1
|
Łukaszewicz-Zając M, Mroczko B. Claudins-Promising Biomarkers for Selected Gastrointestinal (GI) Malignancies? Cancers (Basel) 2023; 16:152. [PMID: 38201579 PMCID: PMC10778544 DOI: 10.3390/cancers16010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Despite recent() improvements in diagnostic ability() and treatment() strategies for patients() with neoplastic disease(), gastrointestinal (GI) cancers(), such() as colorectal, gastric, pancreatic, and oesophageal cancers(), are still common() malignancies and the leading() cause() of cancer() deaths worldwide(), with a high frequency of recurrence and metastasis as well as poor patient() prognosis. There is a link() between the secretion of proteolytic enzymes that degrade the extracellular matrix and the pathogenesis of GI tumours. Recent() findings have focused() on the potential() significance() of selected claudins (CLDNs) in the pathogenesis and prognosis of GI cancers(). Tight junctions (TJs) have been proven to play an important role() in maintaining cell() polarity and permeability. A number of authors have recently() revealed that TJ proteins, particularly() selected CLDNs, are related() to inflammation and the development() of various tumours, including GI malignancies. This review() presents general() characteristics and the involvement() of selected CLDNs in the progression() of GI malignancies, with a focus() on the potential() application() of these proteins in the diagnosis() and prognosis of colorectal cancer() (CRC), gastric cancer() (GC), pancreatic cancer() (PC), and oesophageal cancer() (EC). Our review() indicates that selected CLDNs, particularly() CLDN1, 2, 4, 7, and 18, play a significant() role() in the development() of GI tumours and in patient() prognosis. Furthermore, selected CLDNs may be of value() in the design() of therapeutic() strategies for the treatment() of recurrent tumours.
Collapse
Affiliation(s)
- Marta Łukaszewicz-Zając
- Department of Biochemical Diagnostics, Medical University, Waszyngtona 15 a, 15-269 Bialystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University, Waszyngtona 15 a, 15-269 Bialystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University, 15-269 Bialystok, Poland
| |
Collapse
|
2
|
Maesaka F, Kuwada M, Horii S, Kishi S, Fujiwara-Tani R, Mori S, Fujii K, Mori T, Ohmori H, Owari T, Miyake M, Nakai Y, Tanaka N, Bhawal UK, Luo Y, Kondoh M, Fujimoto K, Kuniyasu H. Hypomethylation of CLDN4 Gene Promoter Is Associated with Malignant Phenotype in Urinary Bladder Cancer. Int J Mol Sci 2022; 23:ijms23126516. [PMID: 35742959 PMCID: PMC9224287 DOI: 10.3390/ijms23126516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/04/2022] Open
Abstract
The tight junction (TJ) protein claudin-4 (CLDN4) is overexpressed in bladder urothelial carcinoma (BUC) and correlates with cancer progression. However, the mechanism of CLDN4 upregulation and promotion of malignant phenotype is not clear. Here, we analyzed 157 cases of BUC and investigated the hypomethylation of CpG island in the CLDN4 promoter DNA and its correlation with cancer progression. In hypomethylated cases, CLDN4 expression, cell proliferation, stemness, and epithelial-mesenchymal transition were increased. Treatment of three human BUC cell lines with the demethylating agent aza-2′-deoxycytidine (AZA) led to excessive CLDN4 expression, and, specifically, to an increase in CLDN4 monomer that is not integrated into the TJ. The TJ-unintegrated CLDN4 was found to bind integrin β1 and increase stemness, drug resistance, and metastatic ability of the cells as well as show an anti-apoptosis effect likely via FAK phosphorylation, which reduces upon knockdown of CLDN4. Thus, CLDN4 is overexpressed in BUC by an epigenetic mechanism and the high expression enhances the malignant phenotype of BUC via increased levels of TJ-unintegrated CLDN4. CLDN4 promoter DNA methylation is expected to be a novel indicator of BUC malignant phenotype and a new therapeutic target.
Collapse
Affiliation(s)
- Fumisato Maesaka
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Masaomi Kuwada
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Shohei Horii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Takuya Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
| | - Takuya Owari
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Makito Miyake
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Ujjal Kumar Bhawal
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India
| | - Yi Luo
- Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China;
| | - Masuo Kondoh
- Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University, 6-1 Yamadaoka, Suita 565-0871, Osaka, Japan;
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Nara, Japan; (T.O.); (M.M.); (Y.N.); (N.T.); (K.F.)
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (F.M.); (M.K.); (S.H.); (S.K.); (R.F.-T.); (S.M.); (K.F.); (T.M.); (H.O.); (U.K.B.)
- Correspondence: ; Tel.: +81-744-22-3051; Fax: +81-744-25-7308
| |
Collapse
|
3
|
Koppad S, Basava A, Nash K, Gkoutos GV, Acharjee A. Machine Learning-Based Identification of Colon Cancer Candidate Diagnostics Genes. BIOLOGY 2022; 11:biology11030365. [PMID: 35336739 PMCID: PMC8944988 DOI: 10.3390/biology11030365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023]
Abstract
Simple Summary We developed a predictive approach using different machine learning methods to identify a number of genes that can potentially serve as novel diagnostic colon cancer biomarkers. Abstract Background: Colorectal cancer (CRC) is the third leading cause of cancer-related death and the fourth most commonly diagnosed cancer worldwide. Due to a lack of diagnostic biomarkers and understanding of the underlying molecular mechanisms, CRC’s mortality rate continues to grow. CRC occurrence and progression are dynamic processes. The expression levels of specific molecules vary at various stages of CRC, rendering its early detection and diagnosis challenging and the need for identifying accurate and meaningful CRC biomarkers more pressing. The advances in high-throughput sequencing technologies have been used to explore novel gene expression, targeted treatments, and colon cancer pathogenesis. Such approaches are routinely being applied and result in large datasets whose analysis is increasingly becoming dependent on machine learning (ML) algorithms that have been demonstrated to be computationally efficient platforms for the identification of variables across such high-dimensional datasets. Methods: We developed a novel ML-based experimental design to study CRC gene associations. Six different machine learning methods were employed as classifiers to identify genes that can be used as diagnostics for CRC using gene expression and clinical datasets. The accuracy, sensitivity, specificity, F1 score, and area under receiver operating characteristic (AUROC) curve were derived to explore the differentially expressed genes (DEGs) for CRC diagnosis. Gene ontology enrichment analyses of these DEGs were performed and predicted gene signatures were linked with miRNAs. Results: We evaluated six machine learning classification methods (Adaboost, ExtraTrees, logistic regression, naïve Bayes classifier, random forest, and XGBoost) across different combinations of training and test datasets over GEO datasets. The accuracy and the AUROC of each combination of training and test data with different algorithms were used as comparison metrics. Random forest (RF) models consistently performed better than other models. In total, 34 genes were identified and used for pathway and gene set enrichment analysis. Further mapping of the 34 genes with miRNA identified interesting miRNA hubs genes. Conclusions: We identified 34 genes with high accuracy that can be used as a diagnostics panel for CRC.
Collapse
Affiliation(s)
- Saraswati Koppad
- Department of Computer Science and Engineering, National Institute of Technology Karnataka, Mangalore 575025, India; (S.K.); (A.B.)
| | - Annappa Basava
- Department of Computer Science and Engineering, National Institute of Technology Karnataka, Mangalore 575025, India; (S.K.); (A.B.)
| | - Katrina Nash
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TT, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham B15 2WB, UK
- MRC Health Data Research UK (HDR UK), Midlands Site, Birmingham B15 2TT, UK
- NIHR Experimental Cancer Medicine Centre, Birmingham B15 2TT, UK
- NIHR Biomedical Research Centre, University Hospital Birmingham, Birmingham B15 2TT, UK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TT, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospital Birmingham, Birmingham B15 2WB, UK
- Correspondence: ; Tel.: +44-07403642022
| |
Collapse
|
4
|
Wang G, Wang F, Meng Z, Wang N, Zhou C, Zhang J, Zhao L, Wang G, Shan B. Uncovering potential genes in colorectal cancer based on integrated and DNA methylation analysis in the gene expression omnibus database. BMC Cancer 2022; 22:138. [PMID: 35114976 PMCID: PMC8815138 DOI: 10.1186/s12885-022-09185-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022] Open
Abstract
Background Colorectal cancer (CRC) is major cancer-related death. The aim of this study was to identify differentially expressed and differentially methylated genes, contributing to explore the molecular mechanism of CRC. Methods Firstly, the data of gene transcriptome and genome-wide DNA methylation expression were downloaded from the Gene Expression Omnibus database. Secondly, functional analysis of differentially expressed and differentially methylated genes was performed, followed by protein-protein interaction (PPI) analysis. Thirdly, the Cancer Genome Atlas (TCGA) dataset and in vitro experiment was used to validate the expression of selected differentially expressed and differentially methylated genes. Finally, diagnosis and prognosis analysis of selected differentially expressed and differentially methylated genes was performed. Results Up to 1958 differentially expressed (1025 up-regulated and 993 down-regulated) genes and 858 differentially methylated (800 hypermethylated and 58 hypomethylated) genes were identified. Interestingly, some genes, such as GFRA2 and MDFI, were differentially expressed-methylated genes. Purine metabolism (involved IMPDH1), cell adhesion molecules and PI3K-Akt signaling pathway were significantly enriched signaling pathways. GFRA2, FOXQ1, CDH3, CLDN1, SCGN, BEST4, CXCL12, CA7, SHMT2, TRIP13, MDFI and IMPDH1 had a diagnostic value for CRC. In addition, BEST4, SHMT2 and TRIP13 were significantly associated with patients’ survival. Conclusions The identified altered genes may be involved in tumorigenesis of CRC. In addition, BEST4, SHMT2 and TRIP13 may be considered as diagnosis and prognostic biomarkers for CRC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09185-0.
Collapse
Affiliation(s)
- Guanglin Wang
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feifei Wang
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zesong Meng
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Wang
- Institute of Tumor, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaoxi Zhou
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Juan Zhang
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lianmei Zhao
- Scientific Research Center, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang, 050010, Hebei Province, China
| | - Guiying Wang
- The Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Department of General Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Scientific Research Center, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang, 050010, Hebei Province, China.
| |
Collapse
|
5
|
Liu W. Long non-coding RNA VPS9D1-AS1 promotes growth of colon adenocarcinoma by sponging miR-1301-3p and CLDN1. Hum Cell 2021; 34:1775-1787. [PMID: 34519940 DOI: 10.1007/s13577-021-00604-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
Colon adenocarcinoma is a frequent malignancy among all colon cancer types. Long non-coding RNAs (lncRNAs) are involved in the progression of colon adenocarcinoma. This study aimed to uncover the molecular mechanism of VPS9D1-AS1 in regulating colon adenocarcinoma development. Quantitative reverse transcription-polymerase chain reaction (RT-qPCR) revealed that VPS9D1-AS1 expression was markedly upregulated in colon adenocarcinoma tissues and cell lines. Cell functional experiments showed that knockdown of VPS9D1-AS1 repressed the growth and invasion of colon adenocarcinoma cells but upregulated cell apoptosis. In addition, we confirmed the interaction of VPS9D1-AS1-miR-1301-3p-CLDN1 using a luciferase assay. Downregulation of miR-1301-3p promoted the progression of colon adenocarcinoma cells. In conclusion, VPS9D1-AS1 facilitated cell growth and suppressed apoptosis of colon adenocarcinoma cells by sponging miR-1301-3p and upregulating CLDN1, which may be effective therapeutic strategies for patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Wei Liu
- Department of Gastroenterology, The First Affiliated Hospital, Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
6
|
Gowrikumar S, Primeaux M, Pravoverov K, Wu C, Szeglin BC, Sauvé CEG, Thapa I, Bastola D, Chen XS, Smith JJ, Singh AB, Dhawan P. A Claudin-Based Molecular Signature Identifies High-Risk, Chemoresistant Colorectal Cancer Patients. Cells 2021; 10:cells10092211. [PMID: 34571860 PMCID: PMC8466455 DOI: 10.3390/cells10092211] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Identifying molecular characteristics that are associated with aggressive cancer phenotypes through gene expression profiling can help predict treatment responses and clinical outcomes. Claudins are deregulated in colorectal cancer (CRC). In CRC, increased claudin-1 expression results in epithelial-to-mesenchymal transition and metastasis, while claudin-7 functions as a tumor suppressor. In this study, we have developed a molecular signature based on claudin-1 and claudin-7 associated with poor patient survival and chemoresistance. This signature was validated using an integrated approach including publicly available datasets and CRC samples from patients who either responded or did not respond to standard-of-care treatment, CRC cell lines, and patient-derived rectal and colon tumoroids. Transcriptomic analysis from a patient dataset initially yielded 23 genes that were differentially expressed along with higher claudin-1 and decreased claudin-7. From this analysis, we selected a claudins-associated molecular signature including PIK3CA, SLC6A6, TMEM43, and ASAP-1 based on their importance in CRC. The upregulation of these genes and their protein products was validated using multiple CRC patient datasets, in vitro chemoresistant cell lines, and patient-derived tumoroid models. Additionally, blocking these genes improved 5-FU sensitivity in chemoresistant CRC cells. Our findings propose a new claudin-based molecular signature that associates with poor prognosis as well as characteristics of treatment-resistant CRC including chemoresistance, metastasis, and relapse.
Collapse
Affiliation(s)
- Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.G.); (M.P.); (K.P.); (A.B.S.)
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.G.); (M.P.); (K.P.); (A.B.S.)
| | - Kristina Pravoverov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.G.); (M.P.); (K.P.); (A.B.S.)
| | - Chao Wu
- Department of Surgery, Colorectal Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.W.); (B.C.S.); (C.-E.G.S.); (J.J.S.)
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Bryan C. Szeglin
- Department of Surgery, Colorectal Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.W.); (B.C.S.); (C.-E.G.S.); (J.J.S.)
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Charles-Etienne Gabriel Sauvé
- Department of Surgery, Colorectal Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.W.); (B.C.S.); (C.-E.G.S.); (J.J.S.)
| | - Ishwor Thapa
- College of Information Science & Technology, University of Omaha, Omaha, NE 68182, USA; (I.T.); (D.B.)
| | - Dhundy Bastola
- College of Information Science & Technology, University of Omaha, Omaha, NE 68182, USA; (I.T.); (D.B.)
| | - Xi Steven Chen
- Department of Public Health Sciences, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - J. Joshua Smith
- Department of Surgery, Colorectal Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.W.); (B.C.S.); (C.-E.G.S.); (J.J.S.)
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Amar B. Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.G.); (M.P.); (K.P.); (A.B.S.)
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.G.); (M.P.); (K.P.); (A.B.S.)
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Correspondence: ; Tel.: +1-(402)-559-6587
| |
Collapse
|
7
|
Xu C, Ding YH, Wang K, Hao M, Li H, Ding L. Claudin-7 deficiency promotes stemness properties in colorectal cancer through Sox9-mediated Wnt/β-catenin signalling. J Transl Med 2021; 19:311. [PMID: 34281572 PMCID: PMC8287764 DOI: 10.1186/s12967-021-02983-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/11/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumour of the digestive tract that is characterized by high patient morbidity and mortality rates. Claudin-7 (Cldn7), a tight junction protein, was recently reported to function as a candidate tumour suppressor gene in CRC. Our previous study demonstrated that the large intestine of C57/BL6 mice showed intestinal adenomas and abnormal Ki67 expression and distribution in the intestinal crypt when Cldn7 was knocked out. The aim of this study was to further investigate whether Cldn7 deficiency has non-tight junction functions, affects intestinal stemness properties, promotes CRC and to determine the specific mechanism. METHODS Cell proliferation assays, migration assays, apoptosis assays, tumour sphere formation assays in vitro, and subcutaneous xenograft models in vivo were used to determine the effects of Cldn7 knockdown on the biological characteristics of CRC stem cells. Western blotting, qPCR and immunofluorescence staining were performed to identify the epithelial-mesenchymal transition and the activation of Wnt/β-catenin pathway in CRC stem cells. Cldn7 inducible conditional gene knockout mice and immunohistochemical staining further verified this hypothesis in vivo. The mechanism and target of Cldn7 were determined by performing a chromatin immunoprecipitation (ChIP) assay and coimmunoprecipitation (CoIP) assay. RESULTS Cldn7 knock down in CRC stem cells promoted cell proliferation, migration, and globular growth in serum-free medium and the ability to form xenograft tumours; cell apoptosis was inhibited, while the cellular epithelial-mesenchymal transition was also observed. These changes in cell characteristics were achieved by activating the Wnt/β-catenin pathway and promoting the expression of downstream target genes after β-catenin entry into the nucleus, as observed in CRC cell lines and Cldn7 gene knockout mouse experiments. Using ChIP and CoIP experiments, we initially found that Cldn7 and Sox9 interacted at the protein level to activate the Wnt/β-catenin pathway. CONCLUSIONS Based on our research, Cldn7 deficiency confers stemness properties in CRC through Sox9-mediated Wnt/β-catenin signalling. This result clarifies that Cldn7 plays an inhibitory role in CRC and reveals a possible molecular mechanism, which is conducive to further research on Cldn7 and cancer stem cells.
Collapse
Affiliation(s)
- Chang Xu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
- Department of Hepato-Pancreato-Biliary Surgery, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing
, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu-han Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Mengdi Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Huimin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038 China
| |
Collapse
|
8
|
Popova OP, Kuznetsova AV, Bogomazova SY, Ivanov AA. Claudins as biomarkers of differential diagnosis and prognosis of tumors. J Cancer Res Clin Oncol 2021; 147:2803-2817. [PMID: 34241653 DOI: 10.1007/s00432-021-03725-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
Claudins are a superfamily of transmembrane proteins, the optimal expression and localization of which are important for the normal physiological function of the epithelium and any imbalance may have pathological consequences. Not only insufficient but also excessive production of claudins in cancer cells, as well as their aberrant localization, equally manifest the formation of a malignant phenotype. Many works are distinguished by contradictory data, which demonstrate the action of the same claudins both in the role of tumor-growth suppressors and promoters in the same cancers. The most important possible causes of significant discrepancies in the results of the works are a considerable variability of sampling and the absence of a consistent approach both to the assessment of the immune reactivity of claudins and to the differential analysis of their subcellular localization. Combined, these drawbacks hinder the histological assessment of the link between claudins and tumor progression. In particular, ambiguous expression of claudins in breast cancer subtypes, revealed by various authors in immunohistochemical analysis, not only fails to facilitate the identification of the claudin-low molecular subtype but rather complicates these efforts. Research into the role of claudins in carcinogenesis has undoubtedly confirmed the potential value of this class of proteins as significant biomarkers in some cancer types; however, the immunohistochemical approach to the assessment of claudins still has limitations, needs standardization, and, to date, has not reached a diagnostic or a prognostic value.
Collapse
Affiliation(s)
- Olga P Popova
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, 20, Bld 1, Delegatskaya Street, Moscow, 127473, Russia
| | - Alla V Kuznetsova
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, 20, Bld 1, Delegatskaya Street, Moscow, 127473, Russia.,Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow, 119334, Russia
| | - Svetlana Yu Bogomazova
- Department of Pathology, National Medical Research Treatment and Rehabilitation Centre, Ministry of Health of the Russian Federation, Ivankovskoe shosse, 3, Moscow, 125367, Russia
| | - Alexey A Ivanov
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, 20, Bld 1, Delegatskaya Street, Moscow, 127473, Russia.
| |
Collapse
|
9
|
The correlation of epithelial-mesenchymal transition-related gene expression and the clinicopathologic features of colorectal cancer patients in Taiwan. PLoS One 2021; 16:e0254000. [PMID: 34214117 PMCID: PMC8253430 DOI: 10.1371/journal.pone.0254000] [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] [Received: 02/11/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed malignancy and the second leading cause of cancer-related deaths in the world. It has been the most prevalent malignancy in Taiwan for consecutive thirteen years. Despite the diversity of its etiologic and pathophysiologic factors, a biological process named as epithelial-mesenchymal transition (EMT) is indispensable in the progression of epithelial cancer. Our aim is to investigate the correlation between the expression of 8 EMT-related proteins (E-cadherin, β-catenin, claudin-1, CD44, N-cadherin, fibronectin, vimentin, S100A4) and the clinicopathologic features of CRC in Taiwan, along with the DNA CpG epigenetic status of CD44 gene. In immunohistochemical assessment, decreased expression of E-cadherin is statistically associated with the progression of cancer stage, while decreased expression of claudin-1 as well as increased β-catenin nuclear translocation and N-cadherin expression is statistically associated with the progression of histopathologic grade. E-cadherin, nuclear β-catenin and claudin-1 are also associated with other important prognostic factors, including nodal metastasis, tumor deposits, and elevated serum CA 19-9 levels. In addition, the left-sided colon and rectal cancers show increased nuclear translocation of β-catenin compared to the right-sided colon cancers, while the rectal cancers show increased fibronectin expression compared to the right-sided and left-sided colon cancers. Moreover, vimentin is aberrantly expressed in one case of signet-ring cell carcinoma. The DNA methylation levels of CD44 gene promoter between the tumoral and non-tumorous tissues by NGS comparison showed statistical difference on six CpG sites. However, such difference may not be sufficient because these DNA methylation proportions are too low to inactivate CD44 gene. Our results demonstrate the expression of E-cadherin, claudin-1, and nuclear β-catenin is closely related to the clinicopathologic prognostic determinants of CRC in Taiwan. The DNA methylation level of CD44 gene and its protein expression, however, show no correlation with the clinicopathologic features in CRC.
Collapse
|
10
|
Marincola Smith P, Choksi YA, Markham NO, Hanna DN, Zi J, Weaver CJ, Hamaamen JA, Lewis KB, Yang J, Liu Q, Kaji I, Means AL, Beauchamp RD. Colon epithelial cell TGFβ signaling modulates the expression of tight junction proteins and barrier function in mice. Am J Physiol Gastrointest Liver Physiol 2021; 320:G936-G957. [PMID: 33759564 PMCID: PMC8285585 DOI: 10.1152/ajpgi.00053.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 01/31/2023]
Abstract
Defective barrier function is a predisposing factor in inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). Although TGFβ signaling defects have been associated with IBD and CAC, few studies have examined the relationship between TGFβ and intestinal barrier function. Here, we examine the role of TGFβ signaling via SMAD4 in modulation of colon barrier function. The Smad4 gene was conditionally deleted in the intestines of adult mice and intestinal permeability assessed using an in vivo 4 kDa FITC-Dextran (FD4) permeability assay. Mouse colon was isolated for gene expression (RNA-sequencing), Western blot, and immunofluorescence analysis. In vitro colon organoid culture was utilized to assess junction-related gene expression by qPCR and transepithelial resistance (TER). In silico analyses of human IBD and colon cancer databases were performed. Mice lacking intestinal expression of Smad4 demonstrate increased colonic permeability to FD4 without gross mucosal damage. mRNA/protein expression analyses demonstrate significant increases in Cldn2/Claudin 2 and Cldn8/Claudin 8, and decreases in Cldn3, Cldn4, and Cldn7/Claudin 7 with intestinal SMAD4 loss in vivo without changes in Claudin protein localization. TGFβ1/BMP2 treatment of polarized SMAD4+ colonoids increases TER. Cldn2, Cldn4, Cldn7, and Cldn8 are regulated by canonical TGFβ signaling, and TGFβ-dependent regulation of these genes is dependent on nascent RNA transcription (Cldn2, Cldn4, Cldn8) but not nascent protein translation (Cldn4, Cldn8). Human IBD/colon cancer specimens demonstrate decreased SMAD4, CLDN4, CLDN7, and CLDN8 and increased CLDN2 compared with healthy controls. Canonical TGFβ signaling modulates the expression of tight junction proteins and barrier function in mouse colon.NEW & NOTEWORTHY We demonstrate that canonical TGFβ family signaling modulates the expression of critical tight junction proteins in colon epithelial cells, and that expression of these tight junction proteins is associated with maintenance of colon epithelial barrier function in mice.
Collapse
Affiliation(s)
- Paula Marincola Smith
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Yash A Choksi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Nicholas O Markham
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David N Hanna
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jinghuan Zi
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Connie J Weaver
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jalal A Hamaamen
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keeli B Lewis
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jing Yang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna L Means
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - R Daniel Beauchamp
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
11
|
Suárez-Arriaga MC, Méndez-Tenorio A, Pérez-Koldenkova V, Fuentes-Pananá EM. Claudin-Low Breast Cancer Inflammatory Signatures Support Polarization of M1-Like Macrophages with Protumoral Activity. Cancers (Basel) 2021; 13:2248. [PMID: 34067089 PMCID: PMC8125772 DOI: 10.3390/cancers13092248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
We previously reported that triple-negative breast cancer (BRCA) cells overexpress the cytokines GM-CSF, G-CSF, MCP-1, and RANTES, and when monocytes were 3-D co-cultured with them, M1-like macrophages were generated with the ability to induce aggressive features in luminal BRCA cell lines. These include upregulation of mesenchymal and stemness markers and invasion. In this study, we stimulated peripheral blood monocytes with the four cytokines and confirmed their capacity to generate protumoral M1-like macrophages. Using the METABRIC BRCA database, we observed that GM-CSF, MCP-1, and RANTES are associated with triple-negative BRCA and reduced overall survival, particularly in patients under 55 years of age. We propose an extended M1-like macrophage proinflammatory signature connected with these three cytokines. We found that the extended M1-like macrophage signature coexists with monocyte/macrophage, Th1 immune response, and immunosuppressive signatures, and all are enriched in claudin-low BRCA samples, and correlate with reduced patient overall survival. Furthermore, we observed that all these signatures are also present in mesenchymal carcinomas of the colon (COAD) and bladder (BLCA). The claudin-low tumor subtype has an adverse clinical outcome and remains poorly understood. This study places M1 macrophages as potential protumoral drivers in already established cancers, and as potential contributors to claudin-low aggressiveness and poor prognosis.
Collapse
Affiliation(s)
- Mayra Cecilia Suárez-Arriaga
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
- Laboratorio de Biotecnología y Bioinformática Genómica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Alfonso Méndez-Tenorio
- Laboratorio de Biotecnología y Bioinformática Genómica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Vadim Pérez-Koldenkova
- Laboratorio Nacional de Microscopía Avanzada, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Ezequiel M. Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| |
Collapse
|
12
|
Ouban A. Expression of Claudin-1 in laryngeal squamous cell carcinomas (LSCCs) and its significance. Histol Histopathol 2021; 36:437-446. [PMID: 33629735 DOI: 10.14670/hh-18-320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND A large body of scientific evidence points to the important roles of tight junction proteins in tumor development, progression and dissemination. The larynx has only a few studies, analyzing the role of this group of junctional proteins in its oncogenesis. In this study, the author sheds some light on the expression and possible role of claudin-1 in laryngeal squamous cell carcinomas. MATERIALS AND METHODS This study analyzed the expression of claudin-1, using immunohistochemistry, in a tissue microarray of 80 cases of laryngeal squamous cell cancers. Clinicopathological parameters were analyzed according to claudin-1 expression in the tissue microarray. Furthermore, the expression of slug/snail1, an Epithelial-Mesenchymal Transition (EMT) linked protein, was analyzed by immunohistochemistry in the same microarray, and the expressions of the two proteins were assessed for correlation. RESULTS A significant majority of laryngeal squamous cell cancers exhibited positive expression of claudin-1 proteins. The majority of those tumors expressed claudin-1 in their cytoplasm. The overall majority of those same tumors also exhibited a cytoplasmic shift of the slug-snail-1 protein from the nuclei to the cytoplasm. There was also evidence of correlation of the two proteins' expressions in the cytoplasm of laryngeal tumors. CONCLUSION The above may suggest a role for claudin-1 in the development and progression of laryngeal squamous cell carcinoma. Overall, claudin-1's aberrant expression in laryngeal cancer is in line with evidence seen in other head and neck cancers. Its co-expression with slug/snail-1 in LSCC patients should be investigated further to understand the nature of the relationship of the two proteins in LSCC and their possible contribution to its development and progression.
Collapse
Affiliation(s)
- Abderrahman Ouban
- Department of Pathology, College of Medicine , Alfaisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
13
|
Expression of Tight Junction Proteins Is Altered in Bladder Cancer. ACTA ACUST UNITED AC 2020; 2020:6341256. [PMID: 33282635 PMCID: PMC7685791 DOI: 10.1155/2020/6341256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022]
Abstract
Bladder cancer (BC) is one of the tumors which occur most frequently in urological system, but less is known about the expression of tight junction proteins and its clinical significance in BC. In this study, expression of claudin-4, zonula occludens-1 (ZO-1) and zonula occludens-1 nucleic acid-binding protein (ZONAB), in BC tissues, adjacent nontumor tissue (ANTT), and BC cell lines was examined by Western blotting, semiquantitative RT-PCR, and immunohistochemistry, and then, the clinical significance of these proteins was investigated. The mRNA and protein expression of ZONAB were significantly upregulated, while those of ZO-1 was significantly downregulated in some BC cell lines and tissues in comparison with nontumor urothelial cell lines and ANTT. High expression rate of ZO-1 and ZONAB had negative correlation in BC tissues and was also correlated with muscle-invasive lesions in BC tissues. In conclusion, the expression of tight junction proteins is significantly altered in BC and ZO-1, and ZONAB interaction might be involved in BC development.
Collapse
|
14
|
Uc PY, Miranda J, Raya-Sandino A, Alarcón L, Roldán ML, Ocadiz-Delgado R, Cortés-Malagón EM, Chávez-Munguía B, Ramírez G, Asomoza R, Shoshani L, Gariglio P, González-Mariscal L. E7 oncoprotein from human papillomavirus 16 alters claudins expression and the sealing of epithelial tight junctions. Int J Oncol 2020; 57:905-924. [PMID: 32945372 PMCID: PMC7473757 DOI: 10.3892/ijo.2020.5105] [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: 01/26/2020] [Accepted: 04/16/2020] [Indexed: 11/24/2022] Open
Abstract
Tight junctions (TJs) are cell-cell adhesion structures frequently altered by oncogenic transformation. In the present study the role of human papillomavirus (HPV) 16 E7 oncoprotein on the sealing of TJs was investigated and also the expression level of claudins in mouse cervix and in epithelial Madin-Darby Canine Kidney (MDCK) cells. It was found that there was reduced expression of claudins -1 and -10 in the cervix of 7-month-old transgenic K14E7 mice treated with 17β-estradiol (E2), with invasive cancer. In addition, there was also a transient increase in claudin-1 expression in the cervix of 2-month-old K14E7 mice, and claudin-10 accumulated at the border of cells in the upper layer of the cervix in FvB mice treated with E2, and in K14E7 mice treated with or without E2. These changes were accompanied by an augmented paracellular permeability of the cervix in 2- and 7-monthold FvB mice treated with E2, which became more pronounced in K14E7 mice treated with or without E2. In MDCK cells the stable expression of E7 increased the space between adjacent cells and altered the architecture of the monolayers, induced the development of an acute peak of transepithelial electrical resistance accompanied by a reduced expression of claudins -1, -2 and -10, and an increase in claudin-4. Moreover, E7 enhances the ability of MDCK cells to migrate through a 3D matrix and induces cell stiffening and stress fiber formation. These observations revealed that cell transformation induced by HPV16 E7 oncoprotein was accompanied by changes in the pattern of expression of claudins and the degree of sealing of epithelial TJs.
Collapse
Affiliation(s)
- Perla Yaceli Uc
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Arturo Raya-Sandino
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Lourdes Alarcón
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - María Luisa Roldán
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Rodolfo Ocadiz-Delgado
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Enoc Mariano Cortés-Malagón
- Research Unit on Genetics and Cancer, Research Division, Hospital Juárez de México, Mexico City 07760, Mexico
| | - Bibiana Chávez-Munguía
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Georgina Ramírez
- Department of Electrical Engineering, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - René Asomoza
- Department of Electrical Engineering, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Liora Shoshani
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Patricio Gariglio
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| |
Collapse
|
15
|
Improved sensitivity in cell surface protein detection by combining chemical labeling with mechanical lysis in a colorectal cancer cell model. Biotechnol Lett 2020; 42:683-695. [DOI: 10.1007/s10529-020-02824-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/26/2020] [Indexed: 10/25/2022]
|
16
|
Kage H, Flodby P, Zhou B, Borok Z. Dichotomous roles of claudins as tumor promoters or suppressors: lessons from knockout mice. Cell Mol Life Sci 2019; 76:4663-4672. [PMID: 31332482 PMCID: PMC6858953 DOI: 10.1007/s00018-019-03238-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/29/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
Abstract
Claudins are a family of integral tight junction proteins that regulate paracellular permeability in polarized epithelia. Overexpression or reduction of claudins can both promote and limit cancer progression, revealing complex dichotomous roles for claudins depending on cellular context. In contrast, recent studies demonstrating tumor formation in claudin knockout mouse models indicate a role for several claudin family members in suppressing tumor initiation. For example, intestine-specific claudin-7 knockout mice spontaneously develop atypical hyperplasia and intestinal adenomas, while claudin-18 knockout mice develop carcinomas in the lung and stomach. Claudin-4, -11, and -15 knockout mice show increased cell proliferation and/or hyperplasia in urothelium, Sertoli cells, and small intestinal crypts, respectively, possibly a precursor to cancer development. Pathways implicated in both cell proliferation and tumorigenesis include Yap/Taz and insulin-like growth factor-1 receptor (IGF-1R)/Akt pathways, among others. Consistent with the tumor suppressive role of claudins shown in mice, in humans, claudin-low breast cancer has been described as a distinct entity with a poor prognosis, and claudin-18-Rho GTPase activating protein 26 (CLDN18-ARHGAP26) fusion protein as a driver gene aberration in diffuse-type gastric cancer due to effects on RhoA. Paradoxically, claudins have also garnered interest as targets for therapy, as they are sometimes aberrantly expressed in cancer cells, which may or may not promote cancer progression. For example, a chimeric monoclonal antibody which targets cells expressing claudin-18.2 through antibody-dependent cell-mediated cytotoxicity has shown promise in multiple phase II studies. In this review, we focus on new findings supporting a tumor suppressive role for claudins during cancer initiation.
Collapse
Affiliation(s)
- Hidenori Kage
- Department of Respiratory Medicine, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Per Flodby
- Division of Pulmonary, Critical Care and Sleep Medicine and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, IRD 620, M/C 9520, Los Angeles, CA, 90089-9520, USA
| | - Beiyun Zhou
- Division of Pulmonary, Critical Care and Sleep Medicine and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, IRD 620, M/C 9520, Los Angeles, CA, 90089-9520, USA
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, IRD 620, M/C 9520, Los Angeles, CA, 90089-9520, USA.
| |
Collapse
|
17
|
Chiang SK, Chang WC, Chen SE, Chang LC. DOCK1 Regulates Growth and Motility through the RRP1B-Claudin-1 Pathway in Claudin-Low Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11111762. [PMID: 31717460 PMCID: PMC6896004 DOI: 10.3390/cancers11111762] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 01/08/2023] Open
Abstract
Dedicator of cytokinesis 1 (DOCK1) is a critical regulator of cancer metastasis. Claudins are transmembrane proteins that play a role in epithelial barrier integrity. Due to a loss or low expression of claudins (CLDN), the claudin-low type of triple-negative breast cancer (TNBC) is characterized by a mesenchymal-like phenotype with strong metastatic potential. In order to elucidate the mechanism of DOCK1 in cancer metastasis, we first analyzed the transcriptomic changes using a clinical database of human TNBC and found that the increase in DOCK1 expression was highly correlated with the poor survival rate of TNBC patients. Interference with DOCK1 expression by shRNA resulted in re-expression of claudin-1 in conjunction with significant inhibition of cell viability and motility of claudin-low breast cancer cells. Accordingly, overexpression of claudin-1 suppressed cell viability and migration. Genetic knockdown and pharmacological blockade of Rac1/Rac2 up-regulated claudin-1. DOCK1 knockdown also caused a decrease in DNA methyltransferase (DNMT) expression and an increase in claudin-1 transcript and promoter activity. Furthermore, RRP1B mediated DOCK1 depletion, which up-regulated claudin-1 expression, cell viability, and motility in claudin-low breast cancer cells. This study demonstrated that DOCK1 mediates growth and motility through down-regulated claudin-1 expression via the RRP1B–DNMT–claudin-1 pathway and that claudin-1 serves as an important effector in DOCK1-mediated cancer progression and metastasis in claudin-low breast cancer cells.
Collapse
Affiliation(s)
- Shih-Kai Chiang
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Shuen-Ei Chen
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan;
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung 40227, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
- Research Center for Sustainable Energy and Nanotechnology, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence: (S.-E.C.); (L.-C.C.); Tel.: 886-4-22870613 (ext. 227) (S.-E.C.); +886-4-22052121 (ext. 7913) (L.-C.C.)
| | - Ling-Chu Chang
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
- Correspondence: (S.-E.C.); (L.-C.C.); Tel.: 886-4-22870613 (ext. 227) (S.-E.C.); +886-4-22052121 (ext. 7913) (L.-C.C.)
| |
Collapse
|
18
|
Fields MA, Del Priore LV, Adelman RA, Rizzolo LJ. Interactions of the choroid, Bruch's membrane, retinal pigment epithelium, and neurosensory retina collaborate to form the outer blood-retinal-barrier. Prog Retin Eye Res 2019; 76:100803. [PMID: 31704339 DOI: 10.1016/j.preteyeres.2019.100803] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023]
Abstract
The three interacting components of the outer blood-retinal barrier are the retinal pigment epithelium (RPE), choriocapillaris, and Bruch's membrane, the extracellular matrix that lies between them. Although previously reviewed independently, this review integrates these components into a more wholistic view of the barrier and discusses reconstitution models to explore the interactions among them. After updating our understanding of each component's contribution to barrier function, we discuss recent efforts to examine how the components interact. Recent studies demonstrate that claudin-19 regulates multiple aspects of RPE's barrier function and identifies a barrier function whereby mutations of claudin-19 affect retinal development. Co-culture approaches to reconstitute components of the outer blood-retinal barrier are beginning to reveal two-way interactions between the RPE and choriocapillaris. These interactions affect barrier function and the composition of the intervening Bruch's membrane. Normal or disease models of Bruch's membrane, reconstituted with healthy or diseased RPE, demonstrate adverse effects of diseased matrix on RPE metabolism. A stumbling block for reconstitution studies is the substrates typically used to culture cells are inadequate substitutes for Bruch's membrane. Together with human stem cells, the alternative substrates that have been designed offer an opportunity to engineer second-generation culture models of the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Ron A Adelman
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA; Department of Surgery, Yale University School of Medicine, PO Box 208062, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
19
|
Wu J, Gao F, Xu T, Li J, Hu Z, Wang C, Long Y, He X, Deng X, Ren D, Zhou B, Dai T. CLDN1 induces autophagy to promote proliferation and metastasis of esophageal squamous carcinoma through AMPK/STAT1/ULK1 signaling. J Cell Physiol 2019; 235:2245-2259. [PMID: 31498437 DOI: 10.1002/jcp.29133] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022]
Abstract
Tight junction is a structural constitution in cell-cell adhesion and play an important role in the maintenance of permeability and integrity of normal epithelial cell barrier. The protein encoded by Claudin 1 (CLDN1), a member of the claudin family, is an integral membrane protein and a component of tight junction strands. CLDN1 has been proved to regulate the proliferation and metastasis of multiple tumors, but little is known about its role in esophageal squamous cell carcinoma (ESCC). Here, we found that CLDN1 was aberrantly increased in ESCC tissues and cell lines, and mainly distributed in the nucleus of tumor cells. Furthermore, we confirmed that CLDN1 promoted the proliferation and metastasis of ESCC by triggering autophagy both in vitro and in vivo. Mechanically, we validated that CLDN1-induced autophagy via increasing Unc-51 like autophagy activating kinase 1 (ULK1) expression through AMP-activated protein kinase (AMPK)/signal transducer and activator of transcription 1 (STAT1) signaling pathway in ESCC cells. Taken together, our findings demonstrated that aberrant expression and distribution of CLDN1 promoted the proliferation and metastasis of esophageal squamous carcinoma by triggering autophagy through AMPK/STAT1/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Jian Wu
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - FengXia Gao
- Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China
| | - Tao Xu
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Jun Li
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Zhi Hu
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Chao Wang
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China.,Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China.,Experiment Medicine Center, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Experiment Medicine Center, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - XueMei He
- Experiment Medicine Center, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Xin Deng
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - DeLian Ren
- Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China
| | - Biao Zhou
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - TianYang Dai
- Department of Cardio-Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| |
Collapse
|
20
|
Huang G, Cheng W, Xi F. Integrated genomic and methylation profile analysis to identify candidate tumor marker genes in patients with colorectal cancer. Oncol Lett 2019; 18:4503-4514. [PMID: 31611959 PMCID: PMC6781519 DOI: 10.3892/ol.2019.10799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022] Open
Abstract
Aberrant genomic expression and methylation serve important roles in cancer development. Integrated analysis of genetic and methylation profiles may identify potential tumor marker genes for colorectal cancer (CRC) prediction. In the current study, DNA methylation and mRNA expression profiles associated with CRC were downloaded from The Cancer Genome Atlas database. Differentially expressed mRNAs and methylated genes between tumor samples and adjacent healthy tissues were identified. Candidate tumor marker genes and prognostic clinical factors were screened according to univariable and multivariable Cox regression analysis. A total of 218 DEGs with aberrant methylation levels were screened from tumor samples. A risk prediction model was constructed based on identified genes and clinical factors. Randomization tests were used to evaluate the performance of the prediction model, including area under the curve (AUC) calculation and cross-validation. Cox regression analysis revealed that eight genes and six prognostic clinical factors were significantly associated with survival outcomes. Functional and pathway enrichment analysis revealed that the eight genes were mainly involved in ‘cell adhesion’, ‘fatty acid metabolism’ and ‘cytokine receptor interaction’ pathways. After combining six clinical factors with eight genes, the accuracy of risk prediction model has been increased intensively. The P-values representing the association between risk grouping and prognosis decreased from 0.009 to 0.001 and the AUC increased from 0.992 to 0.999, indicating that the comprehensive risk prediction model exhibited a good performance for disease prognosis prediction. The current study integrated genomic and methylation profiles and identified eight tumor marker genes in CRC. These candidate genes may improve the prediction accuracy of CRC prognosis.
Collapse
Affiliation(s)
- Guojun Huang
- Department of Oncology, Pidu District People's Hospital, Chengdu, Sichuan 611730, P.R. China
| | - Wang Cheng
- Department of General Surgery, Pidu District People's Hospital, Chengdu, Sichuan 611730, P.R. China
| | - Fu Xi
- Department of Oncology, Pidu District People's Hospital, Chengdu, Sichuan 611730, P.R. China
| |
Collapse
|
21
|
Coati I, Lotz G, Fanelli GN, Brignola S, Lanza C, Cappellesso R, Pellino A, Pucciarelli S, Spolverato G, Guzzardo V, Munari G, Zaninotto G, Scarpa M, Mastracci L, Farinati F, Realdon S, Pilati P, Lonardi S, Valeri N, Rugge M, Kiss A, Loupakis F, Fassan M. Claudin-18 expression in oesophagogastric adenocarcinomas: a tissue microarray study of 523 molecularly profiled cases. Br J Cancer 2019; 121:257-263. [PMID: 31235864 PMCID: PMC6738069 DOI: 10.1038/s41416-019-0508-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Claudin-18 (CLDN18) is a highly specific tight junction protein of the gastric mucosa. An isoform of CLDN18, the Claudin 18.2, has recently emerged as an innovative drug target for metastatic gastric cancer. METHODS We investigated the immunohistochemical profile of CLDN18, p53, p16, E-cadherin, MSH2, MSH6, MLH1, PSM2, HER2, and PDL-1 in a large series of 523 primary gastric carcinomas (GCs; n = 408) and gastro-oesophageal carcinomas (GECs; n = 115) and 135 matched and synchronous nodal metastases. The status of HER2 and EBER by means of chromogenic in situ hybridisation (CISH) was also evaluated. RESULTS High membranous CLDN18 expression was present in 150/510 (29.4%) primary cases and in 45/132 (34.1%) metastases. An abnormal expression (i.e. nuclear and/or cytoplasmic) was observed in 115 (22.5%) primary cases and in 33 (25.0%) metastases. A 38.8% of the cases showed significant CLDN18 intratumoural variability among the different tissue microarray cores obtained from the same tumour. Positive membrane CLDN18 expression was statistically associated with non-antral GCs (p = 0.016), Lauren diffuse type (p = 0.009), and with EBV-associated cancers (p < 0.001). CONCLUSIONS CLDN18 is frequently expressed in gastric and gastro-oesophageal cancers; further studies should investigate the prognostic significance of CLDN18 heterogeneity in order to implement its test into clinical practice.
Collapse
Affiliation(s)
- Irene Coati
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Gábor Lotz
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Giuseppe Nicolò Fanelli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Stefano Brignola
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Cristiano Lanza
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Rocco Cappellesso
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Antonio Pellino
- Unit of Oncology 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Salvatore Pucciarelli
- Department of Surgical Oncology and Gastroenterology Sciences (DiSCOG), Surgery Unit, University of Padua, Padua, Italy
| | - Gaya Spolverato
- Department of Surgical Oncology and Gastroenterology Sciences (DiSCOG), Surgery Unit, University of Padua, Padua, Italy
| | - Vincenza Guzzardo
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Giada Munari
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
- Unit of Oncology 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | | | - Marco Scarpa
- Department of Surgical Oncology and Gastroenterology Sciences (DiSCOG), Surgery Unit, University of Padua, Padua, Italy
| | - Luca Mastracci
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Pathology Unit, University of Genova, Genova, Italy
| | - Fabio Farinati
- Department of Surgical Oncology and Gastroenterology Sciences (DiSCOG), Gastroenterology Unit, University of Padua, Padua, Italy
| | - Stefano Realdon
- Unit of Gastroenterology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Pierluigi Pilati
- Unit of Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Sara Lonardi
- Unit of Oncology 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Nicola Valeri
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Department of Medicine, Royal Marsden Hospital, London, UK
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
- Veneto Cancer Registry, Padua, Italy
| | - Andras Kiss
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Fotios Loupakis
- Unit of Oncology 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy.
| |
Collapse
|
22
|
Zhu L, Han J, Li L, Wang Y, Li Y, Zhang S. Claudin Family Participates in the Pathogenesis of Inflammatory Bowel Diseases and Colitis-Associated Colorectal Cancer. Front Immunol 2019; 10:1441. [PMID: 31316506 PMCID: PMC6610251 DOI: 10.3389/fimmu.2019.01441] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022] Open
Abstract
Claudins are a multigene transmembrane protein family comprising at least 27 members. In gastrointestinal tract, claudins are mainly located in the intestinal epithelia; many types of claudins form a network of strands in tight junction plaques within the intercellular space of neighboring epithelial cells and build paracellular selective channels, while others act as signaling proteins and mediates cell behaviors. Claudin dysfunction may contribute to epithelial permeation disorder and multiple intestinal diseases. Over recent years, the importance of claudins in the pathogenesis of inflammatory bowel diseases (IBD) has gained focus and is being investigated. This review analyzes the expression pattern and regulatory mechanism of claudins based on existing evidence and elucidates the fact that claudin dysregulation correlates with increased intestinal permeability, sustained activation of inflammation, epithelial-to-mesenchymal transition (EMT), and tumor progression in IBD as well as consequent colitis-associated colorectal cancer (CAC), possibly shedding new light on further etiologic research and clinical treatments.
Collapse
Affiliation(s)
| | | | | | | | | | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Wang K, Li T, Xu C, Ding Y, Li W, Ding L. Claudin-7 downregulation induces metastasis and invasion in colorectal cancer via the promotion of epithelial-mesenchymal transition. Biochem Biophys Res Commun 2019; 508:797-804. [PMID: 30528239 DOI: 10.1016/j.bbrc.2018.10.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/07/2018] [Indexed: 02/08/2023]
Abstract
The dysregulation of the tight junctions (TJs) protein claudin-7 is closely related to the development and metastasis of colorectal cancer (CRC). The aim of this study was to investigate the expression of claudin-7 and characterize the relationship between claudin-7 expression and epithelial-mesenchymal transition (EMT) in CRC. In this study, the expression of claudin-7, E-cadherin, vimentin and snail-1 was detected by immunohistochemistry (IHC) in a set of 80 CRC specimens comprising 20 specimens each of well-differentiated, moderately differentiated, poorly differentiated and liver metastases tissues. The correlation between claudin-7 and EMT-related proteins in the stably transfected claudin-7 knockdown HCT116 cell line was analyzed by IHC, immunofluorescence (IF), Western blotting (WB) and nude mouse xenograft models. The results revealed that the expression of claudin-7 was downregulated as CRC tissue differentiation grade decreased, and that low claudin-7 expression corresponded to the downregulation of E-cadherin (r = 0.725, p < 0.001) and upregulation of vimentin (r = -0.376, p = 0.001) and snail-1 (r = -0.599, p < 0.001). Additionally, in the claudin-7 knockdown HCT116 cell line, the staining intensity and expression of E-cadherin was decreased, while the immunoreactivity and expression of vimentin and snail-1 was increased. Futhermore, the result of tumor formation experiment was consistent with CRC tissues. In conclusion, the expression of claudin-7 in CRC is downregulated as differentiation grade decreases. Claudin-7 downregulation may promote the invasion and metastasis of CRC by regulating EMT. Our results provide new perspectives for a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Tengyan Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Cancer Hospital Chinese Academy of Medical Sciences, Peking Union Medical College, 100021, China
| | - Chang Xu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yuhan Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Wenjing Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Binzhou Medical University Hospital, Binzhou City, 256603, Shandong Province, China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
24
|
Wang K, Xu C, Li W, Ding L. Emerging clinical significance of claudin-7 in colorectal cancer: a review. Cancer Manag Res 2018; 10:3741-3752. [PMID: 30288105 PMCID: PMC6159786 DOI: 10.2147/cmar.s175383] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tight junctions (TJs) play an important role in maintaining cell polarity and regulating cell permeability. In recent years, many studies have shown that TJ proteins, especially claudin-7, are closely related to inflammation and the development of various malignant tumors. Claudin-7 plays a significant role in maintaining the physiological functions and pathological conditions of the TJ barrier. The dysregulation of claudin-7 plays a tumor suppressor role or conversely has carcinogenic effects in different target tissues or cells, but the exact underlying mechanism is still unclear. In this review, we will summarize the expression pattern of claudin-7 in tumors, focusing on the expression and regulation of claudin-7 in colorectal cancer and discussing the correlation between claudin-7 and invasion, metastasis and epithelial-mesenchymal transition (EMT) in colorectal cancer. The construction of Cldn7-/- mice and conventional claudin-7 knockout mouse models has helped determine the mechanisms by which claudin-7 promotes tumorigenesis. Elucidation of the expression and subcellular localization of claudin-7 under pathological conditions will help develop claudin-7 as a useful biomarker for detecting and diagnosing cancer, and thus may help combat the occurrence, development, and invasion of cancers.
Collapse
Affiliation(s)
- Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China,
| | - Chang Xu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China,
| | - Wenjing Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China,
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China,
| |
Collapse
|
25
|
Liu N, Wang JQ, Jia SC, Chen YK, Wang JP. Effect of yeast cell wall on the growth performance and gut health of broilers challenged with aflatoxin B1 and necrotic enteritis. Poult Sci 2018; 97:477-484. [PMID: 29211897 DOI: 10.3382/ps/pex342] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/27/2017] [Indexed: 01/15/2023] Open
Abstract
This study aimed to investigate the effect of yeast cell walls (YCW) on the growth performance, visceral lesions, intestinal integrity, enterotoxicity, and bacteria of broilers challenged with aflatoxin B1 (AF) and necrotic enteritis (NE) from 1 to 21 d of age. A total of 576 one-day-old broilers were assigned to a 2 × 2 × 2 design for diets containing AFB1 (0 or 40 μg/kg), NE (challenged or unchallenged), or YCW (0 or 500 mg/kg). The main effect analysis showed that AF depressed (P < 0.01) average daily feed intake (ADFI), average daily body weight gain (ADG), the mRNA profiles of polymeric Ig receptor (pIgR), claudin-1, and occludin, but increased (P < 0.001) liver lesion scores, serum endotoxin, and diamine oxidase (DAO). The NE challenge depressed (P < 0.01) ADFI, ADG, secretory IgA (sIgA), pIgR, claudin-1, occludin, and the populations of Lactobacilli and Bifidobacteria, but increased (P < 0.001) visceral lesions, endotoxins, and DAO. The main effect of YCW on growth performance, visceral lesions, and intestinal integrity was not significant, but decreased (P < 0.01) mortality, endotoxin, DAO, and C. perfringens, and increased (P < 0.05) the populations of Lactobacilli and Bifidobacteria. There were 3-way interactions (P < 0.05) on growth performance, intestinal lesions, integrity, and gut bacteria. Compared with the treatment with the dual challenges, there were pronounced effects (P < 0.05) of YCW on ADFI, ADG, lesions, DAO, pIgR, and Bifidobacteria. The results suggest that with the concurrent challenges of AF and NE, the YCW can partially protect the growth performance and intestinal health of broilers.
Collapse
Affiliation(s)
- N Liu
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471003, China
| | - J Q Wang
- Department of Poultry Science, University of Georgia, Athens 30602, GA, USA
| | - S C Jia
- Department of Animal Science, Texas A&M University, College Station 77843, TX, USA
| | - Y K Chen
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471003, China
| | - J P Wang
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
26
|
Zhang Z, Yu W, Chen S, Chen Y, Chen L, Zhang S. Methylation of the claudin‑3 promoter predicts the prognosis of advanced gastric adenocarcinoma. Oncol Rep 2018; 40:49-60. [PMID: 29749528 PMCID: PMC6059754 DOI: 10.3892/or.2018.6411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/13/2018] [Indexed: 02/07/2023] Open
Abstract
Claudin-3 expression is associated with gastric cancer progression, but the role of epigenetic modifications remains unclear. We investigated methylation of the claudin-3 promoter and expression profiles in gastric adenocarcinoma and their associations with clinicopathological characteristics and prognosis of the patients. A total of 122 patients with advanced gastric cancer [stage IIB-IV, with lymph node (LN) metastasis] were enrolled. Each patient provided 4 tissue samples: normal gastric epithelium, intestinal metaplasia, primary tumor and metastatic LN. Claudin-3 protein expression was examined by immunohistochemistry. Claudin-3 promoter methylation was determined by methylation-specific PCR and verified by bisulfite sequencing PCR. Claudin-3 mRNA expression was measured by real-time PCR in a subset of cases, and its correlation with protein expression was analyzed using Spearman correlation. Kaplan-Meier survival analysis was performed (log-rank test). Factors associated with survival were identified by Cox regression. The strong expression rate of claudin-3 in intestinal metaplasia, primary tumor, metastatic LN and normal gastric epithelium was 91.8, 58.2, 30.3 and 13.9%, respectively. The promoter hypermethylation rate in intestinal metaplasia, primary tumor, normal gastric epithelium and metastatic LN was 5.7, 27.9, 36.9 and 49.2%, respectively. Claudin-3 mRNA and protein expression were positively correlated (P<0.001) with normal gastric epithelium (rs=0.745), intestinal metaplasia (rs=0.876), primary gastric adenocarcinoma (rs=0.915) and metastatic LN (rs=0.819). Claudin-3 mRNA expression was negatively correlated with claudin-3 promoter methylation. Median patient survival was 38, 22 and 11 months in the hypomethylated, partially methylated and hypermethylated groups, respectively (P<0.001). Claudin-3 promoter methylation status (HR: 5.67; 95% CI: 2.27–14.17) but not claudin-3 expression was an independent predictor of survival. Claudin-3 promoter hypermethylation reduces claudin-3 expression and independently predicts poor prognosis.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, P.R. China
| | - Weixing Yu
- Institute of Translational Medicine, Fujian Medical University, Fujian, Fuzhou 350122, P.R. China
| | - Shuqin Chen
- Department of Pathology, Fujian Medical University, Fujian, Fuzhou 350122, P.R. China
| | - Yupeng Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, P.R. China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, P.R. China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, P.R. China
| |
Collapse
|
27
|
Li J, Zhou C, Ni S, Wang S, Ni C, Yang P, Ye M. Methylated claudin-11 associated with metastasis and poor survival of colorectal cancer. Oncotarget 2017; 8:96249-96262. [PMID: 29221203 PMCID: PMC5707097 DOI: 10.18632/oncotarget.21997] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022] Open
Abstract
As one of crucial epigenetic modification, DNA methylation plays an important role during the carcinogenesis of colorectal cancer (CRC). In the current study, we used a human genome methylation array to detect the aberrant methylation genes in CRC. We further identified the hypermethylation of claudin-11 (CLDN11) and proved inverse correlation between CLDN11 methylation and its expression in CRC. In vitro experiments showed debased migration ability of colonic cancer cells in accompany with the converted methylation of CLDN11 after colonic cancer cells treated with demethylation agent, 5-aza-2'-deoxycytidine. Besides, our results also represented that hypermethylation of CLDN11 was associated with increased metastatic potential of CRC and with low progression free survival (PFS) of CRC. In conclusion, our findings supported that the hypermethylated CLDN11 is associated with metastasis of CRC and prognosis of poor survival of CRC.
Collapse
Affiliation(s)
- Jinyun Li
- Department of Oncology and Hematology, Affiliated Hospital, Medical School of Ningbo University, Ningbo, Zhejiang 315000, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital of Ningbo University, Ningbo 315040, Zhejiang, China
| | - Shumin Ni
- Department of Oncology and Hematology, Affiliated Hospital, Medical School of Ningbo University, Ningbo, Zhejiang 315000, China
| | - Shaomin Wang
- Department of Oncology and Hematology, Affiliated Hospital, Medical School of Ningbo University, Ningbo, Zhejiang 315000, China
| | - Chao Ni
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Ping Yang
- Medical School, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Meng Ye
- Department of Oncology and Hematology, Affiliated Hospital, Medical School of Ningbo University, Ningbo, Zhejiang 315000, China
| |
Collapse
|