1
|
Tang HN, Wang MW, Liu XS, Jiao Y. Personalized treatment of perihilar cholangiocarcinoma based on tumor genetic and molecular characteristics. World J Gastrointest Surg 2024; 16:2769-2773. [PMID: 39351571 PMCID: PMC11438824 DOI: 10.4240/wjgs.v16.i9.2769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 09/18/2024] Open
Abstract
This editorial discusses the article written by Tchilikidi et al that was published in the latest edition of the World Journal of Gastrointestinal Surgery. Genetic and molecular profiling of perihilar cholangiocarcinoma (pCCA) has identified a number of key abnormalities that drive tumor growth and spread, including pyruvate kinase M2, proline rich 11, and transcription factor 7, etc. pCCA has specific genetic and molecular features that can be used to develop personalized treatment plans. Personalized treatment approaches offer new opportunities for effectively targeting the underlying drivers of tumor growth and progression. The findings based on tumor genetic and molecular characteristics highlight the importance of developing personalized treatment strategies.
Collapse
Affiliation(s)
- He-Nan Tang
- Department of Outpatient, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Ming-Wei Wang
- Ministry of Health Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130000, Jilin Province, China
| | - Xue-Song Liu
- Department of Gastrointestinal Surgery, Changchun Central Hospital, Changchun 130000, Jilin Province, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
2
|
E M, Ren F, Yu Y, Li H, Shen C. The role of lncRNAKCNQ1OT1/miR-301b/Tcf7 axis in cardiac hypertrophy. Cardiol Young 2024; 34:1493-1505. [PMID: 38456301 DOI: 10.1017/s1047951124000155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Cardiac hypertrophy, acting as a pathologic process of chronic hypertension and coronary disease, and its underlying mechanisms still need to be explored. Long non-coding RNA (LncRNA) potassium voltage-gated channel subfamily Q member 1 Transcript 1 (KCNQ1OT1) has been implicated in myocardial infarction. However, its role in cardiac hypertrophy remains reported. METHOD To explore the regulated effect of lncRNAKCNQ1OT1 and miR-301b in cardiac hypertrophy, gain-and-lose function assays were tested. The expression of lncRNAKCNQ1OT1 and miR-301b were tested by quantitative real time polymerase chain reaction (qRT-PCR). The levels of transcription factor 7 (Tcf7), Proto-oncogene c-myc (c-myc), Brainnatriureticpeptide (BNP) and β-myosin heavy chain (β-MHC) were detected by Western blot. Additionally, luciferase analysis revealed interaction between lncRNAKCNQ1OT1, BNPβ-MHCmiR-301b, and Tcf7. RESULT LncRNAKCNQ1OT1 overexpression significantly induced cardiac hypertrophy. Furthermore, lncRNAKCNQ1OT1 acts as a sponge for microRNA-301b, which exhibited lower expression in cardiac hypertrophy model, indicating an anti-hypertrophic role. Furthermore, the BNP and β-MHC expression increased, as well as cardiomyocyte surface area, with Ang II treatment, while the effect was repealed by miR-301b. Moreover, the protein expression of Tcf7 was inversely regulated by miR-301b and Antisense miRNA oligonucleotides (AMO)-301b. CONCLUSION Our study has shown that overexpression of lncRNAKCNQ1OT1 could promote the development of cardiac hypertrophy by regulating miR-301b and Tcf7. Therefore, inhibition of lncRNAKCNQ1OT1 might be a potential therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Mingyao E
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Feifei Ren
- Nursing Department of Baicheng Hospital, Baicheng Medical College, Baicheng, China
| | - Yanhua Yu
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Haiyan Li
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| | - Chao Shen
- Department of Pharmacology, Baicheng Medical College, Baicheng, China
| |
Collapse
|
3
|
Papoutsoglou P, Pineau R, Leroux R, Louis C, L'Haridon A, Foretek D, Morillon A, Banales JM, Gilot D, Aubry M, Coulouarn C. TGFβ-induced long non-coding RNA LINC00313 activates Wnt signaling and promotes cholangiocarcinoma. EMBO Rep 2024; 25:1022-1054. [PMID: 38332153 PMCID: PMC10933437 DOI: 10.1038/s44319-024-00075-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Cholangiocarcinoma is a devastating liver cancer characterized by high aggressiveness and therapy resistance, resulting in poor prognosis. Long non-coding RNAs and signals imposed by oncogenic pathways, such as transforming growth factor β (TGFβ), frequently contribute to cholangiocarcinogenesis. Here, we explore novel effectors of TGFβ signalling in cholangiocarcinoma. LINC00313 is identified as a novel TGFβ target gene. Gene expression and genome-wide chromatin accessibility profiling reveal that nuclear LINC00313 transcriptionally regulates genes involved in Wnt signalling, such as the transcriptional activator TCF7. LINC00313 gain-of-function enhances TCF/LEF-dependent transcription, promotes colony formation in vitro and accelerates tumour growth in vivo. Genes affected by LINC00313 over-expression in CCA tumours are associated with KRAS and TP53 mutations and reduce overall patient survival. Mechanistically, ACTL6A and BRG1, subunits of the SWI/SNF chromatin remodelling complex, interact with LINC00313 and affect TCF7 and SULF2 transcription. We propose a model whereby TGFβ induces LINC00313 in order to regulate the expression of hallmark Wnt pathway genes, in co-operation with SWI/SNF. By modulating key genes of the Wnt pathway, LINC00313 fine-tunes Wnt/TCF/LEF-dependent transcriptional responses and promotes cholangiocarcinogenesis.
Collapse
Grants
- Recurrent Funding Institut National de la Santé et de la Recherche Médicale (Inserm)
- Recurrent Funding,PhD felloship Université de Rennes 1 (University of Rennes 1)
- PhD fellowship Conseil Régional de Bretagne (Brittany Council)
- R22026NN,R21011NN Ligue Contre le Cancer (French League Against Cancer)
- R21043NN Fondation ARC pour la Recherche sur le Cancer (ARC)
- C18007NS,C20013NS,C20014NS INCa and ITMO Cancer AVIESAN (Alliance Nationale pour les Sciences de la Vie et de la Santé) dans le cadre du Plan cancer (Non-coding RNA in cancerology: fundamental to translational)
- R21095NN French Ministry of Health and the French National Cancer Institute, PRT-K20-136, CHU Rennes, CLCC Eugene Marquis, Rennes
- FIS PI18/01075,PI21/00922,CPII19/00008 Spanish Carlos III Health Institute (ISCIII) [(FIS PI18/01075, PI21/00922, and Miguel Servet Programme CPII19/00008) cofinanced by "Fondo Europeo de Desarrollo Regional" (FEDER)] and CIBERehd (ISCIII)
- HR17-00601 'la Caixa' Foundation ('la Caixa')
- EU/2019/AMMFt/001 AMMF-The Cholangiocarcinoma Charity
- 06119JB PSC Partners US and PSC Supports UK
- 825510/ESCALON European Union Horizon 2020 Research and Innovation Program
- EU TRANSCAN23-002-2023-129,INCa_18688 Institut National Du Cancer (INCa)
Collapse
Affiliation(s)
- Panagiotis Papoutsoglou
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne University, PSL University, Institut Curie, Centre de Recherche, Paris, France
| | - Raphaël Pineau
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Raffaële Leroux
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Corentin Louis
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Anaïs L'Haridon
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Dominika Foretek
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne University, PSL University, Institut Curie, Centre de Recherche, Paris, France
| | - Antonin Morillon
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne University, PSL University, Institut Curie, Centre de Recherche, Paris, France
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, CIBERehd, Ikerbasque, San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - David Gilot
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, SE-48183, Mölndal, Sweden
| | - Marc Aubry
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Cédric Coulouarn
- Inserm, Univ Rennes, OSS (Oncogenesis, Stress, Signaling) laboratory, UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France.
| |
Collapse
|
4
|
Pawaskar R, Huang KZ, Pham H, Nagrial A, Wong M, O’Neill S, Pleass H, Yuen L, Lam VWT, Richardson A, Pang T, Nahm CB. Systematic Review of Preoperative Prognostic Biomarkers in Perihilar Cholangiocarcinoma. Cancers (Basel) 2024; 16:698. [PMID: 38398089 PMCID: PMC10886549 DOI: 10.3390/cancers16040698] [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: 01/18/2024] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Perihilar cholangiocarcinoma (pCCA) is an uncommon malignancy with generally poor prognosis. Surgery is the primary curative treatment; however, the perioperative mortality and morbidity rates are high, with a low 5-year survival rate. Use of preoperative prognostic biomarkers to predict survival outcomes after surgery for pCCA are not well-established currently. This systematic review aimed to identify and summarise preoperative biomarkers associated with survival in pCCA, thereby potentially improving treatment decision-making. The Embase, Medline, and Cochrane databases were searched, and a systematic review was performed using the PRISMA guidelines. English-language studies examining the association between serum and/or tissue-derived biomarkers in pCCA and overall and/or disease-free survival were included. Our systematic review identified 64 biomarkers across 48 relevant studies. Raised serum CA19-9, bilirubin, CEA, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and tumour MMP9, and low serum albumin were most associated with poorer survival; however, the cutoff values used widely varied. Several promising molecular markers with prognostic significance were also identified, including tumour HMGA2, MUC5AC/6, IDH1, PIWIL2, and DNA index. In conclusion, several biomarkers have been identified in serum and tumour specimens that prognosticate overall and disease-free survival after pCCA resection. These, however, require external validation in large cohort studies and/or in preoperatively obtained specimens, especially tissue biopsy, to recommend their use.
Collapse
Affiliation(s)
- Rishaan Pawaskar
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
| | | | - Helen Pham
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Adnan Nagrial
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Mark Wong
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Siobhan O’Neill
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Henry Pleass
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Lawrence Yuen
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Vincent W. T. Lam
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Macquarie University Medical School, Macquarie University, Sydney, NSW 2145, Australia
| | - Arthur Richardson
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Tony Pang
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Christopher B. Nahm
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| |
Collapse
|
5
|
Razaviyan J, Sirati-Sabet M, Tafti A, Hadavi R, Karima S, Rajabibazl M, Mohammadi-Yeganeh S. Inhibition of MiR-155 Using Exosomal Delivery of Antagomir Can Up-Regulate PTEN in Triple Negative Breast Cancer. Endocr Metab Immune Disord Drug Targets 2024; 24:1664-1676. [PMID: 38424419 DOI: 10.2174/0118715303289859240214103350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The most aggressive form of breast cancer (BC) is Triple-Negative BC (TNBC), with the poorest prognosis, accounting for nearly 15% of all cases. Since there is no effective treatment, novel strategies, especially targeted therapies, are essential to treat TNBC. Exosomes are nano-sized microvesicles derived from cells and transport various intracellular cargoes, including microRNAs (miRNAs). MiRNAs, small non-coding RNA, are an influential factor in the development of cancerous transformations in cells. METHOD Bioinformatics analysis of genes related to TNBC revealed that PTEN plays a crucial role in the disease. Relative expression of this gene was analyzed with RT-qPCR in 14 TNBC clinical samples. Electroporation was used to load miRNA antagomir into exosomes extracted from the conditioned medium. Then, the expression of miR-155 and PTEN was evaluated in MDA-MB-231 cells treated with antagomir-loaded exosomes. RESULTS Based on the bioinformatics analysis, miR-155 is a potent inhibitor of PTEN. Following treatment with antagomir-loaded exosomes, RT-qPCR showed significantly reduced miR- 155 and increased PTEN levels in MDA-MB-231 cells. CONCLUSION Based on the results of this study, exosomes can be effectively used as a cargo of oligonucleotides like miRNA mimics and antagomirs in targeted therapies.
Collapse
Affiliation(s)
- Javad Razaviyan
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Sirati-Sabet
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Tafti
- Department of Biotechnology and Molecular Medicine, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Razie Hadavi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Shi A, Zhao L, Sheng G, Zhang G, Tang Y, Li K, Zhang Z. SMAD4 regulates the progression of cholangiocarcinoma by modulating the expression of STING1. J Cell Mol Med 2023; 27:2547-2561. [PMID: 37488750 PMCID: PMC10468663 DOI: 10.1111/jcmm.17857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
SMAD4 is a tumour suppressor and an important regulator of tumour immune scape which is downregulated in cholangiocarcinoma (CCA). STING1 is a vital sensing factor of abnormal DNA; however, the correlation between SMAD4 and STING1 and the role of the SMAD4-STING1 interaction in the progression of CCA have not yet been evaluated. Public database was analysed to reveal the expression of SMAD4 and STING1. A cohort comprising 50 iCCA, 113 pCCA and 119 dCCA patients was assembled for the study. Immunohistochemistry was employed to evaluate the expression levels of STING1 and SMAD4. In vitro transwell and CCK8 assays, along with luciferase reporter assay, were conducted to analyse the potential regulatory mechanisms of SMAD4 on the expression of STING1. Expression of SMAD4 and STING1 were downregulated in CCA tumours and STING1 expression correlated with SMAD4 expression. The overexpression of SMAD4 was found to suppress the migration, invasion and proliferation capabilities of CCA cells; whereas, the knockdown of SMAD4 enhanced these abilities. Furthermore, it was observed that SMAD4 translocated into the nucleus following TGF-β1 stimulation. Knockdown of SMAD4 resulted in the inhibition of STING1 transcriptional activity, whereas the overexpression of SMAD4 promoted the transcriptional activity of STING1. Clinically, low STING1 and SMAD4 expression indicated poor prognosis in CCA, and simultaneously low expression of STING1 and SMAD4 predicts poorer patient survival. SMAD4 regulates the expression of STING1 through its transcription regulating function. Dual low expression of STING1 and SMAD4 had more power in predicting patient survival. These results indicate that SMAD4-silenced CCA may downregulate its STING1 expression to adapt to the immune system.
Collapse
Affiliation(s)
- An‐da Shi
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Li‐ming Zhao
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Guo‐li Sheng
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Ge‐ning Zhang
- Master of Public HealthThe University of QueenslandBrisbaneQueenslandAustralia
| | - Yong‐chang Tang
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Kang‐shuai Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Zong‐li Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| |
Collapse
|
7
|
Duquette D, Harmon C, Zaborowski A, Michelet X, O'Farrelly C, Winter D, Koay HF, Lynch L. Human Granzyme K Is a Feature of Innate T Cells in Blood, Tissues, and Tumors, Responding to Cytokines Rather than TCR Stimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:633-647. [PMID: 37449888 DOI: 10.4049/jimmunol.2300083] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023]
Abstract
NK cells and CD8 T cells use cytotoxic molecules to kill virally infected and tumor cell targets. While perforin and granzyme B (GzmB) are the most commonly studied lytic molecules, less is known about granzyme K (GzmK). However, this granzyme has been recently associated with improved prognosis in solid tumors. In this study, we show that, in humans, GzmK is predominantly expressed by innate-like lymphocytes, as well as a newly identified population of GzmK+CD8+ non- mucosal-associated invariant T cells with innate-like characteristics. We found that GzmK+ T cells are KLRG1+EOMES+IL-7R+CD62L-Tcf7int, suggesting that they are central memory T and effector memory T cells. Furthermore, GzmK+ cells are absent/low in cord blood, suggesting that GzmK is upregulated with immune experience. Surprisingly, GzmK+ cells respond to cytokine stimuli alone, whereas TCR stimulation downregulates GzmK expression, coinciding with GzmB upregulation. GzmK+ cells have reduced IFN-γ production compared with GzmB+ cells in each T cell lineage. Collectively, this suggests that GzmK+ cells are not naive, and they may be an intermediate memory-like or preterminally differentiated population. GzmK+ cells are enriched in nonlymphoid tissues such as the liver and adipose. In colorectal cancer, GzmK+ cells are enriched in the tumor and can produce IFN-γ, but GzmK+ expression is mutually exclusive with IL-17a production. Thus, in humans, GzmK+ cells are innate memory-like cells that respond to cytokine stimulation alone and may be important effector cells in the tumor.
Collapse
Affiliation(s)
- Danielle Duquette
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cathal Harmon
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | | | - Xavier Michelet
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des Winter
- St. Vincent's University Hospital, Dublin, Ireland
| | - Hui-Fern Koay
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Austria
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
8
|
Han PP, Zhang GQ, Li L, Yue L. Downregulation of USP33 inhibits Slit/Robo signaling pathway and is associated with poor patient survival of glioma. J Neurosurg Sci 2023; 67:113-120. [PMID: 32972109 DOI: 10.23736/s0390-5616.20.04929-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Gliomas are the most common malignant tumors in the central nervous system originating from brain glial cells. Although characterized as highly invasive and highly malignant, few molecular targeting therapies have been developed. Ubiquitin Specific Protease 33 (USP33), a gene encoding a deubiquinating enzyme important in a variety of processes, including Slit-dependent cell migration and beta-2 adrenergic receptor signaling, participates in the development of several malignant tumors, however, its role in the development of glioma has not been evaluated. METHODS Real-time quantitative PCR was performed to examine the expression of USP33 in glioma tissues and cell lines. Immunohistochemistry was performed to determine USP33 expressions in glioma tissue microarray. Transwell assay was performed to analyze the effect of USP33 on glioma cell line migration. The Kaplan-Meier method and log rank test were applied to evaluate the prognostic value of USP33 expression. Univariate and multivariate Cox proportional hazard regression models were used to identify the independent prognostic factors associated with overall survival (OS) or disease-free survival. RESULTS The present study demonstrated that USP33 expression was significantly downregulated in glioma tissues. Lower expression of USP33 was associated with a poorer patient disease-free survival and overall survival. In vitro studies revealed that overexpression of USP33 significantly inhibited the migration ability of glioma cells. Mechanistically, USP33 inhibits glioma cell migration by regulating the function of Slit/Robo signaling pathway. CONCLUSIONS Downregulation of USP33 is associated with poor patient survival of glioma. USP33 inhibits glioma cell migration by Slit/Robo signaling pathway. This mechanism may be applied for development of targeting therapy especially for the high-grade glioma.
Collapse
Affiliation(s)
- Peng-Peng Han
- Second Department of Neurology, Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Gui-Qin Zhang
- Second Department of Neurology, Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Lin Li
- Department of Gynecology Ward II, Weifang Yidu Central Hospital, Weifang, China
| | - Liang Yue
- Department of Surgery Teaching and Research Section, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Haike Mingcheng, Chengdu, China -
| |
Collapse
|
9
|
Yang Y, Zhang X. An overview of extrahepatic cholangiocarcinoma: from here to where? Front Oncol 2023; 13:1171098. [PMID: 37197436 PMCID: PMC10183586 DOI: 10.3389/fonc.2023.1171098] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
Extrahepatic cholangiocarcinoma (eCCA) contains perihilar cholangiocarcinoma and distal cholangiocarcinoma both of which can arise at any point of the biliary tree and originate from disparate anatomical sites. Generally, the incidence of eCCA is increasing globally. Though surgical resection is the principal treatment of choice for the early stages of eCCA, optimal survival remains restricted by the high risk of recurrence when most patients are present with unresectable disease or distant metastasis. Furthermore, both intra- and intertumoral heterogeneity make it laborious to determine molecularly targeted therapies. In this review, we mainly focused on current findings in the field of eCCA, mostly including epidemiology, genomic abnormalities, molecular pathogenesis, tumor microenvironment, and other details while a summary of the biological mechanisms driving eCCA may shed light on intricate tumorigenesis and feasible treatment strategies.
Collapse
|
10
|
Li YC, Li KS, Liu ZL, Tang YC, Hu XQ, Li XY, Shi AD, Zhao LM, Shu LZ, Lian S, Yan ZD, Huang SH, Sheng GL, Song Y, Liu YJ, Huan F, Zhang MH, Zhang ZL. Research progress of bile biomarkers and their immunoregulatory role in biliary tract cancers. Front Immunol 2022; 13:1049812. [PMID: 36389727 PMCID: PMC9649822 DOI: 10.3389/fimmu.2022.1049812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Biliary tract cancers (BTCs), including cholangiocarcinoma and gallbladder carcinoma, originate from the biliary epithelium and have a poor prognosis. Surgery is the only choice for cure in the early stage of disease. However, most patients are diagnosed in the advanced stage and lose the chance for surgery. Early diagnosis could significantly improve the prognosis of patients. Bile has complex components and is in direct contact with biliary tract tumors. Bile components are closely related to the occurrence and development of biliary tract tumors and may be applied as biomarkers for BTCs. Meanwhile, arising evidence has confirmed the immunoregulatory role of bile components. In this review, we aim to summarize and discuss the relationship between bile components and biliary tract cancers and their ability as biomarkers for BTCs, highlighting the role of bile components in regulating immune response, and their promising application prospects.
Collapse
|
11
|
Wang J, Ge F, Yuan T, Qian M, Yan F, Yang B, He Q, Zhu H. The molecular mechanisms and targeting strategies of transcription factors in cholangiocarcinoma. Expert Opin Ther Targets 2022; 26:781-789. [PMID: 36243001 DOI: 10.1080/14728222.2022.2137020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/13/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cholangiocarcinoma consists of a cluster of malignant biliary tumors that tend to have a poor prognosis, ranking as the second most prevalent type of liver cancer, and their incidence rate has increased globally recently. The high-frequency driving mutations of cholangiocarcinoma, such as KRAS/IDH1/ARID1A/P53, imply the epigenetic instability of cholangiocarcinoma, leading to the dysregulation of various related transcription factors, thus affecting the occurrence and development of cholangiocarcinoma. Increasingly evidence indicates that the high heterogeneity and malignancy of cholangiocarcinoma are closely related to the dysregulation of transcription factors which promote cell proliferation, invasion, migration, angiogenesis, and drug resistance through reprogrammed transcriptional networks. It is of great significance to further explore and summarize the role of transcription factors in cholangiocarcinoma. AREAS COVERED This review summarizes the oncogenic or tumor suppressive roles of key transcription factors in regulating cholangiocarcinoma progression and the potential targeting strategies of transcription factors in cholangiocarcinoma. EXPERT OPINION Cholangiocarcinoma is a type of cancer highly influenced by transcriptional regulation, specifically transcription factors and epigenetic regulatory factors. Targeting transcription factors could be a potential and important strategy that is likely to impact future cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Jiao Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fujing Ge
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tao Yuan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Meijia Qian
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangjie Yan
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- The Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- The Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Liu Z, Hu C, Zheng L, Liu J, Li K, Li X, Wang Y, Mu W, Chen T, Shi A, Qiu B, Zhang X, Zhang Z, Xu Y. BMI1 promotes cholangiocarcinoma progression and correlates with antitumor immunity in an exosome-dependent manner. Cell Mol Life Sci 2022; 79:469. [PMID: 35932322 PMCID: PMC11071914 DOI: 10.1007/s00018-022-04500-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 07/04/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is a class of malignant tumors originating from bile duct epithelial cells. Due to difficult early diagnosis and limited treatment, the prognosis of CCA is extremely poor. BMI1 is dysregulated in many human malignancies. However, the prognostic significance and oncogenic role of BMI1 in cholangiocarcinoma (CCA) are not well elucidated. METHODS In the present study, we investigated its clinical importance and the potential mechanisms in the progression of CCA. We detected BMI1 expression in a large CCA cohort. We demonstrated that BMI1 was substantially upregulated in CCA tissues and was identified as an independent prognostic biomarker of CCA. Moreover, overexpression of BMI1 promoted CCA proliferation, migration, and invasion. And BMI1 knockdown could inhibit proliferation and metastases of CCA in vitro and in vitro/vivo validation. Interestingly, we found that CCA-derived exosomes contain BMI1 proteins, which can transfer BMI1 between CCA cells. The unique BMI1-containing exosomes promote CCA proliferation and metastasis through autocrine/paracrine mechanisms. In addition, we demonstrated that BMI1 inhibits CD8+T cell-recruiting chemokines by promoting repressive H2A ubiquitination in CCA cells. CONCLUSIONS BMI1 is an unfavorable prognostic biomarker of CCA. Our data depict a novel function of BMI1 in CCA tumorigenesis and metastasis mediated by exosomes. Besides, BMI1 inhibition may augment immune checkpoint blockade to inhibit tumor progression by activating cell-intrinsic immunity of CCA.
Collapse
Affiliation(s)
- Zengli Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Chunxiao Hu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Lijie Zheng
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Jialiang Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Xingyong Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, 11 Wuyingshan Middle Road, Jinan, 250031, Shandong, China
| | - Yue Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Wentao Mu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Anda Shi
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
13
|
Liu P, Wang Y, Duan L. ZFAND5 Is an Independent Prognostic Biomarker of Perihilar Cholangiocarcinoma. Front Oncol 2022; 12:955670. [PMID: 35912230 PMCID: PMC9326020 DOI: 10.3389/fonc.2022.955670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCholangiocarcinoma (CCA) is a highly aggressive malignancy with extremely poor prognosis. Perihilar CCA (pCCA) is the most common subtype of CCA, but its biomarker study is much more lagged behind other subtypes. ZFAND5 protein can interact with ubiquitinated proteins and promote protein degradation. However, the function of ZFAND5 in cancer progression is rarely investigated, and the role of ZFAND5 in pCCA is never yielded.Materials and MethodsIn this study, we established a pCCA cohort consisting of 72 patients. The expression of ZFAND5 in pCCAs, and the paired liver tissues, intrahepatic bile duct tissues and common bile ducts (CBD) tissues were detected with IHC. ZFAND5 mRNA in pCCAs and CBDs was detected with qRT-PCR. The pCCA cohort was divided into ZFAND5low and ZFAND5high subsets according to the IHC score. The correlations between ZFAND5 expression and clinicopathological parameters were assessed bychi-square test. The prognostic significance of ZFAND5 expression and clinicopathological parameters was estimated by univariate analysis with Kaplan-Meier method, and by multivariate analysis with Cox-regression model.ResultsExpression of ZFAND5 in pCCAs was substantially higher than that in interlobular bile ducts and common bile ducts, but lower than that in liver tissues. The ZFAND5low and ZFAND5high subsets accounted for 44.4% and 55.6% of all pCCAs respectively. ZFAND5 high patients had much lower survival rates than the ZFAND5low patients, with the average survival time as 31.2 months and 19.5 months respectively. ZFAND5 was identified as an independent unfavorable prognostic biomarker of pCCA with multivariate analysis.ConclusionZFAND5 expression was up-regulated in pCCAs compared with the CBDs. We identified ZFAND5 as an independent biomarker of pCCA, which could provide more evidence for the molecular classification of pCCA, and help stratify the high-risk patients based on the molecular features.
Collapse
Affiliation(s)
- Pei Liu
- Department of Plastic Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, China
| | - Yijia Wang
- Department of Plastic Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, China
| | - Lingling Duan
- Department of Health Care, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Lingling Duan, ;
| |
Collapse
|
14
|
Liu Z, Liu J, Chen T, Wang Y, Shi A, Li K, Li X, Qiu B, Zheng L, Zhao L, Shu L, Lian S, Huang S, Zhang Z, Xu Y. Wnt-TCF7-SOX9 axis promotes cholangiocarcinoma proliferation and pemigatinib resistance in a FGF7-FGFR2 autocrine pathway. Oncogene 2022; 41:2885-2896. [DOI: 10.1038/s41388-022-02313-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022]
|
15
|
Liu J, Ren G, Li K, Liu Z, Wang Y, Chen T, Mu W, Yang X, Li X, Shi A, Zhao W, Xu B, Chang J, Guo S, Pan C, Zhou T, Zhang Z, Xu Y. The Smad4-MYO18A-PP1A complex regulates β-catenin phosphorylation and pemigatinib resistance by inhibiting PAK1 in cholangiocarcinoma. Cell Death Differ 2022; 29:818-831. [PMID: 34799729 PMCID: PMC8990017 DOI: 10.1038/s41418-021-00897-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Cholangiocarcinoma (CCA), consisting of three subtypes-intrahepatic (iCCA), perihilar (pCCA), and distal (dCCA), is a highly aggressive cancer arising from the bile duct and has an extremely poor prognosis. Pemigatinib is the only FDA-approved targeted drug for CCA, and the CCA treatment options are substantially insufficient considering its poor prognosis and increasing morbidity. Here, we performed next-generation sequencing (NGS) of 15 pCCAs and 16 dCCAs and detected the expression of SMAD4, a frequently mutated gene, in 261 CCAs. By univariate and multivariate analyses, we identified Smad4 as a favorable prognostic biomarker in iCCA and pCCA. With in vitro and in vivo experiments, we demonstrated that Smad4 suppressed CCA proliferation, migration and invasion by inhibiting β-catenin-S675 phosphorylation and intranuclear translocation. We applied LC-MS/MS and multiple biochemical techniques and identified PP1A as the phosphatase in Smad4-mediated dephosphorylation of PAK1-T423, which is responsible for β-catenin-S675 phosphorylation. Moreover, we demonstrated that MYO18A is the PP1-interacting protein of PP1A for substrate recognition in CCA. MYO18A interacts with PP1A via its RVFFR motif and interacts with Smad4 via CC domain. Patients with coexpression of MYO18A and Smad4 have a more favorable prognosis than other patients. Smad4 enhances Pemigatinib efficiency, and Smad4 knockdown results in Pemigatinib resistance. In conclusion, coexpression of Smad4 and MYO18A is a favorable prognostic indicator for iCCA and pCCA. The Smad4-MYO18A-PP1A complex dephosphorylates PAK1-T423 and thus inhibits β-catenin-S675 phosphorylation and its intranuclear localization. Smad4 suppresses CCA proliferation, migration, invasion, and sensitivity to Pemigatinib by governing the phosphorylation and intracellular localization of β-catenin.
Collapse
Affiliation(s)
- Jialiang Liu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guangli Ren
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China ,grid.27255.370000 0004 1761 1174Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kangshuai Li
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zengli Liu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Wang
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianli Chen
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wentao Mu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoqing Yang
- grid.452422.70000 0004 0604 7301Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, China ,Shandong Medicine and Health Key Laboratory of Clinical Pathology, Jinan, China ,Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, China
| | - Xingyong Li
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China ,grid.27255.370000 0004 1761 1174Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Anda Shi
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bowen Xu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianhua Chang
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Sen Guo
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chang Pan
- grid.27255.370000 0004 1761 1174Department of Emergency, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Zhou
- grid.27255.370000 0004 1761 1174Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
16
|
Ou L, Wang X, Cheng S, Zhang M, Cui R, Hu C, Liu S, Tang Q, Peng Y, Chai R, Xie S, Wang S, Huang W, Wang X. Verdinexor, a Selective Inhibitor of Nuclear Exportin 1, Inhibits the Proliferation and Migration of Esophageal Cancer via XPO1/c-Myc/FOSL1 Axis. Int J Biol Sci 2022; 18:276-291. [PMID: 34975332 PMCID: PMC8692140 DOI: 10.7150/ijbs.66612] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal carcinoma (EC) ranks sixth among cancers in mortality worldwide and effective drugs to reduce EC incidence and mortality are lacking. To explore potential anti-esophageal cancer drugs, we conducted drug screening and discovered that verdinexor, a selective inhibitor of nuclear exportin 1 (XPO1/CRM1), has anti-esophageal cancer effects both in vivo and in vitro. However, the mechanism and role of verdinexor in esophageal cancer remain unknown. In the present study, we observed that verdinexor inhibited the proliferation and migration of EC cells in vitro and suppressed tumor growth in vivo. Additionally, we found that verdinexor induced cleavage of PARP and downregulated XPO1, c-Myc, and FOSL1 expression. RNA-sequence analysis and protein-protein interaction (PPI) analysis revealed that verdinexor regulated the XPO1/c-Myc/FOSL1 axis. The results of immunoprecipitation and proximity ligation assays confirmed that verdinexor disrupted the interaction between XPO1 and c-Myc. Overexpression of c-Myc rescued the inhibition of cell proliferation and cell migration caused by verdinexor. Overexpressed FOSL1 restored the inhibited migration by verdinexor. Taken together, verdinexor inhibited cell proliferation and migration of esophageal cancer via XPO1/c-Myc/FOSL1 axis. Our findings provide a new option for the development of anti-esophageal cancer drugs.
Collapse
Affiliation(s)
- Ling Ou
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Xinyou Wang
- The First District of Gastrointestinal Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shumin Cheng
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Min Zhang
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Ruiqin Cui
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Chunxia Hu
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Shiyi Liu
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qian Tang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| | - Yuying Peng
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| | - Ruihuan Chai
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Shaoxiang Wang
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Wei Huang
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
17
|
Wang Y, Li K, Zhao W, Liu Z, Liu J, Shi A, Chen T, Mu W, Xu Y, Pan C, Zhang Z. Aldehyde dehydrogenase 3B2 promotes the proliferation and invasion of cholangiocarcinoma by increasing Integrin Beta 1 expression. Cell Death Dis 2021; 12:1158. [PMID: 34907179 PMCID: PMC8671409 DOI: 10.1038/s41419-021-04451-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022]
Abstract
Aldehyde dehydrogenases (ALDHs) play an essential role in regulating malignant tumor progression; however, their role in cholangiocarcinoma (CCA) has not been elucidated. We analyzed the expression of ALDHs in 8 paired tumor and peritumor perihilar cholangiocarcinoma (pCCA) tissues and found that ALDH3B1 and ALDH3B2 were upregulated in tumor tissues. Further survival analysis in intrahepatic cholangiocarcinoma (iCCA, n = 27), pCCA (n = 87) and distal cholangiocarcinoma (dCCA, n = 80) cohorts have revealed that ALDH3B2 was a prognostic factor of CCA and was an independent prognostic factor of iCCA and pCCA. ALDH3B2 expression was associated with serum CEA in iCCA and dCCA, associated with tumor T stage, M stage, neural invasion and serum CA19-9 in pCCA. In two cholangiocarcinoma cell lines, overexpression of ALDH3B2 promoted cell proliferation and clone formation by promoting the G1/S phase transition. Knockdown of ALDH3B2 inhibited cell migration, invasion, and EMT in vitro, and restrained tumor metastasis in vivo. Patients with high expression of ALDH3B2 also have high expression of ITGB1 in iCCA, pCCA, and dCCA at both mRNA and protein levels. Knockdown of ALDH3B2 downregulated the expression of ITGB1 and inhibited the phosphorylation level of c-Jun, p38, and ERK. Meanwhile, knockdown of ITGB1 inhibited the promoting effect of ALDH3B2 overexpression on cell proliferation, migration, and invasion. ITGB1 is also a prognostic factor of iCCA, pCCA, and dCCA and double-positive expression of ITGB1 and ALDH3B2 exhibits better performance in predicting patient prognosis. In conclusion, ALDH3B2 promotes tumor proliferation and metastasis in CCA by regulating the expression of ITGB1 and upregulating its downstream signaling pathway. The double-positive expression of ITGB1 and ALDH3B2 serves as a better prognostic biomarker of CCA.
Collapse
Affiliation(s)
- Yue Wang
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Kangshuai Li
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Wei Zhao
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Zengli Liu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Jialiang Liu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Anda Shi
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Tianli Chen
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Wentao Mu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Yunfei Xu
- grid.27255.370000 0004 1761 1174Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Chang Pan
- Department of Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China. .,Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China. .,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| |
Collapse
|
18
|
Inhibitory Effect of Astaxanthin on Gene Expression Changes in Helicobacter pylori-Infected Human Gastric Epithelial Cells. Nutrients 2021; 13:nu13124281. [PMID: 34959833 PMCID: PMC8708722 DOI: 10.3390/nu13124281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection promotes gastric carcinogenesis by increasing oxidative stress, inflammation, and dysregulation of cell survival and proliferation of gastric epithelial cells. Astaxanthin (ASTX), a bioactive carotenoid, exhibits antioxidant and anticancer effects by modulating aberrant signaling pathways that lead to dysregulation of cell death and proliferation. To elucidate the molecular mechanism of H. pylori-induced gastric carcinogenesis and to examine the inhibitory effect of ASTX on H. pylori-induced gastric epithelial cell gene expression changes, we performed comparative RNA-sequencing (RNA-Seq) analysis for H. pylori-infected gastric epithelial cells treated with or without ASTX. RNA-Seq results reveal that differentially expressed genes (DEGs) in H. pylori-infected cells were mainly associated with the Wnt/β-catenin signaling pathway, which is related to cell proliferation. ASTX significantly reversed H. pylori-induced transcriptional alterations of the key mediators involved in β-catenin signaling, notably, porcupine (gene symbol, PORCN), spermine oxidase (SMOX), bone morphogenetic protein (BMP) and activin membrane-bound inhibitor (BAMBI), SMAD family member 4 (SMAD4), transforming growth factor-β1 (TGFB1), Fos-like 1 (FOSLI), and c-myc (MYC). We suggest that ASTX may be a potential therapeutic agent that can suppress H. pylori-induced proliferation-associated gene expression changes, in part, by counter-regulating the Wnt/β-catenin signaling pathway.
Collapse
|
19
|
Wang Y, Chen T, Li K, Mu W, Liu Z, Shi A, Liu J, Zhao W, Lian S, Huang S, Pan C, Zhang Z. Recent Advances in the Mechanism Research and Clinical Treatment of Anti-Angiogenesis in Biliary Tract Cancer. Front Oncol 2021; 11:777617. [PMID: 34778094 PMCID: PMC8581488 DOI: 10.3389/fonc.2021.777617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/11/2021] [Indexed: 12/25/2022] Open
Abstract
Biliary tract cancers (BTCs), including cholangiocarcinoma (CCA) and gallbladder cancer (GC), are malignancies originating from the biliary tract with poor prognosis. In the early stage of BTCs, surgery is the only choice for cure. Unfortunately, most patients with BTC are diagnosed at an advanced stage and lose the opportunity for surgery. For many advanced solid tumors, antiangiogenic therapy has achieved encouraging results. While most clinical studies on antiangiogenic therapy in advanced BTCs have shown an excellent disease control rate (DCR), the improvement in overall survival (OS) is controversial. Understanding how the relevant signaling molecules influence the angiogenic response and the functional interaction is necessary for the formulation of new treatment regimens and the selection of enrolled patients. In this review, we aim to summarize and discuss the latest advances in antiangeogenesis for BTCs, mainly focusing on the molecular mechanism of angiogenesis in BTCs and the therapeutic effects from clinical trials. Furthermore, the horizon of antiangiogenesis for BTCs is highlighted.
Collapse
Affiliation(s)
- Yue Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wentao Mu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Anda Shi
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jialiang Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuo Lian
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaohui Huang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chang Pan
- Department of Emergency, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
20
|
Xu L, Wang P, Zhang W, Li W, Liu T, Che X. Dual-Specificity Phosphatase 11 Is a Prognostic Biomarker of Intrahepatic Cholangiocarcinoma. Front Oncol 2021; 11:757498. [PMID: 34660327 PMCID: PMC8513537 DOI: 10.3389/fonc.2021.757498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Background Cholangiocarcinoma (CCA), including intrahepatic (iCCA), perihilar (pCCA), and distal (dCCA) CCA, is a highly aggressive malignancy originating from bile duct. The prognosis of CCA is very poor, and the biomarker study is unsatisfactory compared with other common cancers. Materials and methods In our study, we investigated the expression of dual-specificity phosphatase 11(DUSP11) in eight pairs of iCCAs, pCCAs, and dCCAs, and their corresponding tumor-adjacent tissues, as well as their tumor-adjacent tissues with qPCR. Moreover, we investigated the expression of DUSP11 in 174 cases of CCAs with immunohistochemistry, including 74 iCCAs, 64 pCCAs, and 36 dCCAs. We classified these patients into subsets with low and high expressions of DUSP11, and evaluated the correlations between the DUSP11 subsets and clinicopathological factors. With univariate and multivariate analyses, we assessed the correlation between DUSP11 and the overall survival (OS) rates in these CCA patients. Results In all the CCA subtypes, DUSP11 was elevated in CCAs compared with their paired adjacent tissues. In iCCA, pCCA, and dCCA, the percentages of DUSP11 high expression were 44.59%, 53.85%, and 55.56%, respectively. In iCCA, high DUSP11 expression was significantly associated with an advanced T stage and a poor prognosis. However, the prognostic value of DUSP11 in pCCA and dCCA was not significant. To decrease the statistical error caused by the small sample size of the dCCA cohort, we merged pCCA and dCCA into extracellular CCA (eCCA). In the 101 cases of eCCA, DUSP11 expression was also not significantly associated with the prognosis. Conclusions DUSP11 expression was associated with tumor infiltration and the OS rate in iCCA, but not in pCCA and dCCA. DUSP11 was an independent biomarker of iCCA indicating a poor prognosis. Our results suggested that a high expression of DUSP11 was a post-operational risk factor, and detecting DUSP11 could guide the individual treatment for patients with CCA.
Collapse
Affiliation(s)
- Lin Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Peng Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiran Li
- Department of Oncology Rehabilitation, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Tao Liu
- Department of Oncology Rehabilitation, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.,Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Hirano KI, Hosokawa H, Koizumi M, Endo Y, Yahata T, Ando K, Hozumi K. LMO2 is essential to maintain the ability of progenitors to differentiate into T-cell lineage in mice. eLife 2021; 10:e68227. [PMID: 34382935 PMCID: PMC8360648 DOI: 10.7554/elife.68227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/31/2021] [Indexed: 12/20/2022] Open
Abstract
Notch signaling primarily determines T-cell fate. However, the molecular mechanisms underlying the maintenance of T-lineage potential in pre-thymic progenitors remain unclear. Here, we established two murine Ebf1-deficient pro-B cell lines, with and without T-lineage potential. The latter expressed lower levels of Lmo2; their potential was restored via ectopic expression of Lmo2. Conversely, the CRISPR/Cas9-mediated deletion of Lmo2 resulted in the loss of the T-lineage potential. Introduction of Bcl2 rescued massive cell death of Notch-stimulated pro-B cells without efficient LMO2-driven Bcl11a expression but was not sufficient to retain their T-lineage potential. Pro-B cells without T-lineage potential failed to activate Tcf7 due to DNA methylation; Tcf7 transduction restored this capacity. Moreover, direct binding of LMO2 to the Bcl11a and Tcf7 loci was observed. Altogether, our results highlight LMO2 as a crucial player in the survival and maintenance of T-lineage potential in T-cell progenitors via the regulation of the expression of Bcl11a and Tcf7.
Collapse
Affiliation(s)
- Ken-ichi Hirano
- Department of Immunology, Tokai University School of MedicineIseharaJapan
| | - Hiroyuki Hosokawa
- Department of Immunology, Tokai University School of MedicineIseharaJapan
- Institute of Medical Sciences, Tokai UniversityIseharaJapan
| | - Maria Koizumi
- Department of Immunology, Tokai University School of MedicineIseharaJapan
| | - Yusuke Endo
- Laboratory of Medical Omics Research, Kazusa DNA Research InstituteKisarazuJapan
- Department of Omics Medicine, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Takashi Yahata
- Institute of Medical Sciences, Tokai UniversityIseharaJapan
- Department of Innovative Medical Science, Tokai University School of MedicineIseharaJapan
| | - Kiyoshi Ando
- Institute of Medical Sciences, Tokai UniversityIseharaJapan
- Department of Hematology and Oncology, Tokai University School of MedicineIseharaJapan
| | - Katsuto Hozumi
- Department of Immunology, Tokai University School of MedicineIseharaJapan
| |
Collapse
|
22
|
Hu C, Zhang Y, Zhang M, Li T, Zheng X, Guo Q, Zhang X. Exosomal Cripto-1 Serves as a Potential Biomarker for Perihilar Cholangiocarcinoma. Front Oncol 2021; 11:730615. [PMID: 34434900 PMCID: PMC8380828 DOI: 10.3389/fonc.2021.730615] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Perihilar cholangiocarcinoma (PHCCA) has a poor prognosis, mainly due to diagnosis at an advanced stage. Cripto-1 functions as an oncogene and is highly expressed in several human cancers, however, its clinical application in PHCCA is poorly understood. Herein, we identified that Cripto-1 was released by PHCCA cells via exosomes in vitro and in vivo. Furthermore, an ELISA method was developed to detect exosomal Cripto-1 in the serum of 115 PHCCA patients, 47 cholangitis patients and 65 healthy controls, and it was found that exosomal Cripto-1 was increased in PHCCA patients and associated with metastasis. Compared with traditional serum tumor markers, CA19-9 and CEA, exosomal Cripto-1 demonstrated a larger area under ROC curve for PHCCA diagnosis. The cutoff value of exosomal Cripto-1 was 0.82, achieving a sensitivity of 79.1% and a specificity of 87.5%. As expected, exosomal Cripto-1 levels in immunohistochemically Cripto-1-high cases were significantly elevated compared to in Cripto-1-low cases. When measured 1-week postoperatively, Cripto-1 levels decreased on average from 1.25(0.96-3.26) to 0.85(0.62-1.82). Immunohistochemistry analysis showed Cripto-1 expression was negatively correlated with E-cadherin and was an independent prognostic biomarker for poor survival in PHCCA patients. In conclusion, exosomal Cripto-1 in sera can reflect its expression in the tissue of PHCAA patients and has the potential be a non-invasive biomarker for diagnosis and prognosis of PHCCA.
Collapse
Affiliation(s)
- Chunxiao Hu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yanli Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, China
| | - Mengjiao Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zheng
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, China
| | - Qining Guo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
23
|
Vallejo A, Erice O, Entrialgo-Cadierno R, Feliu I, Guruceaga E, Perugorria MJ, Olaizola P, Muggli A, Macaya I, O'Dell M, Ruiz-Fernandez de Cordoba B, Ortiz-Espinosa S, Hezel AF, Arozarena I, Lecanda F, Avila MA, Fernandez-Barrena MG, Evert M, Ponz-Sarvise M, Calvisi DF, Banales JM, Vicent S. FOSL1 promotes cholangiocarcinoma via transcriptional effectors that could be therapeutically targeted. J Hepatol 2021; 75:363-376. [PMID: 33887357 DOI: 10.1016/j.jhep.2021.03.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/10/2021] [Accepted: 03/25/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Cholangiocarcinoma (CCA) is a neoplasia of the biliary tract driven by genetic, epigenetic and transcriptional mechanisms. Herein, we investigated the role of the transcription factor FOSL1, as well as its downstream transcriptional effectors, in the development and progression of CCA. METHODS FOSL1 was investigated in human CCA clinical samples. Genetic inhibition of FOSL1 in human and mouse CCA cell lines was performed in in vitro and in vivo models using constitutive and inducible short-hairpin RNAs. Conditional FOSL1 ablation was done using a genetically engineered mouse (GEM) model of CCA (mutant KRAS and Trp53 knockout). Follow-up RNA and chromatin immunoprecipitation (ChIP) sequencing analyses were carried out and downstream targets were validated using genetic and pharmacological inhibition. RESULTS An inter-species analysis of FOSL1 in CCA was conducted. First, FOSL1 was found to be highly upregulated in human and mouse CCA, and associated with poor patient survival. Pharmacological inhibition of different signalling pathways in CCA cells converged on the regulation of FOSL1 expression. Functional experiments showed that FOSL1 is required for cell proliferation and cell cycle progression in vitro, and for tumour growth and tumour maintenance in both orthotopic and subcutaneous xenograft models. Likewise, FOSL1 genetic abrogation in a GEM model of CCA extended mouse survival by decreasing the oncogenic potential of transformed cholangiocytes. RNA and ChIP sequencing studies identified direct and indirect transcriptional effectors such as HMGCS1 and AURKA, whose genetic and pharmacological inhibition phenocopied FOSL1 loss. CONCLUSIONS Our data illustrate the functional and clinical relevance of FOSL1 in CCA and unveil potential targets amenable to pharmacological inhibition that could enable the implementation of novel therapeutic strategies. LAY SUMMARY Understanding the molecular mechanisms involved in cholangiocarcinoma (bile duct cancer) development and progression stands as a critical step for the development of novel therapies. Through an inter-species approach, this study provides evidence of the clinical and functional role of the transcription factor FOSL1 in cholangiocarcinoma. Moreover, we report that downstream effectors of FOSL1 are susceptible to pharmacological inhibition, thus providing new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Adrián Vallejo
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain
| | - Oihane Erice
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | | | - Iker Feliu
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain
| | - Elizabeth Guruceaga
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; University of Navarra, Centre for Applied Medical Research, Computational Biology Program, Pamplona, Spain; ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria J Perugorria
- University of the Basque Country, San Sebastian, Spain; Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital -, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Spain
| | - Paula Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital -, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Alexandra Muggli
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Irati Macaya
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain
| | - Michael O'Dell
- University of Rochester Medical Centre, Rochester, NY, USA
| | | | - Sergio Ortiz-Espinosa
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain
| | - Aram F Hezel
- University of Rochester Medical Centre, Rochester, NY, USA
| | - Imanol Arozarena
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Cancer Signalling Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Fernando Lecanda
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, Instituto de Salud Carlos III), Madrid, Spain; University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain
| | - Matias A Avila
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Spain; University of Navarra, Centre for Applied Medical Research, Hepatology Program, Pamplona, Spain
| | - Maite G Fernandez-Barrena
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Spain; University of Navarra, Centre for Applied Medical Research, Hepatology Program, Pamplona, Spain
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | | | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital -, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Spain; Ikerbasque, Basque Foundation for Sciences, Bilbao, Spain
| | - Silve Vicent
- University of Navarra, Centre for Applied Medical Research, Program in Solid Tumours, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, Instituto de Salud Carlos III), Madrid, Spain; University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain.
| |
Collapse
|
24
|
Saikosaponin-D Alleviates Renal Inflammation and Cell Apoptosis in a Mouse Model of Sepsis via TCF7/FOSL1/MMP9 Inhibition. Mol Cell Biol 2021; 41:e0033221. [PMID: 34309413 DOI: 10.1128/mcb.00332-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Evidence exists reporting that Saikosaponin-d can prevent experimental sepsis, and this study aims to illustrate the molecular events underlying its renoprotective effects on lipopolysaccharide (LPS)-induced renal inflammation simulating sepsis. Through network pharmacology analysis and bioinformatics analysis, we identified that saikosaponin-d may influence sepsis development by mediating TCF7. Dual luciferase reporter gene and ChIP assays were used to explore the interactions between TCF7, FOSL1 and MMP9. The experimental data suggested that Saikosaponin-d attenuated LPS-induced renal injury, as evidenced by reduced the production of proinflammatory cytokines as well as cell apoptosis in the renal tissues of LPS-induced mice. Mechanically, Saikosaponin-d inhibited FOSL1 by inhibiting TCF7, which reduced the expression of inflammatory factors in renal cells. TCF7 activated the FOSL1 expression and consequently promoted the expression of MMP9. Also, Saikosaponin-d reduced cell apoptosis and the expression of inflammatory factors by inhibiting the TCF7/FOSL1/MMP9 axis in vivo. In conclusion, Saikosaponin-d suppresses FOSL1 transcription by downregulating TCF7, thereby inhibiting MMP9 expression and ultimately reducing the renal inflammation and cell apoptosis induced by sepsis.
Collapse
|
25
|
Chen A, Liu Z, Wu Q, Li H. Disheveled-associated activator of morphogenesis 2 promotes invasion of colorectal cancer by activating PAK1 and promoting MMP7 expression. Genes Genomics 2021; 43:923-935. [PMID: 33974241 DOI: 10.1007/s13258-021-01111-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Disheveled-associated activator of morphogenesis (DAAM) family, including DAAM1 and DAAM2, is key regulators in Wnt signaling pathway. Although the oncogenic role of Wnt signaling pathway in colorectal cancer (CRC) was investigated in several lines, the expression and function of DAAM in CRC are still obscure. OBJECTIVE To investigate the expression and function of DAAM in CRC. METHODS DAAM1 and DAAM2 expression in high grade dysplasia (HGD), CRCs and corresponding adjacent tissues were detected with qRT-PCR and immunohistochemistry (IHC). The prognostic significance of DAAM1/2 were estimated with univariate and multivariate analyses. Moreover, the correlations between clinicopathological factors and DAAM were evaluated with the χ2 test. With experiments in vitro, we investigated the function of DAAM2 in CRC cell proliferation and invasion, and investigated the underlying mechanism of how DAAM2 facilitated CRC invasion. RESULTS DAAM2, instead of DAAM1, was substantially up-regulated in CRCs compared with paired adjacent normal tissues and HGDs. The ratio of high DAAM1 and DAAM2 expression accounted for 44.83% and 46.31%, respectively. High DAAM2, instead of DAAM1, was a risk factor indicating an unfavorable prognosis of CRC. In multivariate analysis, high DAAM2 was identified as an independent prognostic biomarker of CRC predicting poor prognosis. With experiments in vitro, DAAM2 promoted invasion of CRC cells via activating PAK1 and promoting the expression of MMP7, suggesting an essential role of DAAM2 in CRC invasion. CONCLUSIONS High expression of DAAM2, instead of DAAM1, indicated an unfavorable prognosis of CRC independently, which suggested that detecting DAAM2 can help define the high-risk patients. DAAM2 activated PAK1 and promoted MMP7 expression and facilitated the invasion of CRC cells, indicating that DAAM2 may be a potential drug target of CRC.
Collapse
Affiliation(s)
- Aimei Chen
- Department of Chinese Traditional Medicine, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Zhiqiang Liu
- Department of Chinese Traditional Medicine, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Quanyan Wu
- Department of Chinese Traditional Medicine, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Hailin Li
- Department of General Surgery, Qilu Hospital of Shandong University, #107 Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
26
|
Li X, Xie M, Yin S, Xiong Z, Mao C, Zhang F, Chen H, Jin L, Lan P, Lian L. Identification and Validation of a Six Immune-Related Genes Signature for Predicting Prognosis in Patients With Stage II Colorectal Cancer. Front Genet 2021; 12:666003. [PMID: 34017356 PMCID: PMC8129521 DOI: 10.3389/fgene.2021.666003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/14/2021] [Indexed: 01/20/2023] Open
Abstract
Background Immune-related genes (IRGs) play important roles in the tumor immune microenvironment and can affect the prognosis of cancer. This study aimed to construct a novel IRG signature for prognostic evaluation of stage II colorectal cancer (CRC). Methods Gene expression profiles and clinical data for stage II CRC patients were collected from the Cancer Genome Atlas and Gene Expression Omnibus database. Univariate, multivariate Cox regression, and least absolute shrinkage and selection operator regression were used to develop the IRG signature, namely IRGCRCII. A nomogram was constructed, and the “Cell Type Identification by Estimating Relative Subsets of RNA Transcripts” (CIBERSORT) method was used to estimate immune cell infiltration. The expression levels of genes and proteins were validated by qRT-PCR and immunohistochemistry in 30 pairs of primary stage II CRC and matched normal tissues. Results A total of 466 patients with stage II CRC were included, and 274 differentially expressed IRGs were identified. Six differentially expressed IRGs were detected and used to construct the IRGCRCII signature, which could significantly stratify patients into high-risk and low-risk groups in terms of disease-free survival in three cohorts: training, test, and external validation (GSE39582). Receiver operating characteristics analysis revealed that the area under the curves of the IRGCRCII signature were significantly greater than those of the OncotypeDX colon signature at 1 (0.759 vs. 0.623), 3 (0.875 vs. 0.629), and 5 years (0.906 vs. 0.698) disease-free survival, respectively. The nomogram performed well in the concordance index (0.779) and calibration curves. The high-risk group had a significantly higher percentage of infiltrated immune cells (e.g., M2 macrophages, plasma cells, resting mast cells) than the low-risk group. Finally, the results of qRT-PCR and immunohistochemistry experiments performed on 30 pairs of clinical specimens were consistent with bioinformatics analysis. Conclusion This study developed and validated a novel immune prognostic signature based on six differentially expressed IRGs for predicting disease-free survival and immune status in patients with stage II CRC, which may reflect immune dysregulation in the tumor immune microenvironment.
Collapse
Affiliation(s)
- Xianzhe Li
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minghao Xie
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shi Yin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhizhong Xiong
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chaobin Mao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fengxiang Zhang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huaxian Chen
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Longyang Jin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Lian
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Zhu C, He X, Chen K, Huang Z, Yao A, Tian X, You Y, Zeng M. LncRNA NBR2 aggravates hepatoblastoma cell malignancy and promotes cell proliferation under glucose starvation through the miR-22/TCF7 axis. Cell Cycle 2021; 20:575-590. [PMID: 33651649 DOI: 10.1080/15384101.2021.1885236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatoblastoma (HB) is the most commonly seen pediatric liver malignancy. With frequent mutations in CTNNB1 gene that encodes β-catenin, hepatoblastoma has been considered as a Wnt/β-catenin-activated malignant tumor. Altered glucose metabolism upon nutrient deprivation (glucose starvation) might also be a critical event in hepatoblastoma carcinogenesis. The present study provides a lncRNA NBR2/miR-22/TCF7 axis modulating proliferation, invasion, migration, and apoptosis of hepatoblastoma cells upon glucose starvation through Wnt and downstream TCF7 signaling pathways. The expression of NBR2 is significantly increased within hepatoblastoma tissue samples; moreover, under incubation with 0 mM glucose (glucose starvation), NBR2 expression is significantly upregulated. NBR2 silencing not only inhibited hepatoblastoma cell viability, invasion, and migration under normal culture condition but also promoted the cell apoptosis under glucose starvation. NBR2 silencing in hepatoblastoma cells also decreased TCF7 mRNA expression and TCF7 protein levels, as well as the protein levels of the cell cycle, glucose entrapment, and EMT markers. miR-22 is directly bound to both NBR2 and TCF7; lncRNA NBR2 counteracted miR-22-mediated repression on TCF7 via acting as a ceRNA. The effects of NBR2 silencing on TCF7 expression, hepatoblastoma cell phenotype, and cell cycle, glucose entrapment, and EMT markers were all significantly reversed by miR-22 inhibition. In conclusion, lncRNA NBR2 aggravates hepatoblastoma cell malignancy through competing with TCF7 for miR-22 binding, therefore counteracting miR-22-mediated repression on TCF7. LncRNA NBR2 might be a promising target to inhibit hepatoblastoma cell proliferation under glucose starvation.
Collapse
Affiliation(s)
- Chengguang Zhu
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xiangling He
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Keke Chen
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Zhijun Huang
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Anqi Yao
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xin Tian
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Yalan You
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Minhui Zeng
- Department of Pediatric Hematology and Oncology, Children's Medical Center of Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
28
|
Li Z, Liu J, Chen T, Sun R, Liu Z, Qiu B, Xu Y, Zhang Z. HMGA1-TRIP13 axis promotes stemness and epithelial mesenchymal transition of perihilar cholangiocarcinoma in a positive feedback loop dependent on c-Myc. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:86. [PMID: 33648560 PMCID: PMC7923631 DOI: 10.1186/s13046-021-01890-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/21/2021] [Indexed: 01/04/2023]
Abstract
Background Cholangiocarcinoma is a highly malignant cancer with very dismal prognosis. Perihilar cholangiocarcinoma(pCCA) accounts for more than 50% of all cholangiocarcinoma and is well-characterized for its low rate of radical resection. Effects of radiotherapy and chemotherapy of pCCA are very limited. Methods Here we screened potential biomarkers of pCCA with transcriptome sequencing and evaluated the prognostic significance of HMGA1 in a large cohort pCCA consisting of 106 patients. With bioinformatics and in vitro/vivo experiments, we showed that HMGA1 induced tumor cell stemness and epithelial-mesenchymal-transition (EMT), and thus facilitated proliferation, migration and invasion by promoting TRIP13 transcription. Moreover, TRIP13 was also an unfavorable prognostic biomarker of pCCA, and double high expression of HMGA1/TRIP13 could predict prognosis more sensitively. TRIP13 promoted pCCA progression by suppressing FBXW7 transcription and stabilizing c-Myc. c-Myc in turn induced the transcription and expression of both HMGA1 and TRIP13, indicating that HMGA-TRIP13 axis facilitated pCCA stemness and EMT in a positive feedback pathway. Conclusions HMGA1 and TRIP13 were unfavorable prognostic biomarkers of pCCA. HMGA1 enhanced pCCA proliferation, migration, invasion, stemness and EMT, by inducing TRIP13 expression, suppressing FBXW7 expression and stabilizing c-Myc. Moreover, c-Myc can induce the transcription of HMGA1 and TRIP13, suggesting that HMGA-TRIP13 axis promoted EMT and stemness in a positive feedback pathway dependent on c-Myc. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01890-1.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.,Department of General Surgery, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan, China
| | - Jialiang Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Rongqi Sun
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
29
|
Chen T, Li K, Liu Z, Liu J, Wang Y, Sun R, Li Z, Qiu B, Zhang X, Ren G, Xu Y, Zhang Z. WDR5 facilitates EMT and metastasis of CCA by increasing HIF-1α accumulation in Myc-dependent and independent pathways. Mol Ther 2021; 29:2134-2150. [PMID: 33601056 DOI: 10.1016/j.ymthe.2021.02.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/15/2020] [Accepted: 02/11/2021] [Indexed: 12/30/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive malignancy with extremely poor prognoses. The oncogenic role and prognostic value of c-Myc in CCA is not well elucidated. WD repeat domain 5 (WDR5) is a critical regulatory factor directly interacting with c-Myc to regulate c-Myc recruitment at chromosomal locations, but the interaction of WDR5 and c-Myc in CCA was uncovered. In our study, we detected WDR5 and c-Myc expression in all CCA types, including intrahepatic (iCCA), perihilar (pCCA), and distal (dCCA) CCA, and evaluated their prognostic significance. Consequently, we demonstrated that WDR5 was significantly correlated with poor prognosis of CCA and that WDR5 and c-Myc co-expression was a more sensitive prognostic factor. With in vitro and in vivo experiments and bioinformatics, we showed that WDR5 interacted with the Myc box IIIb (MBIIIb) motif of c-Myc and facilitated Myc-induced HIF1A transcription, thereby promoting the epithelial-mesenchymal transition (EMT), invasion, and metastasis of CCA. Moreover, WDR5 enhanced hypoxia-inducible factor 1 subunit α (HIF-1α) accumulation by binding with histone deacetylase 2 (HDAC2) and increasing histone 3 lysine 4 acetylation (H3K4ac) deacetylation of the prolyl hydroxylase domain protein 2 (PHD2) promoter, resulting in the attenuation of chromatin opening and PHD2 expression, and eventually leading to HIF-1α stabilization and accumulation. In conclusion, WDR5 facilitated EMT and metastasis of CCA by increasing HIF-1α accumulation in a Myc-dependent pathway to promote HIF-1α transcription and a Myc-independent pathway to stabilize HIF-1α.
Collapse
Affiliation(s)
- Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jialiang Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yue Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Rongqi Sun
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhipeng Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of General Surgery, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoming Zhang
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| | - Guangli Ren
- Department of General Surgery, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
30
|
LncRNA MNX1-AS1 promotes progression of intrahepatic cholangiocarcinoma through the MNX1/Hippo axis. Cell Death Dis 2020; 11:894. [PMID: 33093444 PMCID: PMC7581777 DOI: 10.1038/s41419-020-03029-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/13/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Long non-coding RNAs (lncRNAs) have extremely complex roles in the progression of intrahepatic cholangiocarcinoma (ICC) and remain to be elucidated. By cytological and animal model experiments, this study demonstrated that the expression of lncRNA MNX1-AS1 was remarkably elevated in ICC cell lines and tissues, and was highly and positively correlated with motor neuron and pancreas homeobox protein 1 (MNX1) expression. MNX1-AS1 significantly facilitated the proliferation, migration, invasion, and angiogenesis in ICC cells in vitro, and remarkably promoted tumor growth and metastasis in vivo. Further study revealed that MNX1-AS1 promoted the expression of MNX1 via recruiting transcription factors c-Myc and myc-associated zinc finger protein (MAZ). Furthermore, MNX1 upregulated the expression of Ajuba protein via binding to its promoter region, and subsequently, Ajuba protein suppressed the Hippo signaling pathway. Taken together, our results uncovered that MNX1-AS1 can facilitate ICC progression via MNX1-AS1/c-Myc and MAZ/MNX1/Ajuba/Hippo pathway, suggesting that MNX1-AS1 may be able to serve as a potential target for ICC treatment.
Collapse
|
31
|
Shi H, Luo K, Huang W. Bone Marrow Stromal Antigen 2 is a Potential Unfavorable Prognostic Factor for High-Grade Glioma. Onco Targets Ther 2020; 13:8723-8734. [PMID: 32943880 PMCID: PMC7468947 DOI: 10.2147/ott.s258631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/07/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Bone marrow stromal antigen 2 (BST2) is considered as a transmembrane glycoprotein and plays essential roles in innate immunity. It has been recently reported that up-regulation of BST2 was associated with the development of breast carcinoma. However, the clinical significance of BST2 in glioma has not been identified. The purpose of the present study is to explore the expression pattern and the role of BST2 in the progression of high-grade glioma. METHODS Expression levels of BST2 were tested in glioma tissues by analyzing the GEO database and immunohistochemistry staining. The prognostic role of BST2 in glioma was evaluated through univariate and multivariate analyses. In vitro and in vivo assays were conducted to confirm the role of BST2 on promoting glioma proliferation. RESULTS The mRNA level of BST2 was higher in glioma tissues than that in nontumorous brain tissues. High protein level of BST2 was correlated with larger tumor size and advanced WHO grade. Glioma patients with a high BST2 level had worse overall survival. In addition, BST2 was defined as an independent risk factor for glioma prognosis. Cellular and xenograft studies revealed that BST2 can significantly promote glioma proliferation. CONCLUSION Our study revealed that a high BST2 expression level was closely related to the unfavorable clinical features and poor prognosis of high-grade glioma patients. BST2 may serve as an invaluable prognostic indicator and novel therapeutic target for glioma treatment considering its membrane localization.
Collapse
Affiliation(s)
- Haiping Shi
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| | - Ke Luo
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| | - Wei Huang
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, People’s Republic of China
| |
Collapse
|
32
|
Ren N, Tian Z, Sun H, Lu X. Dynamin 2 Is Correlated with Recurrence and Poor Prognosis of Papillary Thyroid Cancer. Med Sci Monit 2020; 26:e924590. [PMID: 32827429 PMCID: PMC7461653 DOI: 10.12659/msm.924590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Papillary thyroid cancer (PTC) is the most common histological type of thyroid cancer. Most PTC patients have favorable outcomes, but 10% of patients still have distant metastases at presentation or during follow-up. Dynamin 2 (DNM2) is the only DNM ubiquitously expressed in human tissues, but its expression and clinical significance in PTC is still unknown. Material/Methods In our study, we investigated the expression of DNM2 in 112 cases of PTC and classified the patients into low and high expression of DNM2. The clinical significance of DNM2 was evaluated by assessing its correlation with the clinicopathological parameters with the chi-square method. The correlations between DNM2 expression and the disease-free survival rate or overall survival rate were assessed with the Kaplan-Meier method and the log-rank test. The independent prognostic factors of PTC were determined by the Cox-regression hazard model. Results Patients with low and high DNM2 expression accounted for 75% and 25% respectively in the 112 patients with PTC. High DNM2 expression was significantly associated with recurrence (P=0.014) and poor prognosis (P=0.004). In addition to tumor stage, DNM2 expression was an independent prognostic biomarker of PTC, indicating an unfavorable prognosis. Conclusions DNM2 was an independent PTC biomarker indicating more likely recurrence and poorer prognosis. Detecting DNM2 expression may help to select the high-risk patients for adjuvant therapy.
Collapse
Affiliation(s)
- Ning Ren
- Department of Otorhinolaryngology, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Zhenmin Tian
- Department of Clinical Laboratory, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Hongmei Sun
- Department of General Surgery, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Xiaofei Lu
- Department of Thyroid and Breast Surgery, The Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China (mainland)
| |
Collapse
|
33
|
Fang X, Zhang D, Zhao W, Gao L, Wang L. Dishevelled Associated Activator Of Morphogenesis (DAAM) Facilitates Invasion of Hepatocellular Carcinoma by Upregulating Hypoxia-Inducible Factor 1α (HIF-1α) Expression. Med Sci Monit 2020; 26:e924670. [PMID: 32772041 PMCID: PMC7437242 DOI: 10.12659/msm.924670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background The dishevelled associated activator of morphogenesis (DAAM) family, consisting of DAAM1 and DAAM2, is an important component of the Wnt signal pathway. Previous studies have suggested that DAAM2 reduces Von Hippel-Lindau (VHL) expression by promoting its ubiquitination, but the correlation between DAAM and HIF-1α in hepatocellular carcinoma (HCC) has not been studied. Material/Methods In our study, expression of DAAM1 and DAAM2 in HCCs and tumor-adjacent liver tissues was assessed with qRT-PCR and immunohistochemistry. Correlations between DAAM1/2 and the clinicopathologic variables were evaluated with the Chi-square test. With univariate and multivariate analysis, we further evaluated the prognostic significance of DAAM1 and DAAM2. Using in vitro experiments, we assessed the functions of DAAM1 and DAAM2 in invasion and proliferation in different HCC cell lines and investigated their underlying mechanisms. Results DAAM1 and 2 overexpression were 18.8% and 48.7%, respectively, of the whole cohort. mRNAs of DAAM2 in HCCs were substantially higher than mRNAs in liver tissues, while DAAM1 mRNA had no marked difference. High DAAM2 expression was notably associated with advanced T stage (P=0.032), TNM stage (P=0.032), and overall survival (OS) rate (P=0.004). DAAM 2 knockdown promoted VHL accumulation and subsequent HIF-1α down-regulation in HCC cells. In HCC specimens, DAAM2 expression was also negatively correlated with VHL and positively associated with HIF-1α. Moreover, HIF-1α was required in DAAM2-induced invasion of HCC cells. Conclusions DAAM2, rather than DAAM1, was able to predict prognosis of HCC. DAAM2 decreased VHL expression and consequently upregulated HIF-1α, eventually facilitating invasion of HCC.
Collapse
Affiliation(s)
- Xiaoxu Fang
- Gastrointestinal Department, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Dandan Zhang
- Urinary Surgery Department, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Wei Zhao
- Department of Ultrasonography, YIDU Central Hospital, Weifang, Shandong, China (mainland)
| | - Longfei Gao
- Dezhou Prison Hospital of Shandong Province, Dezhou, Shandong, China (mainland)
| | - Lanping Wang
- Department of Surgery, 2nd Hospital Sffiliated to Shandong 1st Medical University, Taian, Shandong, China (mainland)
| |
Collapse
|
34
|
Sun H, Zhang M, Li L, Huang Z. ALDH3B1 Is an Independent Prognostic Biomarker of Lung Adenocarcinoma. Technol Cancer Res Treat 2020; 19:1533033820946018. [PMID: 32744175 PMCID: PMC7412899 DOI: 10.1177/1533033820946018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Lung cancer is the leading cause of cancer-related death, and adenocarcinoma is the most common type of lung cancer. Although emerging evidence implicates the role of several aldehyde dehydrogenases in cancer progression, the expression and clinical significance of aldehyde dehydrogenase 3B1 in lung adenocarcinoma has never been studied. Materials: In our study, the expression of aldehyde dehydrogenase 3B1 in 250 cases of lung adenocarcinoma was detected with immunohistochemistry, and the patients were further divided into subgroups with different aldehyde dehydrogenase 3B1 expression. Using real-time polymerase chain reaction, we investigated the aldehyde dehydrogenase 3B1 messenger RNA in 20 lung adenocarcinoma and paired normal lung tissues. With the χ2 test, we evaluated the clinical significance of aldehyde dehydrogenase 3B1 by analyzing its correlation with the clinicopathological factors. Propensity score matching was performed to balance the baseline of cohort. With univariate and multivariate analyses, we screened the prognostic factors of lung adenocarcinoma and identified the independent prognostic factors before and after the propensity score matching. Results: Aldehyde dehydrogenase 3B1 expression was significantly associated with the sex and age of patients, tumor size, and histological grade. High expression of aldehyde dehydrogenase 3B1 predicted the poor prognosis (P = .003). Moreover, male patients (P = .020), large tumor size (P = .009), advanced T stage (P = .001), positive lymphatic invasion (P < .001), and advanced tumor–node–metastasis stage (P < .001) were all the prognostic factors for unfavorable outcome. Aldehyde dehydrogenase 3B1 was an independent prognostic biomarker of lung adenocarcinoma, indicating the poor prognosis. In addition, after balancing the baseline characteristics by propensity score matching, we also demonstrated that aldehyde dehydrogenase 3B1 was an independent prognostic biomarker of lung adenocarcinoma (P = .007). Conclusions: Aldehyde dehydrogenase 3B1 was an independent prognostic biomarker of lung adenocarcinoma, indicating the unfavorable prognosis. Postoperative detection of aldehyde dehydrogenase 3B1 would help stratify the high-risk patients with lung adenocarcinoma and guide individual treatment.
Collapse
Affiliation(s)
- Hongmei Sun
- Department of Gastrointestinal Surgery, YIDU Central Hospital, Weifang, China.,Hongmei Sun and Mingying Zhang contributed equally to the paper
| | - Mingying Zhang
- Department of Child Healthcare, YIDU Central Hospital, Weifang, China.,Hongmei Sun and Mingying Zhang contributed equally to the paper
| | - Li Li
- Department of Respiratory Medicine, YIDU Central Hospital, Weifang, China
| | - Zongwen Huang
- Department of Respiratory Medicine, 970th Hospital of Chinese PLA, Weihai, China
| |
Collapse
|
35
|
Jiang Z, Yan Y, Dong J, Duan L. PD-1 expression on uveal melanoma induces tumor proliferation and predicts poor patient survival. Int J Biol Markers 2020; 35:50-58. [PMID: 32686562 DOI: 10.1177/1724600820943610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Uveal melanoma is one of the most common primary intraocular malignant tumors with poor prognosis and limited treatments. Programmed cell death receptor-1 (PD-1) blockade represents the primary treatment strategy of immune checkpoint inhibition; however, there is a lack of studies on whether PD-1 expression in primary (ocular) uveal melanoma affects tumor progression. METHODS PD-1 expression in 82 cases of primary (ocular) uveal melanoma was detected by immunohistochemistry. The clinical significance of PD-1 expression was evaluated using univariate and multivariate analysis. PD-1 overexpression and knockdown studies were conducted in C918 and Mum-2B cell lines to analyze the effect of PD-1 expression on tumor cell proliferation and intracellular cell signaling transduction. real-time qPCR (RT-qPCR) and western blot analysis were performed to investigate the gene expression level. CCK8 assays were performed to examine the cell proliferation ability. RESULTS High expression of primary (ocular) intratumor PD-1 was associated with poor patient survival. Moreover, PD-1 expression was correlated with the largest tumor diameter. PD-1 expression and optic nerve invasion were independent prognostic risk factors. PD-1 overexpression in uveal melanoma cell lines promoted tumor cell proliferation, while knockdown of PD-1 inhibited cell proliferation capacity. CONCLUSION Our study established the role of PD-1 in the progression of uveal melanoma and provided a new potential treatment selection for uveal melanoma.
Collapse
Affiliation(s)
- Zhongming Jiang
- Department of Ophthalmology, Linyi Central Hospital, Linyi, Shandong, China
| | - Yuexiang Yan
- The Second Division of General Surgery, Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Juan Dong
- Department of Ophthalmology, Sunshine Union Hospital, Weifang, Shandong, China
| | - Lingling Duan
- Department of Health Clinic, Central Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
36
|
Kang Z, Guo L, Zhu Z, Qu R. Identification of prognostic factors for intrahepatic cholangiocarcinoma using long non-coding RNAs-associated ceRNA network. Cancer Cell Int 2020; 20:315. [PMID: 32694937 PMCID: PMC7364620 DOI: 10.1186/s12935-020-01388-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating amount of evidence has highlighted the important roles of long non-coding RNAs (lncRNAs) acting as competing endogenous RNAs (ceRNAs) in tumor pathogenesis. However, the roles of long non coding RNAs (lncRNAs) in the lncRNA-related ceRNA network of intrahepatic cholangiocarcinoma (ICC) still remain enigmatic. The current study aims to identify prognostic factors in the lncRNA-related ceRNA network of ICC. Methods The transcriptome sequencing data of lncRNAs, messenger RNA (mRNA) and microRNA (miR) were downloaded from the SRA and TCGA databases. Differentially expressed lncRNAs (DElncRNAs), DEmiRs and DEmRNAs were identified and adopted to construct an lncRNA-miR-mRNA ceRNA network. ICC-associated DEmRNAs were adopted to construct the protein–protein interaction (PPI) network. The expression of the top 6 genes in the hub module was validated with mRNA transcriptome sequencing data and ICC-related gene expression dataset GSE45001, followed by GO and KEGG pathway enrichment analysis. The relationship between the hub gene-associated ceRNA network and the overall survival of patients with ICC was predicted by conducting a Kaplan–Meier survival analysis. Results Sixty co-expressed DEmRNAs were identified in the ceRNA network. The top 6 hub genes consisted of downregulated FOS, IGF2, FOXO1 and NTF3, upregulated IGF1R, and insignificantly downregulated HGF in ICC tissues, when compared to that of normal adjacent tissues, followed by the successful construction of lncRNA-miR-hub network consisting of 86 ceRNA modules. MME-AS1 and hsa-miR-182 were associated with overall survival in ICC patients. FOS, IGF1R, IGF2, FOXO1, and NTF3 might target “TGF-β signaling pathway”, “the hedgehog signaling pathway”, “retinol metabolism”, or “type II diabetes mellitus” pathways respectively. Conclusion These results indicate that FOS, IGF1R, IGF2, FOXO1, and NTF3 were useful prognostic factors in determining the prognosis of patients with ICC.
Collapse
Affiliation(s)
- Zhichen Kang
- Department of Rehabilitation, the Second Hospital of Jilin University, Changchun, 130022 People's Republic of China
| | - Lixin Guo
- Department of Rehabilitation, the Second Hospital of Jilin University, Changchun, 130022 People's Republic of China
| | - Zhuo Zhu
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, 130022 People's Republic of China
| | - Rongfeng Qu
- Department of Hematology and Oncology, the Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130022 Jilin People's Republic of China
| |
Collapse
|
37
|
Sun QH, Yin ZX, Li Z, Tian SB, Wang HC, Zhang FX, Li LP, Zheng CN, Kong S. miR-874 inhibits gastric cancer cell proliferation by targeting SPAG9. BMC Cancer 2020; 20:522. [PMID: 32503577 PMCID: PMC7275545 DOI: 10.1186/s12885-020-06994-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
Background microRNAs (miRNAs) play essential roles in the development and progression of gastric cancer (GC). Although aberrant miR-874 expression has been reported in various human cancers, its role in GC remains obscure. Methods miR-874 expression was assessed by real-time quantitative polymerase chain reaction (RT-qPCR) in 62 matched GC and adjacent normal tissues, as well as in GC cell lines and immortalized human gastric epithelial cells. CCK8 assay, colony formation assay, and flow cytometry were used to assess the role of miR-874 in GC cell proliferation and apoptosis in vitro. Additionally, to determine the effects of miR-874 on GC cell proliferation and apoptosis in vivo, BALB/c nude mice were injected with GC cells transfected with a miR-874 mimic. The role of miR-874 in SPAG9 expression was assessed by luciferase assay, Western blotting, and RT-qPCR. Results miR-874 was downregulated in GC cell lines and tissues. miR-874 overexpression in GC cells led to inhibition of cell proliferation and induction of apoptosis. Moreover, SPAG9 was identified as a direct miR-874 target, the expression of which was suppressed by miR-874. SPAG9 overexpression markedly promoted GC cell proliferation. Conclusions miR-874 inhibited cell proliferation and induced apoptosis in GC cells. SPAG9 downregulation was crucial for the tumor-suppressive effects of miR-874. Hence, the miR-874/SPAG9 axis could serve as a novel therapeutic target in GC.
Collapse
Affiliation(s)
- Qin Hui Sun
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Zong Xiu Yin
- Department of Respiration Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, China
| | - Zhi Li
- Department of Operating Room, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, China
| | - Shu Bo Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road No.324, Jinan, 250021, China
| | - Hong Chang Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road No.324, Jinan, 250021, China
| | - Fang Xu Zhang
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
| | - Le Ping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road No.324, Jinan, 250021, China
| | - Chun Ning Zheng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road No.324, Jinan, 250021, China
| | - Shuai Kong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road No.324, Jinan, 250021, China.
| |
Collapse
|
38
|
Xu X, Wang Y, Xue F, Guan E, Tian F, Xu J, Zhang H. BST2 Promotes Tumor Growth via Multiple Pathways in Hepatocellular Carcinoma. Cancer Invest 2020; 38:329-337. [PMID: 32427495 DOI: 10.1080/07357907.2020.1769125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Bone marrow stromal antigen 2 (BST2) is a transmembrane glycoprotein and plays an essential role in innate immunity. Here we firstly found that BST2 expression was significantly elevated in hepatocellular carcinoma (HCC) tissues. High BST2 was closely related to the larger tumor size and more tumor number. Moreover, HCC patients with higher expression of BST2 had poorer overall survival and BST2 was identified as an independent unfavorable prognosis factor. Finally, we demonstrated that BST2 can promote proliferation capacity of tumor cells. In conclusion, HCC patients with higher BST2 expression were more predisposed to poorer clinical symptoms and unfavorable prognosis.
Collapse
Affiliation(s)
- Xiaoguang Xu
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Yu Wang
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Fangxi Xue
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Encui Guan
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Feng Tian
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Jian Xu
- Department of Gastroenterology, Linyi Central Hospital, Shandong, Linyi, China
| | - Hongjin Zhang
- Department of Endocrinology and Nephrology, The Third People's Hospital of Linyi, Shandong, Linyi, China
| |
Collapse
|
39
|
Liu X, Yang M, Guo Y, Lu X. Annexin A10 is a novel prognostic biomarker of papillary thyroid cancer. Ir J Med Sci 2020; 190:59-65. [PMID: 32451762 DOI: 10.1007/s11845-020-02263-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/16/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The incidence of thyroid cancer (TC) is increasing rapidly worldwide. The target therapy for papillary TC (PTC) is limited, and the studies of PTC prognostic biomarkers are not common. As a new member of annexin A (ANXA) family, the function and clinical significance of ANXA10 in PTC have not been well investigated. METHODS Expressions of all the 12 ANXA members were detected with qPCR in 12 PTC tissues, and the ANXA10 mRNAs in PTCs and their adjacent normal thyroid tissues were compared. The subcellular location and expression of ANXA10 in 121 PTC patients were investigated with immunohistochemistry, which further classified the patients into subgroups with low or high ANXA10. The clinical significance and prognostic value of ANXA10 were estimated by analyzing its correlation with clinical factors and overall survival rates by the chi-squared test, univariate analyses, and multivariate analyses. RESULTS ANXA10 had the highest expression in PTCs among all the ANXA members. Moreover, ANXA10 was significantly upregulated in PTC compared with normal thyroid tissues. The PTC patients with low and high expression of ANXA10 took up 70.25% (85/121) and 29.75% (36/121), respectively. ANXA10 expression was associated with tumor size, differentiation, and overall survival rates of PTC. ANXA10 was an independent prognostic biomarker predicting the poor outcome of PTC. CONCLUSIONS ANXA10 expression was upregulated in PTC, and it was an independent prognostic biomarker of PTC, suggesting that ANXA10 may be a promising target for individual treatment of ANXA10.
Collapse
Affiliation(s)
- Xiaoyun Liu
- Department of Cardiology, YIDU Central Hospital, Weifang, China
| | - Meijing Yang
- Department of Cardiology, YIDU Central Hospital, Weifang, China
| | - Yang Guo
- Department of Cardiology, YIDU Central Hospital, Weifang, China
| | - Xiaofei Lu
- Department of Thyroid and Breast Surgery, Central Hospital affiliated to Shandong First Medical University, # 105 Jiefang Road, Jinan, 250013, China.
| |
Collapse
|
40
|
Li KS, Zhu XD, Liu HD, Zhang SZ, Li XL, Xiao N, Liu XF, Xu B, Lei M, Zhang YY, Shi WK, Cao MQ, Xu YF, Tang ZY, Sun HC. NT5DC2 promotes tumor cell proliferation by stabilizing EGFR in hepatocellular carcinoma. Cell Death Dis 2020; 11:335. [PMID: 32382041 PMCID: PMC7206140 DOI: 10.1038/s41419-020-2549-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
Most hepatocellular carcinoma (HCC) patients are diagnosed at an advanced stage; however, the effect of systemic therapy on advanced HCC remains undetermined. Therefore, new treatment targets must be identified. We analyzed Gene Expression Omnibus datasets from two HCC patient cohorts and found that NT5DC2 was associated with vascular invasion and poor survival. In two hepatoma cell lines, NT5DC2 overexpression promoted HCC cell proliferation and clone formation in vitro and promoted tumor growth in vivo. Coimmunoprecipitation assays and liquid chromatography with tandem mass spectrometry analysis revealed that NT5DC2 bound directly to epidermal growth factor receptor (EGFR). NT5DC2 upregulated EGFR expression by downregulating EGFR ubiquitination and preventing its degradation via the ubiquitin-proteasome pathway but did not upregulate its transcription. EGFR upregulation activated downstream signal transduction, which played a critical role in the protumor effects of NT5DC2. Erlotinib, a small-molecule inhibitor of EGFR, blocked the effect of NT5DC2 in promoting HCC cell proliferation. In a cohort of 79 patients who underwent curative resection for HCC, NT5DC2 expression in the tumors was associated with larger tumors and microvascular invasion. NT5DC2 expression was also independently associated with recurrence-free survival. The present study demonstrated for the first time that NT5DC2 promotes tumor cell proliferation in HCC and may serve as a potential molecular target for treating HCC. EGFR blockage could be used to treat selected patients with NT5DC2 upregulation.
Collapse
Affiliation(s)
- Kang-Shuai Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Hong-Da Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, Jiangsu province, China
| | - Shi-Zhe Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Xiao-Long Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Nan Xiao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Xue-Feng Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Bin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Ming Lei
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Yuan-Yuan Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Wen-Kai Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Man-Qing Cao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Yun-Fei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua Xi Road, 250012, Jinan, China
| | - Zhao-You Tang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 200032, Shanghai, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 200032, Shanghai, China.
| |
Collapse
|
41
|
Niu R, Tang Y, Xi Y, Jiang D. High Expression of Krüppel-like Factor 7 Indicates Unfavorable Clinical Outcomes in Patients with Lung Adenocarcinoma. J Surg Res 2020; 250:216-223. [PMID: 32092599 DOI: 10.1016/j.jss.2019.12.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/08/2019] [Accepted: 12/29/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Krüppel-like factor 7 (KLF7), which belongs to the KLF family of zinc finger transcription factors, plays a critical role in regulating gene expression. It was reported that KLF7 overexpression was closely related to the progression of gastric cancer. However, the role of KLF7 in lung adenocarcinoma (LAC) has not been elucidated. The aim of our study is to investigate the expression pattern of KLF7 and explore whether the KLF7 expression is correlated with unfavorable clinical outcome of patients with LAC. MATERIALS AND METHODS The protein and mRNA levels of KLF7 were examined in LAC tissues by using immunohistochemistry staining and quantitative reverse transcription polymerase chain reaction, respectively. The prognostic role of KLF7 in patients with LAC was assessed using univariate and multivariate analyses. Clinical outcomes were evaluated by Kaplan-Meier analysis and logrank test. The effects of KLF7 on lung cancer cells were investigated through cellular experiments. RESULTS KLF7 expression was elevated in LAC tissues compared with adjacent normal tissues. High protein level of KLF7 was correlated with larger tumor size, positive lymph node metastasis, and advanced TNM stage. Moreover, patients with LAC with higher expression level of KLF7 had poorer overall survival, and KLF7 was identified as an unfavorable independent prognosis factor. Knockdown of KLF7 can suppress the proliferation and invasion abilities of cancer cells. CONCLUSIONS Our studies revealed that high KLF7 expression level was significantly associated with the poorer clinical outcomes of patients with LAC, indicating the potential role of KLF7 as a novel prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Rungui Niu
- Department of Geratology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Yanlei Tang
- Department of Chest Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China.
| | - Daowen Jiang
- Department of Chest Surgery, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Chen MY, Li ZP, Sun ZN, Ma M. USP9X promotes the progression of hepatocellular carcinoma by regulating beta-catenin. Ir J Med Sci 2020; 189:865-871. [PMID: 32065347 DOI: 10.1007/s11845-020-02199-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/05/2020] [Indexed: 01/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is among the malignant tumors with highest mortality. The role of USP9X in the carcinogenesis of HCC has not yet been determined. In this study, USP9X was found significantly highly expressed in the intratumor tissues. Expression of intratumor USP9X was associated with tumor size and microvascular invasion while USP9X is independent risk factor of HCC disease-free survival and overall survival. In vitro studies revealed that knockdown of USP9X significantly inhibited the proliferation of HCC cells. Mechanically, USP9X promotes HCC cell proliferation by regulating the expression of beta-catenin. The results of the present study demonstrated that high expression of USP9X in intratumoral cells is associated with poor HCC prognosis, which may serve as a potential target for an adjuvant therapy.
Collapse
Affiliation(s)
- Mei-Yuan Chen
- Department of General Surgery, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Zi-Ping Li
- Department of Neurology, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Zhao-Na Sun
- Department of Cardiology, Weifang Yidu Central Hospital, Weifang, 262500, Shandong, China
| | - Ming Ma
- Department of Oncology, Linyi People's Hospital, Linyi, 276000, Shandong, China.
| |
Collapse
|
43
|
Sun R, Chen T, Li M, Liu Z, Qiu B, Li Z, Xu Y, Pan C, Zhang Z. PTPN3 suppresses the proliferation and correlates with favorable prognosis of perihilar cholangiocarcinoma by inhibiting AKT phosphorylation. Biomed Pharmacother 2020; 121:109583. [DOI: 10.1016/j.biopha.2019.109583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/17/2019] [Accepted: 10/25/2019] [Indexed: 10/25/2022] Open
|
44
|
Sun R, Liu Z, Qiu B, Chen T, Li Z, Zhang X, Xu Y, Zhang Z. Annexin10 promotes extrahepatic cholangiocarcinoma metastasis by facilitating EMT via PLA2G4A/PGE2/STAT3 pathway. EBioMedicine 2019; 47:142-155. [PMID: 31492557 PMCID: PMC6796529 DOI: 10.1016/j.ebiom.2019.08.062] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Cholangiocarcinoma (CCA), consisting of intrahepatic (IHCCA), perihilar (PHCCA), and distal (DCCA) CCA, is a type of highly aggressive malignancy with a very dismal prognosis. Potential biomarkers and drug targets of CCA are urgently needed. As a new member of the Annexin (ANXA) family, the role of ANXA10 in the progression and prognosis of CCA is unknown. Methods Potential PHCCA biomarkers were screened by transcriptome sequencing of 5 pairs of PHCCA and adjacent tissues. The clinical significance of ANXA10 was evaluated by analyzing its correlation with clinicopathological variables, and the prognostic value of ANXA10 was evaluated with univariate and multivariate analyses. The function of ANXA10 in the epithelial-mesenchymal transition (EMT), proliferation, invasion and metastasis was detected with in vitro and in vivo experiments. Moreover, we screened the key molecule in ANXA10-induced CCA progression by mRNA sequencing and evaluated the correlation between PLA2G4A and ANXA10. The effect of PLA2G4A downstream signaling, including Cyclooxygenase 2, Prostaglandin E2(PGE2) and Signal transducer and activator of transcription 3(STAT3), on EMT and metastasis was further detected with in vitro and in vivo experiments. Findings ANXA10 expression was upregulated in PHCCA and DCCA but not in IHCCA. High ANXA10 expression was significantly associated with poor tumor differentiation and prognosis. ANXA10 promoted the proliferation, migration and invasion of the PHCCA cells. PLA2G4A expression was regulated by ANXA10 and high PLA2G4A predicted poor prognosis in PHCCA and DCCA. ANXA10 facilitated EMT and promoted metastasis by upregulating PLA2G4A expression, thus increasing PGE2 levels and activating STAT3. Interpretation ANXA10 was an independent prognostic biomarker of PHCCA and DCCA but not IHCCA. ANXA10 promoted the progression of PHCCA and facilitated metastasis by promoting the EMT process via the PLA2G4A/PGE2/STAT3 pathway. ANXA10, PLA2G4A and their downstream molecules, such as COX2 and PGE2, may be promising drug targets of PHCCA and DCCA.
Collapse
Affiliation(s)
- Rongqi Sun
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Tianli Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zhipeng Li
- Department of General Surgery, Shandong Provincial Hospital, Jinan, China
| | - Xiaoming Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|