong D, Zhu H, Zeng L, Hu R, Hu J, Ding J. Overexpression of HOXA10 promotes the growth and metastasis of nasopharyngeal carcinoma. Exp Biol Med (Maywood) 2021; 246:2454-2462. [PMID: 34293937 DOI: 10.1177/15353702211030854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Deregulation of HOX transcription factor family has frequently been observed in multiple human cancers; however, their role in nasopharyngeal carcinoma remains largely unclear. In the present study, we found that HOX gene family is consistently upregulated in nasopharyngeal carcinoma and identified HOXA10 as one of the mostly upregulated HOX genes. Importantly, we show that HOXA10 overexpression is associated with transcriptional activation of multiple oncogenes essential for nasopharyngeal carcinoma carcinogenesis, including S-phase kinase-associated protein 2 (SKP2), calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2), and matrix metalloproteinase 1 (MMP1). Mechanistically, the overexpression of SKP2 induces the degradation of cell cycle inhibitor p27, leading to rapid cell cycle progression and cell proliferation. The overexpression of CAMKK2 is associated with enhanced mTOR signaling activity to meet the increased demand for proteins synthesis in rapid growing nasopharyngeal carcinoma cells. Moreover, MMP1 overexpression facilitates nasopharyngeal carcinoma cell migration and invasion and contributes to cancer metastasis and progression. We thus concluded that HOXA10 overexpression promotes the growth and metastasis of nasopharyngeal carcinoma by transcriptionally activating various oncogenic pathways.
Collapse
Affiliation(s)
- Dan Gong
- Department of Oncology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang 330006, China
| | - Hui Zhu
- Department of Breast Cancer Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China
| | - Lei Zeng
- Department of Oncology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang 330006, China
| | - Ronghuan Hu
- Department of Oncology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang 330006, China
| | - Jiali Hu
- Department of Oncology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang 330006, China
| | - Jianwu Ding
- Department of Oncology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.,Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang 330006, China
| |
Collapse
|
16
|
Inhibition of histone deacetylase 1 suppresses pseudorabies virus infection through cGAS-STING antiviral innate immunity. Mol Immunol 2021; 136:55-64. [PMID: 34087624 DOI: 10.1016/j.molimm.2021.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/16/2021] [Accepted: 05/24/2021] [Indexed: 01/19/2023]
Abstract
Pseudorabies virus (PRV) is an enveloped double-stranded DNA virus that is the etiological agent of Aujeszky's disease in pigs. Vaccination is currently available to prevent PRV infection, but there is still an urgent need for new strategies to control this infectious disease. Histone deacetylases (HDACs) are epigenetic regulators that regulate the histone tail, chromatin conformation, protein-DNA interaction and even transcription. Viral transcription and protein activities are intimately linked to regulation by histone acetyltransferases and HDACs that remodel chromatin and regulate gene expression. We reported here that genetic and pharmacological inhibition of HDAC1 significantly influenced PRV replication. Moreover, we demonstrated that inhibition of HDAC1 induced a DNA damage response and antiviral innate immunity. Mechanistically, the HDAC1 inhibition-induced DNA damage response resulted in the release of double-strand DNA into the cytosol to activate cyclic GMP-AMP synthase and the downstream STING/TBK1/IRF3 innate immune signaling pathway. Our results demonstrate that an HDAC1 inhibitor may be used as a new strategy to prevent Aujeszky's disease in pigs.
Collapse
|
17
|
HBO1 overexpression is important for hepatocellular carcinoma cell growth. Cell Death Dis 2021; 12:549. [PMID: 34039960 PMCID: PMC8155027 DOI: 10.1038/s41419-021-03818-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common primary liver malignancy lacking effective molecularly-targeted therapies. HBO1 (lysine acetyltransferase 7/KAT7) is a member of MYST histone acetyltransferase family. Its expression and potential function in HCC are studied. We show that HBO1 mRNA and protein expression is elevated in human HCC tissues and HCC cells. HBO1 expression is however low in cancer-surrounding normal liver tissues and hepatocytes. In HepG2 and primary human HCC cells, shRNA-induced HBO1 silencing or CRISPR/Cas9-induced HBO1 knockout potently inhibited cell viability, proliferation, migration, and invasion, while provoking mitochondrial depolarization and apoptosis induction. Conversely, ectopic overexpression of HBO1 by a lentiviral construct augmented HCC cell proliferation, migration and invasion. In vivo, xenografts-bearing HBO1-KO HCC cells grew significantly slower than xenografts with control HCC cells in severe combined immunodeficient mice. These results suggest HBO1 overexpression is important for HCC cell progression.
Collapse
|
18
|
KDM3B-ETF1 fusion gene downregulates LMO2 via the WNT/β-catenin signaling pathway, promoting metastasis of invasive ductal carcinoma. Cancer Gene Ther 2021; 29:215-224. [PMID: 33828234 DOI: 10.1038/s41417-021-00301-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 01/11/2021] [Accepted: 01/25/2021] [Indexed: 11/08/2022]
Abstract
Breast cancer is the most common malignancy for women, with invasive ductal carcinoma being the largest subtype of breast cancers, accounting for 75-80% of cases. However, the underlying mechanism of invasive ductal carcinoma remains unclear. In this study, we investigate the possible effects KDM3B-ETF1 fusion gene has on breast cancer cell metastasis, invasion and its downstream signaling mediators as revealed from RNA sequence data analysis. As predicted, KDM3B-ETF1 expression was increased in breast cancer tissues and cells. Overexpression of KDM3B-ETF1 in cancer cell lines promoted the growth and invasion of breast cancer cells, while KDM3B-ETF1 knockdown showed the opposite effects on malignant cell growth and invasion both in vivo and in vitro as evidenced by cell counting kit-8, Transwell assay and tumor xenograft in nude mice. On the contrary, LIM Domain Only 2 (LMO2) expression was significantly reduced in breast cancer tissues and cells. According to chromatin immunoprecipitation and Western blot analysis, KDM3B-ETF1 targets LMO2 and reduced the expression of LMO2, leading to an increase in WNT/β-catenin signaling pathway and thus promoting invasion. In conclusion, fusion gene KDM3B-ETF1 inhibits LMO2, activates the Wnt/β-catenin signaling pathway that leads to increased breast cancer cell invasion and metastasis, providing a novel insight into developing therapeutic strategies. These results provide novel insights into the molecular mechanism of invasive ductal carcinomas, which may lead to potential therapeutic targets.
Collapse
|
19
|
A novel DNA methylation-based model that effectively predicts prognosis in hepatocellular carcinoma. Biosci Rep 2021; 41:227938. [PMID: 33634306 PMCID: PMC7955104 DOI: 10.1042/bsr20203945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To build a novel predictive model for hepatocellular carcinoma (HCC) patients based on DNA methylation data. METHODS Four independent DNA methylation datasets for HCC were used to screen for common differentially methylated genes (CDMGs). Gene Ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to explore the biological roles of CDMGs in HCC. Univariate Cox analysis and least absolute shrinkage and selection operator (LASSO) Cox analysis were performed to identify survival-related CDMGs (SR-CDMGs) and to build a predictive model. The importance of this model was assessed using Cox regression analysis, propensity score-matched (PSM) analysis and stratification analysis. A validation group from the Cancer Genome Atlas (TCGA) was constructed to further validate the model. RESULTS Four SR-CDMGs were identified and used to build the predictive model. The risk score of this model was calculated as follows: risk score = (0.01489826 × methylation level of WDR69) + (0.15868618 × methylation level of HOXB4) + (0.16674959 × methylation level of CDKL2) + (0.16689301 × methylation level of HOXA10). Kaplan-Meier analysis demonstrated that patients in the low-risk group had a significantly longer overall survival (OS; log-rank P-value =0.00071). The Cox model multivariate analysis and PSM analysis identified the risk score as an independent prognostic factor (P<0.05). Stratified analysis results further confirmed this model performed well. By analyzing the validation group, the results of receiver operating characteristic (ROC) curve analysis and survival analysis further validated this model. CONCLUSION Our DNA methylation-based prognosis predictive model is effective and reliable in predicting prognosis for patients with HCC.
Collapse
|
20
|
Ma T, Yan B, Hu Y, Zhang Q. HOXA10 promotion of HDAC1 underpins the development of lung adenocarcinoma through the DNMT1-KLF4 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:71. [PMID: 33596966 PMCID: PMC7891037 DOI: 10.1186/s13046-021-01867-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Previous research has highlighted the ability of Homeobox A10 (HOXA10) to the promote proliferation, migration, and epithelial-mesenchymal transformation of various cancers, including lung adenocarcinoma (LAD), which is characterized by an aggressive disease course that exhibits rapid proliferation and migration, with studies suggesting histone deacetylase 1 (HDAC1) to be a downstream mediator of HOXA10. The current study aimed to investigate the mechanism by which HOXA10-mediated HDAC1 influences the development of LAD. Methods The expression patterns of HOXA10, HDAC1, DNA methyltransferase 1 (DNMT1), and Kruppel-like factor 4 (KLF4) were determined. Additionally, the effect of HOXA10, HDAC1, or DNMT1 on invasive phenotypes of LAD was analyzed using depletion experiments. The interactions among HOXA10, HDAC1, DNMT1, and KLF4 were evaluated via chromatin immunoprecipitation, dual luciferase assay or co-immunoprecipitation. Furthermore, the tumorigenic ability of the LAD cells following HOXA10 silencing and/or HDAC1 overexpression in vivo was also investigated. Results In the LAD tissues and cells, HOXA10, HDAC1, and DNMT1 all exhibited high levels of expression, while KLF4 was poorly expressed. HOXA10 silencing inhibited the expression of HDAC1, reduced LAD cell proliferation, migration, and invasion, and promoted the apoptosis. HDAC1 promoted DNMT1 expression through deacetylation, and DNMT1 inhibited the KLF4 expression through DNA methyltransferase. The in vitro findings were further attested through the use of in vivo assays. Conclusion Taken together, the key observations of the current study highlight the role of HOXA10 and HDAC1 in promoting the proliferation and migration of LAD cells. HOXA10-induced upregulation of HDAC1 interacts with DNMT1-KLF4 axis, while the inhibition of HOXA10 or HDAC1 represents a promising anti-tumor therapy target for LAD. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01867-0.
Collapse
Affiliation(s)
- Tiangang Ma
- Department of Respiratory and Critical Care Medicine, the 2nd Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130041, P.R. China
| | - Bingdi Yan
- Department of Respiratory and Critical Care Medicine, the 2nd Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130041, P.R. China
| | - Yanbing Hu
- Department of Ultrasound, the 2nd Hospital of Jilin University, Changchun, 130041, P.R. China
| | - Qinghua Zhang
- Department of Respiratory and Critical Care Medicine, the 2nd Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130041, P.R. China.
| |
Collapse
|
21
|
Song C, Zhou C. HOXA10 mediates epithelial-mesenchymal transition to promote gastric cancer metastasis partly via modulation of TGFB2/Smad/METTL3 signaling axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:62. [PMID: 33563300 PMCID: PMC7874610 DOI: 10.1186/s13046-021-01859-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/28/2021] [Indexed: 01/06/2023]
Abstract
Background Homeobox A10 (HOXA10) belongs to the HOX gene family, which plays an essential role in embryonic development and tumor progression. We previously demonstrated that HOXA10 was significantly upregulated in gastric cancer (GC) and promoted GC cell proliferation. This study was designed to investigate the role of HOXA10 in GC metastasis and explore the underlying mechanism. Methods Immunohistochemistry (IHC) was used to evaluate the expression of HOXA10 in GC. In vitro cell migration and invasion assays as well as in vivo mice metastatic models were utilized to investigate the effects of HOXA10 on GC metastasis. GSEA, western blot, qRT-PCR and confocal immunofluorescence experiments preliminarily analyzed the relationship between HOXA10 and EMT. ChIP-qPCR, dual-luciferase reporter (DLR), co-immunoprecipitation (CoIP), colorimetric m6A assay and mice lung metastasis rescue models were performed to explore the mechanism by which HOXA10 accelerated the EMT process in GC. Results In this study, we demonstrated HOXA10 was upregulated in GC patients and the difference was even more pronounced in patients with lymph node metastasis (LNM) than without. Functionally, HOXA10 promoted migration and invasion of GC cells in vitro and accelerated lung metastasis in vivo. EMT was an important mechanism responsible for HOXA10-involved metastasis. Mechanistically, we revealed HOXA10 enriched in the TGFB2 promoter region, promoted transcription, increased secretion, thus triggered the activation of TGFβ/Smad signaling with subsequent enhancement of Smad2/3 nuclear expression. Moreover, HOXA10 upregulation elevated m6A level and METTL3 expression in GC cells possible by regulating the TGFB2/Smad pathway. CoIP and ChIP-qPCR experiments demonstrated that Smad proteins played an important role in mediating METTL3 expression. Furthermore, we found HOXA10 and METTL3 were clinically relevant, and METTL3 was responsible for the HOXA10-mediated EMT process by performing rescue experiments with western blot and in vivo mice lung metastatic models. Conclusions Our findings indicated the essential role of the HOXA10/TGFB2/Smad/METTL3 signaling axis in GC progression and metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01859-0.
Collapse
Affiliation(s)
- Chenlong Song
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongzhi Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Circular RNA HIPK3 exacerbates diabetic nephropathy and promotes proliferation by sponging miR-185. Gene 2020; 765:145065. [PMID: 32889056 DOI: 10.1016/j.gene.2020.145065] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of the present study was to investigate expression levels of circular RNA HIPK3 (circHIPK3) in mice with diabetic nephropathy (DN) and the role of circHIPK3 in rat mesangial cells (MCs). METHODS Quantitative real-time polymerase chain reaction was performed to detect expression levels of circHIPK3, miR-185, cyclin D1, proliferating cell nuclear antigen (PCNA), transforming growth factor-β1 (TGF-β1), collagen Ⅰ (Col. Ⅰ), and fibronectin (FN) in mice with DN and rat mesangial cells. Luciferase assay was performed to investigate the binding sites of circHIPK3 and miR-185. Silencing cells of circHIPK3 and miR-185 were constructed using cell transfection assay. RESULTS Our results revealed that the levels of 24-hour urinary albumin and urinary 8-hydroxy-2'-deoxyguanosine (8-OH-dG) from diabetic mice increased considerably. Up-regulation of circHIPK3 was observed in the renal tissues of mice with DN. Similarly, circHIPK3 expression in rat mesangial cells increased significantly in a microenvironment of high glucose. A loss-of-function experiment indicated that down-regulation of circHIPK3 inhibited cell proliferation and significantly decreased mRNA abundance of cyclin D1, PCNA, TGF-β1, Col. I, and FN in MCs. Luciferase assay demonstrated that circHIPK3 can specifically sponge miR-185, and silencing of miR-185 can reverse the effects of knocking down circHIPK3 on cell proliferation and mRNA abundance of cyclin D1, PCNA, TGF-β1, Col. I, and FN in MCs. CONCLUSION Overall, circHIPK3 exhibits a promotive function in DN by sponging miR-185 and this evidence suggests that circHIPK3 might be a biomarker or therapeutic target for DN.
Collapse
|
23
|
Huo X, Qi J, Huang K, Bu S, Yao W, Chen Y, Nie J. Identification of an autophagy-related gene signature that can improve prognosis of hepatocellular carcinoma patients. BMC Cancer 2020; 20:771. [PMID: 32807131 PMCID: PMC7433127 DOI: 10.1186/s12885-020-07277-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022] Open
Abstract
Background Autophagy is a programmed cell degradation mechanism that has been associated with several physiological and pathophysiological processes, including malignancy. Improper induction of autophagy has been proposed to play a pivotal role in the progression of hepatocellular carcinoma (HCC). Methods Univariate Cox regression analysis of overall survival (OS) was performed to identify risk-associated autophagy-related genes (ARGs) in HCC data set from The Cancer Genome Atlas (TCGA). Multivariate cox regression was then performed to develop a risk prediction model for the prognosis of 370 HCC patients. The multi-target receiver operating characteristic (ROC) curve was used to determine the model’s accuracy. Besides, the relationship between drug sensitivity and ARGs expression was also examined. Results A total of 62 differentially expressed ARGs were identified in HCC patients. Univariate and multivariate regression identified five risk-associated ARGs (HDAC1, RHEB, ATIC, SPNS1 and SQSTM1) that were correlated with OS in HCC patients. Of importance, the risk-associated ARGs were independent risk factors in the multivariate risk model including clinical parameters such as malignant stage (HR = 1.433, 95% CI = 1.293–1.589, P < 0.001). In addition, the area under curve for the prognostic risk model was 0.747, which indicates the high accuracy of the model in prediction of HCC outcomes. Interestingly, the risk-associated ARGs were also correlated with drug sensitivity in HCC cell lines. Conclusion We developed a novel prognostic risk model by integrating the molecular signature and clinical parameters of HCC, which can effectively predict the outcomes of HCC patients.
Collapse
Affiliation(s)
- Xingxing Huo
- University of Science and Technology of China, Hefei, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.,Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Jian Qi
- University of Science and Technology of China, Hefei, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Kaiquan Huang
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Su Bu
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Wei Yao
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Ying Chen
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Jinfu Nie
- University of Science and Technology of China, Hefei, China. .,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China. .,Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.
| |
Collapse
|
24
|
Li J, Ye M, Zhou C. Expression Profile and Prognostic Values of HOXA Family Members in Laryngeal Squamous Cell Cancer. Front Oncol 2020; 10:368. [PMID: 32296636 PMCID: PMC7136465 DOI: 10.3389/fonc.2020.00368] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
The homeobox A cluster (HOXA) gene family, comprising 11 members, is involved in a wide spectrum of biological functions in human cancers. However, there is little research on the expression profile and prognostic values of HOXA genes in laryngeal squamous cell cancer (LSCC). Based on updated public resources and integrative bioinformatics analysis, we assessed the expression profile and prognostic values of the HOXA family members. Expression and methylation data on HOXA family members were obtained from The Cancer Genome Atlas (TCGA). The prognostic values of HOXA members and clinical features were identified. A gene set enrichment analysis (GSEA) was conducted to explore the mechanism underlying the involvement of HOXA members in LSCC. The associations between tumor immune infiltrating cells (TIICs) and the HOXA family members were evaluated using the Tumor Immune Estimation Resource (TIMER) database. HOXA2 and HOXA4 were downregulated and HOXA7 and HOXA9–13 were upregulated in LSCC. Upregulation of HOXA10, HOXA11, and HOXA13, along with two clinical characteristics (M stage and gender), were associated with a poor LSCC prognosis based on the results of univariate and multivariate Cox proportional hazards regression analyses. Although there were no significant correlations between TIICs and HOXA members, the GSEA results indicated that HOXA members participate in multiple biological processes underlying tumorigenesis. This study comprehensively analyzed the HOXA members, providing insights for further investigation of the HOXA family members as potential targets in LSCC.
Collapse
Affiliation(s)
- Jinyun Li
- Department of Oncology and Hematology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Meng Ye
- Department of Oncology and Hematology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|