1
|
Chen L, Li W, Zai W, Zheng X, Meng X, Yao Q, Li W, Liang Y, Ye M, Zhou K, Liu M, Yang Z, Mao Z, Wei H, Yang S, Shi G, Yuan Z, Yu W. HBV sequence integrated to enhancer acting as oncogenic driver epigenetically promotes hepatocellular carcinoma development. J Exp Clin Cancer Res 2025; 44:155. [PMID: 40405227 PMCID: PMC12096768 DOI: 10.1186/s13046-025-03413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND HBV integration is considered as the main contributor to hepatocellular carcinoma (HCC). However, whether HBV integrated sequences determine genotype pathogenicity and how to block their function during HCC progression remains unclear. METHODS An in vitro HBV-infected PHH model and liver cancer cell lines were established to confirm the pathogenic potential of HBV-SITEs. The roles of HBV-SITE-1 in HCC development were analyzed using cellular phenotypic assays and molecular biology techniques, including the combined analysis of RNA-seq and ChIP-seq. Animal models were also used to evaluate the therapeutic effect of HBV-miR-2 inhibitors. RESULTS We identified nine fragments of HBV Sequences Integrated To Enhancer, termed as "HBV-SITEs". Particularly, a single nucleotide variation (T > G) was embedded at seed sequence of HBV-miR-2 in the highest integrated HBV-SITE-1 between genotypes B and H. Unexpectedly, B-HBV-SITE-1, not H-HBV-SITE-1, could abnormally activate oncogenic genes including TERT and accelerate HCC cell proliferation and migration. Meanwhile, HBV-miR-2 was gradually increased in HBV-infected cells and patient plasma with different HCC stages. Importantly, 227 genes upregulated by HBV, were also activated by HBV-miR-2 through triggering HBV-SITE-1 enhancer. Conversely, enhancer activities were particularly decreased by HBV-miR-2 inhibitors, and further downregulated activated oncogenic genes. Finally, HCC growth was dramatically restrained and HBV-induced transcripts were systematically reduced via injection of HBV-miR-2 inhibitors in animal models. CONCLUSION HBV-SITEs were identified as novel oncogenic elements for HCC, which provides an insightful perspective for the other cancers caused by oncogenic DNA viruses. We demonstrated that the integrated HBV sequence itself acted as oncogenic enhancers and nucleotide variations of HBV genotypes account for particular pathogenic progression, supporting that the viral nucleotide sequences are vital pathogenic substances beyond viral proteins. And modulation of their enhancer activities could be clinically achievable strategy for blocking DNA viruses-related cancer progression in the future.
Collapse
Affiliation(s)
- Lu Chen
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxuan Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangyi Zheng
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianlong Meng
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qunyan Yao
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Liang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mu Ye
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kaicheng Zhou
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengxing Liu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhicong Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhanrui Mao
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyan Wei
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
- Research and Development Department, Shanghai Epicurer Biotechnology Co., Ltd., Shanghai, China.
| | - Guoming Shi
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Wang L, Zhang Y, Yu T, Wu H. The Role and Mechanism of Deubiquitinase USP7 in Tumor-Associated Inflammation. Biomedicines 2024; 12:2734. [PMID: 39767641 PMCID: PMC11726842 DOI: 10.3390/biomedicines12122734] [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: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Deubiquitinating enzymes are a class of proteases that remove ubiquitin tags from proteins, thereby controlling protein stability and function. Tumor inflammation arises from interactions between tumor cells and their microenvironment, which trigger an inflammatory response. The deubiquitinating enzyme USP7 plays a central role in this process. Research suggests that USP7 may modulate various signaling pathways related to inflammatory responses through its deubiquitinating activity, thereby influencing tumor development and progression, including regulating T cell immune activity, improving macrophage anti-tumor activity, and regulating NF-κB signal pathways. Overall, describing the role and mechanism of USP7 in the tumor inflammatory response is of great importance for elucidating the regulatory mechanism of tumor inflammation and developing new therapeutic strategies. This article mainly reviews the structure, function, role, and mechanism of USP7 in the tumor inflammation response.
Collapse
Affiliation(s)
- Luhong Wang
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
- Dalian Key Laboratory of Protein Modification and Disease, Faculty of Medicine, School of Biological Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yong Zhang
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
| | - Tao Yu
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
| | - Huijian Wu
- Dalian Key Laboratory of Protein Modification and Disease, Faculty of Medicine, School of Biological Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
3
|
Liu Y, Wang F, Yan G, Tong Y, Guo W, Li S, Qian Y, Li Q, Shu Y, Zhang L, Zhang Y, Xia Q. CPT1A loss disrupts BCAA metabolism to confer therapeutic vulnerability in TP53-mutated liver cancer. Cancer Lett 2024; 595:217006. [PMID: 38823763 DOI: 10.1016/j.canlet.2024.217006] [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: 03/21/2024] [Revised: 05/12/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Driver genomic mutations in tumors define specific molecular subtypes that display distinct malignancy competence, therapeutic resistance and clinical outcome. Although TP53 mutation has been identified as the most common mutation in hepatocellular carcinoma (HCC), current understanding on the biological traits and therapeutic strategies of this subtype has been largely unknown. Here, we reveal that fatty acid β oxidation (FAO) is remarkable repressed in TP53 mutant HCC and which links to poor prognosis in HCC patients. We further demonstrate that carnitine palmitoyltransferase 1 (CPT1A), the rate-limiting enzyme of FAO, is universally downregulated in liver tumor tissues, and which correlates with poor prognosis in HCC and promotes HCC progression in the de novo liver tumor and xenograft tumor models. Mechanically, hepatic Cpt1a loss disrupts lipid metabolism and acetyl-CoA production. Such reduction in acetyl-CoA reduced histone acetylation and epigenetically reprograms branched-chain amino acids (BCAA) catabolism, and leads to the accumulation of cellular BCAAs and hyperactivation of mTOR signaling. Importantly, we reveal that genetic ablation of CPT1A renders TP53 mutant liver cancer mTOR-addicted and sensitivity to mTOR inhibitor AZD-8055 treatment. Consistently, Cpt1a loss in HCC directs tumor cell therapeutic response to AZD-8055. CONCLUSION: Our results show genetic evidence for CPT1A as a metabolic tumor suppressor in HCC and provide a therapeutic approach for TP53 mutant HCC patients.
Collapse
Affiliation(s)
- Yanfeng Liu
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China.
| | - Fan Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoquan Yan
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Tong
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyun Guo
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songling Li
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yifei Qian
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyu Li
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Shu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhang
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yonglong Zhang
- Central Laboratory, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China; Shanghai Institute of Transplantation, Shanghai, China.
| |
Collapse
|
4
|
Zhang M, Wei T, Guo D. The role of abnormal ubiquitination in hepatocellular carcinoma pathology. Cell Signal 2024; 114:110994. [PMID: 38036196 DOI: 10.1016/j.cellsig.2023.110994] [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: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Primary liver cancer is known for its high incidence and fatality rate. Over the years, therapeutic strategies for primary liver cancer have advanced significantly. Nonetheless, a substantial number of patients have not benefited from these methods, underscoring the pressing need for new and effective treatments for primary liver cancer. Ubiquitination is a critical post-translational modification that enables proteins to fulfill their normal biological functions and maintain their expression stability within cells. Importantly, increasing evidence suggests that the progression of liver cancer cells is often accompanied by disruptions in protein ubiquitination and deubiquitination processes. In this comprehensive review, we have compiled pertinent research about dysregulated ubiquitination in hepatocellular carcinoma (HCC) to broaden our understanding in this field. We elucidate the connections between the ubiquitination proteasome system, deubiquitination, and HCC. Furthermore, we shed light on the role of ubiquitination in cells situated within the tumor microenvironment of HCC including its involvement in mediating the activation of oncogenic pathways, reprogramming metabolic processes, and perturbing normal cellular functions. In conclusion, targeting the dysregulation of ubiquitination in HCC holds promise as a prospective and complementary therapeutic approach to existing treatments.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Tingju Wei
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Danfeng Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
5
|
Huang C, Chen B, Wang X, Xu J, Sun L, Wang D, Zhao Y, Zhou C, Gao Q, Wang Q, Chen Z, Wang M, Zhang X, Xu W, Shen B, Zhu W. Gastric cancer mesenchymal stem cells via the CXCR2/HK2/PD-L1 pathway mediate immunosuppression. Gastric Cancer 2023; 26:691-707. [PMID: 37300724 DOI: 10.1007/s10120-023-01405-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Anti-PD-1 immunotherapy has emerged as an important therapeutic modality in advanced gastric cancer (GC). However, drug resistance frequently develops, limiting its effectiveness. METHODS The role of gastric cancer mesenchymal stem cells (GCMSCs) in anti-PD-1 resistance was evaluated in vivo in NPGCD34+ or NCGPBMC xenograft mouse model. In addition, we investigated CD8+T cell infiltration and effector function by spectral cytometry and IHC. The effects of GCMSCs conditional medium (GCMSC-CM) on GC cell lines were characterized at the level of the proteome, secretome using western blot, and ELISA assays. RESULTS We reported that GCMSCs mediated tolerance mechanisms contribute to tumor immunotherapy tolerance. GCMSC-CM attenuated the antitumor activity of PD-1 antibody and inhibited immune response in humanized mouse model. In GC cells under serum deprivation and hypoxia, GCMSC-CM promoted GC cells proliferation via upregulating PD-L1 expression. Mechanistically, GCMSC-derived IL-8 and AKT-mediated phosphorylation facilitated HK2 nuclear localization. Phosphorylated-HK2 promoted PD-L1 transcription by binding to HIF-1α. What is more, GCMSC-CM also induced lactate overproduction in GC cells in vitro and xenograft tumors in vivo, leading to impaired function of CD8+ T cells. Furthermore, CXCR1/2 receptor depletion, CXCR2 receptor antagonist AZD5069 and IL-8 neutralizing antibody application also significantly reversed GCMSCs mediated immunosuppression, restoring the antitumor capacity of PD-1 antibody. CONCLUSIONS Our findings reveal that blocking GCMSCs-derived IL-8/CXCR2 pathway decreasing PD-L1 expression and lactate production, improving antitumor efficacy of anti-PD-1 immunotherapy, may be of value for the treatment of advanced gastric carcinoma.
Collapse
Affiliation(s)
- Chao Huang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Bin Chen
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xin Wang
- Department of Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juan Xu
- Department of Laboratory Medicine, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Li Sun
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
- Department of Clinical Laboratory, Kunshan First People's Hospital, Kunshan, China
| | - Deqiang Wang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Chenglin Zhou
- Department of Laboratory Medicine, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Qiuzhi Gao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Qianqian Wang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Zhihong Chen
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mei Wang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Xu Zhang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Wenrong Xu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu, China.
| |
Collapse
|
6
|
Michail A, Gkikas D, Stellas D, Kaltezioti V, Politis PK. Prox1 Suppresses the Proliferation of Breast Cancer Cells via Direct Inhibition of c-Myc Gene Expression. Cells 2023; 12:1869. [PMID: 37508533 PMCID: PMC10377922 DOI: 10.3390/cells12141869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is one of the most lethal malignancies in women worldwide and is characterized by rapid growth and low survival rates, despite advances in tumor biology and therapies. Novel therapeutic approaches require new insights into the molecular mechanisms of malignant transformation and progression. To this end, here, we identified Prox1 as a negative regulator of proliferation and tumor-related metabolism in breast cancer. In particular, we showed that breast tumors from human patients exhibited reduced levels of Prox1 expression, while high expression levels of Prox1 were associated with a favorable prognosis in breast cancer patients. Moreover, we experimentally demonstrated that Prox1 was sufficient to strongly suppress proliferation, migration, and the Warburg effect in human breast cancer cells without inducing apoptosis. Most importantly, over-expression of Prox1 inhibited breast tumor growth in vivo in both heterotopic and orthotopic xenograft mouse models. The anti-tumorigenic effect of Prox1 was mediated by the direct repression of c-Myc transcription and its downstream target genes. Consistently, c-Myc over-expression from an artificial promoter that was not targeted by Prox1 reversed Prox1's anti-tumor effects. These findings suggest that Prox1 has a tumor suppressive role via direct transcriptional regulation of c-Myc, making it a promising therapeutic gene for breast cancer.
Collapse
Affiliation(s)
- Artemis Michail
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str., 115 27 Athens, Greece
- Department of Biology, University of Patras, 265 04 Patras, Greece
| | - Dimitrios Gkikas
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str., 115 27 Athens, Greece
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 116 35 Athens, Greece
| | - Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str., 115 27 Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str., 115 27 Athens, Greece
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus
| |
Collapse
|
7
|
Saha G, Roy S, Basu M, Ghosh MK. USP7 - a crucial regulator of cancer hallmarks. Biochim Biophys Acta Rev Cancer 2023; 1878:188903. [PMID: 37127084 DOI: 10.1016/j.bbcan.2023.188903] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Over the course of three decades of study, the deubiquitinase Herpesvirus associated Ubiquitin-Specific Protease/Ubiquitin-Specific Protease 7 (HAUSP/USP7) has gradually come to be recognized as a crucially important molecule in cellular physiology. The fact that USP7 is overexpressed in a number of cancers, including breast, prostate, colorectal, and lung cancers, supports the idea that USP7 is also an important regulator of tumorigenesis. In this review, we discuss USP7's function in relation to the cancer hallmarks described by Hanahan and Weinberg. This post-translational modifier can support increased proliferation, block unfavorable growth signals, stop cell death, and support an unstable cellular genome by manipulating key players in the pertinent signalling circuit. It is interesting to note that USP7 also aids in the stabilization of molecules that support angiogenesis and metastasis. Targeting USP7 has now emerged as a crucial component of USP7 research because pharmacological inhibition of USP7 supports p53-mediated cell cycle arrest and apoptosis. Efficacious USP7 inhibition is currently being investigated in both synthetic and natural compounds, but issues with selectivity and a lack of co-crystal structure have hindered USP7 inhibition from being tested in clinical settings. Moreover, the development of new, more effective USP7 inhibitors and their encouraging implications by numerous groups give us a glimmer of hope for USP7-targeting medications as effective substitutes for hazardous cancer chemotherapeutics.
Collapse
Affiliation(s)
- Gouranga Saha
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India
| | - Srija Roy
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, University of Calcutta, Kolkata, PIN - 743372, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, PIN - 700032, India.
| |
Collapse
|
8
|
Han H, Lin T, Wang Z, Song J, Fang Z, Zhang J, You X, Du Y, Ye J, Zhou G. RNA-binding motif 4 promotes angiogenesis in HCC by selectively activating VEGF-A expression. Pharmacol Res 2023; 187:106593. [PMID: 36496136 DOI: 10.1016/j.phrs.2022.106593] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Increased angiogenesis in the liver plays a critical role in the progression of hepatocellular carcinoma (HCC). However, the molecular mechanism underlying increased angiogenesis in HCC is not well understood. Current study was designed to identify the potential angiogenic effect of RNA-binding motif 4 (RBM4)through a small-scale overexpression screening, followed by comparison of the expression level of RBM4 in cancer and adjacent tissues in multiple malignancies to explore the relationship between RBM4 and CD31 protein expression level and related clinical indicators, and understand the role of RBM4 in the hepatocellular carcinoma. To understand the specific mechanism of RBM4 in detail, transcriptome sequencing, mass spectrometry and multiple molecular cytological studies were performed. These cellular level results were verified by experiments in animal models of nude mice. The increased expression of RBM4 in cancer tissues, suggested its use as a prognostic biomarker. The RBM4 expression was found to be strongly correlated with tumor microvessel density. Mechanistically, RBM4 mediated its effects via interaction with HNRNP-M through the latter's WDR15 domain, which then stabilized RelA/p65 mRNA. Consequently, RBM4 induced the activation of the NF-kB signaling pathway, upregulating the expression of proangiogenic factor VEGF-A. The results confirmed the mechanism by which RBM4 promotes angiogenesis in hepatocellular carcinoma suggesting RBM4 as a crucial promoter of angiogenesis in HCC, helping understand regulation of NF-kB signaling in HCC.
Collapse
Affiliation(s)
- Hexu Han
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Ting Lin
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Zhenyu Wang
- Department of pediatric surgery, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Jingjing Song
- Department of Pediatrics, the Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027 Zhejiang, China
| | - Ziyi Fang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Xiaomin You
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Yanping Du
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Jun Ye
- Center for Translational Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China.
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China.
| |
Collapse
|
9
|
Peng S, Chen Y, Li T, Mao J, Yang P, Zou B, Luo L, Zhang W, Wang W, Xie R, Li J, Zeng L. Hsa-microRNA-370-3p targeting Snail and Twist1 suppresses IL-8/STAT3-driven hepatocellular carcinoma metastasis. Cancer Sci 2022; 113:4120-4134. [PMID: 36083239 DOI: 10.1111/cas.15571] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
The pro-inflammatory factor interleukin-8 (IL-8) is related to poor prognosis in hepatocellular carcinoma (HCC) patients. Interleukin-8 enhanced HCC invasion by upregulating Snail and Twist1, whether this modulation relies on microRNAs (miR) is unclear. In this study, hsa-miR-370-3p was screened as candidate miRNA targeting Snail and Twist1, and its expression was downregulated by IL-8. Luciferase assays and RNA electrophoretic mobility shift assays were used to evaluate the interaction between miR-370-3p and targeted mRNAs. Coimmunoprecipitation, luciferase, and ChIP assays were undertaken to investigate the mechanisms underlying IL-8-mediated modification of miR-370-3p. Gain- and loss-of-function studies, Transwell assays, and a xenograft nude mouse model were used to investigate pro- and antitumor activities. Interleukin-8 and miR-370-3p levels were analyzed for clinical relevance in HCC patients. Our results showed that HCC patients with high levels of IL-8 experienced more metastasis and shorter survival. Interleukin-8 induced epithelial-mesenchymal transition and promoted liver cancer cell migration, invasion, and metastasis both in vitro and in vivo. MicroRNA-370-3p interacted with its cognate mRNA within the 3'-UTR regions of Twist1 and Snail mRNA directly and specifically and attenuated IL-8 protumoral effects on liver cancer cells. Interleukin-8 negatively modulated miR-370-3p through signal transducer and activator of transcription 3 (STAT3) activation by recruiting histone deacetylase 1 (HDAC1) to miR-370-3p promoter. The STAT3 and HDAC antagonists inhibited liver cancer cell migration and invasion. Patients with high miR-370-3p and low IL-8 levels had longer overall survival. In conclusion, our study elucidated a novel axis IL-8/STAT3/miR-370-3p/Twist1 and Snail relying on HDAC1 recruitment, which showed both diagnostic and therapeutic potentials of miR-370-3p in HCC metastasis.
Collapse
Affiliation(s)
- Siqi Peng
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yutong Chen
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Ting Li
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Junjie Mao
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Pengfei Yang
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Lisi Luo
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Weiyu Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Wen Wang
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Rongzhi Xie
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Linjuan Zeng
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
10
|
Wang Y, Luo M, Wang F, Tong Y, Li L, Shu Y, Qiao K, Zhang L, Yan G, Liu J, Ji H, Xie Y, Zhang Y, Gao WQ, Liu Y. AMPK induces degradation of the transcriptional repressor PROX1 impairing branched amino acid metabolism and tumourigenesis. Nat Commun 2022; 13:7215. [PMID: 36433955 PMCID: PMC9700865 DOI: 10.1038/s41467-022-34747-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/04/2022] [Indexed: 11/26/2022] Open
Abstract
Tumour cell metabolic plasticity is essential for tumour progression and therapeutic responses, yet the underlying mechanisms remain poorly understood. Here, we identify Prospero-related homeobox 1 (PROX1) as a crucial factor for tumour metabolic plasticity. Notably, PROX1 is reduced by glucose starvation or AMP-activated protein kinase (AMPK) activation and is elevated in liver kinase B1 (LKB1)-deficient tumours. Furthermore, the Ser79 phosphorylation of PROX1 by AMPK enhances the recruitment of CUL4-DDB1 ubiquitin ligase to promote PROX1 degradation. Downregulation of PROX1 activates branched-chain amino acids (BCAA) degradation through mediating epigenetic modifications and inhibits mammalian target-of-rapamycin (mTOR) signalling. Importantly, PROX1 deficiency or Ser79 phosphorylation in liver tumour shows therapeutic resistance to metformin. Clinically, the AMPK-PROX1 axis in human cancers is important for patient clinical outcomes. Collectively, our results demonstrate that deficiency of the LKB1-AMPK axis in cancers reactivates PROX1 to sustain intracellular BCAA pools, resulting in enhanced mTOR signalling, and facilitating tumourigenesis and aggressiveness.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengjun Luo
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Tong
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linfeng Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Shu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Qiao
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhang
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoquan Yan
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongbin Ji
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
- Children's Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yonglong Zhang
- Central Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Yanfeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
11
|
Zhao J, Guo J, Wang Y, Ma Q, Shi Y, Cheng F, Lu Q, Fu W, Ouyang G, Zhang J, Xu Q, Hu X. Research Progress of DUB Enzyme in Hepatocellular Carcinoma. Front Oncol 2022; 12:920287. [PMID: 35875077 PMCID: PMC9303014 DOI: 10.3389/fonc.2022.920287] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
According to GLOBOCAN 2021 cancer incidence and mortality statistics compiled by the International Agency for Research on Cancer, hepatocellular carcinoma (HCC) is the most common malignancy in the human liver and one of the leading causes of cancer death worldwide. Although there have been great advances in the treatment of HCC, such as regofenib, sorafenib, and lomvatinib, which have been developed and approved for the clinical treatment of advanced or metastatic HCC. However, they only prolong survival by a few months, and patients with advanced liver cancer are susceptible to tumor invasion metastasis and drug resistance. Ubiquitination modification is a type of post-translational modification of proteins. It can affect the physiological activity of cells by regulating the localization, stability and activity of proteins, such as: gene transcription, DNA damage signaling and other pathways. The reversible process of ubiquitination is called de-ubiquitination: it is the process of re-releasing ubiquitinated substrates with the participation of de-ubiquitinases (DUBs) and other active substances. There is growing evidence that many dysregulations of DUBs are associated with tumorigenesis. Although dysregulation of deuquitinase function is often found in HCC and other cancers, The mechanisms of action of many DUBs in HCC have not been elucidated. In this review, we focused on several deubiquitinases (DUBs) associated with hepatocellular carcinoma, including their structure, function, and relationship to hepatocellular carcinoma. hepatocellular carcinoma was highlighted, as well as the latest research reports. Among them, we focus on the USP family and OTU family which are more studied in the HCC. In addition, we discussed the prospects and significance of targeting DUBs as a new strategy for the treatment of hepatocellular carcinoma. It also briefly summarizes the research progress of some DUB-related small molecule inhibitors and their clinical application significance as a treatment for HCC in the future.
Collapse
Affiliation(s)
- Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Yanan Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Qiancheng Ma
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yu Shi
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Feng Cheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Qiliang Lu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wen Fu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | | | - Ji Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Xiaoge Hu, ; Qiuran Xu,
| | - Xiaoge Hu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical CollegeHangzhou, China
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Xiaoge Hu, ; Qiuran Xu,
| |
Collapse
|
12
|
Lee HC, Chang CY, Huang YC, Wu KL, Chiang HH, Chang YY, Liu LX, Hung JY, Hsu YL, Wu YY, Tsai YM. Downregulated ADAMTS1 Incorporating A2M Contributes to Tumorigenesis and Alters Tumor Immune Microenvironment in Lung Adenocarcinoma. BIOLOGY 2022; 11:biology11050760. [PMID: 35625488 PMCID: PMC9139094 DOI: 10.3390/biology11050760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022]
Abstract
Simple Summary Lung cancer is the most dreadful cancer type and has the worst cancer-related clinical outcomes. This study used specimens from the in-house lung cancer cohort and public cohort to verify the roles of downregulated ADAMTS1, a protease remodeling extracellular matrix, to facilitate cancer promotion and progress. Based on the clinical specimens, cell and animal study with the aid of the public databases, we concluded that downregulated expression of ADAMTS1 might promote tumor progression and metastasis and modify the tumor microenvironment in lung cancer. Further investigation would be required for its application in treating lung cancer. Abstract Lung adenocarcinoma (LUAD) still holds the most dreadful clinical outcomes worldwide. Despite advanced treatment strategies, there are still some unmet needs. Next-generation sequencing of large-scale cancer genomics discovery projects combined with bioinformatics provides the opportunity to take a step forward in meeting clinical conditions. Based on in-house and The Cancer Genome Atlas (TCGA) cohorts, the results showed decreased levels of ADAMTS1 conferred poor survival compared with normal parts. Gene set enrichment analyses (GSEA) indicated the negative correlation between ADAMTS1 and the potential roles of epithelial–mesenchymal transition (EMT), metastasis, and poor prognosis in LUAD patients. With the knockdown of ADAMTS1, A549 lung cancer cells exhibited more aggressive behaviors such as EMT and increased migration, resulting in cancer metastasis in a mouse model. The pathway interaction network disclosed the linkage of downregulated α2-macroglobulin (A2M), which regulates EMT and metastasis. Furthermore, immune components analysis indicated a positive relationship between ADAMTS1 and the infiltrating levels of multiple immune cells, especially anticancer CD4+ T cells in LUAD. Notably, ADAMTS1 expression was also inversely correlated with the accumulation of immunosuppressive myeloid-derived suppressor cells and regulatory T cells, implying the downregulated ADAMTS1 mediated immune adjustment to fit the tumor survival disadvantages in LUAD patients. In conclusion, our study indicates that ADAMTS1 interacts with A2M in regulating EMT and metastasis in LUAD. Additionally, ADAMTS1 contributes to poor prognosis and immune infiltration in LUAD patients
Collapse
Affiliation(s)
- Hsiao-Chen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chao-Yuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
| | - Kuan-Li Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Hung-Hsing Chiang
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yung-Yun Chang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Lian-Xiu Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
| | - Jen-Yu Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 807, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Yuan Wu
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Ying-Ming Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.L.); (C.-Y.C.); (Y.-C.H.); (K.-L.W.); (L.-X.L.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Correspondence:
| |
Collapse
|
13
|
Shen Z, Wu J, Gao Z, Zhang S, Chen J, He J, Guo Y, Deng Q, Xie Y, Liu J, Zhang J. High mobility group AT-hook 1 (HMGA1) is an important positive regulator of hepatitis B virus (HBV) that is reciprocally upregulated by HBV X protein. Nucleic Acids Res 2022; 50:2157-2171. [PMID: 35137191 PMCID: PMC8887475 DOI: 10.1093/nar/gkac070] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/10/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic infection with hepatitis B virus (HBV) is associated with liver cirrhosis and hepatocellular carcinoma. Upon infection of hepatocytes, HBV covalently closed circular DNA (cccDNA) exists as histone-bound mini-chromosome, subjected to transcriptional regulation similar to chromosomal DNA. Here we identify high mobility group AT-hook 1 (HMGA1) protein as a positive regulator of HBV transcription that binds to a conserved ATTGG site within enhancer II/core promoter (EII/Cp) and recruits transcription factors FOXO3α and PGC1α. HMGA1-mediated upregulation of EII/Cp results in enhanced viral gene expression and genome replication. Notably, expression of endogenous HMGA1 was also demonstrated to be upregulated by HBV, which involves HBV X protein (HBx) interacting with SP1 transcription factor to activate HMGA1 promoter. Consistent with these in vitro results, chronic hepatitis B patients in immune tolerant phase display both higher intrahepatic HMGA1 protein levels and higher serum HBV markers compared to patients in inactive carrier phase. Finally, using a mouse model of HBV persistence, we show that targeting endogenous HMGA1 through RNA interference facilitated HBV clearance. These data establish HMGA1 as an important positive regulator of HBV that is reciprocally upregulated by HBV via HBx and also suggest the HMGA1-HBV positive feedback loop as a potential therapeutic target.
Collapse
Affiliation(s)
- Zhongliang Shen
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jingwen Wu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zixiang Gao
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shenyan Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingwen Chen
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingjing He
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yifei Guo
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiang Deng
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiming Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity,National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Department of Microbiology and Parasitology, Shanghai Institute of Infectious Diseases and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Kaltezioti V, Foskolou IP, Lavigne MD, Ninou E, Tsampoula M, Fousteri M, Margarity M, Politis PK. Prox1 inhibits neurite outgrowth during central nervous system development. Cell Mol Life Sci 2021; 78:3443-3465. [PMID: 33247761 PMCID: PMC11072475 DOI: 10.1007/s00018-020-03709-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Iosifina P Foskolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matthieu D Lavigne
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Marigoula Margarity
- Laboratory of Human and Animal Physiology, Department of Biology, School of Natural Sciences, University of Patras, 26500, Rio Achaias, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece.
| |
Collapse
|
15
|
Han H, Lin T, Fang Z, Zhou G. RBM23 Drives Hepatocellular Carcinoma by Activating NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6697476. [PMID: 33791378 PMCID: PMC7994101 DOI: 10.1155/2021/6697476] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/10/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide, and angiogenesis has been proven to be significantly involved in its progression. However, the molecular mechanism underlying HCC angiogenesis has not been well researched. In this study, RNA Binding Motif Protein 23 (RBM23) was identified as a novel proangiogenic factor in HCC cell lines and tissues. MATERIALS AND METHODS Firstly, we analyzed the correlation of clinical specimens. In HCC tissues, the levels of RBM23 and microvessel density (MVD) showed a strong positive correlation. Furthermore, data from related cytology experiments showed that the knockdown of RBM23 expression in HCC cells significantly inhibited the tube formation by the human vascular endothelial cells in vitro. The mechanism of this phenomenon was found to be through increasing the mRNA of p65 and enhanced the nuclear accumulation of p65. Consequently, RBM23 activated the NF-κB signaling pathway and promoted expression of the proangiogenic cytokines selectively. Results and Conclusion. In summary, this study revealed that RBM23 promotes the angiogenesis properties of HCC via the NF-κB signaling pathway. It may, therefore, be a potential therapeutic target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hexu Han
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Ting Lin
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Ziyi Fang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Jiangsu 226001, China
| |
Collapse
|
16
|
Zhou TH, Su JZ, Qin R, Chen X, Ju GD, Miao S. Prognostic and Predictive Value of a 15 Transcription Factors (TFs) Panel for Hepatocellular Carcinoma. Cancer Manag Res 2020; 12:12349-12361. [PMID: 33293862 PMCID: PMC7719121 DOI: 10.2147/cmar.s279194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/31/2020] [Indexed: 01/26/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is one of the most devastating diseases worldwide. Limited performance of clinicopathologic parameters as prognostic factors underscores more accurate and effective biomarkers for high-confidence prognosis that guide decision-making for optimal treatment of HCC. The aim of the present study was to establish a novel panel to improve prognosis prediction of HCC patients, with a particular interest in transcription factors (TFs). Materials and Methods A TF-related prognosis model of liver cancer with data from ICGC-LIRP-JI cohort successively were processed by univariate and multivariate Cox regression analysis. Then, for evaluating the prognostic prediction value of the model, receiver operating characteristic (ROC) curve and survival analysis were performed both with internal data from the International Cancer Genome Consortium (ICGC) and external data from The Cancer Genome Atlas (TCGA). Furthermore, we verified the expression of three genes in HCC cell lines by Western blot and qPCR and protein expression level by IHC in liver cancer patients’ sample. Finally, we constructed a TF clinical characteristics nomogram to furtherly predict liver cancer patient survival probability with TCGA cohort. Results By Cox regression analysis, a panel of 15 TFs (ZNF331, MYCN, AHRR, LEF1, ZNF780A, POU1F1, DLX5, ZNF775, PLSCR1, FOXK1, TAL2, ZNF558, SOX9, TCFL5, GSC) was identified to present with powerful predictive performance for overall survival of HCC patients based on internal ICGC cohort and external TCGA cohort. A nomogram that integrates these factors was established, allowing efficient prediction of survival probabilities and displaying higher clinical utility. Conclusion The 15-TF panel is an independent prognostic factor for HCC, and 15 TF-based nomogram might provide implication an effective approach for HCC patient management and treatment.
Collapse
Affiliation(s)
- Tian-Hao Zhou
- Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Jing-Zhi Su
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Rui Qin
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining 272000, People's Republic of China
| | - Xi Chen
- Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan 250000, People's Republic of China
| | - Gao-Da Ju
- Department of Oncology, Beijing Cancer Hospital, Peking University, Beijing 102206, People's Republic of China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining 272000, People's Republic of China
| |
Collapse
|
17
|
Okimoto S, Tashiro H, Iwako H, Kuroda S, Kobayashi T, Hinoi T, Ohdan H. Antithrombin attenuates the progression of hepatocellular carcinoma by regulating neutrophil/interleukin-8 signaling. Hepatol Res 2020; 50:1284-1296. [PMID: 32893935 DOI: 10.1111/hepr.13558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
AIM Inflammation plays an important role in hepatocellular carcinoma (HCC) progression. Here, we examined whether antithrombin (AT) plays a role in attenuating HCC progression, via its anti-inflammatory effects. METHODS HCCs were developed in AT-insufficient (AT+/- ) mice and wild-type (AT+/+ ) mice treated with diethyl nitrosamine and carbon tetrachloride. AT was administered to AT+/- mice. The development of HCC was compared between the three groups. In vitro study, migration assay was performed. The association of the prognosis of patients with HCC and plasma AT values was clinically examined. RESULTS AT suppressed the release of interleukin (IL)-8 from lipopolysaccharide (LPS)-stimulated human neutrophils in vitro. Huh-7 cells that were co-cultured with neutrophils and stimulated with LPS showed significantly enhanced migration; however, Huh-7 cells co-cultured with LPS/AT-stimulated neutrophils showed significantly decreased migration. Moreover, the addition of anti-IL-8 antibodies to LPS-stimulated Huh-7 cells co-cultured with neutrophils also suppressed migration. AT+/- mice (AT plasma activity: 64%) promoted liver cancer, as compared with wild-type mice (AT plasma activity: 135%); AT administration attenuated liver cancer in AT+/- mice. Patients with HCC with a preoperative AT level of ≥70% showed better outcomes after liver resection, as compared with those with an AT level of <70%. IL-8 expression and neutrophil infiltration in HCC tissues were negatively correlated with the AT level. CONCLUSIONS AT attenuates HCC progression by regulating neutrophil/IL-8 signaling.
Collapse
Affiliation(s)
- Sho Okimoto
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Hirotaka Tashiro
- Department of Surgery, National Hospital Organization Kure Medical Center, Kure City, Hiroshima, Japan
| | - Hiroshi Iwako
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Takao Hinoi
- Department of clinical and molecular Genetics, Hiroshima University Hospital, Kasumi, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| |
Collapse
|
18
|
Cai Y, Zheng Q, Yao DJ. Phosphatidylinositol-3,4,5-trisphosphate dependent Rac exchange factor 1 is a diagnostic and prognostic biomarker for hepatocellular carcinoma. World J Clin Cases 2020; 8:3774-3785. [PMID: 32953853 PMCID: PMC7479560 DOI: 10.12998/wjcc.v8.i17.3774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/15/2020] [Accepted: 08/03/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Phosphatidylinositol-3,4,5-trisphosphate dependent Rac exchange factor 1 (P-Rex1) was reported to be a risk factor in several cancers, including breast cancer, lung cancer, and melanoma, but its expression and role in hepatocellular carcinoma (HCC) have not yet been fully studied.
AIM To explore the expression of P-Rex1 in HCC, and further evaluate its potential application in the diagnosis and prognosis of HCC, especially in hepatitis B virus (HBV)-related patients.
METHODS P-Rex1 expression in HCC was evaluated by real-time-quantitative polymerase chain reaction. The expression of P-Rex1 was subjected to correlation analysis with clinical features, such as lymph node invasion, distant metastasis, HBV infection, patient's age and gender. Receiver operating characteristic analysis was used to examine the potential role of P-Rex1 as a diagnostic biomarker in HCC. Kaplan-Meier analysis was used to determine the association between P-Rex1 expression and overall survival, progression-free survival and relapse-free survival. Bioinformatic analysis was used to validate the clinical findings.
RESULTS P-Rex1 expression was significantly increased in HCC tumors than adjacent tissues. The expression of P-Rex1 was higher in HCC patients with lymph node invasion, distant metastasis, HBV infection and positive alpha-fetoprotein, respectively. The receiver operating characteristic analysis showed that P-Rex1 was a diagnostic biomarker with a higher area under the curve value, especially in patients with HBV infection. Survival analysis showed that patients with higher P-Rex1 expression had a favorable survival rate, even in early-stage patients.
CONCLUSION P-Rex1 expression was highly increased in HCC, and the expression level of P-Rex1 was positively correlated with tumor progression. P-Rex1 has a higher efficiency in the diagnosis of HBV-related HCC, and could also be used as a favorable prognostic factor for HCC patients.
Collapse
Affiliation(s)
- Yi Cai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, Sichuan Province, China
| | - Qiao Zheng
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, Sichuan Province, China
| | - De-Jiao Yao
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, Sichuan Province, China
| |
Collapse
|
19
|
Shao Z, Pan Q, Zhang Y. Hepatocellular carcinoma cell-derived extracellular vesicles encapsulated microRNA-584-5p facilitates angiogenesis through PCK1-mediated nuclear factor E2-related factor 2 signaling pathway. Int J Biochem Cell Biol 2020; 125:105789. [PMID: 32522621 DOI: 10.1016/j.biocel.2020.105789] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/22/2020] [Accepted: 06/05/2020] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease characterized by poor liver function with increasing morbidity and poor prognosis. Extracellular vesicles, released by different cells, have been associated with HCC development. Nevertheless, the mechanisms beyond extracellular vesicles in HCC remain uncharacterized. Therefore, the current study aimed to clarify the mechanism of pro-angiogenic microRNA-584-5p in hepatocellular carcinoma. Our results showed that miR-584-5p was highly-expressed in both cancer cells (Hep3B) and their extracellular vesicles. Hep3B and extracellular vesicles were then respectively co-cultured with human vascular endothelial cell line (Ea.hy926), and they both accelerated Ea.hy926 proliferation and migration. Ea.hy926 cells could internalize extracellular vesicles carrying microRNA-584-5p. Of note, microRNA-584-5p could bind to phosphoenolpyruvate carboxykinase 1 to promote nuclear factor E2-related factor 2. Moreover, silencing microRNA-584-5p was found to decline microvessel density, vascular endothelial growth factor A, and tumor growth in vivo and in vitro. Taken altogether, our findings demonstrated that extracellular vesicles-derived microRNA-584-5p promotes angiogenesis by inhibiting PCK1 -mediating NRF2 activation, which highlights the theoretical basis for potential treatments for HCC.
Collapse
Affiliation(s)
- Zigong Shao
- Department of Organ Transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang 110000, PR China; The Key Laboratory of Organ Transplantation of Liaoning Province, Shenyang 110000, PR China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang 110000, PR China; The Key Laboratory of Organ Transplantation of Liaoning Province, Shenyang 110000, PR China
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang 110000, PR China; The Key Laboratory of Organ Transplantation of Liaoning Province, Shenyang 110000, PR China.
| |
Collapse
|
20
|
Yang H, Xuefeng Y, Jianhua X. Systematic review of the roles of interleukins in hepatocellular carcinoma. Clin Chim Acta 2020; 506:33-43. [PMID: 32142718 DOI: 10.1016/j.cca.2020.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer with high morbidity and mortality that is often accompanied by immune system disorders and local lymphocyte infiltration. Tumor-infiltrating lymphocytes, cancer cells, stromal cells, and the numerous cytokines they produce, such as chemokines, interferons, tumor necrosis factors, and interleukins, collectively constitute the tumor microenvironment. As a main type of immune effector, interleukin plays opposing roles in regulating tumor cell progression, adhesion, and migration according to its different subtypes. Many reports have concentrated on the roles that interleukins play in HCC, but understanding them systematically remains challenging. This study reviewed the current data to comprehensively summarize the relationships between HCC progression and human interleukin gene families.
Collapse
Affiliation(s)
- Hu Yang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China; Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, China
| | - Yang Xuefeng
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, China
| | - Xiao Jianhua
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
21
|
Yang X, Wang Z, Kai J, Wang F, Jia Y, Wang S, Tan S, Shen X, Chen A, Shao J, Zhang F, Zhang Z, Zheng S. Curcumol attenuates liver sinusoidal endothelial cell angiogenesis via regulating Glis-PROX1-HIF-1α in liver fibrosis. Cell Prolif 2020; 53:e12762. [PMID: 32119185 PMCID: PMC7106966 DOI: 10.1111/cpr.12762] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/13/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Hepatic sinusoidal angiogenesis owing to dysfunctional liver sinusoidal endothelial cells (LSECs) accompanied by an abnormal angioarchitecture is a symbol related to liver fibrogenesis, which indicates a potential target for therapeutic interventions. However, there are few researches connecting angiogenesis with liver fibrosis, and the deeper mechanism remains to be explored. MATERIALS AND METHODS Cell angiogenesis and angiogenic protein were examined in primary LSECs of rats, and multifarious cellular and molecular assays revealed the efficiency of curcumol intervention in fibrotic mice. RESULTS We found that curcumol inhibited angiogenic properties through regulating their upstream mediator hypoxia-inducible factor-1α (HIF-1α). The transcription activation of HIF-1α was regulated by hedgehog signalling on the one hand, and the protein stabilization of HIF-1α was under the control of Prospero-related homeobox 1 (PROX1) on the other. A deubiquitinase called USP19 could be recruited by PROX1 and involved in ubiquitin-dependent degradation of HIF-1α. Furthermore, our researches revealed that hedgehog signalling participated in the activation of PROX1 transcription probably in vitro. Besides, curcumol was found to ameliorate liver fibrosis and sinusoid angiogenesis via hedgehog pathway in carbon tetrachloride (CCl4 ) induced liver fibrotic mice. The protein expression of key regulatory factors, PROX1 and HIF-1α, was consistent with the Smo, the marker protein of Hh signalling pathway. CONCLUSIONS In this article, we evidenced that curcumol controlling LSEC-mediated angiogenesis could be a promising therapeutic approach for liver fibrosis.
Collapse
Affiliation(s)
- Xiang Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Zhimin Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Jun Kai
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Yan Jia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Shijun Wang
- Shandong University of Traditional Chinese MedicineJinanChina
| | - Shanzhong Tan
- Nanjing Hospital Affiliated to Nanjing University of Chinese MedicineNanjingChina
| | - Xikun Shen
- Suzhou Hospital of Traditional Chinese MedicineSuzhouChina
| | - Anping Chen
- Department of PathologySchool of MedicineSaint Louis UniversitySt. LouisMOUSA
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
- Jiangsu Key Laboratory of Therapeutic Material of Chinese MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
- Jiangsu Key Laboratory of Therapeutic Material of Chinese MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
- Jiangsu Key Laboratory of Therapeutic Material of Chinese MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia MedicaSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
- Jiangsu Key Laboratory of Therapeutic Material of Chinese MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
22
|
Zhang W, Zhang J, Xu C, Zhang S, Bian S, Jiang F, Ni W, Qu L, Lu C, Ni R, Fan Y, Xiao M, Liu J. Ubiquitin-specific protease 7 is a drug-able target that promotes hepatocellular carcinoma and chemoresistance. Cancer Cell Int 2020; 20:28. [PMID: 32002017 PMCID: PMC6986148 DOI: 10.1186/s12935-020-1109-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ubiquitin-specific protease 7 (USP7) is a de-ubiquitin enzyme that plays an essential role in multiple cancers and becomes a target for treatment. However, the role of USP7 and its therapeutic value for HCC remains unclear. METHODS USP7 expression was examined in HCC tissues by western blot and immunohistochemistry. The correlation of USP7 and HCC prognosis was analyzed by Kaplan-Meier survival method. Mass spectrometry was determined and cell proliferation and tumorigenicity assays were conducted in vitro and in vivo treated by P22077 and sgRNA-USP7. RESULTS USP7 expression was significantly increased in HCC and associated with its progression. Interestingly, many HCC cells are sensitive to USP7 inhibition by using P22077. P22077 treatment not only induced cell death but also inhibited cell proliferation and migration in Huh7 and SK-Hep1 cells. In a xenograft model, P22077 efficiently inhibited tumor growth. In chemo-resistant HCC cells, P22077 decreased cell sensitivity to chemotherapy. In addition, mass spectrometry reveals 224 of significantly changed proteins upon P22077 treatment. CONCLUSIONS We demonstrate a critical role of USP7 in HCC devolvement and chemoresistance. Disruption of USP7 function results in dis-regulated several key biological processes and subsequently activates BAX. USP7 might be a novel and drug-able target in HCC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Medical College, Nantong University, Nantong, 226001 China
| | - Jingxin Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Medical College, Nantong University, Nantong, 226001 China
| | - Chenzhou Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Medical College, Nantong University, Nantong, 226001 China
| | - Shiqing Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Medical College, Nantong University, Nantong, 226001 China
| | - Saiyan Bian
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Medical College, Nantong University, Nantong, 226001 China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Wenkai Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Lishuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Jiangsu, 226001 China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Jinxia Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| |
Collapse
|
23
|
Jiang X, Liu G, Hu Z, Chen G, Chen J, Lv Z. cGAMP inhibits tumor growth in colorectal cancer metastasis through the STING/STAT3 axis in a zebrafish xenograft model. FISH & SHELLFISH IMMUNOLOGY 2019; 95:220-226. [PMID: 31586458 DOI: 10.1016/j.fsi.2019.09.075] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023]
Abstract
The leading cause of mortality due to colorectal cancer (CRC) is highly associated with the development of liver metastases. Recently, we described cGAMP that is closely related to the metastatic state wherein the progress of metastatic tumors is associated with favorable outcomes in a zebrafish xenograft model. cGAMP was administered and the expression levels of type-I interferons were induced amongst tumor tissues to illuminate the overall measure of the induced STING/STAT3 axis in colorectal liver metastases. Furthermore, cGAMP-STING dependent STAT3 activation resulted in the inhibition of tumor cell proliferation, viability, and invasion in vitro. The subtotal reduction in tumor growth attributed to a large number of infiltrating inflammatory cells in vivo. We showed that cGAMP inhibited migration through angiogenesis by up-regulating IL-2, TNF-α, and IFN-γ, whereas STAT3 down-regulation inhibited CXCL8, BCL-2, and VEGFA expression. The importance of cGAMP in inhibiting the invasion front of CRC confirmed that the cGAMP dependent activation of STING/STAT3 axis played a key role in the inhibition of tumor progression.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- College of Lifescience and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018, China.
| | - Guangping Liu
- College of Life Sciences, Yan'an University, Yan'an, 716000, China.
| | - Zhiyi Hu
- College of Lifescience and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018, China.
| | - Guiqian Chen
- College of Lifescience and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018, China.
| | - Jianqing Chen
- College of Lifescience and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018, China.
| | - Zhengbing Lv
- College of Lifescience and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018, China.
| |
Collapse
|
24
|
Jiang Y, Han QJ, Zhang J. Hepatocellular carcinoma: Mechanisms of progression and immunotherapy. World J Gastroenterol 2019; 25:3151-3167. [PMID: 31333308 PMCID: PMC6626719 DOI: 10.3748/wjg.v25.i25.3151] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 05/18/2019] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is one of the most common malignancies, and various pathogenic factors can lead to its occurrence and development. Among all primary liver cancers, hepatocellular carcinoma (HCC) is the most common. With extensive studies, an increasing number of molecular mechanisms that promote HCC are being discovered. Surgical resection is still the most effective treatment for patients with early HCC. However, early detection and treatment are difficult for most HCC patients, and the postoperative recurrence rate is high, resulting in poor clinical prognosis of HCC. Although immunotherapy takes longer than conventional chemotherapy to produce therapeutic effects, it persists for longer. In recent years, the emergence of many new immunotherapies, such as immune checkpoint blockade and chimeric antigen receptor T cell therapies, has given new hope for the treatment of HCC.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Antineoplastic Agents, Immunological/therapeutic use
- Cancer Vaccines/therapeutic use
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Disease Progression
- Humans
- Immunotherapy, Adoptive/methods
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/prevention & control
- Receptors, Chimeric Antigen/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- Yu Jiang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Qiu-Ju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
25
|
Dai L, Hu W, Yang Z, Chen D, He B, Chen Y, Zhou L, Xie H, Wu J, Zheng S. Upregulated expression of HOXB7 in intrahepatic cholangiocarcinoma is associated with tumor cell metastasis and poor prognosis. J Transl Med 2019; 99:736-748. [PMID: 30664713 PMCID: PMC6760572 DOI: 10.1038/s41374-018-0150-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Homeobox B7 (HOXB7) protein is reported to be aberrantly expressed in a variety of cancers and to play an important role in multiple cellular processes. However, the specific mechanism by which HOXB7 promotes the malignant progression of intrahepatic cholangiocarcinoma (ICC) remains unclear. Therefore, we used quantitative real-time polymerase chain reaction (PCR) to detect the expression level of HOXB7 in 38 paired ICC tissue samples. Additionally, to assess correlation between HOXB7 and ICC prognosis, we performed immunohistochemistry (IHC) using 122 ICC tissues to detect HOXB7 expression. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to assess ICC cell proliferation, and Transwell assays were performed to estimate the invasion and migration abilities of ICC cells. The capillary tube formation assay was applied to explore the angiogenic effects of HOXB7. A xenograft tumor model was established in nude mice to assess the role of HOXB7 in tumor growth and lung metastasis. The results showed higher expression of HOXB7 in ICC tissues than in noncancerous tissues, and this increased expression was significantly associated with a poor prognosis. In addition, HOXB7 overexpression enhanced capillary tube formation, invasion and migration of ICC cells in vitro, whereas HOXB7 knockdown produced the opposite results in vitro. Moreover, the role of HOXB7 in promoting tumor growth and metastasis was verified in vivo. Further investigation revealed that the expression levels of MMP2, MMP9, VEGFa, and IL8 were elevated by HOXB7 and that the ERK pathway was activated. Our results demonstrate the prognostic value of HOXB7 and its role in metastasis and angiogenesis in ICC. HOXB7 upregulated MMP2, MMP9, VEGFa, and IL8 expression via the ERK pathway to accelerate the malignant progression of ICC.
Collapse
Affiliation(s)
- Longfei Dai
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Wendi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Zhenjie Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Diyu Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Bin He
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Yunhao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lin Zhou
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, 310000, China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, 310000, China
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, 310000, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, 310000, China.
| |
Collapse
|
26
|
Du S, Liao S, Liu S, Xin Y. TM6SF2 E167K Variant Overexpression Promotes Expression of Inflammatory Cytokines in the HCC Cell Line HEPA 1-6. J Clin Transl Hepatol 2019; 7:27-31. [PMID: 30944816 PMCID: PMC6441636 DOI: 10.14218/jcth.2018.00055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 12/27/2022] Open
Abstract
Background and Aims: Accumulated evidence has shown that chronic liver inflammation is one of the main risks of hepatocellular carcinoma (HCC), and E167K variant of the transmembrane 6 superfamily member 2 (TM6SF2) plays an important role in the progression of chronic liver diseases and HCC. The aim of this study was to explore effects of the TM6SF2 E167K variant on expression of the inflammatory cytokines TNF-α, IL-2, IL-6 and IL-8 in the HCC cell line HEPA 1-6. Methods: HEPA 1-6 cells were infected with lentivirus containing either the TM6SF2 E167K variant or TM6SF2 wild-type, or control plasmids. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were conducted to analyze the expression of the inflammatory cytokines TNF-α, IL-2, IL-6 and IL-8. A t-test was used for statistical analysis. Results: Compared with the control group and TM6SF2 overexpression group, the relative expression of IL-2 and IL-6 mRNAs were significantly elevated in the TM6SF2 E167K overexpression group (p < 0.05). The relative mRNA expression of IL-8 in the TM6SF2 and TM6SF2 E167K overexpression groups were increased compared to the control group (p < 0.05). No obvious differences were observed for the expression of TNF-α in each group. The expression of TNF-α, IL-2, IL-6 and IL-8 that was tested by western blotting showed the same trends as the qRT-PCR results. Conclusions: In conclusion, the E167K variant of the TM6SF2 gene could promote the expression of inflammatory cytokines IL-2 and IL-6 in HEPA 1-6 cells, suggesting that the TM6SF2 E167K variant may accelerate the progression of HCC.
Collapse
Affiliation(s)
- Shuixian Du
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, China
| | - Songling Liao
- Department of Gastroenterology, Dalian Medical University, Dalian, China
| | - Shousheng Liu
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, China
- Digestive Disease Key Laboratory of Qingdao, Qingdao, China
| | - Yongning Xin
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, China
- Department of Gastroenterology, Dalian Medical University, Dalian, China
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
- Digestive Disease Key Laboratory of Qingdao, Qingdao, China
- *Correspondence to: Yongning Xin, Department of Infectious Disease, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong 266011, China. Tel: +86-532-82789463, Fax: +86-532-85968434, E-mail:
| |
Collapse
|
27
|
Cheng Q, Tong TJ, Li Z, Hu SH, Chen DB, Wang SQ, Zhu JY. Paradoxical effects of cellular senescence-inhibited gene involved in hepatocellular carcinoma migration and proliferation by ERK pathway and mesenchymal-like markers. Onco Targets Ther 2019; 12:2035-2046. [PMID: 30936720 PMCID: PMC6421901 DOI: 10.2147/ott.s188449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cellular senescence-inhibited gene (CSIG) strongly prolongs the progression of replicative senescence. However, roles and mechanisms of CSIG in tumor progression have not been studied widely. METHODS Roles of CSIG in migration and proliferation of SMMC7721 and Huh7 cells were analyzed by transwell or cell viability assays, respectively. Tumorigenicity assays were used to study whether CSIG knockdown could affect SMMC7721 proliferation in vivo. Next, Western blotting and RT-PCR were preformed to evaluate the effects of CSIG on P-ERK cascade and epithelial mesenchymal transformation markers. Then, the location and expression of CSIG protein was detected by immunofluorescence and Western blotting, respectively. Finally, the Cancer Genome Atlas dataset was used to analyze CSIG mRNA levels in hepatocellular carcinoma (HCC) and adjacent non-tumor tissues. RESULTS In this study, we found that CSIG overexpression promoted SMMC7721 cell migration, and CSIG knockdown suppressed tumorigenicity of SMMC7721 cells. In contrast to expectation, CSIG up-regulation could significantly inhibit Huh7 cell growth and migration. CSIG could promote P-ERK activation and levels of mesenchymal-like markers in SMMC7721 cells, whereas CSIG suppressed P-ERK activation and levels of mesenchymal-like markers in Huh7 cells. CSIG protein was located in nucleoli as well as nucleoplasm of SMMC7721 cells, whereas CSIG protein was mainly expressed in the nucleoli rather than nucleoplasm of Huh7 cells. Finally, due to individual differences, raised or down-regulated trends of CSIG in HCC as compared with adjacent non-tumor tissues are different among various patient populations. CONCLUSION In summary, these results indicate that CSIG might play different roles in SMMC7721 and Huh7 cells through regulating P-ERK pathway and mesenchymal-like markers. The differential distribution of CSIG might be an important factor that causes its different functions in SMMC7721 and Huh7 cells. CSIG might play different roles in various patient populations.
Collapse
Affiliation(s)
- Qian Cheng
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| | - Tan-Jun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Zhao Li
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| | - Shi-Hua Hu
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| | - Ding-Bao Chen
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| | - Si-Qi Wang
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| | - Ji-Ye Zhu
- Peking University Institute of Organ Transplantation, Peking University Center of Liver Cancer Diagnosis and Treatment, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China,
| |
Collapse
|
28
|
Schlereth SL, Karlstetter M, Hos D, Matthaei M, Cursiefen C, Heindl LM. Detection of Pro- and Antiangiogenic Factors in the Human Sclera. Curr Eye Res 2018; 44:172-184. [PMID: 30358460 DOI: 10.1080/02713683.2018.1540704] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Avascular tissues can be used to identify antilymph- or antihemangiogenic factors. The human sclera-the outer covering layer of the eye, lacks lymphatic vessels and contains only a superficial network of blood vessels and was used here to identify endogenous antiangiogenic factors. METHODS Expression levels of a panel of 96 known pro- and antiangiogenic factors were analyzed in 12 scleral or conjunctival control samples from normal human donors using real-time PCR. In vitro, scleral homogenate was cocultured with blood- and lymphatic endothelial cells (BECs and LECs) and immunohistochemistry was performed of scleral fibroblasts and BECs. RESULTS Three antiangiogenic factors were significantly upregulated in the human sclera compared to the conjunctiva, including FBLN5 (fibulin 5), SERPINF1 (serpin peptidase inhibitor, clade F, member 1 = pigment epithelium derived factor) and TIMP2 (Tissue inhibitor of metalloproteinases 2). Six proangiogenic factors were significantly downregulated in the sclera, including FLT4 (Fms-related tyrosine kinase 4=VEGF-R3), HGF (hepatocyte growth factor), KIT (CD117 / c-kit), PROX1 (prospero homeobox 1), SEMA3F (semaphorin-3F) and TGFA (transforming growth factor alpha). In vitro, scleral homogenate inhibited the growth of both BECs and LECs. Immunohistochemistry labeling of three major antiangiogenic factors from scleral tissue confirmed TIMP3 and PEDF expression both in scleral fibroblasts and in blood endothelial cells, whereas TIMP2 was not detectable. CONCLUSION Balancing anti- and proangiogenic factors actively regulates human scleral avascularity, inhibits endothelial cell growth in vitro, and thus may help maintaining the vascular privilege of the inner eye.
Collapse
Affiliation(s)
- Simona L Schlereth
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Marcus Karlstetter
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Deniz Hos
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Mario Matthaei
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Claus Cursiefen
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Ludwig M Heindl
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| |
Collapse
|
29
|
Hallmarks of Cancer-Related Newly Prognostic Factors of Oral Squamous Cell Carcinoma. Int J Mol Sci 2018; 19:ijms19082413. [PMID: 30115834 PMCID: PMC6121568 DOI: 10.3390/ijms19082413] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancer, including oral squamous cell carcinoma (OSCC), is the sixth leading malignancy worldwide. OSCC is an aggressive tumor and its prognosis has exhibited little improvement in the last three decades. Comprehensive elucidation of OSCC's molecular mechanism is imperative for early detection and treatment, improving patient survival. Based on broadly accepted notions, OSCC arises from multiple genetic alterations caused by chronic exposure to carcinogens. In 2011, research revealed 10 key alterations fundamental to cancer cell development: sustaining proliferative signaling, evading growth suppressors, avoiding immune destruction, activating invasion and metastasis, tumor-promoting inflammation, enabling replicative immortality, inducing angiogenesis, genome instability and mutation, resisting cell death, and deregulating energetics. This review describes molecular pathological findings on conventional and novel hallmarks of OSCC prognostic factors. In addition, the review summarizes the functions and roles of several molecules as novel OSCC prognosticators.
Collapse
|
30
|
Zhou C, Chen W, Sun J, Atyah M, Yin Y, Zhang W, Guo L, Ye Q, Dong Q, Shi Y, Ren N. Low expression of WW domain-containing oxidoreductase associates with hepatocellular carcinoma aggressiveness and recurrence after curative resection. Cancer Med 2018; 7:3031-3043. [PMID: 29905011 PMCID: PMC6051234 DOI: 10.1002/cam4.1591] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
WW domain-containing oxidoreductase (WWOX), which has a protein-interaction domain and is regarded to be a tumor suppressor, has been known to play an important role in anti-angiogenesis and cancer progression. This study aimed to investigate prognostic values of WWOX expression in hepatocellular carcinoma (HCC) patients after hepatectomy. Additionally, we intended to formulate a valuable prognostic nomogram for HCCs. 182 HCC patients who underwent hepatectomy from January 2009 to January 2010 were enrolled in our study. qRT-PCR, Western blot, and immunohistochemistry on tissue microarrays were then used to determine the expression levels of WWOX. An evaluation of the role of WWOX expression levels in the prognosis and outcome of patients was established. A decrease in the expression of WWOX was found when compared to adjacent tumor-free tissues, which led to worse overall survival (OS) and recurrence-free survival (RFS) and, therefore, was considered as an independent negative factor in the prognosis of HCC. Two nomograms, comprising WWOX, alpha-fetoprotein (AFP), tumor size, and γ-glutamyltransferase (γ-GT), were constructed to obtain superior discriminatory abilities than conventional staging systems in terms of C-index and clinical net benefit on decision curve analysis (DCA) for OS and RFS. Our data suggest that WWOX expression is strongly related to HCC post-resection aggressiveness and recurrence. Additional advanced and accurate predictive model through the incorporation of WWOX into nomogram could help predict OS or RFS for HCC patients.
Collapse
Affiliation(s)
- Chenhao Zhou
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Wanyong Chen
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Department of SurgeryInstitute of Fudan‐Minhang Academic Health SystemMinhang BranchZhongshan HospitalFudan UniversityShanghaiChina
| | - Jialei Sun
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Manar Atyah
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Yirui Yin
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Wentao Zhang
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Lei Guo
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Qinghai Ye
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Qiongzhu Dong
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Yi Shi
- Biomedical Research CentreZhongshan HospitalFudan UniversityShanghaiChina
| | - Ning Ren
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Department of SurgeryInstitute of Fudan‐Minhang Academic Health SystemMinhang BranchZhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
31
|
Hao Q, He L, Zhou J, Yuan Y, Ma X, Pang Z, Li W, Zhang Y, Zhang W, Zhang C, Li M. A dimeric thymosin beta 4 with novel bio-activity protects post-ischemic cardiac function by accelerating vascular endothelial cell proliferation. Int J Cardiol 2018; 261:146-154. [PMID: 29550018 DOI: 10.1016/j.ijcard.2018.03.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/24/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thymosin beta 4 (Tβ4) is a 43-amino-acid peptide with protective properties in myocardium injury. Previously, we produced a recombinant human dimeric Tβ4 (DTβ4). Here, the cardioprotective effects of DTβ4 and the molecular mechanisms underlying its enhanced activity were investigated. METHODS AND RESULTS Echocardiography measurements showed that the cardioprotective effect of DTβ4 in myocardial infarction mice was significantly higher than that of wild-type Tβ4. Corresponding in vitro analyses demonstrated that the enhanced cardioprotection provided by DTβ4 was largely due to increased stimulation of angiogenesis. HPLC analysis, western blotting and qRT-PCR indicated that the enhanced pro-angiogenesis activity of DTβ4 was independent of the protein half-life and the known downstream pathways of wild-type Tβ4. Transcriptome deep sequencing (RNA-seq), BrdU incorporation assays, flow cytometry analysis and RNA interference demonstrated that the enhanced angiogenic activity of DTβ4 depended on MALAT1 (metastasis-associated lung adenocarcinoma transcript 1)-induced proliferation of vascular endothelial cells, which has not been reported for wild-type Tβ4. Moreover, transcription factor activation screening, luciferase promoter reporter assay and immunoprecipitation assay demonstrated that DTβ4 enhanced MALAT1 transcription by inhibiting the degradation of prospero-related homeobox 1 (PROX1). CONCLUSION This study demonstrates the potential applications and the novel bioactivity of the Tβ4 dimer. Moreover, to construct the dimer represents a new method for production of bioactive peptides that may have novel activities.
Collapse
Affiliation(s)
- Qiang Hao
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Lei He
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Jiming Zhou
- Department of Cardiology, 153 Central Hospital of People's Liberation Army, Zhengzhou, China; Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Yuan Yuan
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Xiaowen Ma
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Zhijun Pang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Weina Li
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Yingqi Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China
| | - Wei Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| | - Cun Zhang
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| | - Meng Li
- Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xian, China.
| |
Collapse
|
32
|
Xiao P, Long X, Zhang L, Ye Y, Guo J, Liu P, Zhang R, Ning J, Yu W, Wei F, Yu J. Neurotensin/IL-8 pathway orchestrates local inflammatory response and tumor invasion by inducing M2 polarization of Tumor-Associated macrophages and epithelial-mesenchymal transition of hepatocellular carcinoma cells. Oncoimmunology 2018; 7:e1440166. [PMID: 29900041 DOI: 10.1080/2162402x.2018.1440166] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated that neurotensin (NTS) induces local inflammation and promotes tumor invasion in hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms are not clear. In this study, positive correlations between NTS and interleukin (IL)-8 were identified at both the mRNA and protein levels in 71 fresh HCC tissues and 100 paraffin-embedded HCC tissues. Furthermore, significant correlations were determined among the co-expression of NTS and IL-8, infiltration of inflammatory cells and enhanced epithelial-mesenchymal transition (EMT) of HCC cells. NTS-induced IL-8 production was associated with activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways rather than the protein kinase C (PKC) and phosphoinositide-3 kinase (PI3K) pathways, whose specific antagonists significantly inhibited activation of the NTS/IL-8 pathway. IL-8, which promoted EMT and HCC invasion both in vitro and in vivo, was produced by NTS-induced HCC cells and was effectively attenuated by blocking IL-8 receptors in vitro. Moreover, HCC-derived IL-8 attracted more CD68+ tumor-associated macrophages (TAMs) and CD66b+ polymorphonuclear neutrophils (PMNs) to the local microenvironment, displaying enhanced cytokine secretion and phagocytosis. IL-8 stimulated the M2 polarization of TAMs, which promoted the EMT and invasive potential of HCC cells. Blockage of the IL-8 receptor, NTR1 receptor or both significantly reduced HCC metastases in tumor-bearing mouse models via inhibiting EMT. In summary, aberrant activation of the NTS/IL-8 pathway in HCC dramatically stimulated the invasive potential of HCC cells. HCC-derived IL-8 promoted a pro-oncogenic inflammatory microenvironment by inducing M2-type TAMs and indirectly promoting EMT, which might be a valuable therapeutic target to prevent tumor progression.
Collapse
Affiliation(s)
- Pei Xiao
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xinxin Long
- Department of Oncology, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Lijie Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Jincheng Guo
- Bioinformatics Research Group & Health Big-Data, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Pengpeng Liu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Rui Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Junya Ning
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| |
Collapse
|
33
|
Gao T, Ma C, Li Y, Ju J, Kang X, Cai Y, Sun M. High Expression of Prospero-Related Homeobox-1 (PROX1) Is Associated With Poor Prognosis in Patients With Salivary Adenoid Cystic Carcinoma. J Oral Maxillofac Surg 2018; 76:1440-1446. [PMID: 29406257 DOI: 10.1016/j.joms.2017.12.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/28/2017] [Accepted: 12/30/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Prospero-related homeobox-1 (PROX1) plays an important role in the invasion and metastasis of many human cancers. However, the expression pattern of PROX1 in salivary adenoid cystic carcinoma (SACC) remains unclear. The aim of this study was to investigate PROX1 expression and its prognostic value in SACC. MATERIALS AND METHODS PROX1 expression was determined by immunohistochemistry (IHC) in SACC tissue specimens. Correlations between PROX1 expression and clinicopathologic features were investigated. The Kaplan-Meier method was used to analyze the correlation between PROX1 expression and survival. Independent prognostic factors associated with overall survival (OS) were analyzed using Cox regression analysis. RESULTS The IHC data showed that the PROX1 positivity rate in SACC tissue specimens was significantly higher than that in normal salivary gland tissues (71.1 vs 13.3%; P < .05). PROX1 expression was detected mainly in the nucleolus. In addition, PROX1 expression was correlated with perineural invasion, local regional recurrence, and distant metastasis of patients with SACC (P < .05), and no significant association was found between PROX1 expression and other clinicopathologic parameters. Data indicated that patients with positive PROX1 expression had poor OS compared with those with negative PROX1 expression (P = .0005). Multivariate analysis showed that PROX1 expression, local regional recurrence, and distant metastasis were independent prognostic factors for OS. CONCLUSIONS These findings showed that PROX1 expression was statistically higher in SACC specimens. Positive expression of PROX1 might serve as a potential predictor of prognosis in SACC.
Collapse
Affiliation(s)
- Tao Gao
- Attending Physician, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University; Department of Oral and Maxillofacial Surgery, The First Hospital of Yulin, Stomatological Hospital of Yulin, Shaanxi, China
| | - Chao Ma
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yun Li
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jun Ju
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Xi'an; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an; Department of Otolaryngology Head and Neck Surgery, Navy General Hospital, Beijing, China
| | - Xiangfeng Kang
- Resident, Department of Pediatrics, The First Hospital of Yulin, Shaanxi, China
| | - Yuanlin Cai
- Resident, Department of Emergency Medicine, The First Hospital of Yulin, Shaanxi, China
| | - Moyi Sun
- Professor, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|