1
|
Wang Y, Wang W, Wang H, Qin L, Zhang M, Zhang Y, Wang Y, Hao C, Qu M, Wang G. Clinical prognostic significance of xeroderma pigmentosum group C and IFN‑γ in non‑small cell lung cancer. Oncol Lett 2024; 27:259. [PMID: 38646492 PMCID: PMC11027110 DOI: 10.3892/ol.2024.14392] [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: 10/20/2023] [Accepted: 02/28/2024] [Indexed: 04/23/2024] Open
Abstract
Lung cancer is the most common cancer in the world due to its high incidence and recurrence. Genetic instability is one of the main factors leading to its occurrence, development and poor prognosis. Decreased xeroderma pigmentosum group C (XPC) expression notably enhances the stem cell properties of lung cancer cells and increases their proliferation and migration. Additionally, patients with lung cancer and low XPC expression had a poor prognosis. The purpose of the present study was to analyze the effect of XPC and IFN-γ on the clinical prognosis of patients with non-small cell lung cancer (NSCLC). Lung adenocarcinoma specimens were collected from a total of 140 patients with NSCLC. Additionally, from these 140 patients, 48 paracarcinoma tissue specimens were also collected, which were later used to construct tissue microarrays. The expression of XPC and IFN-γ in cancer tissues and in paraneoplastic tissues was detected using immunohistochemistry. The prognosis and overall survival of patients were determined through telephone follow-up. The results showed a positive correlation between expression of XPC and IFN-γ in NSCLC. Additionally, high expression of both markers was associated with a favorable prognosis in patients with NSCLC. The aforementioned findings suggest that the expression of XPC and IFN-γ has prognostic value in clinical practice and is expected to become a marker for clinical application.
Collapse
Affiliation(s)
- Yongming Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Weiyu Wang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Department of Otolaryngology, and Head and Neck Surgery, Shandong Provincial ENT Hospital, Jinan, Shandong 250022, P.R. China
| | - Huaijie Wang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Liya Qin
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Meijia Zhang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Yong Zhang
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Yubing Wang
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Changcheng Hao
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
| | - Meihua Qu
- Department of Thoracic Surgery, Translational Medical Center, Weifang Second People's Hospital (Weifang Respiratory Disease Hospital), Weifang, Shandong 261041, P.R. China
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Sciences and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Gongchao Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
2
|
Lee JY, Bhandare RR, Boddu SHS, Shaik AB, Saktivel LP, Gupta G, Negi P, Barakat M, Singh SK, Dua K, Chellappan DK. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer. Biomed Pharmacother 2024; 173:116275. [PMID: 38394846 DOI: 10.1016/j.biopha.2024.116275] [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: 11/24/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Tumour suppressor genes play a cardinal role in the development of a large array of human cancers, including lung cancer, which is one of the most frequently diagnosed cancers worldwide. Therefore, extensive studies have been committed to deciphering the underlying mechanisms of alterations of tumour suppressor genes in governing tumourigenesis, as well as resistance to cancer therapies. In spite of the encouraging clinical outcomes demonstrated by lung cancer patients on initial treatment, the subsequent unresponsiveness to first-line treatments manifested by virtually all the patients is inherently a contentious issue. In light of the aforementioned concerns, this review compiles the current knowledge on the molecular mechanisms of some of the tumour suppressor genes implicated in lung cancer that are either frequently mutated and/or are located on the chromosomal arms having high LOH rates (1p, 3p, 9p, 10q, 13q, and 17p). Our study identifies specific genomic loci prone to LOH, revealing a recurrent pattern in lung cancer cases. These loci, including 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), 17p13 (TP53), exhibit a higher susceptibility to LOH due to environmental factors such as exposure to DNA-damaging agents (carcinogens in cigarette smoke) and genetic factors such as chromosomal instability, genetic mutations, DNA replication errors, and genetic predisposition. Furthermore, this review summarizes the current treatment landscape and advancements for lung cancers, including the challenges and endeavours to overcome it. This review envisages inspired researchers to embark on a journey of discovery to add to the list of what was known in hopes of prompting the development of effective therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates.
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada, Chebrolu, Guntur, Andhra Pradesh 522212, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Lakshmana Prabu Saktivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli 620024, India
| | - Gaurav Gupta
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Muna Barakat
- Department of Clinical Pharmacy & Therapeutics, Applied Science Private University, Amman-11937, Jordan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara 144411, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
3
|
Nasrallah NA, Wiese BM, Sears CR. Xeroderma Pigmentosum Complementation Group C (XPC): Emerging Roles in Non-Dermatologic Malignancies. Front Oncol 2022; 12:846965. [PMID: 35530314 PMCID: PMC9069926 DOI: 10.3389/fonc.2022.846965] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Xeroderma pigmentosum complementation group C (XPC) is a DNA damage recognition protein essential for initiation of global-genomic nucleotide excision repair (GG-NER). Humans carrying germline mutations in the XPC gene exhibit strong susceptibility to skin cancer due to defective removal via GG-NER of genotoxic, solar UV-induced dipyrimidine photoproducts. However, XPC is increasingly recognized as important for protection against non-dermatologic cancers, not only through its role in GG-NER, but also by participating in other DNA repair pathways, in the DNA damage response and in transcriptional regulation. Additionally, XPC expression levels and polymorphisms likely impact development and may serve as predictive and therapeutic biomarkers in a number of these non-dermatologic cancers. Here we review the existing literature, focusing on the role of XPC in non-dermatologic cancer development, progression, and treatment response, and highlight possible future applications of XPC as a prognostic and therapeutic biomarker.
Collapse
Affiliation(s)
- Nawar Al Nasrallah
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Benjamin M. Wiese
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Catherine R. Sears
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Pulmonary Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, United States
- *Correspondence: Catherine R. Sears,
| |
Collapse
|
4
|
Wang W, Ma S, Ding Z, Yang Y, Wang H, Yang K, Cai X, Li H, Gao Z, Qu M. XPC Protein Improves Lung Adenocarcinoma Prognosis by Inhibiting Lung Cancer Cell Stemness. Front Pharmacol 2021; 12:707940. [PMID: 34803670 PMCID: PMC8595099 DOI: 10.3389/fphar.2021.707940] [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: 06/29/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022] Open
Abstract
Objective: Xeroderma Pigmentosum Complementation Group C (XPC) is a protein involving in nucleotide excision repair (NER). XPC also plays an important role in the lung cancer occurrence with the mechanism remian unclear up to date. Studies showed that the increased stemness of lung cancer cells is related to the recurrence and metastasis of lung cancer. This study aimed to study and analyze the correlation of XPC with lung cancer stem cell biomarkers expression and the overall survival (OS) of lung adenocarcinoma patients. Methods: 140 cases of clinical lung adenocarcinoma tissue samples and 48 cases of paired paracancerous tissue samples were made into tissue microarray. Immunohistochemistry (IHC) was used to detect the expression of XPC and CD133 in cancer and paracancerous tissues. Semi-quantitative analysis and statistics were performed by Pannoramic Digital Slide Scanner. The expression of XPC and CD133 in fresh tissues was verified by Western blotting assay. siXPC was used to knock down XPC in lung cancer cell lines to study the effect of XPC on the expression of lung cancer stem cell biomarkers and the ability of cell invasion. And shXPC was used to knockdown XPC in A549 and H1650 to study the effect of XPC on the expression of lung cancer stem cell biomarkers. Results: IHC and Western blotting results showed that XPC expression significantly decreased, while CD133 expression significantly increased in cancer tissues comparing to paracancerous tissues (P XPC < 0.0001, P CD133 = 0.0395). The high level of XPC in cancer was associated with a better prognosis (Log-rank p = 0.0577) in lung adenocarcinoma patients. Downregulation of XPC in lung cancer cells showed increased expression of cancer stem cell biomarkers and the increased cell invasion abilities. Conclusion: It is suggested that XPC can exert the ability of anti-tumor formation, tumor invasion and metastasis inhibition, and prognostic survival improvement in lung adenocarcinoma patients by regulating the stemness of lung cancer cells.
Collapse
Affiliation(s)
- Weiyu Wang
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China.,Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Shengyao Ma
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China.,College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhenyu Ding
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yang Yang
- School of Public Health, Qingdao University, Qingdao, China
| | - Huaijie Wang
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Kunning Yang
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaoshan Cai
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hanyue Li
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China.,Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Zhiqin Gao
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Meihua Qu
- Translational Medical Center, Weifang Second People's Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang, China.,Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of Life Science and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
5
|
Hsa_circ_0069244 acts as the sponge of miR-346 to inhibit non-small cell lung cancer progression by regulating XPC expression. Hum Cell 2021; 34:1490-1503. [PMID: 34228324 DOI: 10.1007/s13577-021-00573-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/22/2021] [Indexed: 10/20/2022]
Abstract
Circular RNAs (circRNAs) play a significant role in the progression of diverse malignancies. Here, we aimed to probe the function and mechanism of circ_0069244 in non-small cell lung cancer (NSCLC). In the present study, circ_0069244 was selected from the circRNA microarray datasets (GSE112214). Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to examine circ_0069244, miR-346 and XPC complex subunit, DNA damage recognition and repair factor (XPC) expression levels. Kaplan-Meier curve was employed to analyze the association between circ_0069244 expression and overall survival of NSCLC patients. Cell counting kit-8 (CCK-8) and 5-Bromo-2'-deoxyuridine (BrdU) experiments were utilized to examine the proliferation of NSCLC cells. Scratch healing and Transwell experiments were executed to examine the migration of NSCLC cells. Western blot was conducted to detect XPC expression at protein level in NSCLC cells. Bioinformatics analysis, dual-luciferase reporter gene and RNA immunoprecipitation (RIP) experiments predicted and validated the targeting relationships of circ_0069244 and miR-346, as well as miR-346 and 3'untranslated region (UTR) of XPC mRNA, respectively. We reported that circ_0069244 was remarkably down modulated in NSCLC and was linked to shorter survival and poor tumor histological grade in NSCLC patients. Functionally, circ_0069244 repressed NSCLC cell proliferation and migration. Furthermore, miR-346-5p was unveiled to be a downstream target of circ_0069244, and miR-346-5p specifically modulated XPC expression. Rescue experiments indicated that the inhibitory effect of circ_0069244 was abolished by co-expression of miR-346-5p mimics. Taken together, circ_0069244 restrained NSCLC progression by modulating the miR-346-5p/XPC axis.
Collapse
|
6
|
Chen JC, Ko JC, Taso YC, Cheng HH, Chen TY, Yen TC, Lin YW. Downregulation of Xeroderma Pigmentosum Complementation Group C Expression by 17-Allylamino-17-Demethoxygeldanamycin Enhances Bevacizumab-Induced Cytotoxicity in Human Lung Cancer Cells. Pharmacology 2020; 106:154-168. [PMID: 33202406 DOI: 10.1159/000509052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/29/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Xeroderma pigmentosum complementation group C (XPC) protein is an important DNA damage recognition factor involved in nucleotide excision repair and regulation of non-small-cell lung cancer (NSCLC) cell proliferation and viability. 17-Allylamino-17-demethoxygeldanamycin (17-AAG) blocks ATP binding to heat shock protein 90 (Hsp90), resulting in destabilization of Hsp90-client protein complexes. Vascular endothelial growth factor (VEGF) is a potent angiogenic growth factor expressed by many types of tumors. Bevacizumab (Avastin) is a humanized monoclonal antibody against human VEGF used as an antiangiogenesis agent in the therapy of many cancers, proving successful in increasing objective tumor response rate and prolonging overall survival in NSCLC patients. METHODS After the bevacizumab and/or 17-AAG treatment, the expressions of XPC mRNA were determined by quantitative real-time PCR analysis. Protein levels of XPC and phospho-AKT were determined by Western blot analysis. We used specific XPC small interfering RNA and PI3K inhibitor (LY294002) to examine the role of the AKT-XPC signal in regulating the chemosensitivity of bevacizumab and 17-AAG. Cell viability was assessed by the MTS assay and trypan blue exclusion assay. RESULTS In this study, bevacizumab decreased XPC expression in human lung squamous cell carcinoma H520 and H1703 cells via AKT inactivation. Enhancement of AKT activity by transfection with constitutively active AKT vectors increased XPC expression and cell survival after treatment with bevacizumab. In addition, 17-AAG synergistically enhanced bevacizumab-induced cytotoxicity and cell growth inhibition in H520 and H1703 cells, associated with downregulation of XPC expression and inactivation of AKT. DISCUSSION/CONCLUSION Together, these results may provide a rationale to combine bevacizumab with Hsp90 inhibitors in future to enhance therapeutic effects for lung cancer.
Collapse
Affiliation(s)
- Jyh-Cheng Chen
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Jen-Chung Ko
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Yong-Cing Taso
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Hsiang-Hung Cheng
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Tzu-Ying Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Ting-Chuan Yen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Yun-Wei Lin
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan,
| |
Collapse
|
7
|
Zebian A, Shaito A, Mazurier F, Rezvani HR, Zibara K. XPC beyond nucleotide excision repair and skin cancers. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2019; 782:108286. [DOI: 10.1016/j.mrrev.2019.108286] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/23/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022]
|
8
|
Nourmohammadi B, Tafsiri E, Rahimi A, Nourmohammadi Z, Daneshvar Kakhaki A, Cho W, Karimipoor M. Expression of miR-9 and miR-200c, ZEB1, ZEB2 and E-cadherin in Non-Small Cell Lung Cancers in Iran. Asian Pac J Cancer Prev 2019; 20:1633-1639. [PMID: 31244281 PMCID: PMC7021597 DOI: 10.31557/apjcp.2019.20.6.1633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) exert a critical influence on physiological and pathological processes through post-transcriptional modification of their mRNA targets. They play important roles in tumorigenesis and are considered to be potential diagnostic and prognostic biomarkers with various cancers. MiR-200c and miR-9 are regulatory elements that can have dual impacts as oncogenes and/or tumor suppressor genes. MiR-200c regulates two transcription factors, ZEB1 and ZEB2, while miR-9 is a regulatory factor for the E-cadherin protein which has a critical function in cell-cell junctions and is inhibited by two transcription factors ZEB1 and ZEB2. In this study, expression levels of miR-200c and miR-9, ZEB-1, ZEB-2 and E-cadherin were assessed in 30 non-small cell lung cancers (NSCLCs) by real-time qPCR. MiR-9 was down-regulated significantly in tumor tissues compared to normal adjacent tissues, while there was no significant change in expression level of miR-200c. On the other hand, ZEB1 demonstrated significant increase and ZEB2a decrease at the mRNA level. These results indicate roles for miR-9 and ZEB1 in genesis of lung cancer, although clinico-pathological associations were not evident. Further studies are necessary to assess implications for treatment of lung cancer.
Collapse
Affiliation(s)
- Bahareh Nourmohammadi
- Department of Molecular Medicine, Biotechnology Research center, Pasteur Institute of Iran, Tehran, Iran. ,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elham Tafsiri
- Department of Molecular Medicine, Biotechnology Research center, Pasteur Institute of Iran, Tehran, Iran.
| | - Amirabbas Rahimi
- Department of Molecular Medicine, Biotechnology Research center, Pasteur Institute of Iran, Tehran, Iran.
| | - Zahra Nourmohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Abolghasem Daneshvar Kakhaki
- Tracheal Diseases Research Center (TDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - William Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Morteza Karimipoor
- Department of Molecular Medicine, Biotechnology Research center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
9
|
Zhou H, Saliba J, Sandusky GE, Sears CR. XPC protects against smoking- and carcinogen-induced lung adenocarcinoma. Carcinogenesis 2019; 40:403-411. [PMID: 30624620 PMCID: PMC6514449 DOI: 10.1093/carcin/bgz003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 01/08/2023] Open
Abstract
Cigarette smoke (CS) contains hundreds of carcinogens and is a potent inducer of oxidative and bulky DNA damage, which when insufficiently repaired leads to activation of DNA damage response and possibly mutations. The DNA repair protein xeroderma pigmentosum group C (XPC) is primed to play an important role in CS-induced DNA damage because of its function in initiating repair of both bulky oxidative DNA damage. We hypothesized that loss of XPC function will increase susceptibility to developing CS- and carcinogen-induced lung cancer through impaired repair of oxidative DNA damage. Mice deficient in XPC (XPC-/-) exposed to chronic CS developed lung tumors whereas their wild-type littermates (XPC+/+) did not. XPC-/- mice treated with the CS-carcinogen urethane developed lung adenocarcinomas representing progressive stages of tumor development, with lung tumor number increased 17-fold compared with XPC+/+ mice. Mice heterozygous for XPC (XPC+/-) demonstrated a gene-dose effect, developing an intermediate number of lung tumors with urethane treatment. Treatment of XPC-/- mice with the carcinogen 3-methylcholanthrene followed by the proliferative agent butylated hydroxytoluene resulted in a 2-fold increase in lung adenocarcinoma development. Finally, tumor number decreased 7-fold in the lungs of XPC-/- mice by concurrent treatment with the antioxidant, N-acetylcysteine. Altogether, this supports a mechanism by which decreased XPC expression promotes lung adenocarcinoma development in response to CS-carcinogen exposure, due in part to impaired oxidative DNA damage repair.
Collapse
Affiliation(s)
- Huaxin Zhou
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine
| | - Jacob Saliba
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catherine R Sears
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine
- The Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
10
|
Liu Y, Bian T, Feng J, Qian L, Li X, Zhang Q, Zhang J, Jiang D, Liu J, Shi J. CtBP1 interacts with SOX2 to promote the growth, migration and invasion of lung adenocarcinoma. Oncol Rep 2019; 42:67-78. [PMID: 31059077 PMCID: PMC6549098 DOI: 10.3892/or.2019.7142] [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: 04/16/2018] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
Carboxyl-terminal binding protein 1 (CtBP1) is overexpressed in many types of solid tumors and has been found to be functionally associated with proliferation, migration, invasion and drug resistance of cancer cells. However, its expression pattern and functions in lung adenocarcinoma remain unclear. In the present study, we observed that the expression of CtBP1 was upregulated in the lung adenocarcinoma tissues of patients with lymph node metastasis and that its overexpression was correlated with tumor differentiation, size and poor overall survival. Silencing of CtBP1 by transfection with shRNA inhibited the proliferation, migration and invasion of A459 lung adenocarcinoma cells in vitro as determined by MTT assay and Transwell assay, respectively. In vivo studies using a lung patient-derived tumor xenograft (PDTX) mouse model implicated CtBP1 expression in lung adenocarcinoma growth, and further in vitro co-immunoprecipitation and depletion experiments indicated that CtBP1 regulated the biological behavior of lung adenocarcinoma cells by interacting with SOX2. Patients with elevated expression of both CtBP1 and SOX2 expression had a significantly shorter overall survival rate than patients with reduced expression of these transcripts, or than patients with elevated expression of only one transcript (P<0.01 in both cases). Taken together, these findings suggest that CtBP1 plays an important role in lung adenocarcinoma and, along with SOX2, may serve as a viable prognostic marker and therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Li Qian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaoli Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Daishan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jian Liu
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jiahai Shi
- Department of Cardio‑Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
11
|
Hruban RH, Gaida MM, Thompson E, Hong SM, Noë M, Brosens LA, Jongepier M, Offerhaus GJA, Wood LD. Why is pancreatic cancer so deadly? The pathologist's view. J Pathol 2019; 248:131-141. [PMID: 30838636 DOI: 10.1002/path.5260] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
The remarkable aggressiveness of pancreatic cancer has never been fully explained. Although clearly multifactorial, we postulate that venous invasion, a finding seen in most pancreatic cancers but not in most cancers of other organs, may be a significant, underappreciated contributor to the aggressiveness of this disease. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthias M Gaida
- Department of General Pathology, The University Hospital of Heidelberg, Heidelberg, Germany
| | - Elizabeth Thompson
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seung-Mo Hong
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Michaël Noë
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lodewijk Aa Brosens
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Jongepier
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Johan A Offerhaus
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laura D Wood
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Wang H, Huang Y, Shi J, Zhi Y, Yuan F, Yu J, Chen Z, Yang J. XPC deficiency leads to centrosome amplification by inhibiting BRCA1 expression upon cisplatin-mediated DNA damage in human bladder cancer. Cancer Lett 2018; 444:136-146. [PMID: 30579971 DOI: 10.1016/j.canlet.2018.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/03/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Xeroderma pigmentosum group C (XPC) is a well-known DNA damage recognition protein. Defects in XPC lead to carcinogenesis and progression of many human cancers. In the current study, we defined a novel, important role of XPC in preventing centrosome amplification during cisplatin-mediated DNA damage response. From experiments with human bladder cancer tissue, urothelial tissue from Xpc knockout mice and XPC-silenced cell lines, we found that attenuated XPC expression was associated with increased centrosome amplification in human bladder cancer. A significant increase in centrosome amplification was observed in XPC-silenced cells upon cisplatin treatment. XPC deficiency leads to reduced BRCA1 expression via upregulating its transcriptional repressor, Pit-1. The BRCA1 downregulation results in more DNA double strand breaks accumulation and persistent activation of the ATM-Chk1/Chk2 signaling, resulting in a prolonged G2/M arrest during which centrosome can over-duplicate and lead to centrosome amplification. XPC complementation in silenced cells could reduce Pit-1 expression, increase BRCA1 expression and recover the status of centrosome amplification. Our study reveals a new function for XPC in preventing chromosomal instability, providing new information on cancer chemotherapy and potential clinical significance for cancer management.
Collapse
Affiliation(s)
- Huanhuan Wang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yaqin Huang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Jiazhong Shi
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yi Zhi
- Department of Urology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Fang Yuan
- Chongqing University Cancer Hospital, Chongqing, PR China
| | - Jin Yu
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Zhiwen Chen
- Urology Institute of People's Liberation Army, Southwest Hospital, The Third Military Medical University, Chongqing, PR China; Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| | - Jin Yang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China.
| |
Collapse
|
13
|
miR-4516 predicts poor prognosis and functions as a novel oncogene via targeting PTPN14 in human glioblastoma. Oncogene 2018; 38:2923-2936. [PMID: 30559405 DOI: 10.1038/s41388-018-0601-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/10/2018] [Accepted: 10/08/2018] [Indexed: 12/30/2022]
Abstract
Glioblastomas (GBMs) are the most aggressive primary brain tumors, with an average survival of less than 15 months. Therefore, there is a critical need to develop novel therapeutic strategies for GBM. This study aimed to assess the prognostic value of miR-4516 and investigate its oncogenic functions and the underlying cellular and molecular mechanisms in GBM. To determine the correlation between miR-4516 expression and overall survival of patients with GBM, total RNAs were isolated from 268 FFPE tumor samples, miR expression was assayed (simultaneously) using the nCounter human miRNA v3a assay followed by univariable and multivariable survival analyses. Further, in vitro and in vivo studies were conducted to define the role of miR-4516 in GBM tumorigenesis and the underlying molecular mechanisms. Upon multivariable analysis, miR-4516 was correlated with poor prognosis in GBM patients (HR = 1.49, 95%CI: 1.12-1.99, P = 0.01). Interestingly, the significance of miR-4516 was retained including MGMT methylation status. Overexpression of miR-4516 significantly enhanced cell proliferation and invasion of GBM cells both in vitro and in vivo. While conducting downstream targeting studies, we found that the tumor-promoting function of miR-4516, in part, was mediated by direct targeting of PTPN14 (protein tyrosine phosphatase, non-receptor type 14) which, in turn, regulated the Hippo pathway in GBM. Taken together, our data suggest that miR-4516 represents an independent negative prognostic factor in GBM patients and acts as a novel oncogene in GBM, which regulates the PTPN14/Hippo pathway. Thus, this newly identified miR-4516 may serve as a new potential therapeutic target for GBM treatment.
Collapse
|
14
|
Chen JC, Wu CH, Peng YS, Zheng HY, Lin YC, Ma PF, Yen TC, Chen TY, Lin YW. Astaxanthin enhances erlotinib-induced cytotoxicity by p38 MAPK mediated xeroderma pigmentosum complementation group C (XPC) down-regulation in human lung cancer cells. Toxicol Res (Camb) 2018; 7:1247-1256. [PMID: 30555679 DOI: 10.1039/c7tx00292k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 09/07/2018] [Indexed: 12/11/2022] Open
Abstract
Astaxanthin has been demonstrated to exhibit a wide range of beneficial effects that include anti-cancer and anti-inflammatory properties. Xeroderma pigmentosum complementation group C (XPC) protein is an important DNA damage recognition factor in nucleotide excision repair and is involved in regulating non-small cell lung cancer (NSCLC) cell proliferation and viability. Erlotinib (TarcevaR) is a selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that has demonstrated clinical activity in NSCLC cells. However, whether astaxanthin and erlotinib could induce synergistic cytotoxicity in NSCLC cells through modulating XPC expression is unknown. In this study, we found that p38 MAPK activation by astaxanthin decreased XPC expression in two human lung adenocarcinoma A549 and H1975 cells. Inactivation of p38 MAPK by pharmacological inhibitor SB203580 or the specific small interfering RNA (siRNA) rescued the astaxanthin-reduced XPC mRNA and protein levels. Enforced expression of XPC cDNA or inhibiting the p38 MAPK activity reduced the cytotoxicity and cell growth inhibition of astaxanthin. In contrast, knockdown of XPC using siRNA enhanced the cytotoxic effects of astaxanthin. Moreover, astaxanthin synergistically enhanced cytotoxicity and cell growth inhibition of erlotinib in NSCLC cells, which were associated with the down-regulation of XPC expression and activation of p38 MAPK. Our findings suggested that the astaxanthin induced p38 MAPK mediated XPC down-regulation enhanced the erlotinib-induced cytotoxicity in A549 and H1975 cells.
Collapse
Affiliation(s)
- Jyh-Cheng Chen
- Department of Food Science , National Chiayi University , Chiayi , Taiwan
| | - Chia-Hung Wu
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yi-Shuan Peng
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Hao-Yu Zheng
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yuan-Cheng Lin
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Peng-Fang Ma
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Ting-Chuan Yen
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Tzu-Ying Chen
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yun-Wei Lin
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| |
Collapse
|
15
|
Pei G, Lan Y, Lu W, Ji L, Hua ZC. The function of FAK/CCDC80/E-cadherin pathway in the regulation of B16F10 cell migration. Oncol Lett 2018; 16:4761-4767. [PMID: 30214608 DOI: 10.3892/ol.2018.9159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/16/2018] [Indexed: 12/23/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase involved in the development and progression of cancer. However, the regulatory role of FAK in cell migration remains unclear. The aim of the present study was to investigate the mechanism underlying the regulation of melanoma cell migration by FAK. The effect of FAK knockdown on gene expression in B16F10 cells was examined by gene chip analysis. The expression levels of coiled-coil domain containing 80 (CCDC80) and epithelial (E)-cadherin were analyzed by reverse transcription quantitative polymerase chain reaction and western blotting. Wound healing and transwell assays were used to monitor B16F10 cell migration. It was identified that the knockdown of FAK increased the expression levels of CCDC80 and E-cadherin, while the overexpression of CCDC80 elevated E-cadherin expression. Concurrently, upregulation of CCDC80 inhibited the migration of B16F10 cells, and downregulation of CCDC80 promoted the migration of B16F10 cells. The clinical data from the Oncomine database also revealed that the mRNA level of FAK was increased while the mRNA levels of CCDC80 and E-cadherin were decreased in patients with melanoma compared with normal controls. Taken together, the results of the present study suggest that the regulation of B16F10 melanoma cell migration by FAK is potentially mediated by CCDC80.
Collapse
Affiliation(s)
- Guoshun Pei
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Yan Lan
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Weijie Lu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Lina Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories, Inc., Changzhou, Jiangsu 213164, P.R. China
| |
Collapse
|
16
|
Ji W, Yu Y, Li Z, Wang G, Li F, Xia W, Lu S. FGFR1 promotes the stem cell-like phenotype of FGFR1-amplified non-small cell lung cancer cells through the Hedgehog pathway. Oncotarget 2017; 7:15118-34. [PMID: 26936993 PMCID: PMC4924774 DOI: 10.18632/oncotarget.7701] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/29/2016] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cell-like phenotype is critical for tumor formation and treatment resistance. FGFR1 is found to be amplified in non-small cell lung cancer, particularly in the lung squamous cell cancer (LSCC). Whether FGFR1 contributes to the maintenance of stem cell-like phenotype of FGFR1-amplified lung cancer cells remains elusive. In this study, treatment with FGFR1 inhibitor AZD4547 suppressed the growth of tumor spheres and reduced ALDH positive proportion in FGFR1-amplified lung cancer cells in vitro, as well as inhibited the growth of oncospheres and parental cells in xenograft models. Knockdown of FGFR1 recaptured the similar effect as AZD4547 in vitro. Furthermore, activation of FGFR1 and subsequently its downstream ERK signaling enhanced the expression and transcriptional activity of GLI2, which could be blocked by FGFR1 inhibitor/silencing or ERK inhibitor. Knockdown of GLI2 directly inhibited the stem-like phenotype of FGFR1-amilified cells, whereas overexpression of GLI2 sufficiently rescued the phenotype caused by FGFR1 knockdown. Notably we also identified a correlation between FGFR1 and GLI2 expressions from clinical data, as well as an inverse relationship with progression free survival (PFS). Together our study suggests that the FGFR1/GLI2 axis promotes the lung cancer stem cell-like phenotype. These results support a rational strategy of combination of FGFR1 and GLI inhibitors for treatment of FGFR1-amplified lung cancers, especially LSCC.
Collapse
Affiliation(s)
- Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China.,State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guan Wang
- Genomics Center, WuXiAppTec Co., Ltd., Shanghai 200131, China
| | - Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
17
|
Wang Y, Wang H, Pan T, Li L, Li J, Yang H. STIM1 silencing inhibits the migration and invasion of A549 cells. Mol Med Rep 2017; 16:3283-3289. [PMID: 28713917 PMCID: PMC5547937 DOI: 10.3892/mmr.2017.7010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 05/12/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to explore the effects of stromal interaction molecule 1 (STIM1) knockdown on the migration, invasion and metastasis of A549 cells in vitro and in vivo. Western blotting and immunohistochemistry were used to detect protein expression levels. Wound healing and Transwell invasion assays were used to assess the migratory and invasive abilities of A549 cells transfected with STIM1-specific short hairpin (sh)RNA (shSTIM1). In addition, a tail vein metastatic assay was performed. The results demonstrated that the frequency of STIM1 high-expression was significantly increased in metastatic lung cancer tissues (72.2%) compared with in non-metastatic lung cancer tissues (33.0%). STIM1 knockdown inhibited A549 cell migration and invasion in vitro and tumor metastasis in vivo. The protein expression levels of Snail1, Vimentin, matrix metalloproteinase (MMP) 2 and MMP9 were markedly decreased in A549-shSTIM1 compared with in A549 cells transfected with control shRNA (shcon). In addition, the protein expression levels of E-cadherin were markedly increased in A549-shSTIM1 cells compared with in A549-shcon cells. These results suggested that STIM1 knockdown may inhibit the migration and invasion of A549 cells in vitro, and metastasis in vivo.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Teng Pan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
18
|
Jiang Y, Wu Y, Hardie WJ, Zhou X. Mast cell chymase affects the proliferation and metastasis of lung carcinoma cells in vitro. Oncol Lett 2017; 14:3193-3198. [PMID: 28927065 DOI: 10.3892/ol.2017.6487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 03/14/2017] [Indexed: 12/15/2022] Open
Abstract
Metastasis of lung carcinoma cells is a major cause of organ failure and mortality of patients with lung cancer. Lung mast cells are a type of immune cell which reside in the respiratory mucosa. High numbers of mast cells are associated with the majority of common types of cancer; however, the effects of mast cells on cancer remain unclear. In the present study, the effects of mast cell chymase (MCC) on the proliferation and adhesion of the lung carcinoma cell lines A549 and H520 was investigated. After 24 h of treatment, the highest dose of MCC (50 mU/ml) decreased the proliferation rate of A549 and H520 cells, whereas the lowest dose of MCC (5 mU/ml) resulted in a small increase in the viability. A549 cells treated with MCC lost adhesion ability in a MCC dose-dependent manner; however, these detached cells were able to regrow when transferred to a fresh culture. The protein expression of epithelial (E-) cadherin, p53 and p21 in A549 lung carcinoma cells were detected by western blot analysis. The results of the present study revealed that, following 24 h of treatment, the expression level of E-cadherin was decreased, the p53 tumor suppressor protein was expressed in limited quantities and the expression of p21 was decreased. Zymography was used to examine the effects of MCC on the expression and activation of matrix metalloproteinase-9 (MMP-9) in A549 and H520 cells. The expression of MMP-9 in the two cell lines was time- and MCC dose-dependent. The results of the present study demonstrated that MCC stimulated lung carcinoma cell proliferation and adhesion, as well as regulated E-cadherin expression and the cell cycle, all of which are associated with cancer metastasis. Therefore, MCC may be a potential candidate for lung carcinoma therapy.
Collapse
Affiliation(s)
- Yuan Jiang
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Yudan Wu
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - William James Hardie
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Xiaoying Zhou
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| |
Collapse
|
19
|
Bu L, Li W, Ming Z, Shi J, Fang P, Yang S. Inhibition of TrxR2 suppressed NSCLC cell proliferation, metabolism and induced cell apoptosis through decreasing antioxidant activity. Life Sci 2017; 178:35-41. [PMID: 28414076 DOI: 10.1016/j.lfs.2017.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/30/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
AIMS This study aims to analyze the effect of thioredoxin reductase 2 (TrxR2) on lung cancer cell proliferation, apoptosis, invasion and migration in vitro. MAIN METHODS Real-time PCR was used to measure the expression of TrxR2 in NSCLC tumor tissues. After pAd-TrxR2 or shRNA-TrxR2 was transfected into A549 or NCI-H1299 cells, the cell proliferation was measured by CCK-8 method; cell apoptosis was measured by flow cytometry; cell invasion and migration was measured by Transwell method. The production of ROS was measured by DCFH-DA method; the activity of SOD, CAT and GSH-Px was measured by relative ELISA kit. KEY FINDINGS The results showed that TrxR2 was up-regulated in NSCLC tumor tissues. Inhibition of TrxR2 suppressed NSCLC cell proliferation and induced apoptosis, and inhibited cell invasion and migration. However, overexpression of TrxR2 showed the opposite effect. Furthermore, when cells were transfected with shRNA-TrxR2, the production of ROS was significantly increased, and SOD, CAT and GSH-Px activity was decreased. Conversely, pAd-TrxR2 transfection showed the opposite effect. SIGNIFICANCE Taken together, our results suggest that TrxR2 acts as an oncogenic gene in the context of lung cancer progression. The inhibition of TrxR2 suppressed lung cancer cell proliferation, invasion and migration and induced cell apoptosis by inducing ROS production and decreasing antioxidant activity. TrxR2 may be a potential target for NSCLC treatment.
Collapse
Affiliation(s)
- Lina Bu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China; Department of Respiratory Medicine, Xi'an No.3 Hospital, Xi'an, 710018, Shaanxi, China
| | - Wei Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China
| | - Zongjuan Ming
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China
| | - Jie Shi
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China
| | - Ping Fang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China
| | - Shuanying Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710003, Shaanxi, China.
| |
Collapse
|
20
|
Chen Y, Fang L, Zhang J, Li G, Ma M, Li C, Lyu J, Meng QH. Blockage of Glyoxalase I Inhibits Colorectal Tumorigenesis and Tumor Growth via Upregulation of STAT1, p53, and Bax and Downregulation of c-Myc and Bcl-2. Int J Mol Sci 2017; 18:ijms18030570. [PMID: 28282916 PMCID: PMC5372586 DOI: 10.3390/ijms18030570] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/26/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
GlyoxalaseI (GLOI) is an enzyme that catalyzes methylglyoxal metabolism. Overexpression of GLOI has been documented in numerous tumor tissues, including colorectal cancer (CRC). The antitumor effects of GLOI depletion have been demonstrated in some types of cancer, but its role in CRC and the mechanisms underlying this activity remain largely unknown. Our purpose was to investigate the antitumor effects of depleted GLOI on CRC in vitro and in vivo. RNA interference was used to deplete GLOI activity in four CRC cell lines. The cells' proliferation, apoptosis, migration, and invasion were assessed by using the Cell Counting Kit-8, plate colony formation assay, flow cytometry, and transwell assays. Protein and mRNA levels were analyzed by western blot and quantitative real-time PCR (qRT-PCR), respectively. The antitumor effect of GLOI depletion in vivo was investigated in a SW620 xenograft tumor model in BALB/c nude mice. Our results show that GLOI is over-expressed in the CRC cell lines. GLOI depletion inhibited the proliferation, colony formation, migration, and invasion and induced apoptosis of all CRC cells compared with the controls. The levels of signal transducer and activator of transcription 1 (STAT1), p53, and Bcl-2 assaciated X protein (Bax) were upregulated by GLOI depletion, while cellular homologue of avian myelocytomatosis virus oncogene (c-Myc) and B cell lymphoma/lewkmia-2 (Bcl-2) were downregulated. Moreover, the growth of SW620-induced CRC tumors in BALB/c nude mice was significantly attenuated by GLOI depletion. The expression levels of STAT1, p53, and Bax were increased and those of c-Myc and Bcl-2 were decreased in the GLOI-depleted tumors. Our findings demonstrate that GLOI depletion has an antitumor effect through the STAT1 or p53 signaling pathways in CRC, suggesting that GLOI is a potential therapeutic target.
Collapse
Affiliation(s)
- Yuan Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Lei Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jiali Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Gefei Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Mengni Ma
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Changxi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jianxin Lyu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Qing H Meng
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Wang YP, Guo PT, Zhu Z, Zhang H, Xu Y, Chen YZ, Liu F, Ma SP. Pleomorphic adenoma gene like-2 induces epithelial-mesenchymal transition via Wnt/β-catenin signaling pathway in human colorectal adenocarcinoma. Oncol Rep 2017; 37:1961-1970. [PMID: 28259923 PMCID: PMC5367359 DOI: 10.3892/or.2017.5485] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 02/15/2017] [Indexed: 12/20/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a critical step in the acquisition of metastatic and invasive power for tumor cells. Colorectal adenocarcinoma (CRC) is a common cancer where metastasis is directly linked to patient survival. Recent studies show that pleomorphic adenoma gene like-2 (PLAGL2) could induce tumor EMT and is an independent predictive factor associated with poor prognosis in cancer. In the present study, we confirmed the role of PLAGL2 in the prognosis of CRC patients and provide molecular evidence of PLAGL2 promoted EMT in CRC cell line SW480. We found that PLAGL2 expression was upregulated in the paraffin-embedded CRC tissues compared to borderline or benign tissues. Experimental EMT induced by PLAGL2 plasmid transfection proved PLAGL2 protein overexpression could enhance the cell scratch wound-healing and Transwell ability and significantly upregulated mesenchymal marker proteins, N-cadherin and vimentin and concurrently downregulated epithelial marker of E-cadherin. Subsequently, through western blot assay, we found that PLAGL2 could activate the Wnt-signaling component β-catenin in the nuclei. More CRC cell metastasis to the lungs was observed when the PLAGL2 overexpressing SW480 cells were injected into the tail vein of rats, compared with the cell control and PLAGL2 silence group. Our findings indicated that PLAGL2 might be a very upstream key molecule regulating EMT involved in Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yong-Peng Wang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Peng-Tao Guo
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Zhi Zhu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yan Xu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yu-Ze Chen
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Fang Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Si-Ping Ma
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
22
|
Sun CC, Li SJ, Yuan ZP, Li DJ. MicroRNA-346 facilitates cell growth and metastasis, and suppresses cell apoptosis in human non-small cell lung cancer by regulation of XPC/ERK/Snail/E-cadherin pathway. Aging (Albany NY) 2016; 8:2509-2524. [PMID: 27777383 PMCID: PMC5115903 DOI: 10.18632/aging.101080] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 10/04/2016] [Indexed: 01/02/2023]
Abstract
Determinants of growth and metastasis in cancer remain of great interest to define. MicroRNAs (miRNAs) have frequently emerged as tumor metastatic regulator by acting on multiple signaling pathways. Here we report the definition of miR-346 as a novel oncogenic microRNA that facilitates non-small cell lung cancer (NSCLC) cell growth and metastasis. XPC, an important DNA damage recognition factor in nucleotide excision repair was defined as a target for down-regulation by miR-346, functioning through direct interaction with the 3'-UTR of XPC mRNA. Blocking miR-346 by an antagomiR was sufficient to inhibit NSCLC cell growth and metastasis, an effect that could be phenol-copied by RNAi-mediated silencing of XPC. In vivo studies established that miR-346 overexpression was sufficient to promote tumor growth by A549 cells in xenografts mice, relative to control cells. Overall, our results defined miR-346 as an oncogenic miRNA in NSCLC, the levels of which contributed to tumor growth and invasive aggressiveness.
Collapse
Affiliation(s)
- Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| | - Shu-Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
- Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, 430071 Wuhan, P. R. China
| | - Zhan-Peng Yuan
- Department of Toxicology, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| | - De-Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| |
Collapse
|
23
|
Qu MH, Han C, Srivastava AK, Cui T, Zou N, Gao ZQ, Wang QE. miR-93 promotes TGF-β-induced epithelial-to-mesenchymal transition through downregulation of NEDD4L in lung cancer cells. Tumour Biol 2016; 37:5645-51. [PMID: 26581907 PMCID: PMC6528179 DOI: 10.1007/s13277-015-4328-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022] Open
Abstract
The level of microRNA-93 (miR-93) in tumors has been recently reported to be negatively correlated with survival of lung cancer patients. Considering that the most devastating aspect of lung cancer is metastasis, which can be promoted by transforming growth factor-β (TGF-β)-induced epithelial-to-mesenchymal transition (EMT), we sought to determine whether miR-93 is involved in this process. Here, we report that a previously unidentified target of miR-93, neural precursor cell expressed developmentally downregulated gene 4-like (NEDD4L), is able to mediate TGF-β-mediated EMT in lung cancer cells. miR-93 binds directly to the 3'-UTR of the NEDD4L messenger RNA (mRNA), leading to a downregulation of NEDD4L expression at the protein level. We next demonstrated that the downregulation of NEDD4L enhanced, while overexpression of NEDD4L reduced TGF-β signaling, reflected by increased phosphorylation of SMAD2 in the lung cancer cell line after TGF-β treatment. Furthermore, overexpression of miR-93 in lung cancer cells promoted TGF-β-induced EMT through downregulation of NEDD4L. The analysis of publicly available gene expression array datasets indicates that low NEDD4L expression correlates with poor outcomes among patients with lung cancer, further supporting the oncogenic role of miR-93 in lung tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Mei-Hua Qu
- Department of Pharmacology, Key Lab of Applied Pharmacology in Universities of Shandong, Weifang Medical University, 7166 Baotong West St, Weifang, 261053, Shandong, China.
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA.
| | - Chunhua Han
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Amit Kumar Srivastava
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Tiantian Cui
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Ning Zou
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Zhi-Qin Gao
- Department of Pharmacology, Key Lab of Applied Pharmacology in Universities of Shandong, Weifang Medical University, 7166 Baotong West St, Weifang, 261053, Shandong, China
| | - Qi-En Wang
- Department of Radiology, The Ohio State University Wexner Medical Center, Room 1014, 460 W. 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
24
|
Mayer C, Darb-Esfahani S, Meyer AS, Hübner K, Rom J, Sohn C, Braicu I, Sehouli J, Hänsch GM, Gaida MM. Neutrophil Granulocytes in Ovarian Cancer - Induction of Epithelial-To-Mesenchymal-Transition and Tumor Cell Migration. J Cancer 2016; 7:546-54. [PMID: 27053953 PMCID: PMC4820731 DOI: 10.7150/jca.14169] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/22/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Ovarian cancer (OvCa) is a highly aggressive malignoma with a tumor-promoting microenvironment. Infiltration of polymorphonuclear neutrophils (PMN) is frequently seen, raising the question of their impact on tumor development. In that context, effects of PMN on human ovarian cancer cells were assessed. METHODS Human epithelial ovarian cancer cells were incubated with human PMN, lysate of PMN, or neutrophil elastase. Morphological alterations were observed by time-lapse video-microscopy, and the underlying molecular mechanism was analyzed by flow cytometry and Western blotting. Functional alternations were assessed by an in vitro wound healing assay. In parallel, a large cohort of n=334 primary OvCa tissue samples of various histological subtypes was histologically evaluated. RESULTS Co-cultivation of cancer cells with either PMN or PMN lysate causes a change of the polygonal epithelial phenotype of the cells towards a spindle shaped morphology, causing a cribriform cell growth. The PMN-induced alteration could be attributed to elastase, a major protease of PMN. Elastase-induced shape change was most likely due to the degradation of membranous E-cadherin, which results in loss of cell contacts and polarity. Moreover, in response to elastase, epithelial cytokeratins were downmodulated, in parallel with a nuclear translocation of β-catenin. These PMN-elastase induced alterations of cells are compatible with an epithelial-to-mesenchymal transition (EMT) of the cancer cells. Following EMT, the cells displayed a more migratory phenotype. In human biopsies, neutrophil infiltration was seen in 72% of the cases. PMN infiltrates were detected preferentially in areas with low E-cadherin expression. CONCLUSION PMN in the microenvironment of OvCa can alter tumor cells towards a mesenchymal and migratory phenotype.
Collapse
Affiliation(s)
- Christine Mayer
- 1. Department of Gynecology and Obstetrics, University Hospital Heidelberg, Germany
| | - Silvia Darb-Esfahani
- 2. Institute of Pathology, Charité, University Hospital Berlin, Germany;; 3. Tumorbank Ovarian Cancer Network (TOC), Berlin, Germany
| | | | - Katrin Hübner
- 5. BioQuant, COS Heidelberg, University Heidelberg, Germany
| | - Joachim Rom
- 1. Department of Gynecology and Obstetrics, University Hospital Heidelberg, Germany
| | - Christof Sohn
- 1. Department of Gynecology and Obstetrics, University Hospital Heidelberg, Germany
| | - Ioana Braicu
- 3. Tumorbank Ovarian Cancer Network (TOC), Berlin, Germany;; 6. Department of Gynecology, Charité University Hospital Berlin, Germany
| | - Jalid Sehouli
- 3. Tumorbank Ovarian Cancer Network (TOC), Berlin, Germany;; 6. Department of Gynecology, Charité University Hospital Berlin, Germany
| | - G Maria Hänsch
- 7. Institute of Immunology, University Hospital Heidelberg, Germany
| | - Matthias M Gaida
- 4. Institute of Pathology, University Hospital Heidelberg, Germany
| |
Collapse
|
25
|
TGF-β signaling links E-cadherin loss to suppression of nucleotide excision repair. Oncogene 2015; 35:3293-302. [PMID: 26477308 PMCID: PMC4837109 DOI: 10.1038/onc.2015.390] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 01/04/2023]
Abstract
E-cadherin is a cell adhesion molecule best known for its function in suppressing tumor progression and metastasis. Here we show that E-cadherin promotes nucleotide excision repair through positively regulating the expression of xeroderma pigmentosum complementation group C (XPC) and DNA damage-binding protein 1 (DDB1). Loss of E-cadherin activates the E2F4 and p130/107 transcription repressor complexes to suppress the transcription of both XPC and DDB1 through activating the TGF-β pathway. Adding XPC or DDB1, or inhibiting the TGF-β pathway, increases the repair of UV-induced DNA damage in E-cadherin-inhibited cells. In mouse skin and skin tumors UVB radiation down-regulates E-cadherin. In sun-associated premalignant and malignant skin neoplasia, E-cadherin is down-regulated in association with reduced XPC and DDB1 levels. These findings demonstrate a crucial role of E-cadherin in efficient DNA repair of UV-induced DNA damage, identify a new link between epithelial adhesion and DNA repair, and suggest a mechanistic link of early E-cadherin loss in tumor initiation.
Collapse
|
26
|
Abstract
XPC has long been considered instrumental in DNA damage recognition during global genome nucleotide excision repair (GG-NER). While this recognition is crucial for organismal health and survival, as XPC's recognition of lesions stimulates global genomic repair, more recent lines of research have uncovered many new non-canonical pathways in which XPC plays a role, such as base excision repair (BER), chromatin remodeling, cell signaling, proteolytic degradation, and cellular viability. Since the first discovery of its yeast homolog, Rad4, the involvement of XPC in cellular regulation has expanded considerably. Indeed, our understanding appears to barely scratch the surface of the incredible potential influence of XPC on maintaining proper cellular function. Here, we first review the canonical role of XPC in lesion recognition and then explore the new world of XPC function.
Collapse
|