1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Yoo M, Kayastha N, Kwon O, Man W, Cai L, Schachner M. Analysis of the functional sequences in the promoter region of the human adhesion molecule close homolog of L1. Int J Neurosci 2022; 132:483-489. [PMID: 33054469 DOI: 10.1080/00207454.2020.1822357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/13/2020] [Accepted: 09/04/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Close Homolog of L1 (CHL1) is a member of the L1 family of cell adhesion molecules. CHL1 gene is located on human chromosome 3 and has been linked to several pathologies, including 3p deletion syndrome, schizophrenia, and tumor growth and metastasis. OBJECTIVE The goal of the present study was to determine which region of the CHL1 promoter is most competent in driving CHL1 gene expression. Methods: Five candidate DNA fragments in the promoter regions were selected by screening across six species for evolutionary conserved sequences. The activity of these five promoter regions was quantitatively evaluated using a GFP reporter gene in transfection experiments, performed in C6 glioma cells. RESULTS Of the five promoter regions tested, three drove reporter GFP expression, with the conserved region 6 (CR6, Gene ID AC066595.5, 25851-26850) being the most active for transcription. CONCLUSION The identification of the CR6 activity provides a better understanding of the regulatory mechanisms underlying CHL1 expression. It may help future discovery of therapeutic strategies that involve influencing critical promoter regions to drive transcriptional regulation of the mammalian CHL1 gene.HIGHLIGHTSConserved regions of CHL1 promoter sequences were identified by in-silico analysis.Five conserved regions were tested for gene regulatory activity using a reporter assay.Conserved regions CR5, CR6 and CR7 show gene regulatory function in a reporter assay.Co-transfection of CR5 and CR6 yielded the highest reporter activity.The core region of CR6 (CR6core) was identified as a cis-acting element.In-tandem promoter CR5core-CR6core was the best in a reporter assay.
Collapse
Affiliation(s)
- Myungsik Yoo
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Neha Kayastha
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Ohyoon Kwon
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Wai Man
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Melitta Schachner
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
3
|
Karstens KF, Bellon E, Polonski A, Wolters-Eisfeld G, Melling N, Reeh M, Izbicki JR, Tachezy M. Expression and serum levels of the neural cell adhesion molecule L1-like protein (CHL1) in gastrointestinal stroma tumors (GIST) and its prognostic power. Oncotarget 2020; 11:1131-1140. [PMID: 32284790 PMCID: PMC7138165 DOI: 10.18632/oncotarget.27525] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/03/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Diagnosis of gastrointestinal stroma tumors (GIST) is based on the histological evaluation of tissue specimens. Reliable systemic biomarkers are lacking. We investigated the local expression of the neural cell adhesion molecule L1-like protein (CHL1) in GIST and determined whether soluble CHL1 proteoforms could serve as systemic biomarkers. MATERIAL AND METHODS Expression of CHL1 was analyzed in primary tumor specimens and metastases. 58 GIST specimens were immunohistochemically stained for CHL1 on a tissue microarray (TMA). Systemic CHL1 levels were measured in sera derived from 102 GIST patients and 91 healthy controls by ELISA. Results were statistically correlated with clinicopathological parameters. RESULTS CHL1 expression was detected in GIST specimens. Reduced tissue expression was significantly associated with advanced UICC stages (p = 0.036) and unfavorable tumor localization (p = 0.001). CHL1 serum levels are significantly elevated in GIST patients (p < 0.010). Elevated CHL1 levels were significantly associated with larger tumors (p = 0.023), advanced UICC stage (p = 0.021), and an increased Fletcher score (p = 0.041). Moreover, patients with a higher CHL1 serum levels displayed a significantly shortened recurrence free survival independent of other clinicopathological variables. CONCLUSION Local CHL1 expression and serum CHL1 levels show a reverse prognostic behavior, highlighting the relevance of proteolytic shedding of the molecule. The results of the study indicate a potential role of serum CHL1 as a diagnostic and prognostic marker in GIST.
Collapse
Affiliation(s)
- Karl-Frederick Karstens
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Eugen Bellon
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Adam Polonski
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| |
Collapse
|
4
|
Wachowiak R, Mayer S, Suttkus A, Martynov I, Lacher M, Melling N, Izbicki JR, Tachezy M. CHL1 and NrCAM are Primarily Expressed in Low Grade Pediatric Neuroblastoma. Open Med (Wars) 2019; 14:920-927. [PMID: 31989042 PMCID: PMC6972343 DOI: 10.1515/med-2019-0109] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 10/19/2019] [Indexed: 12/28/2022] Open
Abstract
Background Neural cell adhesion molecules like close homolog of L1 protein (CHL1) and neuronal glia related cell adhesion molecule (NrCAM) play an important role in development and regeneration of the central nervous system. However, they are also associated with cancerogenesis and progression in adult malignancies, thus gain increasing importance in cancer research. We therefore studied the expression of CHL1 and NrCAM according to the course of disease in children with neuroblastoma. Methods CHL1 and NrCAM expression levels were histologically assessed by tissue microarrays from surgically resected neuroblastoma specimens of 56 children. Expression of both markers was correlated to demographics as well as clinical data including metastatic dissemination and survival. Results CHL1 was expressed in 9% and NrCAM in 51% of neuroblastoma tissue samples. Expression of CHL1 was higher in patients with low Hughes grade 1a/b (p=0.01). NrCAM was more often detected in patients with a low International Staging System (INSS) score 1/2 (p=0.04). Conclusion CHL1 and NrCAM expression was associated with low-grade pediatric neuroblastoma. These adhesion molecules may play a role in early tumor development of neuroblastoma.
Collapse
Affiliation(s)
- Robin Wachowiak
- Department of Pediatric Surgery, University Hospital Leipzig, Liebigstrasse 20 A, 04103 Leipzig, Germany
| | - Steffi Mayer
- Department of Pediatric Surgery, University Hospital Leipzig, Liebigstrasse 20 A, 04103 Leipzig, Germany
| | - Anne Suttkus
- Department of Pediatric Surgery, University Hospital Leipzig, Liebigstrasse 20 A, 04103 Leipzig, Germany
| | - Illya Martynov
- Department of Pediatric Surgery, University Hospital Leipzig, Liebigstrasse 20 A, 04103 Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University Hospital Leipzig, Liebigstrasse 20 A, 04103 Leipzig, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, 20246, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, 20246, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, 20246, Germany
| |
Collapse
|
5
|
Chen J, Jiang C, Fu L, Zhu CL, Xiang YQ, Jiang LX, Chen Q, Liu WM, Chen JN, Zhang LY, Liu M, Chen C, Tang H, Wang B, Tsao SW, Kwong DLW, Guan XY. CHL1 suppresses tumor growth and metastasis in nasopharyngeal carcinoma by repressing PI3K/AKT signaling pathway via interaction with Integrin β1 and Merlin. Int J Biol Sci 2019; 15:1802-1815. [PMID: 31523184 PMCID: PMC6743306 DOI: 10.7150/ijbs.34785] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/19/2019] [Indexed: 01/24/2023] Open
Abstract
Deletion of Chromosome 3p is one of the most frequently detected genetic alterations in nasopharyngeal carcinoma (NPC). We reported the role of a novel 3p26.3 tumor suppressor gene (TSG) CHL1 in NPC. Down-regulation of CHL1 was detected in 4/6 of NPC cell lines and 71/95 (74.7%) in clinical tissues. Ectopic expressions of CHL1 in NPC cells significantly inhibit colony formation and cell motility in functional study. By up-regulating epithelial markers and down-regulating mesenchymal markers CHL1 could induce mesenchymal-epithelial transition (MET), a key step in preventing tumor invasion and metastasis. CHL1 could also cause the inactivation of RhoA/Rac1/Cdc42 signaling pathway and inhibit the formation of stress fiber, lamellipodia, and filopodia. CHL1 could co-localize with adhesion molecule Integrin-β1, the expression of CHL1 was positively correlated with Integrin-β1 and another known tumor suppressor gene (TSG) Merlin. Down-regulation of Integrin-β1 or Merlin was significantly correlated with the poor survival rate of NPC patients. Further mechanistic studies showed that CHL1 could directly interact with integrin-β1 and link to Merlin, leading to the inactivation of integrin β1-AKT pathway. In conclusion, CHL1 is a vital tumor suppressor in the carcinogenesis of NPC.
Collapse
Affiliation(s)
- Juan Chen
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;,Department of Clinical Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University
| | - Chen Jiang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Research Centre, Shenzhen University school of Medicine, Shenzhen, China
| | - Cai-Lei Zhu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal, Sun Yat-Sen Cancer Center, Guangzhou, China
| | - Ling-Xi Jiang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qian Chen
- Departments of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai Man Liu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin-Na Chen
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li-Yi Zhang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ming Liu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Science and Technology of Huazhong University, Wuhan, China
| | - Hong Tang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bo Wang
- Department of Clinical Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University
| | - Sai Wah Tsao
- Departments of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;,State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, China;,✉ Corresponding author: Xin-Yuan Guan, Department of Clinical Oncology, The University of Hong Kong, Room L10-56, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, Tel: 852-3917-9782, E-Mail: ; or Dora Lai-Wan Kwong, Department of Clinical Oncology, University of Hong Kong, 1/F, Professorial Block, Queen Mary Hospital, Hong Kong, Tel: 852-28554521, E-mail:
| |
Collapse
|
6
|
Li W, Xia S, Aronova A, Min IM, Verma A, Scognamiglio T, Gray KD, Ullmann TM, Liang H, Moore MD, Elemento O, Zarnegar R, Fahey TJ. CHL1 expression differentiates Hürthle cell carcinoma from benign Hürthle cell nodules. J Surg Oncol 2018; 118:1042-1049. [PMID: 30311656 DOI: 10.1002/jso.25214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Hürthle cell carcinoma (HCC) is an unusual and relatively rare type of differentiated thyroid cancer. Currently, cytologic analysis of fine-needle aspiration biopsy is limited in distinguishing benign Hürthle cell neoplasms from malignant ones. The aim of this study was to determine whether differences in the expression of specific genes could differentiate HCC from benign Hürthle cell nodules by evaluating differential gene expression in Hürthle cell disease. METHODS Eighteen benign Hürthle cell nodules and seven HCC samples were analyzed by whole-transcriptome sequencing. Bioinformatics analysis was carried out to identify candidate differentially expressed genes. Expression of these candidate genes was re-examined by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression was quantified by immunohistochemistry. RESULTS Close homolog of L1 (CHL1) was identified as overexpressed in HCC. CHL1 was found to have greater than 15-fold higher expression in fragments per kilobase million in HCC compared with benign Hurthle cell tumors. This was confirmed by qRT-PCR. Moreover, the immunoreactivity score of the CHL1 protein was significantly higher in HCC compared with benign Hürthle cell nodules. CONCLUSIONS CHL1 expression may represent a novel and useful prognostic biomarker to distinguish HCC from benign Hürthle cell disease.
Collapse
Affiliation(s)
- Wei Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Shujun Xia
- Ultrasound Department, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anna Aronova
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Irene M Min
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Akanksha Verma
- Department of Physiology and Biophysics, Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Theresa Scognamiglio
- Department of Pathology, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Katherine D Gray
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Timothy M Ullmann
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Heng Liang
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Maureen D Moore
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Rasa Zarnegar
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Thomas J Fahey
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| |
Collapse
|
7
|
de Cristofaro T, Di Palma T, Soriano AA, Monticelli A, Affinito O, Cocozza S, Zannini M. Candidate genes and pathways downstream of PAX8 involved in ovarian high-grade serous carcinoma. Oncotarget 2018; 7:41929-41947. [PMID: 27259239 PMCID: PMC5173106 DOI: 10.18632/oncotarget.9740] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/16/2016] [Indexed: 12/26/2022] Open
Abstract
Understanding the biology and molecular pathogenesis of ovarian epithelial cancer (EOC) is key to developing improved diagnostic and prognostic indicators and effective therapies. Although research has traditionally focused on the hypothesis that high-grade serous carcinoma (HGSC) arises from the ovarian surface epithelium (OSE), recent studies suggest that additional sites of origin exist and a substantial proportion of cases may arise from precursor lesions located in the Fallopian tubal epithelium (FTE). In FTE cells, the transcription factor PAX8 is a marker of the secretory cell lineage and its expression is retained in 96% of EOC. We have recently reported that PAX8 is involved in the tumorigenic phenotype of ovarian cancer cells. In this study, to uncover genes and pathways downstream of PAX8 involved in ovarian carcinoma we have determined the molecular profiles of ovarian cancer cells and in parallel of Fallopian tube epithelial cells by means of a silencing approach followed by an RNA-seq analysis. Interestingly, we highlighted the involvement of pathways like WNT signaling, epithelial-mesenchymal transition, p53 and apoptosis. We believe that our analysis has led to the identification of candidate genes and pathways regulated by PAX8 that could be additional targets for the therapy of ovarian carcinoma.
Collapse
Affiliation(s)
- Tiziana de Cristofaro
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Tina Di Palma
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Amata Amy Soriano
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Antonella Monticelli
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Ornella Affinito
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Sergio Cocozza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Mariastella Zannini
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| |
Collapse
|
8
|
CHL1 gene polymorphisms increase lung cancer susceptibility. Oncotarget 2018; 9:13545-13550. [PMID: 29568376 PMCID: PMC5862597 DOI: 10.18632/oncotarget.24057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/16/2017] [Indexed: 01/12/2023] Open
Abstract
Lung cancer represents a complex and malignant cancer. Close Homologue of L1 (CHL1) gene plays a crucial role in the progress of cancer. The aim of this study is to explore the association between CHL1 rs425366 polymorphism and lung cancer susceptibility in northeast of China. A hospital-based case-control study was carried out to collect relative characteristics. Logistic regression analysis was conducted to analyze the relationship between single nucleotide polymorphisms and lung cancer susceptibility. The results suggested that there was statistically significant difference between GT genotype and TT genotype of rs425366 and lung cancer susceptibility. In stratified analysis, TT genotype of rs425366 may increase the risk of lung adenocarcinoma. We also found that non-smoking individuals carrying T allele were more likely to develop lung cancer. Overall, our study may indicate that CHL1 gene may increase lung cancer susceptibility in northeast of China.
Collapse
|
9
|
Baharudin R, Ab Mutalib NS, Othman SN, Sagap I, Rose IM, Mohd Mokhtar N, Jamal R. Identification of Predictive DNA Methylation Biomarkers for Chemotherapy Response in Colorectal Cancer. Front Pharmacol 2017; 8:47. [PMID: 28243201 PMCID: PMC5303736 DOI: 10.3389/fphar.2017.00047] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022] Open
Abstract
Resistance to 5-Fluorouracil (5-FU) is a major obstacle to the successful treatment of colorectal cancer (CRC) and posed an increased risk of recurrence. DNA methylation has been suggested as one of the underlying mechanisms for recurrent disease and its contribution to the development of drug resistance remains to be clarified. This study aimed to determine the methylation phenotype in CRC for identification of predictive markers for chemotherapy response. We performed DNA methylation profiling on 43 non-recurrent and five recurrent CRC patients using the Illumina Infinium HumanMethylation450 Beadchip assay. In addition, CRC cells with different genetic backgrounds, response to 5-FU and global methylation levels (HT29 and SW48) were treated with 5-FU and DNA methylation inhibitor 5-aza-2′-deoxycytidine (5-azadC). The singular and combined effects of these two drug classes on cell viability and global methylation profiles were investigated. Our genome-wide methylation study on the clinical specimens showed that recurrent CRCs exhibited higher methylation levels compared to non-recurrent CRCs. We identified 4787 significantly differentially methylated genes (P < 0.05); 3112 genes were hyper- while 1675 genes were hypomethylated in the recurrent group compared to the non-recurrent. Fifty eight and 47 of the significantly hypermethylated and hypomethylated genes have an absolute recurrent/non-recurrent methylation difference of ≥20%. Most of the hypermethylated genes were involved in the MAPK signaling pathway which is a key regulator for apoptosis while the hypomethylated genes were involved in the PI3K-AKT signaling pathway and proliferation process. We also demonstrate that 5-azadC treatment enhanced response to 5-FU which resulted in significant growth inhibition compared to 5-FU alone in hypermethylated cell lines SW48. In conclusion, we found the evidence of five potentially biologically important genes in recurrent CRCs that could possibly serve as a new potential therapeutic targets for patients with chemoresistance. We postulate that aberrant methylation of CCNEI, CCNDBP1, PON3, DDX43, and CHL1 in CRC might be associated with the recurrence of CRC and 5-azadC-mediated restoration of 5-FU sensitivity is mediated at least in part by MAPK signaling pathway.
Collapse
Affiliation(s)
- Rashidah Baharudin
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| | | | - Sri N Othman
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| | - Ismail Sagap
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| | - Isa M Rose
- Department of Clinical Oral Biology, Faculty of Dentistry, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Yang Z, Xie Q, Hu CL, Jiang Q, Shen HF, Schachner M, Zhao WJ. CHL1 Is Expressed and Functions as a Malignancy Promoter in Glioma Cells. Front Mol Neurosci 2017; 10:324. [PMID: 29089868 PMCID: PMC5650976 DOI: 10.3389/fnmol.2017.00324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/25/2017] [Indexed: 02/06/2023] Open
Abstract
The cell adhesion molecule with homology to L1CAM (close homolog of L1) (CHL1) is a member of the cell adhesion molecule L1 (L1CAM) gene family. Although CHL1 expression and function have been reported in several tumors, the roles of CHL1 in the development of glioma remain unclear. In the present study, we investigated the effects of CHL1 on proliferation indexes and activation of Akt1 and Erk signaling by siRNA in U-87 MG human glioblastoma and human U251 and SHG-44 glioma cells. We found that siRNA targeting CHL1 significantly down-regulated the expression of CHL1 mRNA and protein accompanied by reduced cell proliferation and transmigration invasion in all three cell lines. Down-regulating CHL1 expression also reduced cell survival, as measured by the Bax/Bcl-2 ratio, and increased activation of caspase-3. In subcutaneous U-87 MG cell xenograft tumors in nude mice, intratumoral administration of siRNA targeting CHL1 treatment significantly down-regulated CHL1 expression in vivo, accompanied by increased levels of activated caspase-3. Our combined results confirmed for the first time that in contrast to findings about CHL1 in most other cancer types, CHL1 functions in promoting cell proliferation, metastasis and migration in human glioma cells both in vitro and in vivo. These results indicate that CHL1 is a therapeutic target in the clinical management of glioma/glioblastoma.
Collapse
Affiliation(s)
- Zhai Yang
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Qing Xie
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Cheng-Liang Hu
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Qiong Jiang
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Hui-Fan Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Melitta Schachner Wei-Jiang Zhao
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- *Correspondence: Melitta Schachner Wei-Jiang Zhao
| |
Collapse
|
11
|
Horpaopan S, Spier I, Zink AM, Altmüller J, Holzapfel S, Laner A, Vogt S, Uhlhaas S, Heilmann S, Stienen D, Pasternack SM, Keppler K, Adam R, Kayser K, Moebus S, Draaken M, Degenhardt F, Engels H, Hofmann A, Nöthen MM, Steinke V, Perez-Bouza A, Herms S, Holinski-Feder E, Fröhlich H, Thiele H, Hoffmann P, Aretz S. Genome-wide CNV analysis in 221 unrelated patients and targeted high-throughput sequencing reveal novel causative candidate genes for colorectal adenomatous polyposis. Int J Cancer 2014; 136:E578-89. [PMID: 25219767 DOI: 10.1002/ijc.29215] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/04/2014] [Accepted: 09/03/2014] [Indexed: 12/27/2022]
Abstract
To uncover novel causative genes in patients with unexplained adenomatous polyposis, a model disease for colorectal cancer, we performed a genome-wide analysis of germline copy number variants (CNV) in a large, well characterized APC and MUTYH mutation negative patient cohort followed by a targeted next generation sequencing (NGS) approach. Genomic DNA from 221 unrelated German patients was genotyped on high-resolution SNP arrays. Putative CNVs were filtered according to stringent criteria, compared with those of 531 population-based German controls, and validated by qPCR. Candidate genes were prioritized using in silico, expression, and segregation analyses, data mining and enrichment analyses of genes and pathways. In 27% of the 221 unrelated patients, a total of 77 protein coding genes displayed rare, nonrecurrent, germline CNVs. The set included 26 candidates with molecular and cellular functions related to tumorigenesis. Targeted high-throughput sequencing found truncating point mutations in 12% (10/77) of the prioritized genes. No clear evidence was found for autosomal recessive subtypes. Six patients had potentially causative mutations in more than one of the 26 genes. Combined with data from recent studies of early-onset colorectal and breast cancer, recurrent potential loss-of-function alterations were detected in CNTN6, FOCAD (KIAA1797), HSPH1, KIF26B, MCM3AP, YBEY and in three genes from the ARHGAP family. In the canonical Wnt pathway oncogene CTNNB1 (β-catenin), two potential gain-of-function mutations were found. In conclusion, the present study identified a group of rarely affected genes which are likely to predispose to colorectal adenoma formation and confirmed previously published candidates for tumor predisposition as etiologically relevant.
Collapse
|
12
|
Sui WG, He HY, Yan Q, Chen JJ, Zhang RH, Dai Y. ChIP-seq analysis of histone H3K9 trimethylation in peripheral blood mononuclear cells of membranous nephropathy patients. ACTA ACUST UNITED AC 2013; 47:42-9. [PMID: 24345872 PMCID: PMC3932972 DOI: 10.1590/1414-431x20132809] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 08/27/2013] [Indexed: 12/19/2022]
Abstract
Membranous nephropathy (MN), characterized by the presence of diffuse thickening of
the glomerular basement membrane and subepithelial in situ immune
complex disposition, is the most common cause of idiopathic nephrotic syndrome in
adults, with an incidence of 5-10 per million per year. A number of studies have
confirmed the relevance of several experimental insights to the pathogenesis of human
MN, but the specific biomarkers of MN have not been fully elucidated. As a result,
our knowledge of the alterations in histone methylation in MN is unclear. We used
chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) to
analyze the variations in a methylated histone (H3K9me3) in peripheral blood
mononuclear cells from 10 MN patients and 10 healthy subjects. There were 108 genes
with significantly different expression in the MN patients compared with the normal
controls. In MN patients, significantly increased activity was seen in 75 H3K9me3
genes, and decreased activity was seen in 33, compared with healthy subjects. Five
positive genes, DiGeorge syndrome critical region gene 6 (DGCR6), sorting nexin 16
(SNX16), contactin 4 (CNTN4), baculoviral IAP repeat containing 3 (BIRC3), and
baculoviral IAP repeat containing 2 (BIRC2), were selected and quantified. There were
alterations of H3K9me3 in MN patients. These may be candidates to help explain
pathogenesis in MN patients. Such novel findings show that H3K9me3 may be a potential
biomarker or promising target for epigenetic-based MN therapies.
Collapse
Affiliation(s)
- W G Sui
- 181st Hospital, Nephrology Department, Guangxi Key Laboratory of Metabolic Diseases Research, GuilinGuangxi, China, Guangxi Key Laboratory of Metabolic Diseases Research, Nephrology Department, 181st Hospital, Guilin, Guangxi, China
| | - H Y He
- Guangxi Normal University, The Life Science College, GuilinGuangxi, China, The Life Science College, Guangxi Normal University, Guilin, Guangxi, China
| | - Q Yan
- 181st Hospital, Nephrology Department, Guangxi Key Laboratory of Metabolic Diseases Research, GuilinGuangxi, China, Guangxi Key Laboratory of Metabolic Diseases Research, Nephrology Department, 181st Hospital, Guilin, Guangxi, China
| | - J J Chen
- 181st Hospital, Nephrology Department, Guangxi Key Laboratory of Metabolic Diseases Research, GuilinGuangxi, China, Guangxi Key Laboratory of Metabolic Diseases Research, Nephrology Department, 181st Hospital, Guilin, Guangxi, China
| | - R H Zhang
- Guangxi Normal University, The Life Science College, GuilinGuangxi, China, The Life Science College, Guangxi Normal University, Guilin, Guangxi, China
| | - Y Dai
- Jinan University, Shenzhen People's Hospital, The Second Clinical Medical College, Clinical Medical Research Center, ShenzhenGuangdong, China, Clinical Medical Research Center, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
He LH, Ma Q, Shi YH, Ge J, Zhao HM, Li SF, Tong ZS. CHL1 is involved in human breast tumorigenesis and progression. Biochem Biophys Res Commun 2013; 438:433-8. [PMID: 23906755 DOI: 10.1016/j.bbrc.2013.07.093] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 07/23/2013] [Indexed: 12/28/2022]
Abstract
Neural cell adhesion molecules (CAM) play important roles in the development and regeneration of the nervous system. The L1 family of CAMs is comprised of L1, Close Homolog of L1 (CHL1, L1CAM2), NrCAM, and Neurofascin, which are structurally related trans-membrane proteins in vertebrates. Although the L1CAM has been demonstrated play important role in carcinogenesis and progression, the function of CHL1 in human breast cancer is limited. Here, we found that CHL1 is down-regulated in human breast cancer and related to lower grade. Furthermore, overexpression of CHL1 suppresses proliferation and invasion in MDA-MB-231 cells and knockdown of CHL1 expression results in increased proliferation and invasion in MCF7 cells in vitro. Finally, CHL1 deficiency promotes tumor formation in vivo. Our results may provide a strategy for blocking breast carcinogenesis and progression.
Collapse
Affiliation(s)
- Li-Hong He
- Medical Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Quinn MCJ, Wojnarowicz PM, Pickett A, Provencher DM, Mes-Masson AM, Davis EC, Tonin PN. FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome. Int J Oncol 2013; 42:912-20. [PMID: 23354471 DOI: 10.3892/ijo.2013.1797] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/05/2012] [Indexed: 11/06/2022] Open
Abstract
The frequent loss of chromosome 17 in epithelial ovarian carcinomas (EOC), particularly high-grade serous carcinomas (HGSC), has been attributed to the disruption of TP53 (at 17p13.1) and other chromosome 17 genes suspected to play a role in tumour suppressor pathways. In a transcriptome analysis of HGSC, we showed underexpression of a number of chromosome 17 genes, which included FKBP10 (at 17q21.1) and collagen I α 1 (COL1A1; at 17q21.33). FKBP10 codes for the immunophilin FKBP65 and is suspected to act as a chaperone for COL1A1. We have investigated FKBP10 (gene) and FKBP65 (protein) expression in HGSC samples and EOC cell lines that differ in their tumourigenic potential. COL1A1 expression was also investigated given the purported function of FKBP65. RT-PCR analysis verified underexpression of FKBP10 and COL1A1 in HGSCs (n=14) and six tumourigenic EOC cell lines, relative to normal ovarian surface epithelial cells and a non-tumourigenic EOC cell line. Immunohistochemistry analyses of 196 HGSC samples using tissue microarrays revealed variable staining intensities in the epithelial tumour component where only 7.8% and 1.0% of samples stained intensely for FKBP65 and COL1A1, respectively. Variable staining intensities were also observed for the stromal component where 23.6% and 24.1% stained intensely for FKBP65 and COL1A1, respectively. There was no significant correlation of staining intensity of either protein with disease stage. Staining of FKBP65 was clearly visible in normal epithelial cells of the ovarian surface and fallopian tube. There was a significant correlation between absence of FKBP65 staining in the epithelial cell component of the tumour and prolonged overall survival (p<0.001). Our results suggest that underexpression of FKBP65 protein is characteristic of HGSCs and that this expression profile may be linked to molecular pathways associated with an unfavourable outcome in cancer patients.
Collapse
Affiliation(s)
- Michael C J Quinn
- Research Centre of the University of Montreal Hospital Centre/Montreal Cancer Institute, Montreal, QC, Canada
| | | | | | | | | | | | | |
Collapse
|
15
|
Chen J, Fu L, Zhang LY, Kwong DL, Yan L, Guan XY. Tumor suppressor genes on frequently deleted chromosome 3p in nasopharyngeal carcinoma. CHINESE JOURNAL OF CANCER 2012; 31:215-22. [PMID: 22360856 PMCID: PMC3777521 DOI: 10.5732/cjc.011.10364] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 01/30/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is among the most common malignancies in southern China. Deletion of genomic DNA, which occurs during the complex pathogenesis process for NPC, represents a pivotal mechanism in the inactivation of tumor suppressor genes (TSGs). In many circumstances, loss of TSGs can be detected as diagnostic and prognostic markers in cancer. The short arm of chromosome 3 (3p) is a frequently deleted chromosomal region in NPC, with 3p21.1-21.2 and 3p25.2-26.1 being the most frequently deleted minimal regions. In recent years, our research group and others have focused on the identification and characterization of novel target TSGs at 3p, such as RASSF1A, BLU, RBMS3, and CHL1, in the development and progression of NPC. In this review, we summarize recent findings of TSGs at 3p and discuss some of these genes in detail. A better understanding of TSGs at 3p will significantly improve our understanding of NPC pathogenesis, diagnosis, and treatment.
Collapse
Affiliation(s)
- Juan Chen
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P. R. China;
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P. R. China;
| | - Li Fu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P. R. China;
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China.
| | - Li-Yi Zhang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P. R. China;
| | - Dora L. Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P. R. China;
| | - Li Yan
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China.
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P. R. China;
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China.
| |
Collapse
|
16
|
Birch AH, Arcand SL, Oros KK, Rahimi K, Watters AK, Provencher D, Greenwood CM, Mes-Masson AM, Tonin PN. Chromosome 3 anomalies investigated by genome wide SNP analysis of benign, low malignant potential and low grade ovarian serous tumours. PLoS One 2011; 6:e28250. [PMID: 22163003 PMCID: PMC3232202 DOI: 10.1371/journal.pone.0028250] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/04/2011] [Indexed: 02/01/2023] Open
Abstract
Ovarian carcinomas exhibit extensive heterogeneity, and their etiology remains unknown. Histological and genetic evidence has led to the proposal that low grade ovarian serous carcinomas (LGOSC) have a different etiology than high grade carcinomas (HGOSC), arising from serous tumours of low malignant potential (LMP). Common regions of chromosome (chr) 3 loss have been observed in all types of serous ovarian tumours, including benign, suggesting that these regions contain genes important in the development of all ovarian serous carcinomas. A high-density genome-wide genotyping bead array technology, which assayed >600,000 markers, was applied to a panel of serous benign and LMP tumours and a small set of LGOSC, to characterize somatic events associated with the most indolent forms of ovarian disease. The genomic patterns inferred were related to TP53, KRAS and BRAF mutations. An increasing frequency of genomic anomalies was observed with pathology of disease: 3/22 (13.6%) benign cases, 40/53 (75.5%) LMP cases and 10/11 (90.9%) LGOSC cases. Low frequencies of chr3 anomalies occurred in all tumour types. Runs of homozygosity were most commonly observed on chr3, with the 3p12-p11 candidate tumour suppressor region the most frequently homozygous region in the genome. An LMP harboured a homozygous deletion on chr6 which created a GOPC-ROS1 fusion gene, previously reported as oncogenic in other cancer types. Somatic TP53, KRAS and BRAF mutations were not observed in benign tumours. KRAS-mutation positive LMP cases displayed significantly more chromosomal aberrations than BRAF-mutation positive or KRAS and BRAF mutation negative cases. Gain of 12p, which harbours the KRAS gene, was particularly evident. A pathology review reclassified all TP53-mutation positive LGOSC cases, some of which acquired a HGOSC status. Taken together, our results support the view that LGOSC could arise from serous benign and LMP tumours, but does not exclude the possibility that HGOSC may derive from LMP tumours.
Collapse
Affiliation(s)
- Ashley H. Birch
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Suzanna L. Arcand
- The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kathleen K. Oros
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
| | - Kurosh Rahimi
- Department of Pathology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - A. Kevin Watters
- Department of Pathology, McGill University and McGill University Health Centre (MUHC), Montréal, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Institut du cancer de Montréal, Montreal, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, Canada
| | - Celia M. Greenwood
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Institut du cancer de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Patricia N. Tonin
- Department of Human Genetics, McGill University, Montreal, Canada
- The Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Medicine, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
17
|
Senchenko VN, Krasnov GS, Dmitriev AA, Kudryavtseva AV, Anedchenko EA, Braga EA, Pronina IV, Kondratieva TT, Ivanov SV, Zabarovsky ER, Lerman MI. Differential expression of CHL1 gene during development of major human cancers. PLoS One 2011; 6:e15612. [PMID: 21408220 PMCID: PMC3049765 DOI: 10.1371/journal.pone.0015612] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 11/17/2010] [Indexed: 11/19/2022] Open
Abstract
Background CHL1 gene (also known as CALL) on 3p26.3 encodes a one-pass trans-membrane cell adhesion molecule (CAM). Previously CAMs of this type, including L1, were shown to be involved in cancer growth and metastasis. Methodology/Principal Findings We used Clontech Cancer Profiling Arrays (19 different types of cancers, 395 samples) to analyze expression of the CHL1 gene. The results were further validated by RT-qPCR for breast, renal and lung cancer. Cancer Profiling Arrays revealed differential expression of the gene: down-regulation/silencing in a majority of primary tumors and up-regulation associated with invasive/metastatic growth. Frequent down-regulation (>40% of cases) was detected in 11 types of cancer (breast, kidney, rectum, colon, thyroid, stomach, skin, small intestine, bladder, vulva and pancreatic cancer) and frequent up-regulation (>40% of cases) – in 5 types (lung, ovary, uterus, liver and trachea) of cancer. Using real-time quantitative PCR (RT-qPCR) we found that CHL1 expression was decreased in 61% of breast, 60% of lung, 87% of clear cell and 89% papillary renal cancer specimens (P<0.03 for all the cases). There was a higher frequency of CHL1 mRNA decrease in lung squamous cell carcinoma compared to adenocarcinoma (81% vs. 38%, P = 0.02) without association with tumor progression. Conclusions/Significance Our results suggested that CHL1 is involved in the development of different human cancers. Initially, during the primary tumor growth CHL1 could act as a putative tumor suppressor and is silenced to facilitate in situ tumor growth for 11 cancer types. We also suggested that re-expression of the gene on the edge of tumor mass might promote local invasive growth and enable further metastatic spread in ovary, colon and breast cancer. Our data also supported the role of CHL1 as a potentially novel specific biomarker in the early pathogenesis of two major histological types of renal cancer.
Collapse
Affiliation(s)
- Vera N. Senchenko
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S. Krasnov
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A. Dmitriev
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna V. Kudryavtseva
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina A. Anedchenko
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Eleonora A. Braga
- Laboratory of Molecular Diagnosis, Russian State Genetics Center GosNIIgenetika, Moscow, Russia
| | - Irina V. Pronina
- Laboratory of Molecular Diagnosis, Russian State Genetics Center GosNIIgenetika, Moscow, Russia
| | | | - Sergey V. Ivanov
- Cardiothoracic Surgery Department, NYU Langone Medical Center, New York, New York, United States of America
| | - Eugene R. Zabarovsky
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Microbiology, Tumor and Cell Biology, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institute, Stockholm, Sweden
- * E-mail:
| | - Michael I. Lerman
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
18
|
Wolterink S, Moldenhauer G, Fogel M, Kiefel H, Pfeifer M, Lüttgau S, Gouveia R, Costa J, Endell J, Moebius U, Altevogt P. Therapeutic antibodies to human L1CAM: functional characterization and application in a mouse model for ovarian carcinoma. Cancer Res 2010; 70:2504-15. [PMID: 20215505 DOI: 10.1158/0008-5472.can-09-3730] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent work has identified L1CAM (CD171) as a novel marker for human carcinoma progression. Functionally, L1CAM promotes tumor cell invasion and motility, augments tumor growth in nude mice, and facilitates experimental tumor metastasis. These functional features qualify L1 as an interesting target molecule for tumor therapy. Here, we generated a series of novel monoclonal antibodies (mAb) to the L1CAM ectodomain that were characterized by biochemical and functional means. All novel mAbs reacted specifically with L1CAM and not with the closely related molecule CHL1, whereas antibodies to the COOH terminal part of L1CAM (mAb2C2, mAb745H7, pcytL1) showed cross-reactivity. Among the novel mAbs, L1-9.3 was selected and its therapeutic potential was analyzed in various isotype variants in a model of SKOV3ip cells growing i.p. in CD1 nude mice. Only therapy with the IgG2a variant efficiently prolonged survival and reduced tumor burden. This was accompanied by an increased infiltration of F4/80-positive monocytic cells. Clodronate pretreatment of tumor-bearing animals led to the depletion of monocytes and abolished the therapeutic effect of L1-9.3/IgG2a. Expression profiling of tumor-derived mRNA revealed that L1-9.3/IgG2a therapy induced altered expression of cellular genes associated with apoptosis and tumor growth. Our results establish that anti-L1 mAb therapy acts via immunologic and nonimmunologic effector mechanism to block tumor growth. The novel antibodies to L1CAM could become helpful tools for the therapy of L1-positive human carcinomas.
Collapse
Affiliation(s)
- Silke Wolterink
- Tumor Immunology Program, Translational Immunology Unit (D015), German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|