1
|
Wang C, Zhang J, Wang H, Chen R, Lu M. Family with sequence similarity 83, member A (FAM83A) inhibits ferroptosis via the Wnt/β-catenin pathway in lung squamous cell cancer. Cell Death Discov 2024; 10:332. [PMID: 39033191 PMCID: PMC11271298 DOI: 10.1038/s41420-024-02101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
The function of Family With Sequence Similarity 83, Member A (FAM83A) in lung squamous cell carcinoma (LUSC) is largely unknown. Here, we detected its prognostic and regulation roles in LUSC. Bioinformatics methods were applied initially to predict the expression level and prognostic value of FAM83A mRNA in LUSC. In vitro experiments, such as western blot, colony formation and cell viability assay, lipid Reactive oxygen species (ROS), malondialdehyde (MDA), reduced glutathione (GSH)/oxidized glutathione disulfide (GSSG), and 4-hydroxy-2-nonenal (4-HNE) assay, were used to investigate its mechanism. In vivo experiments were further conducted to validate the mechanism. Results from TCGA and Oncomine databases revealed significantly higher FAM83A mRNA expression levels in LUSC than in normal lung tissue. TCGA and GEO databases and our database revealed that FAM83A expression level was an independent prognostic factor for both overall survival and progression-free survival. Besides, FAM83A was significantly associated with a higher ability of growth and clonogenicity. Mechanistically, in vitro and in vivo experiments revealed that FAM83A could promote LUSC cell growth by inhibiting ferroptosis via activating the Wnt/β-catenin signaling pathway. The rescue experiment demonstrated that inhibition of the Wnt/β-catenin pathway counteracted the function of FAM83A. FAM83A is overexpressed in LUSC and could serve as a prognosis prediction biomarker for LUSC. FAM83A promotes LUSC cell growth by inhibiting ferroptosis via activating the Wnt/β-catenin signaling pathway, which provides a new potential therapeutic target for LUSC treatment.
Collapse
Affiliation(s)
- Cong Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences; Department of Radiation Oncology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Zhang
- Department of Drug Inspection, Tai'an Institute For Food And Drug Control (Tai'an Fiber Inspection Institute), Tai'an, China
| | - Hongjiao Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ruixue Chen
- Department of Encephalopathy (II), Xintai Hospital of Traditional Chinese Medicine, Tai'an, China
| | - Ming Lu
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
2
|
Liu H, Yao J, Liu Y, Wu L, Tan Z, Hu J, Chen S, Zhang X, Cheng S. Diagnostic value of immune-related biomarker FAM83A in differentiating malignant from benign pleural effusion in lung adenocarcinoma. Discov Oncol 2024; 15:242. [PMID: 38914812 PMCID: PMC11196556 DOI: 10.1007/s12672-024-01109-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Malignant pleural effusion (MPE) is frequently observed in patients with advanced lung adenocarcinoma (LUAD). Pleural fluid cytology is a less invasive procedure compared to pleural biopsy. Therefore, it is crucial to identify novel effective biomarkers for LUAD-associated pleural fluid cytology. METHODS The RNA sequencing (RNA-Seq) and clinical data of LUAD cases were downloaded from TCGA and OncoSG databases. Differential gene expression analysis, survival analysis and immune cell infiltration analysis were performed on the LUAD datasets. The expression levels of FAM83A, TFF-1, and NapsinA in 94 paired LUAD and adjacent normal tissues, and in the pleural effusion specimens of 40 LUAD and 21 non-neoplastic patients were evaluated by immunohistochemistry. RESULTS FAM83A expression levels were significantly different between the LUAD and normal tissue datasets, and correlated with overall or disease-free survival, and histological grade of the tumors. Furthermore, the in-situ expression of FAM83A was higher in 89/94 LUAD tissues compared to the paired normal tissues. FAM83A expression was significantly correlated with immune cell infiltration, and showed a positive association with macrophage infiltration. In addition, FAM83A staining was positive in 37 LUAD pleural effusion samples, and negative in 20 non-neoplastic pleural effusion samples. The expression pattern of FAM83A in the pleural effusion of LUAD patients was relatively consistent with that of TFF-1 and NapsinA, and even stronger in some specimens that were weakly positive or negative for TTF1/NapsinA. CONCLUSIONS FAM83A is a promising immune-related biomarker in LUAD biopsy specimens and pleural fluid, and can distinguish between malignant and benign pleural effusion.
Collapse
Affiliation(s)
- Hangfeng Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Jia Yao
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610051, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610051, China
| | - Yulan Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Liping Wu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Zhiwei Tan
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Jie Hu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Shigao Chen
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Xiaolin Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
| | - Shuanghua Cheng
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
| |
Collapse
|
3
|
Hu X, Zou S, Shi X, Zhang Q, Li Y, Wang M, Li T, Zhang X, Li G. FAM83B promotes cell proliferation via regulating the expression of CDK4/CDK6/CCND1 complex in laryngeal squamous cell carcinoma. Heliyon 2024; 10:e29933. [PMID: 38707417 PMCID: PMC11066311 DOI: 10.1016/j.heliyon.2024.e29933] [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: 12/27/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024] Open
Abstract
FAM83B, as one of the FAM83 family members, has been closely involved in cell transformation, and a growing number of scholars have been studied its role in tumours over the years. Whereas the effect and potential mechanism of FAM83B in laryngeal squamous cell carcinoma (LSCC) have not been investigated. In this research, we discovered that the expression quantity of FAM83B was remarkably higher in LSCC tissues (79.65 ± 35.98) than in matched adjacent tissues (59.34 ± 32.59) by tissue microarrays and immunohistochemistry. Furthermore, expression of FAM83B was knocked down in HEP-2 and TU177 cell lines via lentivirus, and in the course of intracorporal and extracorporeal experiments, FAM83B knockdown showed the inhibition of tumour growth, migration, and invasion ability. Moreover, cell cycle assay showed that FAM83B knockdown leads to an apparent accumulation of cells in the G1 phase, indicating that FAM83B knockdown can inhibit cell proliferation. Meanwhile, western blotting (WB) demonstrated that FAM83B knockdown led to a significant reduction in CDK4/CDK6/CCND1 protein expression, which may have decelerated cell cycle progression. Collectively, this study demonstrates that FAM83B serves as an oncogene in LSCC, promoting cell proliferation by controlling the protein expression of CDK4, CDK6, and CCND1, thus inducing a transference of the G1 stage to S stage in cell-cycle of LSCC cells. These results provide an academic foundation for elucidating the mechanism of LSCC occurrence and evolution and for developing treatment strategies for LSCC.
Collapse
Affiliation(s)
- Xiaoling Hu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Siwei Zou
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Xiaoyu Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Qiangwei Zhang
- Department of Otolaryngology, Shanxi Provincial People's Hospital / the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yanfei Li
- Department of Otolaryngology, Shanxi Provincial People's Hospital / the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Mengya Wang
- Department of Otolaryngology, Shanxi Provincial People's Hospital / the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Tongli Li
- Department of Otolaryngology, Shanxi Provincial People's Hospital / the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Xuanping Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Guodong Li
- Department of Otolaryngology, Shanxi Provincial People's Hospital / the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| |
Collapse
|
4
|
Duan N, Hua Y, Yan X, He Y, Zeng T, Gong J, Fu Z, Li W, Yin Y. Unveiling Alterations of Epigenetic Modifications and Chromatin Architecture Leading to Lipid Metabolic Reprogramming during the Evolutionary Trastuzumab Adaptation of HER2-Positive Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309424. [PMID: 38460162 PMCID: PMC11095153 DOI: 10.1002/advs.202309424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/08/2024] [Indexed: 03/11/2024]
Abstract
Secondary trastuzumab resistance represents an evolutionary adaptation of HER2-positive breast cancer during anti-HER2 treatment. Most current studies have tended to prioritize HER2 and its associated signaling pathways, often overlooking broader but seemingly less relevant cellular processes, along with their associated genetic and epigenetic mechanisms. Here, transcriptome data is not only characterized but also examined epigenomic and 3D genome architecture information in both trastuzumab-sensitive and secondary-resistant breast cancer cells. The findings reveal that the global metabolic reprogramming associated with trastuzumab resistance may stem from genome-wide alterations in both histone modifications and chromatin structure. Specifically, the transcriptional activities of key genes involved in lipid metabolism appear to be regulated by variant promoter H3K27me3 and H3K4me3 modifications, as well as promoter-enhancer interactions. These discoveries offer valuable insights into how cancer cells adapt to anti-tumor drugs and have the potential to impact future diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Ningjun Duan
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yijia Hua
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Xueqi Yan
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yaozhou He
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Tianyu Zeng
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Jue Gong
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Ziyi Fu
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Wei Li
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yongmei Yin
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| |
Collapse
|
5
|
Zhao C, Li X, Zhang R, Lyu H, Xiao S, Guo D, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Sense and anti-sense: Role of FAM83A and FAM83A-AS1 in Wnt, EGFR, PI3K, EMT pathways and tumor progression. Biomed Pharmacother 2024; 173:116372. [PMID: 38432129 DOI: 10.1016/j.biopha.2024.116372] [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: 01/05/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
An increasing number of studies have shown that FAM83A, a member of the family with sequence similarity 83 (FAM83), which consists of eight members, is a key tumor therapeutic target involved in multiple signaling pathways. It has been reported that FAM83A plays essential roles in the regulation of Wnt/β-catenin, EGFR, MAPK, EMT, and other signaling pathways and physiological processes in models of pancreatic cancer, lung cancer, breast cancer, and other malignant tumors. Moreover, the expression of FAM83A could be significantly affected by multiple noncoding RNAs that are dysregulated in malignant tumors, the dysregulation of which is essential for the malignant process. Among these noncoding RNAs, the most noteworthy is the antisense long noncoding (Lnc) RNA of FAM83A itself (FAM83A-AS1), indicating an outstanding synergistic carcinogenic effect between FAM83A and FAM83A-AS1. In the present study, the specific mechanisms by which FAM83A and FAM83A-AS1 cofunction in the Wnt/β-catenin and EGFR signaling pathways were reviewed in detail, which will guide subsequent research. We also described the applications of FAM83A and FAM83A-AS1 in tumor therapy and provided a certain theoretical basis for subsequent drug target development and combination therapy strategies.
Collapse
Affiliation(s)
- Chenshu Zhao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Xiaowen Li
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Hao Lyu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Cefan Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| | - Jingfeng Tang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
6
|
Jiang Y, Yu J, Zhu T, Bu J, Hu Y, Liu Y, Zhu X, Gu X. Involvement of FAM83 Family Proteins in the Development of Solid Tumors: An Update Review. J Cancer 2023; 14:1888-1903. [PMID: 37476189 PMCID: PMC10355199 DOI: 10.7150/jca.83420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
FAM83 family members are a group of proteins that have been implicated in various solid tumors. In this updated review, we mainly focus on the cellular localization, molecular composition, and biological function of FAM83 family proteins in solid tumors. We discussed the factors that regulate abnormal protein expression and alterations in the functional activities of solid tumor cells (including non-coding microRNAs and protein modifiers) and potential mechanisms of tumorigenesis (including the MAPK, WNT, and TGF-β signaling pathways). Further, we highlighted the application of FAM83 family proteins in the diagnoses and treatment of different cancers, such as breast, lung, liver, and ovarian cancers from two aspects: molecular marker diagnosis and tumor drug resistance. We described the overexpression of FAM83 genes in various human malignant tumor cells and its relationship with tumor proliferation, migration, invasion, transformation, and drug resistance. Moreover, we explored the prospects and challenges of using tumor treatments based on the FAM83 proteins. Overall, we provide a theoretical basis for harnessing FAM83 family proteins as novel targets in cancer treatment. We believe that this review opens up open new directions for solid tumor treatment in clinical practice.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Jiahui Yu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Tong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Yueting Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Xudong Zhu
- Department of General Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| |
Collapse
|
7
|
Chi H, Gao X, Xia Z, Yu W, Yin X, Pan Y, Peng G, Mao X, Teichmann AT, Zhang J, Tran LJ, Jiang T, Liu Y, Yang G, Wang Q. FAM family gene prediction model reveals heterogeneity, stemness and immune microenvironment of UCEC. Front Mol Biosci 2023; 10:1200335. [PMID: 37275958 PMCID: PMC10235772 DOI: 10.3389/fmolb.2023.1200335] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Background: Endometrial cancer (UCEC) is a highly heterogeneous gynecologic malignancy that exhibits variable prognostic outcomes and responses to immunotherapy. The Familial sequence similarity (FAM) gene family is known to contribute to the pathogenesis of various malignancies, but the extent of their involvement in UCEC has not been systematically studied. This investigation aimed to develop a robust risk profile based on FAM family genes (FFGs) to predict the prognosis and suitability for immunotherapy in UCEC patients. Methods: Using the TCGA-UCEC cohort from The Cancer Genome Atlas (TCGA) database, we obtained expression profiles of FFGs from 552 UCEC and 35 normal samples, and analyzed the expression patterns and prognostic relevance of 363 FAM family genes. The UCEC samples were randomly divided into training and test sets (1:1), and univariate Cox regression analysis and Lasso Cox regression analysis were conducted to identify the differentially expressed genes (FAM13C, FAM110B, and FAM72A) that were significantly associated with prognosis. A prognostic risk scoring system was constructed based on these three gene characteristics using multivariate Cox proportional risk regression. The clinical potential and immune status of FFGs were analyzed using CiberSort, SSGSEA, and tumor immune dysfunction and rejection (TIDE) algorithms. qRT-PCR and IHC for detecting the expression levels of 3-FFGs. Results: Three FFGs, namely, FAM13C, FAM110B, and FAM72A, were identified as strongly associated with the prognosis of UCEC and effective predictors of UCEC prognosis. Multivariate analysis demonstrated that the developed model was an independent predictor of UCEC, and that patients in the low-risk group had better overall survival than those in the high-risk group. The nomogram constructed from clinical characteristics and risk scores exhibited good prognostic power. Patients in the low-risk group exhibited a higher tumor mutational load (TMB) and were more likely to benefit from immunotherapy. Conclusion: This study successfully developed and validated novel biomarkers based on FFGs for predicting the prognosis and immune status of UCEC patients. The identified FFGs can accurately assess the prognosis of UCEC patients and facilitate the identification of specific subgroups of patients who may benefit from personalized treatment with immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xinrui Gao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wanying Yu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xisheng Yin
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yifan Pan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Gaoge Peng
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xinrui Mao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Alexander Tobias Teichmann
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Zhang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, SD, United States
| | - Lisa Jia Tran
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tianxiao Jiang
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yunfei Liu
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Tu P, Li X, Cao L, Zhong M, Xie Z, Wu Z. Machine learning and BP neural network revealed abnormal B cell infiltration predicts the survival of lung cancer patients. Front Oncol 2022; 12:882018. [PMID: 36303835 PMCID: PMC9592816 DOI: 10.3389/fonc.2022.882018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/29/2022] [Indexed: 11/26/2022] Open
Abstract
FAM83A gene is related to the invasion and metastasis of various tumors. However, the abnormal immune cell infiltration associated with the gene is poorly understood in the pathogenesis and prognosis of NSCLC. Based on the TCGA and GEO databases, we used COX regression and machine learning algorithms (CIBERSORT, random forest, and back propagation neural network) to study the prognostic value of FAM83A and immune infiltration characteristics in NSCLC. High FAM83A expression was significantly associated with poor prognosis of NSCLC patients (p = 0.00016), and had excellent prognostic independence. At the same time, the expression level of FAM83A is significantly related to the T, N, and Stage. Subsequently, based on machine learing strategies, we found that the infiltration level of naive B cells was negatively correlated with the expression of FAM83A. The low infiltration of naive B cells was significantly related to the poor overall survival rate of NSCLC (p = 0.0072). In addition, Cox regression confirmed that FAM83A and naive B cells are risk factors for the prognosis of NSCLC patients. The nomogram combining FAM83A and naive B cells (C-index = 0.748) has a more accurate prognostic ability than the Stage (C-index = 0.651) system. Our analysis shows that abnormal infiltration of naive B cells associated with FAM83A is a key factor in the prognostic prediction of NSCLC patients.
Collapse
Affiliation(s)
- Pinghua Tu
- *Correspondence: Pinghua Tu, ; Zhanling Wu,
| | | | | | | | | | | |
Collapse
|
9
|
Overexpression of FAM83A Is Associated with Poor Prognosis of Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:8767333. [PMID: 36245969 PMCID: PMC9556212 DOI: 10.1155/2022/8767333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 12/03/2022]
Abstract
Family with sequence similarity 83, member A (FAM83A) plays an essential and fundamental role in the proliferation, progression, and apoptosis of many malignant tumors, including lung cancer. This study aimed to determine the expression pattern of FAM83A in lung adenocarcinoma (LUAD) and its correlation with the prognosis of cancer and the survival of the patients. Bioinformatics analysis, immunohistochemistry, and Western blotting were used to explore and detect the expression of FAM83A in LUAD cells. The mechanism of FAM83A in proliferation and migration was examined. The correlation between FAM83A expression and survival rate was assessed by the Kaplan-Meier and Cox regression. FAM83A expression was elevated in LUAD tissues and was related to shorter overall survival (P < 0.05). A significant increase in FAM83A protein was observed in the LUAD tissue (P < 0.05). Compared with patients with early-stage tumors (stage I-II), those with advanced stage tumors (stage III-IV) had significantly higher FAM83A expression levels (P < 0.05). Downregulation of FAM83A led to a reduction in cell proliferation, a decrease in migration ability, and diminished epithelial-mesenchymal transition (EMT) in the lung cancer cell lines. Overexpression of FAM83A was associated with early lymph node metastasis and poor overall survival among LUAD patients. The findings indicated that FAM83A may play a critical role in promoting the LUAD progression and thus might serve as a novel prognostic marker in LUAD.
Collapse
|
10
|
Jin Y, Yu J, Jiang Y, Bu J, Zhu T, Gu X, Zhu X. Comprehensive analysis of the expression, prognostic significance, and function of FAM83 family members in breast cancer. World J Surg Oncol 2022; 20:172. [PMID: 35650627 PMCID: PMC9158143 DOI: 10.1186/s12957-022-02636-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The FAM83 family plays a key role in tumorigenesis and cancer progression. However, the role of the FAM83 family in the development of breast tumors is unclear to date. This report explores the expression, prognostic significance, and function of the FAM83 family members in breast cancer using public databases. METHODS UALCAN database was used to explore the expression of FAM83 family members in breast cancer. Furthermore, we validated the expression of FAM83 family members in twenty pairs of breast cancer and normal tissues by RT-PCR. Kaplan-Meier plotter database was used to explore the prognostic significance of FAM83 family members in breast cancer. GeneMANIA and DAVID databases were used for functional and pathway enrichment analysis of genes co-expressed with FAM83A, FAM83D, FAM83F, and FAM83G. MEXPRESS and UALCAN databases were used to analyze the level of DNA promoter methylation of FAM83A, FAM83D, FAM83F, and FAM83G in breast cancer. TIMER database was utilized to explore the relationships between immune cell infiltration and FAM83A, FAM83D, FAM83F, and FAM83G expression. RESULTS Among FAM83 family members, FAM83A, FAM83D, FAM83F, and FAM83G were higher expressed in breast cancer than in normal tissues. We also validated the significant high expression of FAM83A, FAM83D, FAM83F, and FAM83G mRNA in breast cancer than in normal samples. Their increased expression has an adverse prognostic effect on breast cancer patients. These genes co-expressed with FAM83A, FAM83D, FAM83F, and FAM83G might take part in cell proliferation, G2/M transition of the mitotic cell cycle, regulation of apoptosis process and other cancer-related biological processes. In addition, they were mainly enriched in the Hippo signaling pathway, Hedgehog signaling pathway, PI3K/AKT signaling pathway, and other cancer-related pathways. We also found that promoter DNA methylation might regulate the expression of FAM83A, FAM83D, FAM83F, and FAM83G mRNA in most CpG islands. At last, we found the expression of FAM83A, FAM83D, FAM83F, and FAM83G mRNA was significantly related to immune cell infiltration. CONCLUSIONS FAM83A, FAM83D, FAM83F, and FAM83G were highly expressed in breast cancer tissues and had an adverse effect on the survival outcomes of breast cancer patients. Also, they were involved in breast cancer-related signal pathways. Therefore, they might serve as potential therapeutic targets for breast cancer clinical treatment.
Collapse
Affiliation(s)
- Yi Jin
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Jiahui Yu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Yi Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Tong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.
| | - Xudong Zhu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| |
Collapse
|
11
|
Marino N, German R, Podicheti R, Rockey P, Sandusky GE, Temm CJ, Nakshatri H, Addison RJ, Selman B, Althouse SK, Storniolo AMV. FAM83A is a potential biomarker for breast cancer initiation. Biomark Res 2022; 10:8. [PMID: 35183258 PMCID: PMC8858535 DOI: 10.1186/s40364-022-00353-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/31/2022] [Indexed: 11/10/2022] Open
Abstract
Background Family with sequence similarity 83 member A (FAM83A) presents oncogenic properties in several cancers including breast cancer. Recently, we reported FAM83A overexpression in normal breast tissues from women at high risk of breast cancer. We now hypothesize that FAM83A is a key factor in breast cancer initiation. Methods Immunohistochemical staining was used to evaluate FAM83A protein levels in both a normal breast tissue microarray (TMA, N = 411) and a breast tumor TMA (N = 349). EGFR staining and its correlation with FAM83A expression were also assessed. Lentivirus-mediated manipulation of FAM83A expression in primary and hTERT-immortalized breast epithelial cells was employed. Biological and molecular alterations upon FAM83A overexpression/downregulation and FAM83A’s interaction partners were investigated. Results TMA analysis revealed a 1.5-fold increase in FAM83A expression level in breast cancer cases as compared with normal breast tissues (p < 0.0001). FAM83A protein expression was directly correlated with EGFR level in both normal and breast cancer tissues. In in vitro assays, exogenous expression of FAM83A in either primary or immortalized breast epithelial cells promoted cell viability and proliferation. Additionally, Ingenuity Pathway Analysis (IPA) revealed that FAM83A overexpression in primary cells affected the expression of genes involved in cellular morphology and metabolism. Mass spectrometry analysis identified DDX3X and LAMB3 as potential FAM83A interaction partners in primary cells, while we detected FAM83A interaction with cytoskeleton reorganization factors, including LIMA1, MYH10, PLEC, MYL6 in the immortalized cells. Conclusions This study shows that FAM83A promotes metabolic activation in primary breast epithelial cells and cell proliferation in both primary and immortalized cells. These findings support its role in early breast oncogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s40364-022-00353-9.
Collapse
|
12
|
Marino N, German R, Podicheti R, Rusch DB, Rockey P, Huang J, Sandusky GE, Temm CJ, Althouse S, Nephew KP, Nakshatri H, Liu J, Vode A, Cao S, Storniolo AMV. Aberrant epigenetic and transcriptional events associated with breast cancer risk. Clin Epigenetics 2022; 14:21. [PMID: 35139887 PMCID: PMC8830042 DOI: 10.1186/s13148-022-01239-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Genome-wide association studies have identified several breast cancer susceptibility loci. However, biomarkers for risk assessment are still missing. Here, we investigated cancer-related molecular changes detected in tissues from women at high risk for breast cancer prior to disease manifestation. Disease-free breast tissue cores donated by healthy women (N = 146, median age = 39 years) were processed for both methylome (MethylCap) and transcriptome (Illumina's HiSeq4000) sequencing. Analysis of tissue microarray and primary breast epithelial cells was used to confirm gene expression dysregulation. RESULTS Transcriptomic analysis identified 69 differentially expressed genes between women at high and those at average risk of breast cancer (Tyrer-Cuzick model) at FDR < 0.05 and fold change ≥ 2. Majority of the identified genes were involved in DNA damage checkpoint, cell cycle, and cell adhesion. Two genes, FAM83A and NEK2, were overexpressed in tissue sections (FDR < 0.01) and primary epithelial cells (p < 0.05) from high-risk breasts. Moreover, 1698 DNA methylation changes were identified in high-risk breast tissues (FDR < 0.05), partially overlapped with cancer-related signatures, and correlated with transcriptional changes (p < 0.05, r ≤ 0.5). Finally, among the participants, 35 women donated breast biopsies at two time points, and age-related molecular alterations enhanced in high-risk subjects were identified. CONCLUSIONS Normal breast tissue from women at high risk of breast cancer bears molecular aberrations that may contribute to breast cancer susceptibility. This study is the first molecular characterization of the true normal breast tissues, and provides an opportunity to investigate molecular markers of breast cancer risk, which may lead to new preventive approaches.
Collapse
Affiliation(s)
- Natascia Marino
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA. .,Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Rana German
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, 47405, USA
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, 47405, USA
| | - Pam Rockey
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | - Jie Huang
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, 47405, USA
| | - George E Sandusky
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Constance J Temm
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sandra Althouse
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kenneth P Nephew
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Bloomington, IN, 47405, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jun Liu
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, 47405, USA
| | - Ashley Vode
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | - Sha Cao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Anna Maria V Storniolo
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA.,Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
13
|
CircMIIP Contributes to Non-Small Cell Lung Cancer Progression by Binding miR-766-5p to Upregulate FAM83A Expression. Lung 2022; 200:107-117. [PMID: 35013754 DOI: 10.1007/s00408-021-00500-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/20/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Circular RNA migration and invasion inhibitory protein (circMIIP) is reported to be upregulated in non-small cell lung cancer (NSCLC) tissues compared with normal tissues. However, the role and working mechanism of circMIIP in NSCLC progression remain largely unclear. METHODS Cell proliferation ability was analyzed by colony formation assay, cell counting kit-8 (CCK-8) assay, and 5-ethynyl-2'-deoxyuridine assay. Cell apoptosis was assessed by flow cytometry. Transwell assays were performed to analyze the migration and invasion abilities of NSCLC cells. The interaction between microRNA-766-5p (miR-766-5p) and circMIIP or family with sequence similarity 83A (FAM83A) was validated by dual-luciferase reporter assay and RNA immunoprecipitation assay. Xenograft tumor model was established to analyze the role of circMIIP on tumor growth in vivo. RESULTS CircMIIP was highly expressed in NSCLC tissues and cell lines. CircMIIP knockdown restrained the proliferation, migration and invasion and induced the apoptosis of NSCLC cells. CircMIIP acted as a molecular sponge for miR-766-5p, and circMIIP silencing-mediated anti-tumor effects were largely overturned by the knockdown of miR-766-5p in NSCLC cells. miR-766-5p interacted with the 3' untranslated region (3'UTR) of FAM83A, and FAM83A overexpression largely reversed miR-766-5p accumulation-induced anti-tumor effects in NSCLC cells. CircMIIP competitively bound to miR-766-5p to elevate the expression of FAM83A in NSCLC cells. CircMIIP knockdown significantly restrained xenograft tumor growth in vivo. CONCLUSION CircMIIP promoted cell proliferation, migration and invasion and suppressed cell apoptosis in NSCLC cells through mediating miR-766-5p/FAM83A axis.
Collapse
|
14
|
Zhang T, Lai S, Cai Y, Huang Z, Li Y, Chen S, Zhang Z, Ye Z, Lai X, Zhai E, Cai S, Chen J. Comprehensive Analysis and Identification of Prognostic Biomarkers and Therapeutic Targets Among FAM83 Family Members for Gastric Cancer. Front Cell Dev Biol 2021; 9:719613. [PMID: 34869310 PMCID: PMC8640971 DOI: 10.3389/fcell.2021.719613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/15/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the most common and poor prognosis malignancy in the world. The Family with sequence similarity 83 (FAM83) comprises of eight members of A–H. Accumulating evidence confirmed important roles for FAM83 family in tumorigenesis and progression. However, the prognostic values of FAM83 family in GC still have not been clarified. Methods: ONCOMINE, UALCAN, GEPIA, THE HUMAN PROTEIN ATLAS, Kaplan–Meier Plotter, cBioPortal, DAVID, STRING and TIMER databases and R software were adopted in this study. Results: In this study, we demonstrated that the mRNA levels of FAM83 B/C/D/H were significantly up-regulated in stomach adenocarcinoma (STAD), but the protein level of FAM83G/H were remarkable lowly in STAD. Next, FAM83C/D/G/H were significantly associated with tumor stages in STAD patients. Then, the mutation rate of FAM83 family members in STAD patients was 46%, and the highest mutation rate was FAM83H (23%). Furthermore, the functions of FAM83 family and their 259 co-expression genes were primarily related to Shigellosis, RNA degradation and Ribosome biogenesis in eukaryotes pathway. Besides, we have established the prognostic model of FAM83 family in STAD, including the prognostic model of STAD patients (FAM83C/D/G), STAD with lymph node metastasis (FAM83C/D/G/H) and STAD with ERBB2 high expression (FAM83G/H). FAM83C/D high expression with a poor prognosis, while FAM83G/H high expression with a favorable prognosis of STAD. Additionally, we found that the expression of FAM83C/D/G/H were significantly correlated with the infiltration of six types of immune cells [B cells, CD8+T cells, CD4+T cells, macrophages and Myeloid dendritic cells (DC)], whereas CD4+T cells and Macrophage cells have higher risk scores (HR > 1) when FAM83C lowly expression and FAM83D highly expression. The risk score of NK cells was significantly reduced when FAM83G lowly expression and FAM83H highly expression (HR < 1). Conclusion: These findings suggested that FAM83C/D/G/H might play key roles in STAD tumorigenesis and progression, and FAM83C/D might be risk factors but FAM83G/H might be favorable prognostic factors for STAD patients. In addition, CD4+T cells and Macrophage cells may be the promoters of FAM83D in progression of STAD, while NK cells may promote the protective effect of FAM83H on STAD patients.
Collapse
Affiliation(s)
- Tianhao Zhang
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shurong Lai
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Yuzhi Cai
- Department of Hepatobiliary Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Zhixin Huang
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Sile Chen
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Zhimei Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Ye
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoling Lai
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Ertao Zhai
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Shirong Cai
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Jianhui Chen
- Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Metsiou DN, Kozaniti FK, Deligianni DD. Engineering Breast Cancer Cells and hUMSCs Microenvironment in 2D and 3D Scaffolds: A Mechanical Study Approach of Stem Cells in Anticancer Therapy. Bioengineering (Basel) 2021; 8:bioengineering8110189. [PMID: 34821755 PMCID: PMC8615245 DOI: 10.3390/bioengineering8110189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Cell biomechanics plays a major role as a promising biomarker for early cancer diagnosis and prognosis. In the present study, alterations in modulus of elasticity, cell membrane roughness, and migratory potential of MCF-7 (ER+) and SKBR-3 (HER2+) cancer cells were elucidated prior to and post treatment with conditioned medium from human umbilical mesenchymal stem cells (hUMSCs-CM) during static and dynamic cell culture. Moreover, the therapeutic potency of hUMSCs-CM on cancer cell’s viability, migratory potential, and F-actin quantified intensity was addressed in 2D surfaces and 3D scaffolds. Interestingly, alterations in ER+ cancer cells showed a positive effect of treatment upon limiting cell viability, motility, and potential for migration. Moreover, increased post treatment cell stiffness indicated rigid cancer cells with confined cell movement and cytoskeletal alterations with restricted lamellipodia formation, which enhanced these results. On the contrary, the cell viability and the migratory potential were not confined post treatment with hUMSCs-CM on HER2+ cells, possibly due to their intrinsic aggressiveness. The increased post treatment cell viability and the decreased cell stiffness indicated an increased potency for cell movement. Hence, the therapy had no efficacy on HER2+ cells.
Collapse
|
16
|
Ji H, Song H, Wang Z, Jiao P, Xu J, Li X, Du H, Wu H, Zhong Y. FAM83A promotes proliferation and metastasis via Wnt/β-catenin signaling in head neck squamous cell carcinoma. J Transl Med 2021; 19:423. [PMID: 34641907 PMCID: PMC8507380 DOI: 10.1186/s12967-021-03089-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/26/2021] [Indexed: 11/23/2022] Open
Abstract
This research aimed to investigate the expression and function of FAM83A in the proliferation and metastasis in head and neck squamous cell carcinoma (HNSCC). FAM83A mRNA and protein expressions in HNSCC were detected in primary HNSCC samples and cell lines. The associations between FAM83A expression and clinicopathologic variables were evaluated through tissue microarrays. Besides, FAM83A knockdown and overexpression cell lines were constructed to assess cell growth and metastasis in vitro and the relationship between FAM83A and epithelial-mesenchymal transition (EMT). Furthermore, two models of xenograft tumors in nude mice were used to assess the tumorigenicity and metastasis ability of FAM83A in vivo. In the present study, overexpression of FAM83A in HNSCC samples was significantly associated with tumor size, lymph node status and clinical tumor stages. Mechanically, FAM83A could promote HNSCC cell growth and metastasis by inducing EMT via activating Wnt/β-catenin signaling pathway. Rescue experiment demonstrated the inhibition of β-catenin could counteract the function of FAM83A. Also, the FAM83A knockdown could suppress tumor growth and distant metastasis in the xenograft animal models of HNSCC. In conclusion, this study identifies FAM83A as an oncogene of HNSCC. This study provides new insights into the molecular pathways that contribute to EMT in HNSCC. We revealed a previously unknown FAM83A-Wnt–β-catenin signaling axis involved in the EMT of HNSCC. There may be a potential bi-directional signaling loop between FAM83A and Wnt/β-catenin signaling pathway in HNSCC.
Collapse
Affiliation(s)
- Huan Ji
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyang Song
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China.,Department of General Dentistry, Department of Oral Medicine, The Affiliated Stomatological Hospital of Nanjing Medical University, #136 Hanzhong Road, Nanjing, 210029, Jiangsu, China
| | - Zeyu Wang
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Pengfei Jiao
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Jiani Xu
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xuan Li
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hongming Du
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Heming Wu
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China. .,Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.
| | - Yi Zhong
- Jiangsu Province Key Laboratory of Oral Diseases, School of Stomatology, Nanjing Medical University, Nanjing, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, School of Stomatology, Nanjing Medical University, Nanjing, China. .,Department of General Dentistry, Department of Oral Medicine, The Affiliated Stomatological Hospital of Nanjing Medical University, #136 Hanzhong Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
17
|
Owolabi KM, Shikongo A. Fractal Fractional Operator Method on HER2+ Breast Cancer Dynamics. INTERNATIONAL JOURNAL OF APPLIED AND COMPUTATIONAL MATHEMATICS 2021; 7:85. [DOI: 10.1007/s40819-021-01030-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/24/2021] [Indexed: 04/01/2025]
|
18
|
Liu T, Chen J, Du Q, Liu J, Chen M, Ooi S, Huang J, Wang W, Yao S. Family with sequence similarity 83 member A promotes tumor cell proliferation and metastasis and predicts poor prognosis in cervical cancer. Pathol Res Pract 2021; 222:153450. [PMID: 33962175 DOI: 10.1016/j.prp.2021.153450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/09/2022]
Abstract
Family with sequence similarity 83 member A (FAM83A) is a member of the FAM83 family and is proven to have oncogenic properties in several cancers. However, the mechanisms of FAM83A in human cervical cancer (CC) progression are unknown. Here, we found that FAM83A is highly expressed in CC tissues and cell lines through western blot and qRT-PCR. We utilized GEO datasets to assess FAM83A expression in CC in comparison to the normal cervical tissue (NCT) (GSE6791), and similarly, in lymph node positive CC compared to the lymph node negative CC (GSE26511). Immunohistochemistry (IHC) was used to quantify FAM83A expression in 20 NCT and 105 CC patient samples. FAM83A expression is upregulated in early-stage CC and correlates with aggressive clinicopathologic features. Moreover, both our hospital's and TCGA datasets revealed that patients of early-stage CC with higher FAM83A expression had a poorer prognosis. Subsequently, CCK-8 and transwell assays verified that FAM83A promotes proliferation, migration, and invasion of CC cells. Additionally, Gene Set Enrichment Analysis (GSEA) revealed that FAM83A is not only involved in cell development, differentiation, and proliferation but is also correlated with cell junction assembly and cell matrix adhesion. It might also be affiliated with the regulation of tumor necrosis factor-mediated signaling pathway and the regulation of the ErbB signaling pathway in CC. These results indicate that FAM83A promotes tumor cell proliferation, migration, and metastasis. Our study provides novel evidence FAM83A may act as a promising therapeutic target for CC.
Collapse
Affiliation(s)
- Tianyu Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Jian Chen
- Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Ming Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Shiyin Ooi
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Jiaming Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
19
|
Zhao M, Scott S, Evans KW, Yuca E, Saridogan T, Zheng X, Wang H, Korkut A, Cruz Pico CX, Demirhan M, Kirby B, Kopetz S, Diala I, Lalani AS, Piha-Paul S, Meric-Bernstam F. Combining Neratinib with CDK4/6, mTOR, and MEK Inhibitors in Models of HER2-positive Cancer. Clin Cancer Res 2021; 27:1681-1694. [PMID: 33414137 PMCID: PMC8075007 DOI: 10.1158/1078-0432.ccr-20-3017] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/16/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Neratinib is an irreversible, pan-HER tyrosine kinase inhibitor that is FDA approved for HER2-overexpressing/amplified (HER2+) breast cancer. In this preclinical study, we explored the efficacy of neratinib in combination with inhibitors of downstream signaling in HER2+ cancers in vitro and in vivo. EXPERIMENTAL DESIGN Cell viability, colony formation assays, and Western blotting were used to determine the effect of neratinib in vitro. In vivo efficacy was assessed with patient-derived xenografts (PDX): two breast, two colorectal, and one esophageal cancer (with HER2 mutations). Four PDXs were derived from patients who received previous HER2-targeted therapy. Proteomics were assessed through reverse phase protein arrays and network-level adaptive responses were assessed through Target Score algorithm. RESULTS In HER2+ breast cancer cells, neratinib was synergistic with multiple agents, including mTOR inhibitors everolimus and sapanisertib, MEK inhibitor trametinib, CDK4/6 inhibitor palbociclib, and PI3Kα inhibitor alpelisib. We tested efficacy of neratinib with everolimus, trametinib, or palbociclib in five HER2+ PDXs. Neratinib combined with everolimus or trametinib led to a 100% increase in median event-free survival (EFS; tumor doubling time) in 25% (1/4) and 60% (3/5) of models, respectively, while neratinib with palbociclib increased EFS in all five models. Network analysis of adaptive responses demonstrated upregulation of EGFR and HER2 signaling in response to CDK4/6, mTOR, and MEK inhibition, possibly providing an explanation for the observed synergies with neratinib. CONCLUSIONS Taken together, our results provide strong preclinical evidence for combining neratinib with CDK4/6, mTOR, and MEK inhibitors for the treatment of HER2+ cancer.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen Scott
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kurt W Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erkan Yuca
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Turcin Saridogan
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heping Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christian X Cruz Pico
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mehmet Demirhan
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryce Kirby
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Identification of Prognostic and Therapeutic Biomarkers among FAM83 Family Members for Pancreatic Ductal Adenocarcinoma. DISEASE MARKERS 2021; 2021:6682697. [PMID: 33747255 PMCID: PMC7943308 DOI: 10.1155/2021/6682697] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 01/11/2023]
Abstract
Family with sequence similarity 83 (FAM83) members were shown recently to have oncogenic effect in a variety of cancer types, but the biological roles and prognostic value of FAM83 family in pancreatic ductal adenocarcinoma remain unknown. In the current study, the clinical significance and molecular function of the FAM83 family were assessed by multiple bioinformatics analysis. Besides, potential associations between differentially expressed genes (DEGs) of FAM83 family and antitumor immunity were evaluated using TIMER and TISIDB analyses. As the results show, FAM83A, FAM83D, FAM83E, and FAM83H were significantly upregulated in PDAC and were identified as DEGs. Higher expression of FAM83A, FAM83B, FAM83D, FAM83E, and FAM83H were associated with advanced tumor stage or worse patient prognosis. Importantly, the overexpression of DEGs was found to be significantly correlated with activated KRAS and loss of SMAD4, which are important drivers for PDAC. Further, FAM83A, FAM83D, and FAM83H were associated with CD8+ T cell, Gamma Delta T cell, and CD4+ T cell infiltration in PDAC and FAM83H was found closely correlated with some immunomodulators including immunoinhibitors, immunostimulators, and MHC molecules. In conclusion, FAM83A, FAM83D, FAM83E, and FAM83H have significant prognostic value in PDAC and they may play important roles in regulating tumor progression and the immune cell infiltration.
Collapse
|
21
|
Rong L, Li H, Li Z, Ouyang J, Ma Y, Song F, Chen Y. FAM83A as a Potential Biological Marker Is Regulated by miR-206 to Promote Cervical Cancer Progression Through PI3K/AKT/mTOR Pathway. Front Med (Lausanne) 2020; 7:608441. [PMID: 33344485 PMCID: PMC7746878 DOI: 10.3389/fmed.2020.608441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Background and Objective: Chemotherapy and radiotherapy are effective treatment options for cervical cancer (CC), but their efficacy is limited by short survival rate of about 5 years particularly for advance stage CC. Bioinformatics analysis combined with experimental in vivo and in vitro data can identify potential markers of tumorigenesis and cancer progression to improve CC prognosis and survival rate of the patients. This study aims to investigate the prognostic value of family with sequence similarity 83, member A (FAM83A) gene and miR-206 in promoting CC progression and the involved genetic signaling pathways. Method: This was a bioinformatic analysis study based on RNA sequencing data of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and verification by in vivo and in vitro experimental data. It was designed to evaluate whether the aberrantly expressed gene signatures could serve as new potential biomarker to improve prognosis prediction in CC. The TCGA RNA sequencing data [306 cervical squamous cell carcinoma (SCC) and endocervical adenocarcinoma samples and 13 adjacent samples] and GEO data (GSE9750 and GSE52903 datasets) were integrated and performed a bioinformatics analysis. Results: The results showed that CC-associated FAM83A gene serves as a key regulator of CC development and progression. Functionally, we observed that FAM83A is significantly overexpressed in CC, which is linked to poor overall survival as well as disease-free survival in CC patients. The in-vitro and in-vivo assessments performed after silencing FAM83A revealed that cell proliferation was significantly inhibited and the S-phase cell cycle arrest was induced. Mechanistically, FAM83A plays a role in PI3K/AKT signaling, and its downstream molecules could promote CC cell proliferation. Furthermore, functionality assessments by in-vitro luciferase reporter system and immunoblot analysis showed that miR-206 was the upstream of FAM83A and negatively correlated with FAM83A. Conclusion: The miR-206/FAM83A/PI3K/AKT signaling pathway possibly serves as a critical effector in CC progression indicating the potential prognostic value of FAM83A gene as a novel biomarker for CC progression.
Collapse
Affiliation(s)
- Li Rong
- Chongqing Public Health Medical Center, Chongqing, China
| | - Haiyu Li
- Chongqing Public Health Medical Center, Chongqing, China
| | - Zhaodong Li
- Chongqing Public Health Medical Center, Chongqing, China
| | - Jing Ouyang
- Chongqing Medical University, Chongqing, China
| | - Yongping Ma
- Chongqing Public Health Medical Center, Chongqing, China
| | - Fangzhou Song
- Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Liu C, Jiang Y, Han B. miR-613 Suppresses Chemoresistance and Stemness in Triple-Negative Breast Cancer by Targeting FAM83A. Cancer Manag Res 2020; 12:12623-12633. [PMID: 33324107 PMCID: PMC7733386 DOI: 10.2147/cmar.s276316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/10/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is the most aggressive malignancy of breast cancer, which represents about 20% of all cases. The prognosis of TNBC remains unfavorable due to the lack of targeted therapy and chemoresistance. The aim of this study is to investigate the role of miR-613 in TNBC. Material and Methods Quantitative RT-PCT was used to explore the expression of miR-613 in breast cancer clinical samples and cell lines. MTT, colony formation assay, spheroid formation assay and xenograft tumor growth assay were used to investigate the role of miR-613 in vitro and in vivo. Cell apoptosis and surface marker expression were measured by flow cytometry. Dual-luciferase reporter assay was used to explore the function of miR-613 in regulating FAM83A 3ʹUTR. Immunohistochemical staining was used to investigate the expression of FAM83A in TNBC tissues. Results We found that miR-613 expression was significantly downregulated in breast cancer tissues and was even lower in TNBC compared with that in other types of breast cancer. A similar result was found in breast cancer cell lines. Further analysis indicated that miR-613 could suppress TNBC cell growth, chemoresistance and stem-cell-like phenotype. Moreover, we also demonstrated that miR-613 suppressed tumorigenesis in vivo. Mechanically, we explored the downstream target of miR-613 and identified that miR-613 could directly bind to the 3ʹUTR of FAM83A, which contributed to the miR-613 mediated tumor suppression. The expression of miR-613 and FAM83A was negatively correlated. Restoring the expression of FAM83A attributed to the chemoresistance and stemness of TNBC cells. Conclusion We demonstrated that loss of miR-613 was critical for TNBC malignancy and restoring its expression could be served as a potential approach for TNBC treatment.
Collapse
Affiliation(s)
- Cuiping Liu
- Department of Breast Surgery, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia 024005, People's Republic of China
| | - Yizhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Baosan Han
- Department of Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
| |
Collapse
|
23
|
Involvement of MicroRNA-1-FAM83A Axis Dysfunction in the Growth and Motility of Lung Cancer Cells. Int J Mol Sci 2020; 21:ijms21228833. [PMID: 33266425 PMCID: PMC7700477 DOI: 10.3390/ijms21228833] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the most prevalent types of cancer and the leading cause of cancer-related deaths worldwide. Among all cancers, lung cancer has the highest incidence, accompanied by a high mortality rate at the advanced stage. Favorable prognostic biomarkers can effectively increase the survival rate in lung cancer. Our results revealed FAM83A (Family with sequence similarity 83, member A) overexpression in lung cancer tissues compared with adjacent normal tissues. Furthermore, high FAM83A expression was closely associated with poor lung cancer survival. Here, through siRNA transfection, we effectively inhibited FAM83A expression in the lung cancer cell lines H1355 and A549. FAM83A knockdown significantly suppressed the proliferation, migration, and invasion ability of these cells. Furthermore, FAM83A knockdown could suppress Epidermal growth factor receptor (EGFR)/Mitogen-activated protein kinase (MAPK)/Choline kinase alpha (CHKA) signaling activation in A549 and H1355. By using a bioinformatics approach, we found that FAM83A overexpression in lung cancer may result from miR-1-3p downregulation. In summary, we identified a novel miR-1-FAM83A axis could partially modulate the EGFR/choline phospholipid metabolism signaling pathway, which suppressed lung cancer growth and motility. Our findings provide new insights for the development of lung cancer therapeutics.
Collapse
|
24
|
Long noncoding RNA FAM83A-AS1 facilitates hepatocellular carcinoma progression by binding with NOP58 to enhance the mRNA stability of FAM83A. Biosci Rep 2020; 39:220808. [PMID: 31696213 PMCID: PMC6851519 DOI: 10.1042/bsr20192550] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/10/2019] [Accepted: 10/15/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC), as one of the commonest cancers globally, is a primary malignancy in human liver with a characteristic of high mortality rate. Long noncoding RNAs (lncRNAs) are confirmed to be implicated with multiple cancers including HCC. LncRNA FAM83A-AS1 has also been validated as an oncogene in lung cancer, but its mechanism in HCC is poorly understood. Our research is intended to investigate the underlying mechanism of FAM83A-AS1 in HCC. In the present study, we found the abundantly increased expression level of FAM83A-AS1 in HCC tissues and cells. FAM83A-AS1 inhibition hampered cell proliferation, migration and elevated cell apoptosis in HCC. Moreover, FAM83A-AS1 could positively regulate FAM83A, and FAM83A could also promote the progression of HCC. In addition, FAM83A-AS1 and FAM83A were both verified to bind with NOP58, and FAM83A-AS1 enhanced the mRNA stability of FAM83A by binding with NOP58. In rescue assays, the suppressed influence of down-regulated FAM83A-AS1#1 on cell proliferation, migration as well as the accelerated influence of FAM83A-AS1#1 knockdown on cell apoptosis could be partially recovered by overexpression of FAM83A. In conclusion, FAM83A-AS1 facilitated HCC progression by binding with NOP58 to enhance the stability of FAM83A. These findings offer a novel biological insight into HCC treatment.
Collapse
|
25
|
Xu J, Lu W. FAM83A exerts tumor‑suppressive roles in cervical cancer by regulating integrins. Int J Oncol 2020; 57:509-521. [PMID: 32626940 PMCID: PMC7307588 DOI: 10.3892/ijo.2020.5078] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Family with sequence similarity 83 member A (FAM83A) has been recently observed to be upregulated in various types of cancer and hypothesized to be serve as an oncogene. The present study aimed to determine the functional roles and the underlying molecular mechanism of FAM83A in cervical cancer. The results demonstrated that although FAM83A expression was increased in cervical cancer compared with normal tissues, the expression levels of FAM83A were decreased in patients with advanced FIGO stage, deep stromal invasion, poor differentiation and/or lymph node metastasis and negatively associated with short survival time of patients with cervical cancer. FAM83A knockdown promoted cell proliferative, migratory and invasive abilities of CaSki and HeLa cells. A mouse xenograft model demonstrated that FAM83A knockdown promoted tumor growth in vivo. Mechanistically, RNA sequencing results revealed that knockdown of FAM83A increased the transcription of genes mainly associated with oncogenesis-associated pathways. In addition, FAM83A knockdown increased the protein levels of α1, α3, α5, β4 and β5 integrins in vitro and in vivo, and the expression of FAM83A was also negatively associated with the levels of these proteins in human cervical cancer tissue samples. In conclusion, the results of the present study suggested that FAM83A may exert a tumor-suppressive role in cervical cancer by suppressing the expression of integrins, which may offer new insight into the biological basis of cervical cancer.
Collapse
Affiliation(s)
- Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Weiguo Lu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
26
|
Zhang J, Sun G, Mei X. Elevated FAM83A expression predicts poorer clincal outcome in lung adenocarcinoma. Cancer Biomark 2020; 26:367-373. [PMID: 31594212 DOI: 10.3233/cbm-190520] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Family with sequence similarity 83 member A (FAM83A) can promote tumor cell proliferation and facilitate epidermal growth factor tyrosine kinase inhibitor resistance in some malignant tumors, but its role in lung cancer has not been directly explored. OBJECTIVE We investigated FAM83A expression in lung adenocarcinoma (LUAD) and its significance in clinicopathologic characteristics and prognosis of the disease. PATIENTS AND METHODS We analyzed the mRNA expression of FAM83A in LUAD and normal (or adjacent) lung tissues from Oncomine database firstly. Then, we detected FAM83A protein expression in five paired fresh LUAD and adjacent lung tissue specimens from patients in our hospital by Western blotting. In addtion, FAM83A expression in 86 paraffin-embedded archived LUAD samples was evaluated by Immunohistochemistry, and the correlations between FAM83A expression and clinicopathologic characteristics and prognosis of the patients were analyzed. RESULTS Oncomine data analysis manifested that FAM83A mRNA expression was increased in LUAD. Western blotting revealed higher FAM83A expression in fresh LUAD tissues than in the adjacent lung tissues (P= 0.036). Immunohistochemistry analysis on 86 paraffin samples further demonstrated that the LUAD tissue had higher FAM83A expression than adjacent lung tissue (P< 0.001). The correlation analysis revealed that advanced stage tumors (stage III-IV) had higher FAM83A expression than early stage tumors (stage I-II) (P= 0.004). High FAM83A expression was significantly associated with lymphnode involvement and clinical staging (P= 0.008 and 0.008 respectively). Univariate and multivariate Cox regression analysis manifested that FAM83A expression was an independent predictive factor for poor survival. Kaplan-Meier survival curves showed that patients with higher FAM83A expression had shorter overall survival than those with lower FAM83A expressions (P= 0.002). CONCLUSION FAM83A is upregulated in advanced LUAD and is related to unfavorible prognosis. FAM83A might be a novel diagnostic and prognositic biomarker for LUAD.
Collapse
Affiliation(s)
- Junqiang Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaodong Mei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
27
|
Hu H, Wang F, Wang M, Liu Y, Wu H, Chen X, Lin Q. FAM83A is amplified and promotes tumorigenicity in non-small cell lung cancer via ERK and PI3K/Akt/mTOR pathways. Int J Med Sci 2020; 17:807-814. [PMID: 32218702 PMCID: PMC7085261 DOI: 10.7150/ijms.33992] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity 83A (FAM83A) is a newly-found over-expressed oncogene in several types of cancers and associates with poor prognosis. However, the role that FAM83A may play in the carcinogenesis of non-small cell lung cancer (NSCLC) still needs to be defined. The present study aimed to investigate the function of FAM83A in NSCLC progression and to investigate the possible mechanism. Analysis of Gene Expression Omnibus (GEO) database and rt-PCR showed up-regulated expression of FAM83A in NSCLC. GEO and the Cancer Genome Atlas (TCGA) data analysis revealed that high expression level of FAM83A in NSCLC was associated with poor prognosis. In vitro experiments showed that depleting FAM83A by siRNA/shRNA significantly inhibited cell proliferation and induced cell apoptosis. Cell motility was also retarded after silencing FAM83A, as demonstrated by Transwell assay. FAM83A depletion in A549 cells also inhibited subcutaneous tumor growth and lung metastasis in vivo. Western blotting showed that silencing FAM83A decreased the phosphorylation of ERK and PI3K/Akt/mTOR. On the other hand, overexpressing FAM83A in vitro enhanced cell proliferation and invasiveness, which was repressed by PI3K inhibitor and ERK inhibitor separately. Taken together, our study suggests that FAM83A promotes tumorigenesis of NSCLC at least partly via ERK and PI3K/Akt/mTOR pathways, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Haiyang Hu
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Fajiu Wang
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, No. 41 Xibei Road, Ningbo 315010, China
| | - Muyun Wang
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yuanyuan Liu
- Department of Otorhinolaryngology Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Han Wu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Xi Chen
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Qiang Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| |
Collapse
|
28
|
Liu C, Peng X, Li Y, Liu S, Hou R, Zhang Y, Zuo S, Liu Z, Luo R, Li L, Fang W. Positive feedback loop of FAM83A/PI3K/AKT/c-Jun induces migration, invasion and metastasis in hepatocellular carcinoma. Biomed Pharmacother 2019; 123:109780. [PMID: 31901550 DOI: 10.1016/j.biopha.2019.109780] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/13/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
FAM83A is part of an 8-member protein family of unknown function and is reported to be a cancer-promoting and treatment-resistance factor in several cancers. However, its role in hepatocellular carcinoma (HCC) remains unclear. Analysis of the Cancer Genome Atlas (TCGA) showed that FAM83A mRNA expression is upregulated in HCC, as are the protein expression levels in both HCC cell lines and tissues. Clinical data have demonstrated that high FAM83A expression is positively correlated with poor progression-free survival time, thus suggesting its cancer-promoting potential. Functional analyses showed that FAM83A overexpression promoted HCC cell migration and invasion in vitro and suppressed sorafenib sensitivity. Inhibiting FAM83A reversed these results. A pulmonary metastasis model further confirmed that FAM83A promoted HCC cell metastasis in vivo. Mechanistic analyses indicated that FAM83A activated the PI3K/AKT signaling pathway, its downstream c-JUN protein, and epithelial-to-mesenchymal transition (EMT)-related protein levels, including downregulation of E-cadherin and upregulation of Vimentin and N-cadherin. Interestingly, c-JUN induced FAM83A expression by directly binding to its promoter region and thus forming a positive-feedback loop for FAM83A/PI3K/AKT/c-JUN. In conclusion, we demonstrated that FAM83A, as a cancer-metastasis promoter, accelerates migration, invasion and metastasis by activating the PI3K/AKT/c-JUN pathway and inducing its self-expression via feedback, thus forming a FAM83A/PI3K/AKT/c-JUN positive-feedback loop to activate EMT signaling and finally promote HCC migration, invasion and metastasis.
Collapse
Affiliation(s)
- Chen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China; The First Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Xuemei Peng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Yonghao Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Shu Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Rentao Hou
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Yewei Zhang
- Department of General Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Shi Zuo
- Department of General Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Zhan Liu
- Department of Gastroenterology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, 410002, China
| | - Rongcheng Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| | - Libo Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| |
Collapse
|
29
|
Zhang J, Sun M, Hao M, Diao K, Wang J, Li S, Cao Q, Mi X. FAM53A Affects Breast Cancer Cell Proliferation, Migration, and Invasion in a p53-Dependent Manner. Front Oncol 2019; 9:1244. [PMID: 31799197 PMCID: PMC6874147 DOI: 10.3389/fonc.2019.01244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity 53-member A (FAM53A) is an uncharacterized protein with a suspected but unclear role in tumorigenesis. In this study, we examined its role in breast cancer. Immunohistochemical staining of specimens from 199 cases of breast cancer demonstrated that FAM53A levels were negatively correlated with p53 status. In the p53 wild-type breast cancer cell line MCF-7, FAM53A overexpression inhibited cell migration, invasion, and proliferation, downregulated the expression of Snail, cyclin D1, RhoA, RhoC, and MMP9, and decreased mitogen-activated protein kinase kinase (MEK) and extracellular-signal regulated kinase (ERK) phosphorylation. Concurrently, it upregulated E-cadherin and p21 expression levels. Interestingly, opposite trends were observed in the p53-null breast cancer cell line MDA-MB-231. The MEK inhibitor PD98059 reduced the biological effects of FAM53A knockdown in MCF-7 cells and FAM53A overexpression in MDA-MB-231 cells, suggesting that FAM53A affects breast cancer through the MEK-ERK pathway. Silencing TP53 in MCF-7 cells and stably expressing wild-type p53 in MDA-MB-231 cells confirmed that the effects of FAM53A signaling through the MEK/ERK pathway depended on the p53 status of the cells. These results suggest that FAM53A acts as a tumor suppressor in p53-positive breast cancer by modulating the MEK-ERK pathway, but may be a potential candidate for targeted anticancer therapies in p53-negative breast cancer.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Mingfang Sun
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Miaomiao Hao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Kexin Diao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jian Wang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shiping Li
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qixue Cao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoyi Mi
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
30
|
Parameswaran N, Bartel CA, Hernandez-Sanchez W, Miskimen KL, Smigiel JM, Khalil AM, Jackson MW. A FAM83A Positive Feed-back Loop Drives Survival and Tumorigenicity of Pancreatic Ductal Adenocarcinomas. Sci Rep 2019; 9:13396. [PMID: 31527715 PMCID: PMC6746704 DOI: 10.1038/s41598-019-49475-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinomas (PDAC) are deadly on account of the delay in diagnosis and dearth of effective treatment options for advanced disease. The insurmountable hurdle of targeting oncogene KRAS, the most prevalent genetic mutation in PDAC, has delayed the availability of targeted therapy for PDAC patients. An alternate approach is to target other tumour-exclusive effector proteins important in RAS signalling. The Family with Sequence Similarity 83 (FAM83) proteins are oncogenic, tumour-exclusive and function similarly to RAS, by driving the activation of PI3K and MAPK signalling. In this study we show that FAM83A expression is significantly elevated in human and murine pancreatic cancers and is essential for the growth and tumorigenesis of pancreatic cancer cells. Elevated FAM83A expression maintains essential MEK/ERK survival signalling, preventing cell death in pancreatic cancer cells. Moreover, we identified a positive feed-forward loop mediated by the MEK/ERK-activated AP-1 transcription factors, JUNB and FOSB, which is responsible for the elevated expression of oncogenic FAM83A. Our data indicates that targeting the MEK/ERK-FAM83A feed-forward loop opens up additional avenues for clinical therapy that bypass targeting of oncogenic KRAS in aggressive pancreatic cancers.
Collapse
Affiliation(s)
- Neetha Parameswaran
- Department of Pathology, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Courtney A Bartel
- Department of Pathology, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Wilnelly Hernandez-Sanchez
- Department of Pharmacology, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Kristy L Miskimen
- Department of Epidemiology and Biostatistics, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Ahmad M Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Wolstein Research Building, Cleveland, OH, 44106, USA.
| |
Collapse
|
31
|
Zhang JT, Lin YC, Xiao BF, Yu BT. Overexpression of Family with Sequence Similarity 83, Member A (FAM83A) Predicts Poor Clinical Outcomes in Lung Adenocarcinoma. Med Sci Monit 2019; 25:4264-4272. [PMID: 31175804 PMCID: PMC6580865 DOI: 10.12659/msm.910804] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background The aim of this study was to explore the expression levels of family with sequence similarity 83, member A (FAM83A) in lung adenocarcinoma (LUAD) and investigate its clinical prognostic value. Material/Methods Bioinformatics mining methods were used to predict the differential expression levels of FAM83A mRNA in LUAD and normal lung tissues based on the TCGA and Oncomine databases. Immunohistochemical staining was performed to demonstrate the FAM83A protein expression levels in 83 cases of LUAD combined with paired normal lung tissues. The correlation between clinicopathologic factors and FAM83A differential expression levels in LUAD was explored by the chi-square test. Kaplan-Meier univariate and Cox multivariate survival analyses were performed to investigate the clinical prognostic value of FAM83A expression in LUAD patients. Results Results from TCGA and Oncomine databases revealed that FAM83A mRNA expression level was significantly higher in LUAD than that in normal lung tissues (both P<0.05). Immunohistochemical findings demonstrated that the high positive rate of FAM83A in LUAD was 73.49% (61/83), while that of matched normal lung tissues was only 22.89% (19/83). Moreover, LUAD patients with FAM83A mRNA or high protein levels had dramatically lower OS times than those with FAM83A mRNA or low protein levels (All P<0.05). Lastly, Cox multivariate survival analysis showed that FAM83A differential expression level (low vs. high) was the only independent factor predicting the prognosis of LUAD patients (P=0.001). Conclusions FAM83A was overexpressed in LUAD, and FAM83A overexpression could be used as an independent factor of poor prognosis in LUAD patients.
Collapse
Affiliation(s)
- Jing-Tao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Ye-Chun Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Bu-Fan Xiao
- First Clinical Medical College, Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Ben-Tong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
32
|
Richtmann S, Wilkens D, Warth A, Lasitschka F, Winter H, Christopoulos P, Herth FJF, Muley T, Meister M, Schneider MA. FAM83A and FAM83B as Prognostic Biomarkers and Potential New Therapeutic Targets in NSCLC. Cancers (Basel) 2019; 11:E652. [PMID: 31083571 PMCID: PMC6562954 DOI: 10.3390/cancers11050652] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 11/17/2022] Open
Abstract
Although targeted therapy has improved the survival rates in the last decade, non-small-cell lung cancer (NSCLC) is still the most common cause of cancer-related death. The challenge of identifying new targets for further effective therapies still remains. The FAMily with sequence similarity 83 (FAM83) members have recently been described as novel oncogenes in numerous human cancer specimens and shown to be involved in epidermal growth factor receptor (EGFR) signaling. Here, gene expression of FAM83A and B was analyzed in a cohort of 362 NSCLC patients using qPCR. We further investigated relations in expression and their prognostic value. Functional assays in NSCLC cell lines were performed to evaluate FAM83A and B involvement in proliferation, anchorage-independent growth, migration, and the EGFR pathway. We observed a highly increased gene expression level of FAM83A (ø = 68-fold) and FAM83B (ø = 20-fold) which resulted in poor survival prognosis (p < 0.0001 and p = 0.002). Their expression was influenced by EGFR levels, pathway signaling, and mutation status. Both genes affected cell proliferation, and FAM83A depletion resulted in reduced migration and anchorage-independent growth. The results support the hypothesis that FAM83A and B have different functions in different histological subtypes of NSCLC and might be new therapeutic targets.
Collapse
Affiliation(s)
- Sarah Richtmann
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
| | - Dennis Wilkens
- Microbial Energy Conversion and Biotechnology, Department of Biology, Technische Universität Darmstadt, D-64287 Darmstadt, Germany.
| | - Arne Warth
- Institute of Pathology, Heidelberg University Hospital, D-69120 Heidelberg, Germany.
| | - Felix Lasitschka
- Institute of Pathology, Heidelberg University Hospital, D-69120 Heidelberg, Germany.
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
- Department of Surgery, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
| | - Petros Christopoulos
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
| | - Felix J F Herth
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
- Department of Pneumology and Critical Care Medicine, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
| | - Michael Meister
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
| | - Marc A Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), D-69120 Heidelberg, Germany.
| |
Collapse
|
33
|
Wang Y, Lu T, Wo Y, Sun X, Li S, Miao S, Dong Y, Leng X, Jiao W. Identification of a putative competitive endogenous RNA network for lung adenocarcinoma using TCGA datasets. PeerJ 2019; 7:e6809. [PMID: 31065463 PMCID: PMC6485208 DOI: 10.7717/peerj.6809] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
The mechanisms underlying the oncogenesis and progression of lung adenocarcinoma (LUAD) are currently unclear. The discovery of competitive endogenous RNA (ceRNA) regulatory networks has provided a new direction for the treatment and prognosis of patients with LUAD. However, the mechanism of action of ceRNA in LUAD remains elusive. In the present study, differentially expressed mRNAs, microRNAs (miRs) and long non-coding RNAs from the cancer genome atlas database were screened. CeRNAs for LUAD were then identified using online prediction software. Among the ceRNAs identified, family with sequence similarity 83 member A (FAM83A), miR-34c-5p, KCNQ1OT1 and FLJ26245 were observed to be significantly associated with the overall survival of patients with LUAD. Of note, FAM83A has potential significance in drug resistance, and may present a candidate biomarker for the prognosis and treatment of patients with LUAD.
Collapse
Affiliation(s)
- Yuanyong Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tong Lu
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yang Wo
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiao Sun
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shicheng Li
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shuncheng Miao
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanting Dong
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoliang Leng
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|