1
|
Maliko M, Su FH, Kamiza AB, Su MJ, Yeh CC. The association between hepatic viral infections and cancers: a cross-sectional study in the Taiwan adult population. Clin Exp Med 2024; 24:20. [PMID: 38279980 PMCID: PMC10821961 DOI: 10.1007/s10238-023-01292-x] [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: 10/04/2023] [Accepted: 12/28/2023] [Indexed: 01/29/2024]
Abstract
BACKGROUND Hepatitis B (HBV) and hepatitis C (HCV) viruses are diseases of global public health concern and are associated with liver cancer. Recent studies have revealed associations between hepatic viral infections and extrahepatic cancers. This study aimed to explore the associations between hepatitis B and C viruses and cancer at baseline in the Taiwan Biobank database while controlling for a wide range of confounding variables. METHODS In a cross-sectional study of adults aged > 20 years, we compared the distribution of demographic factors, lifestyle, and comorbidities between viral and nonviral hepatic groups using the chi-square test. Univariate and multivariate logistic regressions were performed to observe the associations between hepatitis B and C viral infections and cancers by estimating the odds ratio (OR) and 95% confidence interval (CI). Multivariate regression analysis was adjusted for sociodemographic factors, lifestyle, and comorbidities. RESULTS From the database, 2955 participants were identified as having HCV infection, 15,305 as having HBV infection, and 140,108 as the nonviral group. HBV infection was associated with an increased likelihood of liver cancer (adjusted OR (aOR) = 6.60, 95% CI = 3.21-13.57, P < 0.001) and ovarian cancer (aOR = 4.63, 95% CI = 1.98-10.83, P = 0.001). HCV infection was observed to increase the likelihood of liver cancer (aOR = 4.90, 95% CI = 1.37-17.53, P = 0.015), ovarian cancer (aOR = 8.50, 95% CI = 1.78-40.69, P = 0.007), and kidney cancer (aOR = 12.89, 95% CI = 2.41-69.01, P = 0.003). CONCLUSION Our findings suggest that hepatic viral infections are associated with intra- and extrahepatic cancers. However, being cross-sectional, causal inferences cannot be made. A recall-by-genotype study is recommended to further investigate the causality of these associations.
Collapse
Affiliation(s)
- Moreen Maliko
- School of Public Health, College of Public Health, Taipei Medical University, 10F Biomedical Technology Building, No.301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235603, Taiwan
| | - Fu-Hsiung Su
- Department of Family Medicine, Cardinal Tien Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Abram Bunya Kamiza
- Faculty of Health Sciences, Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- The African Computational Genomic (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda
| | - Ming-Jang Su
- School of Public Health, College of Public Health, Taipei Medical University, 10F Biomedical Technology Building, No.301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235603, Taiwan
- Department of Clinical Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235041, Taiwan
| | - Chih-Ching Yeh
- School of Public Health, College of Public Health, Taipei Medical University, 10F Biomedical Technology Building, No.301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235603, Taiwan.
- Department of Public Health, College of Public Health, China Medical University, Taichung, 406, Taiwan.
- Cancer Center, Wan Fang Hospital, Taipei Medical University, New Taipei City, 116, Taiwan.
- Master Program in Applied Epidemiology, College of Public Health, Taipei Medical University, New Taipei City, 235603, Taiwan.
| |
Collapse
|
2
|
Qin Y, Ni P, Zhang Q, Wang X, Du X, Yin Z, Wang L, Ye L, Chen L. Hbxip is essential for murine embryogenesis and regulates embryonic stem cell differentiation through activating mTORC1. Development 2022; 149:275503. [DOI: 10.1242/dev.200527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/09/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
HBXIP, also named LAMTOR5, has been well characterized as a transcriptional co-activator in various cancers. However, the role of Hbxip in normal development remains unexplored. Here, we demonstrated that homozygous knockout of Hbxip leads to embryonic lethality, with retarded growth around E7.5, and that depletion of Hbxip compromises the self-renewal of embryonic stem cells (ESCs), with reduced expression of pluripotency genes, reduced cell proliferation and decreased colony-forming capacity. In addition, both Hbxip−/− ESCs and E7.5 embryos displayed defects in ectodermal and mesodermal differentiation. Mechanistically, Hbxip interacts with other components of the Ragulator complex, which is required for mTORC1 activation by amino acids. Importantly, ESCs depleted of Ragulator subunits, Lamtor3 or Lamtor4, displayed differentiation defects similar to those of Hbxip−/− ESCs. Moreover, Hbxip−/−, p14−/− and p18−/− mice, lacking subunits of the Ragulator complex, also shared similar phenotypes, embryonic lethality and retarded growth around E7-E8. Thus, we conclude that Hbxip plays a pivotal role in the development and differentiation of the epiblast, as well as the self-renewal and differentiation of ESCs, through activating mTORC1 signaling.
Collapse
Affiliation(s)
- Yan Qin
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Peiling Ni
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Qingye Zhang
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Xiao Wang
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Xiaoling Du
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Zixi Yin
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Lingling Wang
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Lihong Ye
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| | - Lingyi Chen
- Institute of Translational Medicine, Tianjin Union Medical Center, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University , Tianjin 300071 , China
| |
Collapse
|
3
|
Zaniani NR, Oroujalian A, Valipour A, Peymani M. LAMTOR5 expression level is a biomarker for colorectal cancer and lncRNA LAMTOR5-AS1 predicting miRNA sponging effect. Mol Biol Rep 2021; 48:6093-6101. [PMID: 34374893 DOI: 10.1007/s11033-021-06623-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Strong evidence indicated that high expression of HBXIP (also known as LAMTOR5) promotes cancer cells proliferation and helps cancer progression. Long non-coding RNAs (lncRNA) have also a crucial role in developing cancer. In this study, we aimed to determine the expression of LAMTOR5 and its nearby lncRNA, LAMTOR5-AS1 and investigate their potential as a biomarker in colorectal cancer (CRC) patients. METHODS 75 tissues of colorectal tumors and non-tumor adjacent normal sampled in this study. After RNA procedure then RT-qPCR was applied for expression analysis. Moreover, in silico investigation also enrolled for predicting sponging effect of lncRNA with miRNAs. RESULTS LAMTOR5 transcription level significantly overexpressed (p value < 0.001) and has shown a diagnostic potential (AUC = 0.8) in CRC. LAMTOR5-AS1 did not indicate any remarkable expression change overall, but showed a significant overexpressed in elderly patients (> 60) with CRC (p value < 0.0097). Moreover, the correlation analysis between LAMTOR5 and LAMTOR5-AS1 revealed a significant association in CRC (p value = 0.0074) which can be partly explained by its predicting act as a mediator with sponging effect on hsa-miR-let-7b-3p and hsa-miR-20a-5p. CONCLUSION LAMTOR5 gene can be considered as prognostic biomarker for CRC. LAMTOR5-AS5 which is a nearby lncRNA of this gene could play a regulatory impact through its sponging effect on hsa-miR-let-7b-3p and hsa-miR-20a-5p which both have shown a significant impact on overall survival rate in CRC patients in high expression levels.
Collapse
Affiliation(s)
- Najmeh Riahi Zaniani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Andisheh Oroujalian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Ali Valipour
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| |
Collapse
|
4
|
Choe JW, Hyun JJ, Kim B, Han KD. Influence of Metabolic Syndrome on Cancer Risk in HBV Carriers: A Nationwide Population Based Study Using the National Health Insurance Service Database. J Clin Med 2021; 10:jcm10112401. [PMID: 34072289 PMCID: PMC8198770 DOI: 10.3390/jcm10112401] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Hepatitis B virus (HBV) infection and metabolic syndrome (MS) are known independent risk factors for hepatocellular carcinoma (HCC) and other extrahepatic organ malignancies. The purpose of this study was to investigate whether MS and HBV have synergistic effects on cancers and to examine whether increasing the number of MS components could lead to higher risk of cancer development. MATERIALS AND METHODS We evaluated data from 1,504,880 HBV-infected adults who underwent a regular HCC screening program provided by the Korean National Health Insurance Service between 2009 and 2016. RESULTS The prevalence of MS in Korean HBV patients was 38.7% (582,449/1,504,880). Among individuals with HBV infection, the presence of MS was associated with an increased risk for the majority of malignancies except for HCC (HR = 0.862, p-value < 0.05). The presence of a higher number of MS components was associated with a significantly increased risk of developing cancers in most organs; only HCC was negatively associated with an increasing number of MS components (p < 0.01). CONCLUSIONS Our data show that the presence of MS increases the risk for most malignancies, excluding HCC. Moreover, we found that as the number of MS components increased, the risk for most cancers also increased; this trend was reversed in HCC.
Collapse
Affiliation(s)
- Jung Wan Choe
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan 15355, Korea;
| | - Jong Jin Hyun
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan 15355, Korea;
- Correspondence: (J.J.H.); (K.-D.H.)
| | - Bongseong Kim
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Kyung-Do Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Correspondence: (J.J.H.); (K.-D.H.)
| |
Collapse
|
5
|
Zhang J, Sun B, Ruan X, Hou X, Zhi J, Meng X, Zheng X, Gao M. Oncoprotein HBXIP promotes tumorigenesis through MAPK/ERK pathway activation in non-small cell lung cancer. Cancer Biol Med 2021; 18:105-119. [PMID: 33628588 PMCID: PMC7877173 DOI: 10.20892/j.issn.2095-3941.2020.0098] [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: 03/09/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: The oncoprotein, hepatitis B X-interacting protein (HBXIP), has been reported to play an important role in human malignancies. However, its functions in non-small cell lung cancer (NSCLC) are poorly understood. The goal of the present study was to identify the role of HBXIP in the regulation of NSCLC development. Methods: The level of HBXIP expression in NSCLC tissue was assessed by immunohistochemical and Western blot analyses, and its relationships with clinicopathological features and outcomes were statistically evaluated. The effects of HBXIP on NSCLC cell progression were assessed through cell viability, colony formation, and flow cytometry analyses in vitro. The mechanism by which HBXIP regulated the MAPK pathway was studied by Western blot, immunofluorescence, and immunoprecipitation assays. In addition, in vivo experiments were performed to evaluate the progression of NSCLC and ERK signaling pathway activation after HBXIP knockdown. Results: HBXIP was overexpressed in human NSCLC and was correlated with the invasiveness of NSCLC. The high expression of HBXIP in NSCLC was significantly correlated with gender (P = 0.033), N stage (P = 0.002), and tumor-node-metastasis stage (P = 0.008). In vitro experiments using an NSCLC cell line revealed that HBXIP knockdown resulted in the suppression of cell proliferation and colony formation, which was consistent with the enhanced cell cycle arrest in G1 phase. The results of a mechanistic investigation suggested that binding of HBXIP to MEK1 protein promoted MAPK/ERK signaling pathway activation in NSCLC by preventing the proteasome-mediated degradation of MEK1. In addition, the results obtained using in vivo subcutaneous tumor xenografts confirmed that HBXIP deficiency decreased MEK1 protein levels and NSCLC tumor growth. Conclusions: Taken together, our results showed that the HBXIP-MEK interaction promoted oncogenesis via the MAPK/ERK pathway, which may serve as a novel therapeutic target for cancers in which MAPK/ERK signaling is a dominant feature.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Breast Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bei Sun
- Department of Outpatient Office, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xianhui Ruan
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiukun Hou
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jingtai Zhi
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiangrui Meng
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ming Gao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
6
|
Darvishian M, Butt ZA, Wong S, Yoshida EM, Khinda J, Otterstatter M, Yu A, Binka M, Rossi C, McKee G, Pearce M, Alvarez M, Wong J, Cook D, Grennan T, Buxton J, Tyndall M, Woods R, Krajden M, Bhatti P, Janjua NZ. Elevated risk of colorectal, liver, and pancreatic cancers among HCV, HBV and/or HIV (co)infected individuals in a population based cohort in Canada. Ther Adv Med Oncol 2021; 13:1758835921992987. [PMID: 33633801 PMCID: PMC7887683 DOI: 10.1177/1758835921992987] [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: 09/02/2020] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Introduction: Studies of the impact of hepatitis C virus (HCV), hepatitis B virus (HBV) and HIV mono and co-infections on the risk of cancer, particularly extra-hepatic cancer, have been limited and inconsistent in their findings. Methods: In the British Columbia Hepatitis Testers Cohort, we assessed the risk of colorectal, liver, and pancreatic cancers in association with HCV, HBV and HIV infection status. Using Fine and Gray adjusted proportional subdistribution hazards models, we assessed the impact of infection status on each cancer, accounting for competing mortality risk. Cancer occurrence was ascertained from the BC Cancer Registry. Results: Among 658,697 individuals tested for the occurrence of all three infections, 1407 colorectal, 1294 liver, and 489 pancreatic cancers were identified. Compared to uninfected individuals, the risk of colorectal cancer was significantly elevated among those with HCV (Hazard ration [HR] 2.99; 95% confidence interval [CI] 2.55–3.51), HBV (HR 2.47; 95% CI 1.85–3.28), and HIV mono-infection (HR 2.30; 95% CI 1.47–3.59), and HCV/HIV co-infection. The risk of liver cancer was significantly elevated among HCV and HBV mono-infected and all co-infected individuals. The risk of pancreatic cancer was significantly elevated among individuals with HCV (HR 2.79; 95% CI 2.01–3.70) and HIV mono-infection (HR 2.82; 95% CI 1.39–5.71), and HCV/HBV co-infection. Discussion/Conclusion: Compared to uninfected individuals, the risk of colorectal, pancreatic and liver cancers was elevated among those with HCV, HBV and/or HIV infection. These findings highlight the need for targeted cancer prevention and diligent clinical monitoring for hepatic and extrahepatic cancers in infected populations.
Collapse
Affiliation(s)
- Maryam Darvishian
- BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Zahid A Butt
- School of Public Health and Health Systems, University of Waterloo, Waterloo, ON, Canada
| | - Stanley Wong
- BC Centre for Disease Control, Vancouver, Canada
| | | | | | | | - Amanda Yu
- BC Centre for Disease Control, Vancouver, Canada
| | | | | | - Geoff McKee
- University of British Columbia, Vancouver, Canada
| | - Margo Pearce
- BC Centre for Disease Control, Vancouver, Canada
| | | | - Jason Wong
- BC Centre for Disease Control, Vancouver, Canada
| | - Darrel Cook
- BC Centre for Disease Control, Vancouver, Canada
| | - Troy Grennan
- BC Centre for Disease Control, Vancouver, Canada
| | - Jane Buxton
- BC Centre for Disease Control, Vancouver, Canada
| | - Mark Tyndall
- University of British Columbia, Vancouver, Canada
| | - Ryan Woods
- Cancer Control Research, BC Cancer Research Centre, Vancouver, Canada
| | - Mel Krajden
- BC Centre for Disease Control, Vancouver, Canada
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer Research Centre, Vancouver, Canada
| | | |
Collapse
|
7
|
Schweer D, McCorkle JR, Rohr J, Tsodikov OV, Ueland F, Kolesar J. Mithramycin and Analogs for Overcoming Cisplatin Resistance in Ovarian Cancer. Biomedicines 2021; 9:70. [PMID: 33445667 PMCID: PMC7828137 DOI: 10.3390/biomedicines9010070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is a highly deadly malignancy in which recurrence is considered incurable. Resistance to platinum-based chemotherapy bodes a particularly abysmal prognosis, underscoring the need for novel therapeutic agents and strategies. The use of mithramycin, an antineoplastic antibiotic, has been previously limited by its narrow therapeutic window. Recent advances in semisynthetic methods have led to mithramycin analogs with improved pharmacological profiles. Mithramycin inhibits the activity of the transcription factor Sp1, which is closely linked with ovarian tumorigenesis and platinum-resistance. This article summarizes recent clinical developments related to mithramycin and postulates a role for the use of mithramycin, or its analog, in the treatment of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- David Schweer
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology Lexington, University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA; (D.S.); (F.U.)
| | - J. Robert McCorkle
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (J.R.); (O.V.T.)
| | - Jurgen Rohr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (J.R.); (O.V.T.)
| | - Oleg V. Tsodikov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (J.R.); (O.V.T.)
| | - Frederick Ueland
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology Lexington, University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA; (D.S.); (F.U.)
| | - Jill Kolesar
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology Lexington, University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA; (D.S.); (F.U.)
| |
Collapse
|
8
|
Xiu M, Zeng X, Shan R, Wen W, Li J, Wan R. The oncogenic role of HBXIP. Biomed Pharmacother 2020; 133:111045. [PMID: 33378953 DOI: 10.1016/j.biopha.2020.111045] [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: 10/06/2020] [Revised: 11/14/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B X-interacting protein (HBXIP) is a conserved protein of 19 kDa that was originally identified as a binding partner of hepatitis B virus X protein. Emerging evidence indicates that HBXIP is highly expressed in a variety of cancers and is correlated with poor clinical outcomes in cancer patients. HBXIP plays a critical role in cancer progression, but the underlying mechanisms are still unclear. In this review, we primarily focus on publications investigating HBXIP in cancer research, including its expression and clinical significance in cancer patients, its role as a coactivator of transcription factors in cancer cells, its inhibitory effects on the mitochondrial cytochrome c-caspase apoptotic pathway, as well as its roles in promoting mitosis and drug resistance in cancer cells, its regulatory effects on cancer metabolism, and its relationships with other signaling pathways or microRNAs in cancer. This review aims to compile and summarize existing knowledge of the functions of HBXIP in cancer, which provides a comprehensive reference for future studies on the oncogenic mechanisms of HBXIP.
Collapse
Affiliation(s)
- Mengxi Xiu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China; Second Clinical Medical College, Nanchang University, China
| | - Xiaohong Zeng
- Imaging Department, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Renfeng Shan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Wu Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Jianfeng Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Renhua Wan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China.
| |
Collapse
|
9
|
Hong CY, Sinn DH, Kang D, Paik SW, Guallar E, Cho J, Gwak GY. Incidence of extrahepatic cancers among individuals with chronic hepatitis B or C virus infection: A nationwide cohort study. J Viral Hepat 2020; 27:896-903. [PMID: 32340080 DOI: 10.1111/jvh.13304] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/25/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
This study examined the association between chronic HBV or HCV infection and the risk of extrahepatic cancers. A total of 537 103 adults aged ≥20 years without history of cancer were identified from the Korean National Health Insurance Service-National Sample Cohort between 2003 and 2013. The difference in cancer incidence was compared between those with and without chronic HBV or HCV infection. During 3 854 130 person-years of follow-up (median follow-up: 8.0 years), 19 089 participants developed cancer. After adjusting for sex, body mass index, smoking, drinking, income percentile, residential area and comorbidities, hazard ratios (HRs) for incident extrahepatic cancer were significantly higher in participants with chronic HBV infection (HR: 1.27, 95% confidence interval [CI]: 1.20-1.35), HCV infection (HR: 1.31, 95% CI: 1.16-1.48) or HBV/HCV dual infection (HR: 1.41, 95% CI: 1.31-1.72) compared to participants without HBV or HCV infection. In chronic HBV infection, the cancer risk was higher for haematologic malignancy [HR (95% CI) = 2.46 (1.92-3.15)], gallbladder [1.55 (1.05-2.29)], pancreas [1.52 (1.07-2.15)], stomach [1.39 (1.22-1.58)], lung [1.27 (1.04-1.55)], colorectum [1.21 (1.03-1.42)] and thyroid cancer [1.20 (1.05-1.36)]. In chronic HCV infection, the cancer risk was higher for testis [10.34 (1.35-79.78)], gallbladder [2.90 (1.62-5.18)], prostate [2.51 (1.65-3.82)] and thyroid cancer [1.46 (1.10-1.93)]. In conclusion, chronic HBV or HCV infection was not only associated with an increased risk of liver cancer, but also associated with an increased risk of multiple extrahepatic cancers.
Collapse
Affiliation(s)
- Chai Yeong Hong
- Quality Management Division, Intro Biopharma Korea, Gyeonggi-do, South Korea
| | - Dong Hyun Sinn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Danbee Kang
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Seung Woon Paik
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Eliseo Guallar
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea.,Departments of Epidemiology and Medicine and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institution, Baltimore, MD, USA
| | - Juhee Cho
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea.,Departments of Epidemiology and Medicine and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institution, Baltimore, MD, USA
| | - Geum-Youn Gwak
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
10
|
Liu Y, Shao Z, Liao Y, Xia X, Huang C, He J, Hu T, Yu C, Jiang L, Liu J, Huang H. Targeting SKP2/Bcr-Abl pathway with Diosmetin suppresses chronic myeloid leukemia proliferation. Eur J Pharmacol 2020; 883:173366. [PMID: 32679184 DOI: 10.1016/j.ejphar.2020.173366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022]
Abstract
Bcr-Abl is the primary cause as well as currently key therapeutic target of chronic myeloid leukemia (CML). SKP2, an E3 ligase, is a downstream factor of Bcr-Abl to motivate the cell cycle transition of CML and also found to bind and activate Bcr-Abl in reverse. Therefore, SKP2/Bcr-Abl pathway is an attractive target for CML treatment. This study aims to identify an inhibitor of the SKP2/Bcr-Abl pathway based on a large screening of the natural products. We demonstrate that Diosmetin, a kind of phytoestrogens, notably downregulates the expression of SKP2, Bcr-Abl phosphorylation, and moderately downregulates the Bcr-Abl level. Furthermore, Diosmetin displays a favorable anti-tumor activity in CML cells and xenograft models. Collectively, our study reveals a natural compound in the treatment of CML on the basis of SKP2/Bcr-Abl signaling pathway.
Collapse
Affiliation(s)
- Yuan Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenlong Shao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuning Liao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaohong Xia
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chuyi Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jinchan He
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Tumei Hu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Cuifu Yu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lili Jiang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Hongbiao Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
11
|
Wong L, Cheung TH, Yim SF, Lao TT. Prevalence and impact of hepatitis B virus infection in ovarian cancer patients in an endemic area-A retrospective cohort study. J Viral Hepat 2020; 27:520-525. [PMID: 31854060 DOI: 10.1111/jvh.13250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/19/2019] [Accepted: 12/07/2019] [Indexed: 12/13/2022]
Abstract
Hepatitis B virus (HBV) infection is associated with many extrahepatic malignancies, but its association with and impact on ovarian cancer has not been examined. We therefore examined the prevalence of HBV infection among women with primary ovarian carcinoma in an endemic area, and whether this impacts the presentation and survival of these patients. In a retrospective study, we reviewed 523 patients presenting with primary ovarian cancer and known HBV status between 1 January 2006 and 31 December 2017. Patients were divided into HBV-positive and negative groups for the comparison of the patient characteristics and presentation, including staging and histological types, and short term (2 years) mortality from ovarian cancer. Among the 10.1% (53/523) patients screened positive for HBV, more of them presented with advanced staging at FIGO stage 3 or above (OR 1.378, 95% CI 1.063-1.787), although there were no significant differences in patient characteristics. Within 24 months from presentation, there were more deaths due to malignancy in the HBV-positive group (73.3% vs 44.2%, OR 1.659, 95% CI 1.135-2.425). On multivariate analysis after adjusting for nulliparity status, previous use of oestrogens, presence of metastases, histological type (epithelial or others) and grading (high grade or not), whether optimal debulking was performed, and chemotherapy, HBV infection was independently associated with increased death within 24 months of presentation (aOR 2.683, 95% CI 1.015-7.091). In conclusion, the findings of this study suggested an adverse effect of chronic HBV infection on survival within two years of presentation in patients with primary ovarian cancer.
Collapse
Affiliation(s)
- Lo Wong
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tak Hong Cheung
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - So Fan Yim
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Terence T Lao
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
12
|
Qiu L, Lu F, Zhang L, Wang G, Geng R, Miao Y. HBXIP Regulates Gastric Cancer Glucose Metabolism and Malignancy Through PI3K/AKT and p53 Signaling. Onco Targets Ther 2020; 13:3359-3374. [PMID: 32368094 PMCID: PMC7183336 DOI: 10.2147/ott.s243250] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/30/2020] [Indexed: 01/27/2023] Open
Abstract
Introduction Hepatitis B X-interacting protein (HBXIP) overexpression is related to the progression of multiple cancers. However, its role in gastric cancer (GC) remains unclear. Materials and Methods HBXIP expression was determined in human GC specimens and cell lines by quantitative polymerase chain reaction (qRT-PCR) and Western blot. The effects of HBXIP depletion or ectopic expression on GC proliferation were evaluated in vitro using the cell counting kit-8 (CCK-8), 5-ethynyl-2ʹ-deoxyuridine (EdU) incorporation, colony formation, and cell cycle assays. The in vivo effects were investigated using a mouse xenograft model. Apoptosis was evaluated by flow cytometry (in vitro) and immunohistochemistry (IHC; in vivo). Cell migration and invasion were evaluated in vitro using wound healing, transwell migration, and matrigel invasion assays; and in vivo by quantifying distant metastases from injection of GC cells in the lateral tail vein. Results Herein, we reported that HBXIP expression was higher in GC than in normal tissues, and this high expression indicated a poorer prognosis. Gain- and loss-of-function assays showed that HBXIP promoted GC proliferation, migration, and invasion, and inhibited apoptosis. High-performance liquid chromatography (HPLC) quantification of glycolytic metabolites revealed that HBXIP promoted glucose metabolic reprogramming. Investigation of the PI3K/AKT and p53 pathways highlighted their role in this HBXIP-mediated metabolic reprogramming. Conclusion Our results indicate that the up-regulation of HBXIP leads to GC progression by positively regulating glucose metabolism. Therefore, HBXIP is a potential target for the treatment of GC.
Collapse
Affiliation(s)
- Lei Qiu
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Feng Lu
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Lili Zhang
- Emergency Department, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Gang Wang
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| | - Rui Geng
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yongchang Miao
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, People's Republic of China
| |
Collapse
|
13
|
Vellingiri B, Iyer M, Devi Subramaniam M, Jayaramayya K, Siama Z, Giridharan B, Narayanasamy A, Abdal Dayem A, Cho SG. Understanding the Role of the Transcription Factor Sp1 in Ovarian Cancer: from Theory to Practice. Int J Mol Sci 2020; 21:E1153. [PMID: 32050495 PMCID: PMC7038193 DOI: 10.3390/ijms21031153] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 12/23/2022] Open
Abstract
Ovarian cancer (OC) is one of the deadliest cancers among women contributing to high risk of mortality, mainly owing to delayed detection. There is no specific biomarker for its detection in early stages. However, recent findings show that over-expression of specificity protein 1 (Sp1) is involved in many OC cases. The ubiquitous transcription of Sp1 apparently mediates the maintenance of normal and cancerous biological processes such as cell growth, differentiation, angiogenesis, apoptosis, cellular reprogramming and tumorigenesis. Sp1 exerts its effects on cellular genes containing putative GC-rich Sp1-binding site in their promoters. A better understanding of the mechanisms underlying Sp1 transcription factor (TF) regulation and functions in OC tumorigenesis could help identify novel prognostic markers, to target cancer stem cells (CSCs) by following cellular reprogramming and enable the development of novel therapies for future generations. In this review, we address the structure, function, and biology of Sp1 in normal and cancer cells, underpinning the involvement of Sp1 in OC tumorigenesis. In addition, we have highlighted the influence of Sp1 TF in cellular reprogramming of iPSCs and how it plays a role in controlling CSCs. This review highlights the drugs targeting Sp1 and their action on cancer cells. In conclusion, we predict that research in this direction will be highly beneficial for OC treatment, and chemotherapeutic drugs targeting Sp1 will emerge as a promising therapy for OC.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, India; (M.I.); (K.J.)
| | - Mohana Devi Subramaniam
- Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai 600006, India;
| | - Kaavya Jayaramayya
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, India; (M.I.); (K.J.)
| | - Zothan Siama
- Department of Zoology, School of Life-science, Mizoram University, Aizawl 796004, Mizoram, India;
| | - Bupesh Giridharan
- R&D Wing, Sree Balaji Medical College and Hospital (SBMCH), BIHER, Chromepet, Chennai 600044, Tamil Nadu, India;
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India;
| | - Ahmed Abdal Dayem
- Molecular & Cellular Reprogramming Center, Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea;
| | - Ssang-Goo Cho
- Molecular & Cellular Reprogramming Center, Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea;
| |
Collapse
|
14
|
Zhou J, Li Y, Li D, Liu Z, Zhang J. Oncoprotein LAMTOR5 Activates GLUT1 Via Upregulating NF-κB in Liver Cancer. Open Med (Wars) 2019; 14:264-270. [PMID: 30847404 PMCID: PMC6401392 DOI: 10.1515/med-2019-0022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023] Open
Abstract
Objective Accumulating reports reveal that serving as an oncogenic factor LAMTOR5 is involved in the progression of many specific cancers. Glucose transporter 1 (GLUT1) is frequently identified in many cancers. However, it remains unexplored whether GLUT1 plays a role in LAMTOR5-enhanced liver cancer. Here, we aim to decipher the function of LAMTOR5 in the regulation of GLUT1 in liver cancer. Methods The effect of LAMTOR5 on GLUT1 was analyzed using Western blotting and RT-PCR assay. Dose-increased over-expression or silencing of LAMTOR5 was performed through transient transfection. LAMTOR5-activated GLUT1 promoter was revealed by luciferase reporter assay. The regulation of GLUT1 by LAMTOR5/NF-κB was examined via Western blotting and luciferase reporter assays. Results The data showed that in liver cancer cells under the administration with dose-increased LAMTOR5, the level of mRNA and protein of GLUT1 was obviously raised. Our data revealed that the activities of GLUT1 promoter were induced by LAMTOR5. Then, we found that the elevation of GLUT 1 mediated by LAMTOR5 slowed when the inhibitor or siRNAs of NF-κB was introduced into the liver cancer cells. Conclusion. LAMTOR5 is responsible for the activation of GLUT1 via transcription factor NF-κB in liver cancer.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Endocrinology, General Hospital of Tianjin Medical University, Tianjin 300052, Tianjin, China
| | - Yajun Li
- Department of Internal Medicine, Tsinghua University Hospital, Beijing 100084, Beijing, China
| | - Danhua Li
- Graduate Admission Office Tsinghua University, Tsinghua University, Beijing 100084, Beijing, China
| | - Zhi Liu
- Department of Radiology, Affiliated Hospital of North China University of Science and Technology, Tangshan 063000, Tangshan, China
| | - Jie Zhang
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin 300052, Tianjin, China
| |
Collapse
|
15
|
Jiang Y, Wang D, Ren H, Shi Y, Gao Y. Oncogenic HBXIP enhances ZEB1 through Sp1 to accelerate breast cancer growth. Thorac Cancer 2018; 9:1664-1670. [PMID: 30273966 PMCID: PMC6275833 DOI: 10.1111/1759-7714.12878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/22/2018] [Accepted: 08/25/2018] [Indexed: 12/11/2022] Open
Abstract
Background There is abundant evidence to indicate that HBXIP functions as an oncoprotein and transcription co‐activator during the development and promotion of cancers. In multiple cancers, ZEB1 serves as a transcription activator to regulate gene expression. We explored the roles of ZEB1 in HBXIP‐induced breast cancer growth. Methods HBXIP regulation of ZEB1 was evaluated by reverse transcription PCR and immunoblotting. The stimulation of ZEB1 promoter by HBXIP and/or Sp1 was tested using luciferase reporter gene analysis. The alteration of cell proliferation mediated by HBXIP‐induced ZEB1 was tested using methyl‐thiazolyl‐tetrazolium and 5‐Ethynyl‐2′‐deoxyuridine (EdU) incorporation analysis. ZEB1 and HBXIP expression in human breast cancer tissues was analyzed using quantitative real‐time PCR. The relationship between HBXIP and ZEB1 was confirmed by Pearson's correlation coefficient. Results We observed dose‐dependent upregulation of ZEB1 by HBXIP in breast cancer cells. HBXIP can activate the ZEB1 promoter by interacting with transcription factor Sp1. Cell viability and EdU incorporation analysis showed that HBXIP could drive cell proliferation by enhancing ZEB1 in breast cancer. Using quantitative real‐time PCR, ZEB1 overexpression and a positive relationship between ZEB1 and HBXIP were observed in clinical breast cancer samples. Conclusion Oncogenic HBXIP controls the transcription regulation of ZEB1 by co‐activating Sp1, thereby accelerating breast cancer growth.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dan Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hui Ren
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Shi
- Department of Breast-Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yufei Gao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Li H, Wang Z, Li Y, Fang R, Wang H, Shi H, Zhang X, Zhang W, Ye L. Hepatitis B X-interacting protein promotes the formation of the insulin gene-transcribing protein complex Pdx-1/Neurod1 in animal pancreatic β-cells. J Biol Chem 2017; 293:2053-2065. [PMID: 29259128 DOI: 10.1074/jbc.m117.809582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/04/2017] [Indexed: 12/26/2022] Open
Abstract
The activation of insulin gene transcription depends on multiple nuclear proteins, including the transcription factors PDX-1 and NEUROD1, which form a transcriptional complex. We recently reported that hepatitis B X-interacting protein (HBXIP, also termed LAMTOR5) can modulate glucose metabolism reprogramming in cancer cells. However, the physiological role of HBXIP in the modulation of glucose metabolism in normal tissues is poorly understood. Here, we report that Hbxip is an essential regulator of the effect of the Pdx-1/Neurod1 complex on insulin gene transcription in murine pancreatic β-cells in vitro and in vivo We found that pancreatic β-cell-specific Hbxip-knockout mice displayed higher fasting blood glucose levels and impaired glucose tolerance. Furthermore, Hbxip was involved in the regulation of insulin in the pancreas islets and increased insulin gene expression in rat pancreatic β-cells. Mechanistically, Hbxip stimulated insulin enhancer activity by interacting with Pdx-1 and recruiting Neurod1 to Pdx-1. Functionally, we provide evidence that Hbxip is required for Pdx-1/Neurod1-mediated insulin expression in rat pancreatic β-cells. Collectively, these results indicate that Hbxip is involved in the transcription of insulin by increasing the levels of the Pdx-1/Neurod1 complex in animal pancreatic β-cells. Our finding provides the insight into the mechanism by which Hbxip stimulates the transcription of the insulin gene.
Collapse
Affiliation(s)
- Hang Li
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Zhen Wang
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Yinghui Li
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Runping Fang
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Huawei Wang
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Hui Shi
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Xiaodong Zhang
- Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiying Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| | - Lihong Ye
- From the State Key Laboratory of Medicinal Chemical Biology, Departments of Biochemistry and
| |
Collapse
|
17
|
Wang Y, Li N, Che S, Jin T, Piao J, Liu S, Lin Z. HBXIP suppression reduces cell proliferation and migration and its overexpression predicts poor prognosis in non-small-cell lung cancer. Tumour Biol 2017; 39:1010428317709675. [DOI: 10.1177/1010428317709675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging evidence has demonstrated that the high expression of HBXIP has been correlated with many cancers. With evaluation of the functional role of HBXIP in non-small-cell lung cancer, the primary aim of this study is to investigate the correlation between HBXIP expression and the prognosis of non-small-cell lung cancer patients. The protein levels of HBXIP were detected using western blotting in non-small-cell lung cancer cells. Cell proliferation and migration assays were measured to evaluate the function of HBXIP in non-small-cell lung cancer cells. A total of 120 non-small-cell lung cancer patients with strict follow-up and 60 adjacent non-tumor lung tissues were selected for immunohistochemical staining of the HBXIP protein. The localization of the HBXIP protein was detected in A549 non-small-cell lung cancer cells using immunofluorescence staining. The correlation between HBXIP expression and the clinicopathological features of non-small-cell lung cancer patients was analyzed by a chi-squared and Fisher’s exact test. The overall survival rates of all of the non-small-cell lung cancer patients were calculated using the Kaplan–Meier method, and univariate and multivariate analyses were performed using the Cox proportional hazards regression model. In function, we showed that suppression of HBXIP decreased A549 cell proliferation and migration. HBXIP protein showed a mainly cytoplasmic staining pattern in non-small-cell lung cancer using immunohistochemical staining in paraffin-embedded non-small-cell lung cancer tissues and immunofluorescence staining in A549 cells. The HBXIP protein had strong positive staining in the non-small-cell lung cancer tissues, which was significantly higher than the percentage of adjacent non-tumor tissues. The overexpression of HBXIP was closely correlated with histological grade, clinical stage, lymph node metastasis, and lower overall survival rates of patients with non-small-cell lung cancer. Moreover, multivariate analysis suggested that HBXIP emerged as a significant independent prognostic factor along with clinical stage in patients with non-small-cell lung cancer. In conclusion, a high level of expression of HBXIP is associated with the progression of non-small-cell lung cancer and may be a useful biomarker for poor prognostic evaluation and a potential molecular therapy target for patients with non-small-cell lung cancer.
Collapse
Affiliation(s)
- Yixuan Wang
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Nan Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Shuanlong Che
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
| | - Tiefeng Jin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Junjie Piao
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Shuangping Liu
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Zhenhua Lin
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Yanbian University, Yanji, China
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| |
Collapse
|
18
|
Mou H, Guo P, Li X, Zhang C, Jiang J, Wang L, Wang Q, Yuan Z. Nitidine chloride inhibited the expression of S phase kinase-associated protein 2 in ovarian cancer cells. Cell Cycle 2017; 16:1366-1375. [PMID: 28594256 DOI: 10.1080/15384101.2017.1327490] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Nitidine chloride (NC) has been reported to exert its anti-tumor activity in various types of human cancers. However, the molecular mechanism of NC-mediated tumor suppressive function is largely unclear. In the current study, we used several approaches such as MTT, FACS, RT-PCR, Western blotting analysis, invasion assay, transfection, to explore the molecular basis of NC-triggered anti-cancer activity. We found that NC inhibited cell growth, induced cell apoptosis, caused cell cycle arrest in ovarian cancer cells. Emerging evidence has demonstrated that Skp2 plays an important oncogenic role in ovarian cancer. Therefore, we also explored whether NC exerts its biologic function via downregulation of Skp2 in ovarian cancer cells. We observed that NC significantly inhibited the expression of Skp2 in ovarian cancer cells. Notably, overexpression of Skp2 abrogated the anti-cancer activity induced by NC in ovarian cancer cells. Consistently, downregulation of Skp2 expression enhanced the sensitivity of ovarian cancer cells to NC treatment. Thus, inactivation of Skp2 by NC could be a novel strategy for the treatment of human ovarian cancer.
Collapse
Affiliation(s)
- Huaping Mou
- a Department of Gynecology , Second People Hospital of Sichuan Province , Yibin City , China
| | - Ping Guo
- b Department of Oncology , Second People Hospital of Sichuan Province , Yibin City , China.,c Department of Hematology , First Affiliated Hospital of Southwest Medical University , Sichuan , Luzhou , China
| | - Xiaoming Li
- c Department of Hematology , First Affiliated Hospital of Southwest Medical University , Sichuan , Luzhou , China
| | - Chuanli Zhang
- b Department of Oncology , Second People Hospital of Sichuan Province , Yibin City , China
| | - Jing Jiang
- a Department of Gynecology , Second People Hospital of Sichuan Province , Yibin City , China
| | - Lishuai Wang
- b Department of Oncology , Second People Hospital of Sichuan Province , Yibin City , China
| | - Qiu Wang
- b Department of Oncology , Second People Hospital of Sichuan Province , Yibin City , China
| | - Zhiping Yuan
- b Department of Oncology , Second People Hospital of Sichuan Province , Yibin City , China.,c Department of Hematology , First Affiliated Hospital of Southwest Medical University , Sichuan , Luzhou , China
| |
Collapse
|
19
|
Wang Y, Sun J, Li N, Che S, Jin T, Liu S, Lin Z. HBXIP overexpression is correlated with the clinical features and survival outcome of ovarian cancer. J Ovarian Res 2017; 10:26. [PMID: 28388957 PMCID: PMC5384129 DOI: 10.1186/s13048-017-0322-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/29/2017] [Indexed: 01/13/2023] Open
Abstract
Background Accumulated evidence has demonstrated that Mammalian hepatitis B X-interacting protein (HBXIP) has broad roles in cancer. Although HBXIP is associated with a variety of cancers, the HBXIP protein expression level and its clinical significance in ovarian cancer have not yet been determined. The aim of this study is to investigate the association between HBXIP expression and the clinicopathological features of ovarian cancer patients to determine whether HBXIP may be correlated with a poor prognosis in ovarian cancer patients. Methods HBXIP protein expression was assessed in a well-characterized series of ovarian cancer tissue samples (n = 120) with long-term follow-up, using immunohistochemistry to determine the location pattern and expression of HBXIP in ovarian cancer. The localization of HBXIP was detected in SKOV-3 ovarian cancer cells using immunofluorescence (IF) staining. The relationship between high HBXIP expression and the clinicopathological features of ovarian cancer patients was analyzed by Chi-square and Fisher’s exact test. Overall survival (OS) rates of all the ovarian cancer patients were calculated using the Kaplan-Meier method, and univariate and multivariate analyses were performed using the Cox proportional hazards regression model. Results IF staining revealed strongly positive signals for HBXIP in both cytoplasm and nucleus, but mainly in the cytoplasm of SKOV-3 ovarian cancer cells. High HBXIP expression was predominantly observed in ovarian cancer tissues but not the adjacent non-tumor ovarian tissues. The strongly positive rate of HBXIP expression was 60.0% (72/120) in ovarian cancer and was significantly higher than in adjacent non-tumor tissues (17.4%, 4/23) (P = 0.000). High HBXIP expression was positively correlated with the occurrence of lymph node metastases (P = 0.025), histological grade (P = 0.036) and clinical stage (P = 0.003). The patients with high HBXIP expression had lower overall survival (OS) rates. Moreover, multivariate analysis indicated that HBXIP, in addition to the clinical stage, was a significant independent prognostic factor in patients with ovarian cancer. Conclusions High-level expression of HBXIP is associated with the progression of ovarian cancer and may be an effective biomarker for poor prognostic evaluation as well as a potential molecular therapy target for ovarian cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s13048-017-0322-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yixuan Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Yanbian University, Yanji, 133002, Jilin, China.,Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Jie Sun
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Yanbian University, Yanji, 133002, Jilin, China.,Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Nan Li
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Yanbian University, Yanji, 133002, Jilin, China
| | - Shuanlong Che
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Tiefeng Jin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Shuangping Liu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Yanbian University, Yanji, 133002, Jilin, China. .,Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China.
| | - Zhenhua Lin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Yanbian University, Yanji, 133002, Jilin, China. .,Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, Jilin, China.
| |
Collapse
|
20
|
Zou W, Ma X, Yang H, Hua W, Chen B, Cai G. Hepatitis B X-interacting protein promotes cisplatin resistance and regulates CD147 via Sp1 in ovarian cancer. Exp Biol Med (Maywood) 2017; 242:497-504. [PMID: 28056551 DOI: 10.1177/1535370216685007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is the highest mortality rate of all female reproductive malignancies. Drug resistance is a major cause of treatment failure in malignant tumors. Hepatitis B X-interacting protein acts as an oncoprotein, regulates cell proliferation, and migration in breast cancer. We aimed to investigate the effects and mechanisms of hepatitis B X-interacting protein on resistance to cisplatin in human ovarian cancer cell lines. The mRNA and protein levels of hepatitis B X-interacting protein were detected using RT-PCR and Western blotting in cisplatin-resistant and cisplatin-sensitive tissues, cisplatin-resistant cell lines A2780/CP and SKOV3/CP, and cisplatin-sensitive cell lines A2780 and SKOV3. Cell viability and apoptosis were measured to evaluate cellular sensitivity to cisplatin in A2780/CP cells. Luciferase reporter gene assay was used to determine the relationship between hepatitis B X-interacting protein and CD147. The in vivo function of hepatitis B X-interacting protein on tumor burden was assessed in cisplatin-resistant xenograft models. The results showed that hepatitis B X-interacting protein was highly expressed in ovarian cancer of cisplatin-resistant tissues and cells. Notably, knockdown of hepatitis B X-interacting protein significantly reduced cell viability in A2780/CP compared with cisplatin treatment alone. Hepatitis B X-interacting protein and cisplatin cooperated to induce apoptosis and increase the expression of c-caspase 3 as well as the Bax/Bcl-2 ratio. We confirmed that hepatitis B X-interacting protein up-regulated CD147 at the protein expression and transcriptional levels. Moreover, we found that hepatitis B X-interacting protein was able to activate the CD147 promoter through Sp1. In vivo, depletion of hepatitis B X-interacting protein decreased the tumor volume and weight induced by cisplatin. Taken together, these results indicate that hepatitis B X-interacting protein promotes cisplatin resistance and regulated CD147 via Sp1 in ovarian cancer cell lines. Impact statement We found that hepatitis B X-interacting protein (HBXIP) was able to activate the CD147 promoter through Sp1. In vivo, depletion of HBXIP decreased the tumor volume and weight induced by CP. Taken together, these results indicate that HBXIP promotes cisplatin resistance and regulated CD147 via Sp1 in ovarian cancer cell lines.
Collapse
Affiliation(s)
- Wei Zou
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Xiangdong Ma
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Hong Yang
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Wei Hua
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Biliang Chen
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Guoqing Cai
- Department of Obstetrics and Gynecology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, P.R. China
| |
Collapse
|
21
|
Kamiza AB, Su FH, Wang WC, Sung FC, Chang SN, Yeh CC. Chronic hepatitis infection is associated with extrahepatic cancer development: a nationwide population-based study in Taiwan. BMC Cancer 2016; 16:861. [PMID: 27821099 PMCID: PMC5100218 DOI: 10.1186/s12885-016-2918-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023] Open
Abstract
Background Hepatitis B virus (HBV) and hepatitis C virus (HCV) are the major causes of chronic hepatitis infection (CHI). This longitudinal cohort study investigated the association of CHI with hepatic and extrahepatic cancer development in Taiwan. Methods Patients with HBV infection and HCV infection were identified from the Taiwan National Health Insurance Research Database. A Cox proportional hazard model was used to calculate hazard ratios (HRs) and 95 % confidence intervals (CIs) for determining the association between CHI and cancer development. Results The patients with HBV infection exhibited an increased risk of colorectal cancer (HR: 1.36, 95 % CI: 1.09–1.70), liver cancer (HR: 21.47, 95 % CI: 18.0–25.6), gallbladder and extrahepatic bile duct cancer (HR: 2.05, 95 % CI: 1.07–3.91), pancreatic cancer (HR: 2.61, 95 % CI: 1.47–4.61), kidney cancer (HR: 1.72, 95 % CI: 1.10–2.68), ovarian cancer (HR: 2.31, 95 % CI: 1.21–4.39), and non-Hodgkin’s lymphoma (HR: 2.10, 95 % CI: 1.25–3.52). The patients with HCV infection exhibited an increased risk of liver cancer (HR: 25.10, 95 % CI: 20.9–30.2), gallbladder and extrahepatic bile duct cancer (HR: 2.60, 95 % CI: 1.42–4.73), ovarian cancer (HR: 5.15, 95 % CI: 1.98–13.4), and non-Hodgkin’s lymphoma (HR: 2.30, 95 % CI: 1.34–3.96). Conclusion The present population-based study revealed that in addition to its association with primary liver cancer, CHI is associated with an increased risk of extrahepatic cancer.
Collapse
Affiliation(s)
- Abram Bunya Kamiza
- School of Public Health, College of Public Health and Nutrition, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Fu-Hsiung Su
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 11031, Taiwan.,Department of Family Medicine, Taipei Medical University Hospital, No. 252 Wu-Hsing Street, Taipei, 11031, Taiwan.,Master Program in Long-Term Care, College of Nursing, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 11031, Taiwan.,School of Medicine, Flinders University, Bedford Park, Australia
| | - Wen-Chang Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Fung-Chang Sung
- Management Office for Health Data, China Medical University Hospital, No. 2 Yude Road, Taichung, 40402, Taiwan.,Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Shih-Ni Chang
- Management Office for Health Data, China Medical University Hospital, No. 2 Yude Road, Taichung, 40402, Taiwan.,Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Chih-Ching Yeh
- School of Public Health, College of Public Health and Nutrition, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 11031, Taiwan. .,Department of Public Health, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| |
Collapse
|
22
|
Li X, Liu S. Suppression of HBXIP Reduces Cell Proliferation, Migration and InvasionIn Vitro, and TumorigenesisIn Vivoin Human Urothelial Carcinoma of the Bladder. Cancer Biother Radiopharm 2016; 31:311-316. [PMID: 27831760 DOI: 10.1089/cbr.2016.2038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Xiaogang Li
- Department of Urology, The Affiliated Hospital of YanBian University, Yanbian, China
| | - Shuangping Liu
- Department of Pathology, YanBian University, Yanbian, China
| |
Collapse
|
23
|
Extracellular matrix protein ITGBL1 promotes ovarian cancer cell migration and adhesion through Wnt/PCP signaling and FAK/SRC pathway. Biomed Pharmacother 2016; 81:145-151. [PMID: 27261588 DOI: 10.1016/j.biopha.2016.03.053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 12/15/2022] Open
Abstract
Despite the advances in cancer treatment and the progresses in tumor biological, ovarian cancer remains a bad situation. In current study, we found a novel extracellular matrix protein, ITGBL1, which is highly expressed in ovarian cancer tissues by immunohistochemistry examination. The expression pattern of ITGBL1 in malignant tissues inspired us to investigate its role in ovarian cancer progression. Both loss- and gain-function assays revealed that ITGBL1 could promote ovarian cancer cell migration and adhesion. As it's a secreted protein, we further used recombinant ITGBL1 protein treated cancer cells and found that ITGBL1 promotes cell migration and adhesion in a concentration dependent manner. Furthermore, we found that ITGBL1 not only influences the activity of Wnt/PCP signaling but also affects FAK/src pathway in vitro. Taken together, our results suggest that highly expressed ITGBL1 could promotes cancer cell migration and adhesion in ovarian cancer and as a secreted protein, ITGBL1 might be a novel biomarker for ovarian cancer diagnosis.
Collapse
|
24
|
Uddin S, Bhat AA, Krishnankutty R, Mir F, Kulinski M, Mohammad RM. Involvement of F-BOX proteins in progression and development of human malignancies. Semin Cancer Biol 2016; 36:18-32. [PMID: 26410033 DOI: 10.1016/j.semcancer.2015.09.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/15/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022]
|
25
|
Li Y, Wang Z, Shi H, Li H, Li L, Fang R, Cai X, Liu B, Zhang X, Ye L. HBXIP and LSD1 Scaffolded by lncRNA Hotair Mediate Transcriptional Activation by c-Myc. Cancer Res 2015; 76:293-304. [PMID: 26719542 DOI: 10.1158/0008-5472.can-14-3607] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 09/19/2015] [Indexed: 11/16/2022]
Abstract
c-Myc is regarded as a transcription factor, but the basis for its function remains unclear. Here, we define a long noncoding RNA (lncRNA)/protein complex that mediates the transcriptional activation by c-Myc in breast cancer cells. Among 388 c-Myc target genes in human MCF-7 breast cancer cells, we found that their promoters could be occupied by the oncoprotein HBXIP. We confirmed that the HBXIP expression correlated with expression of the c-Myc target genes cyclin A, eIF4E, and LDHA. RNAi-mediated silencing of HBXIP abolished c-Myc-mediated upregulation of these target genes. Mechanistically, HBXIP interacted directly with c-Myc through the leucine zippers and recruited the lncRNA Hotair along with the histone demethylase LSD1, for which Hotair serves as a scaffold. Silencing of HBXIP, Hotair, or LSD1 was sufficient to block c-Myc-enhanced cancer cell growth in vitro and in vivo. Taken together, our results support a model in which the HBXIP/Hotair/LSD1 complex serves as a critical effector of c-Myc in activating transcription of its target genes, illuminating long-standing questions on how c-Myc drives carcinogenesis.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Zhen Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Hui Shi
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Hang Li
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Leilei Li
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Runping Fang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Xiaoli Cai
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Bowen Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China
| | - Xiaodong Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, Institute for Molecular Biology, College of Life Sciences, Nankai University, Tianjin, P.R. China.
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, P.R. China.
| |
Collapse
|
26
|
Li H, Liu Q, Wang Z, Fang R, Shen Y, Cai X, Gao Y, Li Y, Zhang X, Ye L. The oncoprotein HBXIP modulates the feedback loop of MDM2/p53 to enhance the growth of breast cancer. J Biol Chem 2015; 290:22649-61. [PMID: 26229107 DOI: 10.1074/jbc.m115.658468] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Indexed: 12/26/2022] Open
Abstract
MDM2 and p53 form a negative feedback loop, in which p53 as a transcription factor positively regulates MDM2 and MDM2 negatively regulates tumor suppressor p53 through promoting its degradation. However, the mechanism of the feedback loop is poorly understood in cancers. We had reported previously that the oncoprotein hepatitis B X-interacting protein (HBXIP) is a key oncoprotein in the development of cancer. Thus, we supposed that HBXIP might be involved in the event. Here, we observed that the expression levels of HBXIP were positively correlated to those of MDM2 in clinical breast cancer tissues. Interestingly, HBXIP was able to up-regulate MDM2 at the levels of mRNA and protein in MCF-7 breast cancer cells. Mechanically, HBXIP increased the promoter activities of MDM2 through directly binding to p53 in the P2 promoter of MDM2. Strikingly, we identified that the acetyltransferase p300 was recruited by HBXIP to p53 in the promoter of MDM2. Moreover, we validated that HBXIP enhanced the p53 degradation mediated by MDM2. Functionally, the knockdown of HBXIP or/and p300 inhibited the proliferation of breast cancer cells in vitro, and the depletion of MDM2 or overexpression of p53 significantly blocked the HBXIP-promoted growth of breast cancer in vitro and in vivo. Thus, we concluded that highly expressed HBXIP accelerates the MDM2-mediated degradation of p53 in breast cancer through modulating the feedback loop of MDM2/p53, resulting in the fast growth of breast cancer cells. Our findings provide new insights into the mechanism of the acceleration of the MDM2/p53 feedback loop in the development of cancer.
Collapse
Affiliation(s)
- Hang Li
- From the Department of Biochemistry and
| | - Qian Liu
- From the Department of Biochemistry and
| | - Zhen Wang
- From the Department of Biochemistry and
| | | | - Yu Shen
- From the Department of Biochemistry and
| | | | - Yuen Gao
- the Department of Cancer Research, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | | | - Xiaodong Zhang
- the Department of Cancer Research, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lihong Ye
- From the Department of Biochemistry and
| |
Collapse
|
27
|
Wang B, Xu W, Tan M, Xiao Y, Yang H, Xia TS. Integrative genomic analyses of a novel cytokine, interleukin-34 and its potential role in cancer prediction. Int J Mol Med 2014; 35:92-102. [PMID: 25395235 PMCID: PMC4249750 DOI: 10.3892/ijmm.2014.2001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/03/2014] [Indexed: 12/19/2022] Open
Abstract
Interleukin-34 (IL-34) is a novel cytokine, which is composed of 222 amino acids and forms homodimers. It binds to the macrophage colony-stimulating factor (M-CSF) receptor and plays an important role in innate immunity and inflammatory processes. In the present study, we identified the completed IL-34 gene in 25 various mammalian genomes and found that IL-34 existed in all types of vertebrates, including fish, amphibians, birds and mammals. These species have a similar 7 exon/6 intron gene organization. The phylogenetic tree indicated that the IL-34 gene from the primate lineage, rodent lineage and teleost lineage form a species-specific cluster. It was found mammalian that IL-34 was under positive selection pressure with the identified positively selected site, 196Val. Fifty-five functionally relevant single nucleotide polymorphisms (SNPs), including 32 SNPs causing missense mutations, 3 exonic splicing enhancer SNPs and 20 SNPs causing nonsense mutations were identified from 2,141 available SNPs in the human IL-34 gene. IL-34 was expressed in various types of cancer, including blood, brain, breast, colorectal, eye, head and neck, lung, ovarian and skin cancer. A total of 5 out of 40 tests (1 blood cancer, 1 brain cancer, 1 colorectal cancer and 2 lung cancer) revealed an association between IL-34 gene expression and cancer prognosis. It was found that the association between the expression of IL-34 and cancer prognosis varied in different types of cancer, even in the same types of cancer from different databases. This suggests that the function of IL-34 in these tumors may be multidimensional. The upstream transcription factor 1 (USF1), regulatory factor X-1 (RFX1), the Sp1 transcription factor 1, POU class 3 homeobox 2 (POU3F2) and forkhead box L1 (FOXL1) regulatory transcription factor binding sites were identified in the IL-34 gene upstream (promoter) region, which may be involved in the effects of IL-34 in tumors.
Collapse
Affiliation(s)
- Bo Wang
- Department of Medical Oncology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510700, P.R. China
| | - Wenming Xu
- Department of Endocrinology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510700, P.R. China
| | - Miaolian Tan
- Department of Medical Oncology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510700, P.R. China
| | - Yan Xiao
- Department of Medical Oncology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510700, P.R. China
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Tian-Song Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|